WO1995023225A2 - Method and reagent for inhibiting the expression of disease related genes - Google Patents

Method and reagent for inhibiting the expression of disease related genes Download PDF

Info

Publication number
WO1995023225A2
WO1995023225A2 PCT/IB1995/000156 IB9500156W WO9523225A2 WO 1995023225 A2 WO1995023225 A2 WO 1995023225A2 IB 9500156 W IB9500156 W IB 9500156W WO 9523225 A2 WO9523225 A2 WO 9523225A2
Authority
WO
WIPO (PCT)
Prior art keywords
rna
nucleic acid
ribozyme
molecule
acid molecule
Prior art date
Application number
PCT/IB1995/000156
Other languages
French (fr)
Other versions
WO1995023225A3 (en
Inventor
Dan T. Stinchcomb
Bharat Chowrira
Anthony Direnzo
Kenneth G. Draper
Lech W. Dudycz
Susan Grimm
Alexander Karpeisky
Kevin Kisich
Jasenka Matulic-Adamic
James A. Mcswiggen
Anil Modak
Pamela Pavco
Leonid Beigelman
Sean M. Sullivan
David Sweedler
James D. Thompson
Danuta Tracz
Nassim Usman
Francine E. Wincott
Tod Woolf
Original Assignee
Ribozyme Pharmaceuticals, 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
Priority claimed from US08/218,934 external-priority patent/US5639647A/en
Priority claimed from US08/291,932 external-priority patent/US5658780A/en
Priority claimed from US08/292,620 external-priority patent/US5837542A/en
Priority claimed from US08/311,486 external-priority patent/US5811300A/en
Priority claimed from US08/319,492 external-priority patent/US5616488A/en
Priority claimed from US08/321,993 external-priority patent/US5631359A/en
Priority claimed from US08/334,847 external-priority patent/US5693532A/en
Priority claimed from US08/337,608 external-priority patent/US5902880A/en
Priority claimed from US08/357,577 external-priority patent/US5783425A/en
Priority claimed from US08/363,233 external-priority patent/US5714383A/en
Priority to JP7522236A priority Critical patent/JPH09509323A/en
Priority to AU18214/95A priority patent/AU706417B2/en
Priority to EP95909920A priority patent/EP0746614A1/en
Priority to MX9603638A priority patent/MX9603638A/en
Application filed by Ribozyme Pharmaceuticals, Inc. filed Critical Ribozyme Pharmaceuticals, Inc.
Publication of WO1995023225A2 publication Critical patent/WO1995023225A2/en
Publication of WO1995023225A3 publication Critical patent/WO1995023225A3/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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • C12N15/101Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers by chromatography, e.g. electrophoresis, ion-exchange, reverse phase
    • 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
    • C12N15/1131Non-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 against viruses
    • 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
    • C12N15/1136Non-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 against growth factors, growth regulators, cytokines, lymphokines or hormones
    • 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
    • C12N15/1138Non-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 against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2310/121Hammerhead
    • 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
    • C12N2310/122Hairpin
    • 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
    • C12N2310/123Hepatitis delta
    • 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
    • C12N2310/124Type of nucleic acid catalytic nucleic acids, e.g. ribozymes based on group I or II introns
    • C12N2310/1241Tetrahymena
    • 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
    • C12N2310/126Type of nucleic acid catalytic nucleic acids, e.g. ribozymes involving RNAse P
    • 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
    • C12N2310/127DNAzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/333Modified A
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/335Modified T or U
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/336Modified G
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3523Allyl
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3527Other alkyl chain
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • C12N2310/3533Halogen
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • C12N2310/3535Nitrogen
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/38Vector systems having a special element relevant for transcription being a stuffer

Definitions

  • This invention relates to reagents useful as inhibitors of gene expression relating to diseases such as inflammatory or autoimmune disorders, chronic myelogenous leukemia, or respiratory tract illness. Summary of the Invention
  • the invention features novel enzymatic RNA molecules, or ribozymes, and methods for their use for inhibiting the expression of disease related genes, e.g., ICAM-1 , IL-5, relA, TNF- ⁇ , p210 bcr-abl , and respiratory syncytial virus genes.
  • disease related genes e.g., ICAM-1 , IL-5, relA, TNF- ⁇ , p210 bcr-abl , and respiratory syncytial virus genes.
  • ribozymes can be used in a method for treatment of diseases caused by the expression of these genes in man and other animals, including other primates.
  • Ribozymes are RNA molecules having an enzymatic activity which is able to repeatedly cleave other separate RNA molecules in a nucleotide base sequence specific manner. Such enzymatic RNA molecules can be targeted to virtually any RNA transcript, and efficient cleavage has been achieved in vitro. Kim et al., 84 Proc. Natl. Acad. Sci. USA 8788, 1987; Haseloff and Gerlach, 334 Nature 585, 1988; Cech, 260 JAMA 3030, 1988; and Jefferies et al., 17 Nucleic Acids Research 1371 , 1989.
  • RNA phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions.
  • Table 1 summarizes some of the characteristics of these ribozymes.
  • Ribozymes act by first binding to a target RNA. Such binding occurs through the target RNA binding portion of a ribozyme which is held in close proximity to an enzymatic portion of the RNA which acts to cleave the target RNA. Thus, the ribozyme first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA. Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After a ribozyme has bound and cleaved its RNA target it is released from that RNA to search for another target and can repeatedly bind and cleave new targets.
  • ribozyme The enzymatic nature of a ribozyme is advantageous over other technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the effective concentration of ribozyme necessary to effect a therapeutic treatment is lower than that of an antisense oligonucleotide.
  • antisense technology where a nucleic acid molecule simply binds to a nucleic acid target to block its translation
  • the advantage reflects the ability of the ribozyme to act enzymatically.
  • a single ribozyme molecule is able to cleave many molecules of target RNA.
  • the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding, but also on the mechanism by which the molecule inhibits the expression of the RNA to which it binds. That is, the inhibition is caused by cleavage of the RNA target and so specificity is defined as the ration of the rate of cleavage of the targeted RNA over the rate of cleavage of non-targeted RNA. This cleavage mechanism is dependent upon factors additional to those involved in base pairing. Thus, it is thought that the specificity of action of a ribozyme is greater than that of antisense oligonucleotide binding the same RNA site. With their catalytic activity and increased site specificity, ribozymes represent more potent and safe therapeutic molecules than antisense oligonucleotides. Thus, in a first aspect, this invention relates to ribozymes, or enzymatic
  • RNA molecules directed to cleave RNA species encoding ICAM-1 , IL-5, relA, TNF- ⁇ , p210 bcr-abl , or RSV proteins.
  • applicant describes the selection and function of ribozymes capable of cleaving these RNAs and their use to reduce levels of ICAM-1 , IL-5, relA, TNF- ⁇ , p210 bor-abl or RSV proteins in various tissues to treat the diseases discussed herein.
  • Such ribozymes are also useful for diagnostic uses.
  • ribozymes are able to inhibit expression of ICAM-1 , IL-5, rel A, TNF- ⁇ , p21 ⁇ bcr-abl. or RSV genes and that the catalytic activity of the ribozymes is required for their inhibitory effect.
  • Those of ordinary skill in the art will find that it is clear from the examples described that other ribozymes that cleave target ICAM-1 , IL-5, rel A, TNF- ⁇ , p210 b cr-abl , or RSV encoding mRNAs may be readily designed and are within the invention.
  • RNA molecules contain substrate binding domains that bind to accessible regions of their target mRNAs.
  • the RNA molecules also contain domains that catalyze the cleavage of RNA.
  • the ribozymes cleave the target encoding mRNAs, preventing translation and protein accumulation. In the absence of the expression of the target gene, a therapeutic effect may be observed.
  • gene is meant to refer to either the protein coding regions of the cognate mRNA, or any regulatory regions in the RNA which regulate synthesis of the protein or stability of the mRNA; the term also refers to those regions of an mRNA which encode the ORF of a cognate polypeptide product, and the proviral genome.
  • enzymatic RNA molecule an RNA molecule which has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity which is active to specifically cleave RNA in that target. That is, the enzymatic RNA molecule is able to intermolecularly cleave RNA and thereby inactivate a target RNA molecule. This complementarity functions to allow sufficient hybridization of the enzymatic RNA molecule to the target RNA to allow the cleavage to occur. One hundred percent complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention.
  • RNA to a virus is meant to include those naturally occurring viral encoded RNA molecules associated with viral caused diseases in various animals, including humans, cats, simians, and other primates. These viral or viral-encoded RNAs have similar structures and equivalent genes to each other.
  • complementarity it is meant a nucleaic acid that can form hydrogen bond(s) with other RNA sequence by either traditional WatsonCrick or other non-traditional types (for examplke, Hoogsteen type) of base-paired interactions.
  • the enzymatic nucleic acid molecule is formed in a hammerhead or hairpin motif, but may also be formed in the motif of a hepatitis delta virus, group I intron or RNaseP RNA (in associateion with an RNA guide sequence) or Neurospora VS RNA.
  • hammerhead motifs are described by Rossi et al., 1992, Aids Research and Human Retroviruses , 8,183, of hairpin motifs by Hampel and Tritz, 1989 Biochemistry, 28, 4929, EP 0360257 and Hampel et al., 1990, Nucleic Acids Res.
  • hepatitis delta virus motif is described by Perotta and Been, 1992 Biochemistry, 31 16 of the RNaseP motif by Guerrier-Takada et al., 1983 Cell, 35 849, cleavage of RNA.
  • the ribozymes cleave the target encoding mRNAs, preventing translation and protein accumulation. In the absence of the expression of the target gene, a therapeutic effect may be observed.
  • gene is meant to refer to either the protein coding regions of the cognate mRNA, or any regulatory regions in the RNA which regulate synthesis of the protein or stability of the mRNA; the term also refers to those regions of an mRNA which encode the ORF of a cognate polypeptide product, and the proviral genome.
  • enzymatic RNA molecule an RNA molecule which has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity which is active to specifically cleave RNA in that target. That is, the enzymatic RNA molecule is able to intermolecularly cleave RNA and thereby inactivate a target RNA molecule. This complementarity functions to allow sufficient hybridization of the enzymatic RNA molecule to the target RNA to allow the cleavage to occur. One hundred percent complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention.
  • RNA to a virus is meant to include those naturally occurring viral encoded RNA molecules associated with viral caused diseases in various animals, including humans, cats, simians, and other primates. These viral or viral-encoded RNAs have similar structures and equivalent genes to each other.
  • complementarity it is meant a nucleaic acid that can form hydrogen bond(s) with other RNA sequence by either traditional Watson-Crick or other non-traditional types (for examplke, Hoogsteen type) of base-paired interactions.
  • the enzymatic nucleic acid molecule is formed in a hammerhead or hairpin motif, but may also be formed in the motif of a hepatitis delta virus, group I intron or RNaseP RNA (in associateion with an RNA guide sequence) or Neurospora VS RNA.
  • hammerhead motifs are described by Rossi et al., 1992, Aids Research and Human Retroviruses , 8,183, of hairpin motifs by Hampel and Tritz, 1989 Biochemistry, 28, 4929, EP 0360257 and Hampel et al., 1990, Nucleic Acids Res.
  • hepatitis delta virus motif is described by Perotta and Been, 1992 Biochemistry, 31 16 of the RNaseP motif by Guerrier-Takada et al., 1983 Cell, 35 849, expressed in eukaryotic cells from the appropriate DNA or RNA vector.
  • the activity of such ribozymes can be augmented by their release from the primary transcript by a second ribozyme (Draper et al., PCT W093/23569, and Sullivan et al., PCT WO94/02595, both hereby incorporated in their totality by reference herein; Ohkawa, J., et al., 1992, Nucleic Acids Symp. Ser. 27, 15-6; Taira, K.
  • inhibitor is meant that the activity or level of ICAM-1 , Rel A, IL-5, TNF- ⁇ , p210 bcr-abl or RSV encoding mRNA is reduced below that observed in the absense of the ribozyme, and preferably is below that level observed in the presence of an inactive RNA molecule able to bind to the same site on the mRNA, but unable to cleave that RNA.
  • Such ribozymes are useful for the prevention of the diseases and conditions discussed above, and any other diseases or conditions that are related to the level of ICAM-1 , IL-5, Rel A, TNF- ⁇ , p210 bcr-abl or RSV protein or activity in a cell or tissue.
  • diseases or conditions that are related to the level of ICAM-1 , IL-5, Rel A, TNF- ⁇ , p210 bcr-abl or RSV protein or activity in a cell or tissue.
  • related is meant that the inhibition of ICAM-1 , IL-5, Rel A, TNF- ⁇ , P 210 bcr-abl or RSV mRNA translation, and thus reduction in the level of, ICAM-1 , IL-5, Rel A, TNF- ⁇ , p210 bcl-abl or RSV proteins will relieve to some extent the symptoms of the disease or condition.
  • Ribozymes are added directly, or can be complexed with cationic lipids, packaged within liposomes, or otherwise delivered to target cells.
  • the RNA or RNA complexes can be locally administered to relevant tissues through the use of a catheter, infusion pump or stent, with or without their incorporation in biopolymers.
  • the ribozymes have binding arms which are complementary to the sequences in Tables 2,3,6-9, 11 , 13, 15-23, 27, 28, 31 , 33, 34, 36 and 37.
  • ribozymes examples are shown in Tables 4-8, 10, 12, 14-16, 19-22, 24, 26-28, 30, 32, 34 and 36-38.
  • Examples of such ribozymes consist essentially of sequences defined in these Tables. By “consists essentially of” is meant that the active ribozyme contains an enzymatic center equivalent to those in the examples, and binding arms able to bind mRNA such that cleavage at the target site occurs. Other sequences may be present which do not interfere with such cleavage. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity.
  • stem-loop II sequence of hammerhead ribozymes listed in the above identified Tables can be altered (substitution, deletion, and/or insertion) to contain any sequences provided a minimum of two base-paired stem structure can form.
  • stem-loop IV sequence of hairpin ribozymes listed in the above identified Tables can be altered (substitution, deletion, and/or insertion) to contain any sequence, provided a minimum of two base-paired stem structure can form.
  • the sequence listed in the above identified Tables may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables.
  • ribozymes that cleave target molecules and inhibit ICAM-1 , IL-5, Rel A, TNF- ⁇ , p210 bcr-abl or RSV gene expression are expressed from transcription units inserted into DNA, RNA, or viral vectors.
  • Another means of accumulating high concentrations of a ribozyme(s) within cells is to incorporate the ribozyme-encoding sequences into a DNA or RNA expression vector. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III).
  • Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby.
  • Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990 Proc. Natl. Acad. Sci. USA, 87, 6743-7; Gao and Huang 1993 Nucleic Acids Res., 21 2867-72; Lieber et al., 1993 Methods Enzymol., 217, 47-66; Zhou et al., 1990 Mol. Cell.
  • ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992 Antisense Res. Dev., 2, 3-15; Ojwang et al., 1992 Proc. Natl. Acad. Sci. USA, 90, 6340-4; L'Huiller et al., 1992 EMBO J. 11 , 4411-8; Lisziewicz et al., 1993 Proc. Natl. Acad. Sci. U.S.A., 90 8000-4).
  • ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors).
  • plasmid DNA vectors such as adenovirus or adeno-associated virus vectors
  • viral RNA vectors such as retroviral or alphavirus vectors
  • Figure 1 is a diagrammatic representation of the hammerhead ribozyme domain known in the art.
  • Stem II can be ⁇ 2 base-pair long.
  • Figure 2(a) is a diagrammatic representation of the hammerhead ribozyme domain known in the art
  • Figure 2(b) is a diagrammatic representation of the hammerhead ribozyme as divided by Uhlenbeck (1987, Nature, 327, 596-600) into a substrate and enzyme portion
  • Figure 2(c) is a similar diagram showing the hammerhead divided by Haseloff and Gerlach (1988, Nature, 334, 585-591) into two portions
  • Figure 2(d) is a similar diagram showing the hammerhead divided by Jeffries and Symons (1989, N ⁇ cl. Acids. Res., 17, 1371-1371) into two portions.
  • FIG 3 is a diagrammatic representation of the general structure of a hairpin ribozyme.
  • Helix 2 (H2) is provided with a least 4 base pairs (i.e., n is 1 ,2,3 or 4) and helix 5 can be optionally provided of length 2 or more bases (preferably 3-20 bases, i.e., m is from 1-20 or more). Helix 2 and helix 5 may be covalently linked by one or more bases (i.e., r is ⁇ 1 base).
  • Helix 1 , 4 or 5 may also be extended by 2 or more base pairs (e.g., 4-20 base pairs) to stabilize the ribozyme structure, and preferably is a protein binding site.
  • each N and N' independently is any normal or modified base and each dash represents a potential base-pairing interaction. These nucleotides may be modified at the sugar, base or phosphate. Complete base-pairing is not required in the helices, but is preferred.
  • Helix 1 and 4 can be of any size (i.e., o and p is each independently from 0 to any number, e.g. 20) as long as some base-pairing is maintained.
  • Helix 4 can be formed from two separate molecules, i.e., without a connecting loop.
  • the connecting loop when present may be a ribonucleotide with or without modifications to its base, sugar or phosphate, "q" is ⁇ 2 bases.
  • the connecting loop can also be replaced with a non-nucleotide linker molecule.
  • H refers to bases A, U, or C.
  • Y refers to pyrimidine bases.
  • Figure 4 is a representation of the general structure of the hepatitis delta virus ribozyme domain known in the art.
  • Figure 5 is a representation of the general structure of the self-cleaving VS RNA ribozyme domain.
  • Figure 6 is a diagrammatic representation of the genetic map of RSV strain A2.
  • Figure 7 is a diagrammatic representation of the solid-phase synthesis of RNA.
  • Figure 8 is a diagrammatic representation of exocyclic amino protecting groups for nucleic acid synthesis.
  • Figure 9 is a diagrammatic representation of the deprotection of RNA.
  • Figure 10 is a graphical representation of the cleavage of an RNA substrate by ribozymes synthesized, deprotected and purified using the improved methods described herein.
  • Figure 11 is a schematic representation of a two pot deprotection protocol.
  • Base deprotection is carried out with aqueous methyl amine at 65°C for 10 min.
  • the sample is dried in a speed-vac for 2-24 hours depending on the scale of RNA synthesis.
  • Silyl protecting group at the 2'-hydroxyl position is removed by treating the sample with 1.4 M anhydrous HF at 65°C for 1.5 hours.
  • Figure 12 is a schematic representation of a one pot deprotection of RNA synthesized using RNA phosphoramidite chemistry.
  • Anhydrous methyl amine is used to deprotect bases at 65°C for 15 min.
  • the sample is allowed to cool for 10 min before adding TEA•3HF reagent, to the same pot, to remove protecting groups at the 2'-hydroxyI position.
  • the deprotection is carried out for 1.5 hours.
  • Figs. 13a - b is a HPLC profile of a 36 nt long ribozyme, targeted to site B.
  • the RNA is deprotected using either the two pot or the one pot deprotection protocol. The peaks corresponding to full-length RNA is indicated.
  • the sequence for site B is CCUGGGCCAGGGAUUA
  • Figure 14 is a graph comparing RNA cleavage activity of ribozymes deprotected by two pot vs one pot deprotection protocols.
  • Figure 15 is a schematic representation of an improved method of synthesizing RNA containing phosphorothioate linkages.
  • Figure 16 shows RNA cleavage reaction catalyzed by ribozymes containing phosphorothioate linkages.
  • Hammerhead ribozyme targeted to site C is synthesized such that 4 nts at the 5' end contain phosphorothioate linkages.
  • the sequence for site C is UCAUUUUGGCCAUCUC UUCCUUCAGGCGUGG.
  • Figure 17 is a schematic representation of synthesis of 2'-N-phtalimido-nucleoside phosphoramidite.
  • Figure 18 is a diagrammatic representation of a prior art method for the solid-phase synthesis of RNA using silyl ethers, and the method of this invention using SEM as a 2'-protecting group.
  • Figure 19 is a diagrammatic representation of the synthesis of 2'- SEM-protected nucleosides and phosphoramidites useful for the synthesis of RNA.
  • B is any nucleotide base as exemplified in the Figure, P is purine and I is inosine. Standard abbreviations are used throughout this application, well known to those in the art.
  • Figure 20 is a diagrammatic representation of a prior art method for deprotection of RNA using TBDMS protection of the 2'-hydroxyl group.
  • Figure 21 is a diagrammatic representation of the deprotection of RNA having SEM protection of the 2'-hydroxyl group.
  • Figure 22 is a representation of an HPLC chromatogram of a fully deprotected 10-mer of uridylic acid.
  • Figs. 23 - 25 are diagrammatic representations of hammerhead, hairpin or hepatitis delta virus ribozyme containing self-processing RNA transcript.
  • Solid arrows indicate self-processing sites. Boxes indicate the sites of nucleotide substitution.
  • Solid lines are drawn to show the binding sites of primers used in a primer-extension assay. Lower case letters indicate vector sequence present in the RNA when transcribed from a HindIII-linearized plasmid. (23) HH Cassette, transcript containing the hammerhead trans-acting ribozyme linked to a 3' cis-acting hammerhead ribozyme.
  • the structure of the hammerhead ribozyme is based on phylogenetic and mutational analysis (reviewed by Symons, 1992 supra).
  • the trans ribozyme domain extends from nucleotide 1 through 49. After 3'-end processing, the trans-ribozyme contains 2 non-ribozyme nucleotides (UC at positions 50 and 51) at its 3' end.
  • the 3' processing ribozyme is comprised of nucleotides 44 through 96.
  • Roman numerals I, II and III indicate the three helices that contribute to the structure of the 3' cis-acting hammerhead ribozyme (Hertel et al., 1992 Nucleic Acids Res. 20, 3252).
  • the trans-ribozyme contains 5 non-ribozyme nucleotides (UGGCA at positions 50 to 54) at its 3' end.
  • the 3' cis-acting ribozyme is comprised of nucleotides 50 through 115.
  • the transcript named HP(GU) was constructed with a potential wobble base pair between G52 and U77; HP(GC) has a Watson-Crick base pair between G52 and C77.
  • a shortened helix 1 (5 base pairs) and a stable tetraloop (GAAA) at the end of helix 1 was used to connect the substrate with the catalytic domain of the hairpin ribozyme (Feldstein & Bruening, 1993 Nucleic Acids Res.
  • HDV Cassette transcript containing the trans-acting hammerhead ribozyme linked to a 3' cis-acting hepatitis delta virus (HDV) ribozyme.
  • the secondary structure of the HDV ribozyme is as proposed by Been and coworkers (Been et al., 1992 Biochemistry 31 , 11843).
  • the trans-ribozyme domain extends from nucleotides 1 through 48. After 3'-end processing, the trans-ribozyme contains 2 non-ribozyme nucleotides (AA at positions 49 to 50) at its 3' end.
  • the 3' cis-acting HDV ribozyme is comprised of nucleotides 50 through 114.
  • Roman numerals I, II, III & IV, indicate the location of four helices within the 3' cis-acting HDV ribozyme (Perrota & Been, 1991 Nature 350, 434).
  • the ⁇ HDV transcript contains a 31 nucleotide deletion in the HDV portion of the transcript (nucleotides 84 through 115 deleted).
  • Fig. 26 is a schematic representation of a plasmid containing the insert encoding self-processing cassette. The figure is not drawn to scale.
  • Fig. 27 demonstrates the effect of 3' flanking sequences on RNA self-processing in vitro.
  • H Plasmid templates linearized with HindIII restriction enzyme. Transcripts from H templates contain four non-ribozyme nucleotides at the 3' end.
  • N Plasmid templates linearized with Ndel restriction enzyme. Transcripts from ⁇ templates contain 220 non-ribozyme nucleotides at the 3' end.
  • R Plasmid templates linearized with Rcal restriction enzyme. Transcripts from R templates contain 450 non-ribozyme nucleotides at the 3' end.
  • Fig. 28 shows the effect of 3' flanking sequences on the transcleavage reaction catalyzed by a hammerhead ribozyme.
  • Fig. 29 shows R ⁇ A self-processing in OST7-1 cells.
  • In vitro lanes contain full-length, unprocessed transcripts that were added to cellular lysates prior to R ⁇ A extraction. These R ⁇ As were either pre-incubated with MgCl 2 (+) or with DEPC-treated water (-) prior to being hybridized with 5' end-labeled primers.
  • Cellular lanes contain total cellular RNA from cells transfected with one of the four self-processing constructs. Cellular RNA are probed for ribozyme expression using a sequence specific primer-extension assay. Solid arrows indicate the location of primer extension bands corresponding to Full-Length RNA and 3' Cleavage Products.
  • Figs. 30,31 are diagrammatic representations of self-processing cassettes that will release trans-acting ribozymes with defined, stable stemloop structures at the 5' and the 3' end following self-processing. 30, shows various permutations of a hammerhead self-processing cassette. 31 , shows various permutations of a hairpin self-processing cassette.
  • Figs. 32a-b Schematic representation of RNA polymerse III promoter structure. Arrow indicates the transcription start site and the direction of coding region.
  • A, B and C refer to consensus A, B and C box promoter sequences.
  • I refers to intermediate cis-acting promoter sequence.
  • PSE refers to proximal sequence element.
  • DSE refers to distal sequence element.
  • ATF refers to activating transcription factor binding element.
  • ? refers to cis-acting sequence element that has not been fully characterized.
  • EBER Epstein-Barr-virus-encoded-RNA. TATA is a box well known in the art.
  • the A and B box are internal promoter regions necessary for pol III transcription. Arrows indicate the sites of endogenous tRNA processing.
  • the ⁇ 3-5 transcript is a truncated version of tRNA wherein the sequence 3' of B box has been deleted (Adeniyi-Jones et al., 1984 supra). This modification renders the ⁇ 3-5 RNA resistant to endogenous tRNA processing.
  • FIG. 34 Schematic representation of RNA structural motifs inserted into the ⁇ 3-5 RNA.
  • ⁇ 3-5/HHI- a hammerhead (HHI) ribozyme was cloned at the 3' region of ⁇ 3-5 RNA;
  • S3- a stable stem-loop structure was incorporated at the 3' end of the ⁇ 3-5/HHI chimera;
  • S5- stable stem-loop structures were incorporated at the 5' and the 3' ends of ⁇ 3-5/HHI ribozyme chimera;
  • S35- sequence at the 3' end of the ⁇ 3-5/HHI ribozyme chimera was altered to enable duplex formation between the 5' end and a complementary 3' region of the same RNA;
  • S35Plus- in addition to structural alterations of S35, sequences were altered to facilitate additional duplex formation within the non-ribozyme sequence of the ⁇ 3-5/HHI chimera.
  • FIGS. 35 and 36 Northern analysis to quantitate ribozyme expression in T cell lines transduced with ⁇ 3-5 vectors. 35) ⁇ 3-5/HHI and its variants were cloned individually into the DC retroviral vector (Sullenger et al., 1990 supra). Northern analysis of ribozyme chimeras expressed in MT-2 cells was performed. Total RNA was isolated from cells (Chomczynski & Sacchi, 1987 Analytical Biochemistry 162, 156-159), and transduced with various constructs described in Fig. 34. Northern analysis was carried out using standard protocols (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY).
  • Nomenclature is same as in Figure 34. This assay measures the level of expression from the type 2 pol III promoter. 36) Expression of S35 constructs in MT2 cells. S35 (+ribozyme), S35 construct containing HHI ribozyme. S35 (-ribozyme), S35 construct containing no ribozyme.
  • FIG. 37 Ribozyme activity in total RNA extracted from transduced MT-2 cells.
  • Total RNA was isolated from cells transduced with ⁇ 3-5 constructs described in Figs. 35 and 36
  • 5 ⁇ g total RNA and trace amounts of 5' terminus-labeled ribozyme target RNA were denatured separately by heating to 90°C for 2 min in the presence of 50 mM Tris-HCI, pH 7.5 and 10 mM MgCl 2 .
  • RNAs were renatured by cooling the reaction mixture to 37°C for 10-15 min.
  • Cleavage reaction was initiated by mixing the labeled substrate RNA and total cellular RNA at 37°C. The reaction was allowed to proceed for ⁇ 18h, following which the samples were resolved on a 20 % urea-polyacrylamide gel. Bands were visualized by autoradiography.
  • FIGS. 38 and 39 Ribozyme expression and activity levels in S35-transduced clonal CEM cell lines.
  • tRNA i met refers to the endogenous tRNA.
  • S35 refers to the position of the ribozyme band. M, marker lane. 39)
  • Figures 40 and 41 are proposed secondary structures of S35 and S35 containing a desired RNA (HHI), respectively.
  • HHI desired RNA
  • Intramolecular stem refers to the stem structure formed due to an intramolecular base-paired interaction between the 3' sequence and the complementary 5' terminus.
  • the length of the stem ranges from 15-16 base-pairs. Location of the A and the B boxes are shown.
  • Figures 42 and 43 are proposed secondary structures of S35 plus and S35 plus containing HHI ribozyme.
  • Figures 44, 45, 46 and 47 are the nucleotide base sequences of S35, HHIS35, S35 Plus, and HHIS35 Plus respectively.
  • Figs. 48a-b is a general formula for pol III RNA of this invention.
  • Figure 49 is a digrammatic representation of 5T construct.
  • the desired RNA is located 3' of the intramolecular stem.
  • Figures 50 and 51 contain proposed secondary structures of 5T construct alone and 5T contruct containing a desired RNA (HHI ribozyme) respectively.
  • Figure 52 is a diagrammatic representation of TRZ-tRNA chimeras. The site of desired RNA insertion is indicated.
  • Figure 53 shows the general structure of HHITRZ-A ribozyme chimera.
  • a hammerhead ribozyme targeted to site I is inserted into the stem II region of TRZ-tRNA chimera.
  • Figure 54 shows the general structure of HPITRZ-A ribozyme chimera. A hairpin ribozyme targeted to site I is cloned into the indicated region of TRZ-tRNA chimera.
  • Figure 55 shows a comparison of RNA cleavage activity of HHITRZ-A, HHITRZ-B and a chemically synthesized HHI hammerhead ribozymes.
  • Figure 56 shows expression of ribozymes in T cell lines that are stably transduced with viral vectors.
  • M markers; lane 1 , non-transduced CEM cells; lanes 2 and 3, MT2 and CEM cells transduced with retroviral vectors; lanes 4 and 5, MT2 and CEM cells transduced with AAV vectors.
  • Figs. 57a-b Schematic diagram of adeno-associated virus and adenovirues vectors for ribozyme delivery. Both vectors utilize one or more ribozyme encoding transcription units (RZ) based on RNA polymerase II or RNA polymerase III promoters.
  • RZ ribozyme encoding transcription units
  • ITR inverted terminal repeats
  • Pro exogenous promoter
  • stuffer sufficient additional sequences
  • Fig. 58 is a graph showing the effect of arm length variation on the activity of ligated hammerhead (HH) ribozymes. Nomenclature 5/5, 6/6, 7/7, 8/8 and so on refers to the number of base-pairs being formed between the ribozyme and the target.
  • 5/8 means that the HH ribozyme forms 5 bp on the 5' side and 8 bp on the 3' side of the cleavage site for a total of 13 bp.
  • - ⁇ G refers to the free energy of binding calculated for base-paired interactions between the ribozyme and the substrate RNA (Turner and Sugimoto, 1988 Ann. Rev. Biophys. Chem. 17, 167).
  • RPI A is a HH ribozyme with 6/6 binding arms.
  • Figs. 59 and 60 and 61 show cleavage of long substrate (622 nt) by ligated HH ribozymes.
  • Fig. 62 is a diagrammatic representation of a hammerhead ribozyme (HH-H) targeted against a site termed H. Variants of HH-H are also shown that contain either a 2 base-paired stem II (HH-H1 and HH-H2) or a 3 base- paired stem II (HH-H3 and HH-H4).
  • Figs. 63 and 64 show RNA cleavage activity of HH-I and its variants (see Fig.62). 63) cleavage of matched substrate RNA (15 nt). 64) cleavage of long substrate RNA (613 nt).
  • Figs. 65a-b is a schematic representation of a method of this invention to synthesize a full length hairpin ribozyme.
  • No splint strand is required for ligation but rather the two fragments hybridize together at helix 4 prior to ligation. The only prerequisite is that the 3' fragment is phosphorylated at its 5' end and that the 3' end of the 5' fragment have a hydroxyl group.
  • the hairpin ribozyme is targeted against site J.
  • H1 and H2 are intermolecular helices formed between the ribozyme and the substrate.
  • H3 and H4 are intramolecular helices formed within the hairpin ribozyme motif. Arrow indicates the cleavage site.
  • Fig. 66 shows RNA cleavage activity of ligated hairpin ribozymes targeted against site J.
  • Figs. 67a-b is a diagrammatic representation of a Site K Hairpin
  • Ribozyme (HP-K) showing the proposed secondary structure of the hairpin ribozyme•substrate complex as described in the art (Berzal-Herranz et al., 1993 EMBO. J.12, 2567).
  • the ribozyme has been assembled from two fragments (bimolecular ribozyme; Chowrira and Burke, 1992 Nucleic Acids Res. 20, 2835); #H1 and H2 represent intermolecular helix formation between the ribozyme and the substrate.
  • H3 and H4 represent intramolecular helix formation within the ribozyme (intermolecular helix in the case of bimolecular ribozyme).
  • FIG. 68 is a graph showing RNA cleavage by hairpin ribozymes targeted to site K. A plot of fraction of the target RNA uncleaved (fraction uncleaved) as a function of time is shown.
  • HP-K2 (6 bp helix 2) cleaves a 422 target RNA to a greater extent than the HP-K1 (4 bp helix 2).
  • a 422 nt region (containing hairpin site A) was synthesized by PCR using primers that place the T7 RNA promoter upstream of the amplified sequence.
  • Target RNA was transcribed in a standard transcription buffer in the presence of [ ⁇ - 32 P]CTP (Chowrira & Burke, 1991 supra).
  • the reaction mixture was treated with 15 units of ribonuclease-free DNasel, extracted with phenol followed chloroform:isoamyl alcohol (25:1), precipitated with isopropanol and washed with 70% ethanol.
  • the dried pellet was resuspended in 20 ⁇ l DEPC-treated water and stored at -20°C.
  • Unlabeled ribozyme (1 ⁇ M) and internally labeled 422 nt substrate RNA ( ⁇ 10 nM) were denatured and renatured separately in a standard cleavage buffer (containing 50 mM Tris ⁇ HCl pH 7.5 and 10 mM MgCl 2 ) by heating to 90°C for 2 min. and slow cooling to 37°C for 10 min. The reaction was initiated by mixing the ribozyme and substrate mixtures and incubating at 37°C. Aliquots of 5 ⁇ l were taken at regular time intervals, quenched by adding an equal volume of 2X formamide gel loading buffer and frozen on dry ice. The samples were resolved on 5% polyacrylamide sequencing gel and results were quantitatively analyzed by radioanalytic imaging of gels with a Phosphorlmager (Molecular Dynamics, Sunnyvale, CA).
  • Figs. 69a-b is the Site L Hairpin Ribozyme (HP-L) showing proposed secondary structure of the hairpin ribozyme•substrate complex.
  • the ribozyme was assembled from two fragments as described above. The nomenclature is the same as above.
  • Figure 70 shows RNA cleavage by hairpin ribozymes targeted to site L.
  • A plot of fraction of the target RNA uncleaved (fraction uncleaved) as a function of time is shown.
  • HP-L2 (6 bp helix 2) cleaves a 2 KB target RNA to a greater extent than the HP-L1 (4 bp helix 2).
  • a 2 kB region (containing hairpin site L) was synthesized by PCR using primers that place the T7 RNA promoter upstream of the amplified sequence. The cleavage reactions were carried out as described above.
  • Figs. 71 a-b shows a Site M Hairpin Ribozyme (HP-M) with the proposed secondary structure of the hairpin ribozyme•substrate complex. The ribozyme was assembled from two fragments as described above.
  • Figure 72 is a graph showing RNA cleavage by hairpin ribozymes targeted to site M.
  • the ribozymes were tested at both 20°C and at 26°C.
  • a 1.9 KB region (containing hairpin site M) was synthesized by PCR using primers that place the 77 RNA promoter upstream of the amplified sequence. Cleavage reactions were carried out as described above except that 20°C and at 26°C temperatures were used.
  • Figs. 73a-d shows various structural modifications of the present invention.
  • Helix 4 is replaced by a nucleotide loop wherein q is ⁇ 2 bases. Nomenclature is same as described under figure 3.
  • Figs. 74a-b shows Hairpin ribozymes containing nucleotide spacer region "s" at the indicated location, wherein s is ⁇ 1 base. Hairpin ribozymes containing spacer region, can be synthesized as one fragment or can be assembled from multiple fragments. Nomenclature is same as described under figure 3.
  • Figs. 75a-e shows the structures of the 5'-C-alkyl-modified nucleotides.
  • R 1 is as defined above.
  • R is OH, H, O-protecting group, NH, or any group described by the publications discussed above, and those described below.
  • B is as defined in the Figure or any other equivalent nucleotide base.
  • CE is cyanoethyl
  • DMT is a standard blocking group.
  • Figure 76 is a diagrammatic representation of the synthesis of 5'-C-alkyl-D-allose nucleosides and their phosphoramidites.
  • Figure 77 is a diagrammatic representation of the synthesis of 5'-C-alkyl-L-talose nucleosides and their phosphoramidites.
  • Figure 78 is a diagrammatic representation of hammerhead ribozymes targeted to site O containing 5'-C-methyl-L-talo modifications at various positions.
  • Figure 79 shows RNA cleavage activity of HH-O ribozymes. Fraction of target RNA uncleaved as a function of time is shown.
  • Figure 80 is a diagrammatic representation of a position numbered hammerhead ribozyme (according to Hertel et al. Nucleic Acids Res. 1 992, 20, 3252) showing specific substitutions.
  • Figs. 81 a-j shows the structures of various 2'-alkyl modified nucleotides which exemplify those of this invention.
  • R groups are alkyl groups, Z is a protecting group.
  • Figure 82 is a diagrammatic representation of the synthesis of 2'-C-allyl uridine and cytidine.
  • Figure 83 is a diagrammatic representation of the synthesis of 2'-C-methylene and 2'-C-difluoromethylene uridine.
  • Figure 84 is a diagrammatic representation of the synthesis of 2'-C-methylene and 2'-C-difluoromethylene cytidine.
  • Figure 85 is a diagrammatic representation of the synthesis of 2'-C-methylene and 2'-C-difluoromethylene adenosine.
  • Figure 86 is a diagrammatic representation of the synthesis of 2'-C-carboxymethylidine uridine, 2'-C-methoxycarboxymethylidine uridine and derivatized amidites thereof.
  • X is CH 3 or alkyl as discussed above, or another substituent.
  • Figure 87 is a diagrammatic representation of a synthesis of nucleoside 5'-deoxy-5'-difluoromethylphosphonates.
  • Figure 88 is a diagrammatic representation of the synthesis of nucleoside 5'-deoxy-5'-difluoromethylphosphonate 3'-phosphoramidites, dimers and solid supported dimers.
  • Figure 89 is a diagrammatic representation of the synthesis of nucleoside 5'-deoxy-5'-difluoromethylene triphosphates.
  • Figures 90 and 91 are diagrammatic representations of the synthesis of 3'-deoxy-3'-difluoromethylphosphonates and dimers.
  • Figure 92 is a schematic representation of synthesizing RNA phosphoramidite of a nucleotide containing a 2'-hydroxyl group modification of the present invention.
  • Figs. 93a-b describes a method for deprotection of oligonucleotides containing a 2'-hydroxyl group modification of the present invention.
  • Figure 94 is a diagrammatic representation of a hammerhead ribozyme targeted to site N. Positions of 2'-hydroxyl group substitution is indicated.
  • Figure 95 shows RNA cleavage activity of ribozymes containing a 2'-hydroxyl group modification of the present invention.
  • All RNA represents hammerhead ribozyme (HHN) with no 2'-hydroxyl group modifications.
  • U7-ala represents HHN ribozyme containing 2'-NH-alanine modification at the U7 position.
  • U4/U7-ala represents HHA containing 2'-NH-alanine modifications at U4 and U7 positions.
  • U4 lys represents HHA containing 2'-NH-lysine modification at U4 position.
  • U7 lys represents HHA containing 2'-NH-lysine modification at U7 position.
  • U4/U7-lys represents HHN containing 2'-NH-lysine modification at U4 and U7 positions.
  • Figures 96 and 97 are schematic representations of synthesizing
  • RNA with modification of the present invention refers to either a base, modified base or an H.
  • Figure 98 and 99 are schematic representations of synthesizing (solid-phase synthesis) 5' ends of RNA with modification of the present invention.
  • B refers to either a base, modified base or an H.
  • Figures 100 and 101 are general schematic representations of the invention.
  • Fig. 102a-d is a schematic representation of a method of the invention.
  • Fig. 103 is a graph of the results of the experiment diagrammed in figure 104.
  • Figure 104 is a diagrammatic representation of a fusion mRNA used in the experiment diagrammed in Fig. 102.
  • Figure 105 is a diagrammatic representation of a method for selection of useful ribozymes of this invention.
  • Figure 106 generally shows R-loop formation, and an R-loop complex. In addition, it indicates the location at which ligands can be provided to target the R-loop complex to cells using at least three different procedures, such as ligand receptor interaction, lipid or calcium phosphate mediated delivery, or electroporation.
  • Figure 107 shows a method for use of self-processing ribozymes to generate therapeutic ribozymes of unit length. This method is essentially described by Draper et al., PCT WO 93/23509.
  • Figure 108 shows a method of linking ligands like folate, carbohydrate or peptides to R-loop forming RNA.
  • Ribozymes of this invention block to some extent ICAM-1 , IL-5, rel A, TNF- ⁇ , p210 bcr-abI , or RSV genes expression and can be used to treat diseases or diagnose such diseases. Ribozymes will be delivered to cells in culture and to tissues in animal models. Ribozyme cleavage of ICAM-1 , II-5, rel A, TNF- ⁇ ,p210 bcr-abl , or RSV mRNA in these systems may prevent or alleviate disease symptoms or conditions.
  • Target sites Targets for useful ribozymes can be determined as disclosed in
  • RNA folding algorithm It must be established that the sites predicted by the computer-based RNA folding algorithm correspond to potential cleavage sites.
  • Hammerhead or hairpin ribozymes are designed that could bind and are individually analyzed by computer folding (Jaeger et al., 1989 Proc. Natl. Acad. Sci., USA, 86 7706-7710) to assess whether the ribozyme sequences fold into the appropriate secondary structure. Those ribozymes with unfavorable intramolecular interactions between the binding arms and the catalytic core are eliminated from consideration. Varying binding arm lengths can be chosen to optimize activity. Generally, at least 5 bases on each arm are able to bind to, or otherwise interact with, the target RNA.
  • RNA is screened for accessible cleavage sites by the method described generally in Draper et al., PCT WO93/23569 hereby incorporated by reference herein. Briefly, DNA oligonucleotides representing potential hammerhead or hairpin ribozyme cleavage sites are synthesized. A polymerase chain reaction is used to generate a substrate for T7 RNA polymerase transcription from cDNA clones. Labeled RNA transcripts are synthesized in vitro from DNA templates. The oligonucleotides and the labeled trascripts are annealed, RNaseH is added and the mixtures are incubated for the designated times at 37°C. Reactions are stopped and RNA separated on sequencing polyacrylamide gels. The percentage of the substrate cleaved is determined by autoradiographic quantitation using a phosphor imaging system. From these data, hammerhead or hairpin ribozynme sites are chosen as the most accessible.
  • Ribozymes of the hammerhead or hairpin motif are designed to anneal to various sites in the mRNA message.
  • the binding arms are complementary to the target site sequences desribed above.
  • the ribozymes are chemically synthesized.
  • the method of synthesis used follows the procedure for normal RNA synthesis as described in Usman et al., 1987 J. Am. Chem. Soc., 109, 7845 and in Scaringe et al., 1990 Nucleic Acids Res., 18, 5433 and made use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, phosphoramidites at the 3'-end.
  • the average stepwise coupling yeilds are >98%.
  • Inactive ribozymes are synthesized by substituting a U for G5 and a U for A 14 (numbering from Hertel et al., 1992 Nucleic Acids Res., 20, 3252). Hairpin ribozymes are synthesized in two parts and annealed to reconstruct the active ribozyme (Chowrira and Burke, 1992 Nucleic Acids Res., 20, 2835-2840). Ribozymes are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbach, 1989, Methods Enzymol, 180, 51).
  • ribozymes are modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-O-methyl, 2'H (for a review see Usman and Cedergren, 1992 TIBS 17,34). Ribozymes are purified by gel electrophoresis using heneral methods or are purified by high pressure liquid chromatography and are resuspended in water.
  • nuclease resistant groups for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-O-methyl, 2'H (for a review see Usman and Cedergren, 1992 TIBS 17,34).
  • Ribozymes are purified by gel electrophoresis using heneral methods or are purified by high pressure liquid chromatography and are resuspended in water.
  • Ribozymes that cleave ICAM-1 mRNA represent a novel therapeutic approach to inflammatory or autoimmune disorders. ICAM-1 function can be blocked therapeutically using monoclonal antibodies. Ribozymes have the advantage of being generally immunologically inert, whereas significant neutralizing anti-IgG responses can be observed with some monoclonal antibody treatments.
  • Intercellular adhesion molecule-1 is a cell surface protein whose expression is induced by inflammatory mediators. ICAM-1 is required for adhesion of leukocytes to endothelial cells and for several immunological functions including antigen presentation, immunoglobulin production and cytotoxic cell activity. Blocking ICAM-1 function prevents immune cell recognition and activity during transplant rejection and in animal models of rheumatoid arthritis, asthma and reperfusion injury. Cell-cell adhesion plays a pivotal role in inflammatory and immune responses (Springer et al., 1987 Ann. Rev. Immunol. 5, 223-252). Cell adhesion is required for leukocytes to bind to and migrate through vascular endothelial cells.
  • Intercellular adhesion molecule-1 is a 110 kilodalton member of the immunoglobulin superfamily that is involved in all of these cell-cell interactions (Simmons et al., 1988 Nature (London) 331, 624-627). ICAM-1 is expressed on only a limited number of cells and at low levels in the absence of stimulation (Dustin et al., 1986 J. Immunol. 137, 245-254).
  • inflammatory mediators lipopolysaccharide, ⁇ -interferon, tumor necrosis factor- ⁇ , or interleukin-1
  • a variety of cell types endothelial, epithelial, fibroblastic and hematopoietic cells
  • ICAM-1 interleukin-1
  • a variety of cell types endothelial, epithelial, fibroblastic and hematopoietic cells
  • ICAM-1 endothelial, epithelial, fibroblastic and hematopoietic cells
  • Induction occurs via increased transcription of ICAM-1 mRNA (Simmons et al., supra). Elevated expression is detectable after 4 hours and peaks after 16 - 24 hours of induction.
  • ICAM-1 induction is critical for a number of inflammatory and immune responses.
  • antibodies to ICAM-1 block adhesion of leukocytes to cytokine-activated endothelial cells (Boyd,1988 Proc. Natl. Acad. Sci. USA 85, 3095-3099; Dustin and Springer, 1988 J. Cell Biol. 107, 321-331 ).
  • ICAM-1 expression may be required for the extravasation of immune cells to sites of inflammation.
  • Antibodies to ICAM-1 also block T cell killing, mixed lymphocyte reactions, and T cell-mediated B cell differentiation, suggesting that ICAM-1 is required for these cognate cell interactions (Boyd et al., supra).
  • ICAM-1 in antigen presentation is underscored by the inability of ICAM-1 defective murine B cell mutants to stimulate antigen-dependent T cell proliferation (Dang et al., 1990 J. Immunol. 144, 4082-4091). Conversely, murine L cells require transfection with human ICAM-1 in addition to HLA-DR in order to present antigen to human T cells (Altmann et al., 1989 Nature (London) 338, 512-514). In summary, evidence in vitro indicates that ICAM-1 is required for cell-cell interactions critical to inflammatory responses, cellular immune responses, and humoral antibody responses. By engineering ribozyme motifs we have designed several ribozymes directed against ICAM-1 mRNA sequences.
  • ribozymes cleave ICAM-1 target sequences in vitro.
  • the sequence of human, rat and mouse ICAM-1 mRNA can be screened for accessible sites using a computer folding algorithm. Regions of the mRNA that did not form secondary folding structures and that contain potential hammerhead or hairpin ribozyme cleavage sites can be identified. These sites are shown in Tables 2, 3, and 6-9. (All sequences are 5' to 3' in the tables) While rat, mouse and human sequences can be screened and ribozymes thereafter designed, the human targeted sequences are of most utility.
  • Ribozymes will be delivered by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA or RNA vectors described above. Cytokine-induced ICAM-1 expression will be monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. ICAM-1 mRNA levels will be assessed by Northern, by RNAse protection, by primer extension or by quantitative RT-PCR analysis. Ribozymes that block the induction of ICAM-1 protein and mRNA by more than 90% will be identified.
  • ribozymes and/or genes encoding them will be locally delivered to transplant tissue ex vivo in animal models. Expression of the ribozyme will be monitored by its ability to block ex vivo induction of ICAM-1 mRNA and protein. The effect of the anti-ICAM-1 ribozymes on graft rejection will then be assessed. Similarly, ribozymes will be introduced into joints of mice with collagen-induced arthritis or rabbits with Streptococcal cell wall-induced arthritis. Liposome delivery, cationic lipid delivery, or adeno-associated virus vector delivery can be used.
  • One dose is disclosed by Sullivan et al., PCT WO94/02595, incorporated by reference herein.
  • a stable anti-ICAM-1 ribozyme or a gene construct that constitutively expresses the ribozyme may abrogate inflammatory and immune responses in these diseases.
  • ICAM-1 plays a central role in immune cell recognition and function.
  • Ribozyme inhibition of ICAM-1 expression can reduce transplant rejection and alleviate symptoms in patients with rheumatoid arthritis, asthma or other acute and chronic inflammatory disorders.
  • these targets can be therapeutically treated with agents that reduce
  • ICAM-1 expression or function are listed below. This list is not meant to be complete and those in the art will recognize further conditions and diseases that can be effectively treated using ribozymes of the present invention.
  • ICAM-1 is expressed on venules and capillaries of human cardiac biopsies with histological evidence of graft rejection (Briscoe et al., 1991 Transplantation 51 , 537-539).
  • Anti-ICAM antibody inhibits adjuvant-induced arthritis in rats (ligo et al., 1991 J Immunol 147, 4167-71).
  • Anti-ICAM-1 antibody blocks adherence of neutrophils to anoxic endothelial cells (Yoshida et al., 1992 Am J Physiol 262, H1891-8).
  • Anti-ICAM-1 antibody reduces neurological damage in a rabbit model of cerebral stroke (Bowes et al., 1993 Exp Neurol 119, 215-9).
  • Anti-ICAM-1 antibody protects against reperfusion injury in a cat model of myocardial ischemia (Ma et al., 1992 Circulation 86, 937-46).
  • Antibody to ICAM-1 partially blocks eosinophil adhesion to endothelial cells and is overexpressed on inflamed airway endothelium and epithelium in vivo (Wegner et al., 1990 Science 247, 456-9). In a primate model of asthma, anti-ICAM-1 antibody blocks airway eosinophilia
  • ICAM-1 expression correlates with the disease and is reduced by effective immunoglobulin treatment (Leung, et al., 1989 Lancet 2, 1298-302). Soluble ICAM levels are elevated in Kawasaki disease patients; particularly high levels are observed in patients with coronary artery lesions (Furukawa et al., 1992 Arthritis Rheum 35, 672-7; Tsuji, 1992 Arerugi 41 , 1507-14).
  • Circulating LFA-1+ T cells are depleted (presumably due to ICAM-1 mediated extravasation) in Kawasaki disease patients (Furukawa et al., 1993 Scand J Immunol 37, 377-80).
  • Ribozymes that cleave IL-5 mRNA represent a novel therapeutic approach to inflammatory disorders like asthma.
  • the invention features use of ribozymes to treat chronic asthma, e.g., by inhibiting the synthesis of IL-5 in lymphocytes and preventing the recruitment and activation of eosinophils.
  • cytokines besides IL-5 may also be involved in the activation of inflammation in asthmatic patients, including platelet activating factor, IL-1 , IL-3, IL-4, GM-CSF, TNF- ⁇ , gamma interferon, VCAM, ILAM-1 , ELAM-1 and NF- ⁇ B.
  • platelet activating factor IL-1 , IL-3, IL-4, GM-CSF, TNF- ⁇ , gamma interferon, VCAM, ILAM-1 , ELAM-1 and NF- ⁇ B.
  • any cellular receptors which mediate the activities of the cytokines are also good targets for intervention in inflammatory diseases. These targets include, but are not limited to, the IL-1 R and TNF- ⁇ R on keratinocytes, epithelial and endothelial cells in airways.
  • Recent data suggest that certain neuropeptides may play a role in asthmatic symptoms. These peptides include substance P, neurokinin A and calcit
  • Ribozymes of this invention block to some extent IL-5 expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of asthma (Clutterbuck et al., 1989 supra: Garssen et al., 1991 Am. Rev. Respir. Dis. 144, 931-938; Larsen et al., 1992 J. Clin. Invest. 89, 747-752; Mauser et al., 1993 supra). Ribozyme cleavage of IL-5 mRNA in these systems may prevent inflammatory cell function and alleviate disease symptoms.
  • mice and mouse IL-5 mRNA were screened for accessible sites using a computer folding algorithm. Potential hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 11 , 13, and 14, 15. (All sequences are 5' to 3' in the tables.) While mouse and human sequences can be screened and ribozymes thereafter designed, the human targeted sequences are of most utility. However, mouse targeted ribozymes are useful to test efficacy of action of the ribozyme prior to testing in humans. The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme.
  • stem-loop IV sequence of hairpin ribozymes listed in Tables 15 and 16 (5'-CACGUUGUG-3') can be altered (substitution, deletion and/or insertion) to contain any sequence provided, a minimum of two base-paired stem structure can form.
  • the sequences listed in Tables 12, 14 - 16 may be formed of ribonucleotides or other nucleotides or non-nucleotides.
  • Such ribozymes are equivalent to the ribozymes described specifically in the Tables.
  • By engineering ribozyme motifs we have designed several ribozymes directed against IL-5 mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave IL-5 target sequences in vitro is evaluated.
  • Ribozymes will be tested for function in vivo by analyzing IL-5 expression levels. Ribozymes will be delivered to cells by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA or RNA vectors. IL-5 expression will be monitored by biological assays, ELISA, by indirect immunofluoresence, and/or by FACS analysis. IL-5 mRNA levels will be assessed by Northern analysis, RNAse protection or primer extension analysis or quantitative RT-PCR. Ribozymes that block the induction of IL-5 activity and/or IL-5 mRNA by more than 90% will be identified.
  • Interleukin 5 a cytokine produced by CD4+ T helper cells and mast cells, was originally termed B cell growth factor II (reviewed by Takatsu et al., 1988 Immunol. Rev. 102, 107). It stimulates proliferation of activated B cells and induces production of IgM and IgA.
  • IL-5 plays a major role in eosinophil function by promoting differentiation (Clutterbuck et al., 1989 Blood 73, 1504-12), vascular adhesion (Walsh et al., 1990 Immunology 71 , 258-65) and in vitro survival of eosinophils (Lopez et al., 1988 J. Exp. Med. 167, 219-24).
  • Bronchoalveolar lavage cells were screened for production of cytokines using in situ hybridization for mRNA.
  • In situ hybridization signals were detected for IL-2, IL-3, IL-4, IL-5 and GM-CSF.
  • Upregulation of mRNA was observed for IL-4, IL-5 and GM-CSF (Robinson et al., 1993 J. Allergy Clin. Immunol. 92, 313-24).
  • Another study showed that upregulation of IL-5 transcripts from allergen challenged vs. saline challenged asthmatic patients (Krishnaswamy et al., 1993 Am. J. Respir. Cell. Mol. Biol. 9, 279-86).
  • eosinophil phenotype was the same for asthmatic patients and normal individuals. However, eosinophils from asthmatic patients had greater leukotriene C4 producing capacity and migration capacity. There were elevated levels of IL-3, IL-5 and GM-CSF in the circulation of asthmatics but not in normal individuals (Bruijnzeel et al., 1992 Sau. Med. Schuschr. 122, 298-301).
  • Efficacy of antibody to IL-5 was assessed in a guinea pig asthma model.
  • the animals were challenged with ovalbumin and assayed for eosinophilia and the responsiveness to the bronchioconstriction substance P.
  • a 30 mg/kg dose of antibody administered i.p. blocked ovalbumin- induced increased sensitivity to substance P and blocked increases in bronchoalveolar and lung tissue accumulation of eosinophils (Mauser et al., 1993 Am. Rev. Respir. Dis. 148, 1623-7).
  • guinea pigs challenged for eight days with ovalbumin were treated with monoclonal antibody to IL-5.
  • Results obtained from human clinical analysis and animal studies indicate the role of activated T helper cells, cytokines and eosinophils in asthma.
  • the role of IL-5 in eosinophil development and function makes IL-5 a good candidate for target selection.
  • the antibody studies neutralized IL-5 in the circulation thus preventing eosinophilia. Inhibition of the production of IL-5 will achieve the same goal.
  • Asthma - a prominent feature of asthma is the infiltration of eosinophils and deposition of toxic eosinophil proteins (e.g. major basic protein, eosinophil-derived neurotoxin) in the lung.
  • eosinophil proteins e.g. major basic protein, eosinophil-derived neurotoxin
  • a number of T-cell-derived factors like IL-5 are responsible for the activation and maintainance of eosinophils (Kay, 1991 J. Allergy Clin. Immun. 87, 893). Inhibition of IL-5 expression in the lungs can decrease the activation of eosinophils and will help alleviate the symptoms of asthma.
  • Atopy - is characterized by the developement of type I hypersensitive reactions associated with exposure to certain environmental antigens.
  • One of the common clinical manifestations of atopy is eosinophilia (accumulation of abnormally high levels of eosinophils in the blood).
  • Antibodies against IL-5 have been shown to lower the levels of eosinophils in mice (Cook et al., 1993 in Immunopharmacol. Eosinophils ed. Smith and Cook, pp. 193-216, Academic, London, UK)
  • mice Parasitic infection-related eosinophilia- infections with parasites like helminths, can lead to severe eosinophilia (Cook et al., 1993 supra). Animal models for eosinophilia suggest that infection of mice, for example, can lead to blood, peritoneal and/or tissue eosinophilia, all of which seem to be lowered to varying degrees by antibodies directed against IL-5.
  • Pulmonary infiltration eosinophilia- is characterised by accumulation of high levels of eosinophils in pulmonary parenchyma (Gleich, 1990 J. Aller ⁇ v Clin. Immunol. 85, 422). L-Tryptophan-associated eosinophilia-myalgia syndrome
  • EMS EMS- The EMS disease is closely linked to the consumption of L-tryptophan, an essential aminoacid used to treat conditions like insomnia (for review see Varga et al., 1993 J Invest. Dermatol. 100, 97s).
  • Pathologic and histologic studies have demonstrated high levels of eosinophils and mononuclear inflammatory cells in patients with EMS. It appears that IL-5 and transforming growth factor play a significant role in the development of EMS (Varga et al., 1993 supra) by activating eosinophils and other inflammatory cells.
  • ribozymes of the present invention that cleave IL-5 mRNA and thereby IL-5 activity have many potential therapeutic uses, and there are reasonable modes of delivering the ribozymes in a number of the possible indications.
  • Development of an effective ribozyme that inhibits IL-5 function is described above; available cellular and activity assays are numerous, reproducible, and accurate.
  • Ribozymes that cleave rel A mRNA represent a novel therapeutic approach to inflammatory or autoimmune disorders.
  • Inflammatory mediators such as lipopolysaccharide (LPS), interleukin-1 (IL-1) or tumor necrosis factor-a (TNF- ⁇ ) act on cells by inducing transcription of a number of secondary mediators, including other cytokines and adhesion molecules.
  • LPS lipopolysaccharide
  • IL-1 interleukin-1
  • TNF- ⁇ tumor necrosis factor-a
  • Ribozyme therapy due to its extraordinar specificity, is particularly well-suited to target intracellular factors that contribute to disease pathology.
  • ribozymes that cleave mRNA encoded by rel A or TNF- ⁇ may represent novel therapeutics for the treatment of inflammatory and autoimmune disorders.
  • NF- ⁇ B nuclear DNA-binding activity
  • NF- ⁇ B The activity first described as NF- ⁇ B is a heterodimer of p49 or p50 with p65.
  • the p49 and p50 subunits of NF- ⁇ B (encoded by the nf- ⁇ B2 or nf- ⁇ B1 genes, respectively) are generated from the precursors NF- ⁇ B1 (p105) or NF- ⁇ B2 (p100).
  • the p65 subunit of NF- ⁇ B (now termed Rel A ) is encoded by the rel A locus.
  • heterodimers of NF- ⁇ B2/RelA act with Tat-I to activate transcription of the HIV genome, while NF- ⁇ B1/RelA (p50/p65) heterodimers have little effect (J. Liu, N.D. Perkins, R.M. Schmid, G.J. Nabel, J. Virol. 1992 66, 3883-3887).
  • blocking rel A gene expression with antisense oligonucleotides specifically blocks embryonic stem cell adhesion; blocking NF- ⁇ B1 gene expression with antisense oligonucleotides had no effect on cellular adhesion (Narayanan et al., 1993 Mol. Cell. Biol. 13, 3802-3810).
  • NF- ⁇ B A number of specific inhibitors of NF- ⁇ B function in cells exist, including treatment with phosphorothioate antisense oliogonucleotide, treatment with double-stranded NF- ⁇ B binding sites, and over expression of the natural inhibitor MAD-3 (an 1 ⁇ B family member). These agents have been used to show that NF- ⁇ B is required for induction of a number of molecules involved in inflammation, as described below.
  • NF- ⁇ B is required for phorbol ester-mediated induction of IL-6 (I. Kitajima, et al., Science 258, 1792-5 (1992)) and IL-8 (Kunsch and Rosen, 1993 Mol. Cell. Biol. 13, 6137-46).
  • NF- ⁇ B is required for induction of the adhesion molecules ICAM-1 (Eck, et al., 1993 Mol. Cell. Biol. 13, 6530-6536), VCAM-1 (Shu et al., supra), and E-selectin (Read, et al., 1994 J. Exp. Med. 179, 503-512) on endothelial cells.
  • ICAM-1 adhesion molecules
  • VCAM-1 VCAM-1
  • E-selectin Read, et al., 1994 J. Exp. Med. 179, 503-512
  • NF- ⁇ B is involved in the induction of the integrin subunit, CD18, and other adhesive properties of leukocytes (Eck et al., 1993 supra).
  • glucocorticoids may exert their anti-inflammatory effects by inhibiting NF- ⁇ B.
  • the glucocorticoid receptor and p65 both act at NF- ⁇ B binding sites in the ICAM-1 promoter (van de Stolpe, et al., 1994 J. Biol. Chem. 269, 6185-6192).
  • Glucocorticoid receptor inhibits NF- ⁇ B-mediated induction of IL-6 (Ray and Prefontaine, 1994 Proc. Natl Acad. Sci USA 91 , 752-756).
  • Ribozymes of this invention block to some extent NF- ⁇ B expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of restenosis, transplant rejection and rheumatoid arthritis. Ribozyme cleavage of relA mRNA in these systems may prevent inflammatory cell function and alleviate disease symptoms.
  • the sequence of human and mouse re/A mRNA can be screened for accessible sites using a computer folding algorithm. Potential hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 17, 18 and 21-22. (All sequences are 5' to 3' in the tables.) While mouse and human sequences can be screened and ribozymes thereafter designed, the human targetted sequences are of most utility.
  • the ribozymes will be tested for function in vivo by analyzing cytokine-induced VCAM-1 , ICAM-1 , IL-6 and IL-8 expression levels. Ribozymes will be delivered to cells by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA and RNA vectors. Cytokine-induced VCAM-1 , ICAM-1 , IL-6 and IL-8 expression will be monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. Rel A mRNA levels will be assessed by Northern analysis, RNAse protection or primer extension analysis or quantitative RT-PCR. Activity of NF- ⁇ B will be monitored by gel-retardation assays.
  • Ribozymes that block the induction of NF- ⁇ B activity and/or rel A mRNA by more than 50% will be identified.
  • RNA ribozymes and/or genes encoding them will be locally delivered to transplant tissue ex vivo in animal models. Expression of the ribozyme will be monitored by its ability to block ex vivo induction of VCAM-1 , ICAM-1 , IL-6 and IL-8 mRNA and protein. The effect of the anti-rel A ribozymes on graft rejection will then be assessed.
  • ribozymes will be introduced into joints of mice with collagen-induced arthritis or rabbits with Streptococcal cell wall-induced arthritis.
  • Liposome delivery cationic lipid delivery, or adeno-associated virus vector delivery can be used.
  • One dose (or a few infrequent doses) of a stable anti-relA ribozyme or a gene construct that constitutively expresses the ribozyme may abrogate inflammatory and immune responses in these diseases.
  • a therapeutic agent that inhibits cytokine gene expression, inhibits adhesion molecule expression, and mimics the anti-inflammatory effects of glucocorticoids (without inducing steroid-responsive genes) is ideal for the treatment of inflammatory and autoimmune disorders.
  • Disease targets for such a drug are numerous. Target indications and the delivery options each entails are summarized below. In all cases, because of the potential immunosuppressive properties of a ribozyme that cleaves rel A mRNA, uses are limited to local delivery, acute indications, or ex vivo treatment. •Rheumatoid arthritis (RA).
  • a gene therapy approach is logical. Delivery of a ribozyme to inflamed joints is mediated by adenovirus, retrovirus, or adeno-associated virus vectors.
  • the appropriate adenovirus vector can be administered by direct injection into the synovium: high efficiency of gene transfer and expression for several months would be expected (B.J. Roessler, E.D. Allen, J.M. Wilson, J.W. Hartman, B. L. Davidson, J. Clin. Invest. 92, 1085-1092 (1993)). It is unlikely that the course of the disease could be reversed by the transient, local administration of an anti-inflammatory agent. Multiple administrations may be necessary. Retrovirus and adeno-associated virus vectors would lead to permanent gene transfer and expression in the joint. However, permanent expression of a potent anti-inflammatory agent may lead to local immune deficiency.
  • NF- ⁇ B expression of NF- ⁇ B in the vessel wall of pigs causes a narrowing of the luminal space due to excessive deposition of extracellular matrix components. This phenotype is similar to matrix deposition that occurs subsequent to coronary angioplasty.
  • NF- ⁇ B is required for the expression of the oncogene c-myb (F.A. La Rosa, J.W. Pierce, G.E. Soneneshein, Mol. Cell. Biol. 14, 1039-44 (1994)).
  • NF- ⁇ B induces smooth muscle proliferation and the expression of excess matrix components: both processes are thought to contribute to reocclusion of vessels after coronary angioplasty.
  • NF- ⁇ B is required for the induction of adhesion molecules (Eck et al., supra, K. O'Brien, et al., J. Clin. Invest. 92, 945-951 (1993)) that function in immune recognition and inflammatory responses.
  • adhesion molecules Eck et al., supra, K. O'Brien, et al., J. Clin. Invest. 92, 945-951 (1993)
  • At least two potential modes of treatment are possible.
  • transplanted organs are treated ex vivo with ribozymes or ribozyme expression vectors.
  • Transient inhibition of NF- ⁇ B in the transplanted endothelium may be sufficient to prevent transplant-associated vasculitis and may significantly modulate graft rejection.
  • donor B cells are treated ex vivo with ribozymes or ribozyme expression vectors.
  • Recipients would receive the treatment prior to transplant.
  • Treatment of a recipient with B cells that do not express T cell co-stimulatory molecules can induce antigen-specific anergy. Tolerance to the donor's histocompatibility antigens could result; potentially, any donor could be used for any transplantation procedure. •Asthma.
  • Granulocyte macrophage colony stimulating factor (GM-CSF) is thought to play a major role in recruitment of eosinophils and other inflammatory cells during the late phase reaction to asthmatic trauma. Again, blocking the local induction of GM-CSF and other inflammatory mediators is likely to reduce the persistent inflammation observed in chronic asthmatics. Aerosol delivery of ribozymes or adenovirus ribozyme expression vectors is a feasible treatment.
  • ribozymes of the present invention that cleave rel A mRNA and thereby NF- ⁇ B activity have many potential therapeutic uses, and there are reasonable modes of delivering the ribozymes in a number of the possible indications.
  • Development of an effective ribozyme that inhibits NF- ⁇ B function is described above; available cellular and activity assays are number, reproducible, and accurate.
  • Animal models for NF- ⁇ B function (Kitajima, et al., supra) and for each of the suggested disease targets exist and can be used to optimize activity.
  • Example 4 TNF- ⁇
  • Ribozymes that cleave the specific cites in TNF- ⁇ mRNA represent a novel therapeutic approach to inflammatory or autoimmune disorders.
  • Tumor necrosis factor- ⁇ is a protein, secreted by activated leukocytes, that is a potent mediator of inflammatory reactions. Injection of TNF- ⁇ into experimental animals can simulate the symptoms of systemic and local inflammatory diseases such as septic shock or rheumatoid arthritis.
  • TNF- ⁇ was initially described as a factor secreted by activated macrophages which mediates the destruction of solid tumors in mice (Old, 1985 Science 230, 4225-4231). TNF- ⁇ subsequently was found to be identical to cachectin, an agent responsible for the weight loss and wasting syndrome associated with tumors and chronic infections (Beutler, et al., 1985 Nature 316, 552-554). The cDNA and the genomic locus for TNF- ⁇ have been cloned and found to be related to TNF- ⁇ (Shakhov et al., 1990 J. Exp. Med. 171 , 35-47). Both TNF- ⁇ and TNF- ⁇ bind to the same receptors and have nearly identical biological activities.
  • TNF- ⁇ secretion has been detected from monocytes/macrophages, CD4+ and CD8+ T-cells, B-cells, lymphokine activated killer cells, neutrophils, astrocytes, endothelial cells, smooth muscle cells, as well as various non-hematopoietic tumor cell lines (for a review see Turestskaya et al., 1991 in Tumor Necrosis Factor: Structure. Function, and Mechanism of Action B. B. Aggarwal, J. Vilcek, Eds. Marcel Dekker, Inc., pp. 35-60).
  • TNF- ⁇ is regulated transcriptionally and translationally, and requires proteolytic processing at the plasma membrane in order to be secreted (Kriegler et al., 1988 Cell 53, 45-53). Once secreted, the serum half life of TNF- ⁇ is approximately 30 minutes. The tight regulation of TNF- ⁇ is important due to the extreme toxicity of this cytokine. Increasing evidence indicates that overproduction of TNF- ⁇ during infections can lead to severe systemic toxicity and death (Tracey & Cerami, 1992 Am. J. Trop. Med. Hv ⁇ . 47, 2-7).
  • Antisense RNA and Hammerhead ribozymes have been used in an attempt to lower the expression level of TNF- ⁇ by targeting specified cleavage sites [Sioud et al., 1992 J. Mol. Biol. 223; 831 ; Sioud WO 94/10301 ; Kisich and co-workers, 1990 abstract (FASEB J. 4. A1860; 1991 slide presentation (J. Leukocyte Biol. sup.
  • Ribozymes of this invention block to some extent TNF- ⁇ expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of septic shock and rheumatoid arthritis. Ribozyme cleavage of TNF- ⁇ mRNA in these systems may prevent inflammatory cell function and alleviate disease symptoms.
  • the sequence of human and mouse TNF- ⁇ mRNA can be screened for accessible sites using a computer folding algorithm. Hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 23, 25, and 27 - 28. (All sequences are 5' to 3' in the tables.) While mouse and human sequences can be screened and ribozymes thereafter designed, the human targeted sequences are of most utility. However, mouse targeted ribozymes are useful to test efficacy of action of the ribozyme prior to testing in humans. The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme. (In Table 24, lower case letters indicate positions that are not conserved between the human and the mouse TNF- ⁇ sequences.)
  • stem-loop IV sequence of hairpin ribozymes listed in Tables 27 and 28 (5'-CACGUUGUG-3') can be altered (substitution, deletion, and/or insertion) to contain any sequence, provided a minimum of two base-paired stem structure can form.
  • the sequences listed in Tables 24, 26 - 28 may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables or AAV .
  • a transcription unit expressing a ribozyme that cleaves TNF- ⁇ RNA is inserted into a plasmid
  • DNA vector or an adenovirus DNA viral vector or AAV or alpha virus or retroviris vectors have been used to transfer genes to the intact vasculature or to joints of live animals (Willard et al., 1992
  • the adenovirus vector is delivered as recombinant adenoviral particles.
  • DNA may be delivered alone or complexed with vehicles (as described for RNA above).
  • DNA/vehicle complexes, or the recombinant adenovirus particles are locally administered to the site of treatment, e.g., through the use of an injection catheter, stent or infusion pump or are directly added to cells or tissues ex vivo.
  • a transcription unit expressing a ribozyme that cleaves TNF- ⁇ RNA is inserted into a retrovirus vector for sustained expression of ribozyme(s).
  • ribozyme motifs we have designed several ribozymes directed against TNF- ⁇ mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave TNF- ⁇ target sequences in vitro is evaluated.
  • the ribozymes will be tested for function in cells by analyzing bacterial lipopolysaccharide (LPS)-induced TNF- ⁇ expression levels. Ribozymes will be delivered to cells by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA vectors. TNF- ⁇ expression will be monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. TNF- ⁇ mRNA levels will be assessed by Northern analysis, RNAse protection, primer extension analysis or quantitative RT-PCR. Ribozymes that block the induction of TNF- ⁇ activity and/or TNF- ⁇ mRNA by more than 90% will be identified.
  • LPS bacterial lipopolysaccharide
  • RNA ribozymes and/or genes encoding them will be locally delivered to macrophages by intraperitoneal injection. After a period of ribozyme uptake, the peritoneal macrophages are harvested and induced ex vivo with LPS. The ribozymes that significantly reduce TNF- ⁇ secretion are selected. The TNF- ⁇ can also be induced after ribozyme treatment with fixed Streptococcus in the peritoneal cavity instead of ex vivo. In this fashion the ability of TNF- ⁇ ribozymes to block TNF- ⁇ secretion in a localized inflammatory response are evaluated. In addition, we will determine if the ribozymes can block an ongoing inflammatory response by delivering the TNF- ⁇ ribozymes after induction by the injection of fixed Streptococcus.
  • the ribozymes are delivered by intravenous injection.
  • the ability of the ribozymes to inhibit TNF- ⁇ secretion and lethal shock caused by systemic LPS administration are assessed.
  • TNF- ⁇ ribozymes can be introduced into the joints of mice with collagen-induced arthritis.
  • Either free delivery, liposome delivery, cationic lipid delivery, adeno-associated virus vector delivery, adenovirus vector delivery, retrovirus vector delivery or plasmid vector delivery in these animal model experiments can be used to supply ribozymes.
  • One dose (or a few infrequent doses) of a stable anti-TNF- ⁇ ribozyme or a gene construct that constitutively expresses the ribozyme may abrogate tissue damage in these inflammatory diseases.
  • thioglycollate broth (Difco, Detroit, MI.) was injected i.p. into 6 week old female C57bl/6NCR mice 3 days before peritoneal lavage. Mice were maintained as specific pathogen free in autoclaved cages in a laminar flow hood and given sterilized water to minimize "spontaneous" activation of macrophages.
  • the resulting peritoneal exudate cells were obtained by lavage using Hanks balanced salt solution (HBSS) and were plated at 2.5X10 5 /well in 96 well plates (Costar, Cambridge, MA.) with Eagles minimal essential medium (EMEM) containing 10% heat inactivated fetal bovine serum. After adhering for 2 hours the wells were washed to remove non-adherent cells. The resulting cultures were 97% macrophages as determined by morphology and staining for non-specific esterase.
  • HBSS Hanks balanced salt solution
  • EMEM Eagles minimal essential medium
  • ribozymes Transfection of ribozymes into macrophages: The ribozymes were diluted to 2X final concentration, mixed with an equal volume of 11 nM lipofectamine (Life Technologies, Gaithersburg, MD.), and vortexed. 100 ml of lipid:ribozyme complex was then added directly to the cells, followed immediately by 10 ml fetal bovine serum. Three hours after ribozyme addition 100 ml of 1 mg/ml bacterial lipopolysaccaride (LPS) was added to each well to stimulate TNF production.
  • LPS bacterial lipopolysaccaride
  • TNF- ⁇ was sampled at 0, 2, 4, 8, and 24 hours post LPS stimulation and stored at -70°C. Quantitation of TNF- ⁇ was done by a specific ELISA. ELISA plates were coated with rabbit anti-mouse TNF- ⁇ serum at 1 :1000 dilution (Genzyme) followed by blocking with milk proteins and incubation with TNF- ⁇ containing supernatants. TNF- ⁇ was then detected using a murine TNF- ⁇ specific hamster monoclonal antibody (Genzyme). The ELISA was developed with goat anti-hamster IgG coupled to alkaline phosphatase.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide
  • TNF- ⁇ The association between TNF- ⁇ and bacterial sepsis, rheumatoid arthritis, and autoimmune disease make TNF- ⁇ an attractive target for therapeutic intervention [Tracy & Cerami 1992 supra: Williams et al., 1992
  • Septic shock is a complication of major surgery, bacterial infection, and polytrauma characterized by high fever, increased cardiac output, reduced blood pressure and a neutrophilic infiltrate into the lungs and other major organs.
  • Current treatment options are limited to antibiotics to reduce the bacterial load and non-steroidal anti-inflammatories to reduce fever.
  • mortality from septic shock averages 50%, due primarily to multiple organ failure and disseminated vascular coagulation.
  • Septic shock with an incidence of 200,000 cases per year in the United States, is the major cause of death in intensive care units.
  • tissue injury or bacterial products initiate massive immune activation, resulting in the secretion of pro-inflammatory cytokines which are not normally detected in the serum, such as TNF- ⁇ , interleukin-1 ⁇ (IL-1 ⁇ ), ⁇ -interferon (IFN- ⁇ ), interleukin-6 (IL-6), and interleukin-8 (IL-8).
  • cytokines which are not normally detected in the serum, such as TNF- ⁇ , interleukin-1 ⁇ (IL-1 ⁇ ), ⁇ -interferon (IFN- ⁇ ), interleukin-6 (IL-6), and interleukin-8 (IL-8).
  • Other non-cytokine mediators such as leukotriene b4, prostaglandin E2, C3a and C3d also reach high levels (de Boer et al., 1992 Immunopharmacology 24, 135-148).
  • TNF- ⁇ is detected early in the course of septic shock in a large fraction of patients (de Boer et al., 1992 supra).
  • injection of TNF- ⁇ has been shown to induce shock-like symptoms similar to those induced by LPS injection (Beutler et al., 1985 Science 229, 869-871); in contrast, injection of IL-1 ⁇ , IL-6, or IL-8 does not induce shock.
  • Injection of TNF- ⁇ also causes an elevation of IL-1 ⁇ , IL-6, IL-8, PgE2, acute phase proteins, and TxA2 in the serum of experimental animals (de Boer et al., 1992 supra).
  • the lethal effects of LPS can be blocked by preadministration of anti-TNF- ⁇ antibodies. The cumulative evidence indicates that TNF- ⁇ is a key player in the pathogenesis of septic shock, and therefore a good candidate for therapeutic intervention.
  • Rheumatoid Arthritis Rheumatoid arthritis is an autoimmune disease characterized by chronic inflammation of the joints leading to bone destruction and loss of joint function. At the cellular level, autoreactive T- lymphocytes and monocytes are typically present, and the synoviocytes often have altered morphology and immunostaining patterns. RA joints have been shown to contain elevated levels of TNF- ⁇ , IL-1 ⁇ and IL-1 ⁇ , IL-6, GM-CSF, and TGF- ⁇ (Abney et al., 1991 Imm. Rev. 119, 105-123), some or all of which may contribute to the pathological course of the disease.
  • TNF- ⁇ has also been shown to increase osteoclast activity and bone resorbtion, and may have a role in the bone erosion commonly found in the RA joint (Cooper et al., 1992 Clin. Exp. Immunol. 89, 244-250). Elimination of TNF- ⁇ from the rheumatic joint would be predicted to reduce overall inflammation by reducing induction of MHC class II, IL-1 ⁇ / ⁇ , II-6, and GM-CSF, and reducing T-cell activation. Osteoclast activity might also fall, reducing the rate of bone erosion at the joint. Finally, elimination of TNF- ⁇ would be expected to reduce accumulation of scar tissue within the joint by removal of a fibroblast growth factor.
  • Psoriasis is an inflammatory disorder of the skin characterized by keratinocyte hyperproliferation and immune cell infiltrate (Kupper, 1990 J. Clin. Invest. 86, 1783-1789). It is a fairly common condition, affecting 1.5-2.0% of the population. The disorder ranges in severity from mild, with small flaky patches of skin, to severe, involving inflammation of the entire epidermis.
  • the cellular infiltrate of psoriasis includes T-lymphocytes, neutrophils, macrophages, and dermal dendrocytes. The majority of T-lymphocytes are activated CD4+ cells of the T H -1 phenotype, although some CD8+ and CD4-/CD8- are also present. B lymphocytes are typically not found in abundance in psoriatic plaques.
  • cytokines The role of cytokines in the pathogenesis of psoriasis has been investigated. Among those cytokines found to be abnormally expressed were TGF- ⁇ , IL-1 ⁇ , IL-1 ⁇ , IL-1 ra, IL-6, IL-8, IFN- ⁇ , and TNF- ⁇ . In addition to abnormal cytokine production, elevated expression of ICAM-1 , ELAM-1 , and VCAM has been observed (Reeves, 1991 supra). This cytokine profile is similar to that of normal wound healing, with the notable exception that cytokine levels subside upon healing.
  • Keratinocytes themselves have recently been shown to be capable of secreting EGF, TGF- ⁇ , IL-6, and TNF- ⁇ , which could increase proliferation in an autocrine fashion (Oxholm et al., 1991 APMIS 99, 58-64).
  • Keratinocytes secrete IL-1 ⁇ , IL-1 ⁇ , IL-6, IL-8, TNF- ⁇ . These factors activate the endothelium of dermal capillaries, recruiting PMNs, macrophages, and T-cells into the wound site.
  • Dermal dendrocytes near the dermal/epidermal junction remain activated when they should return to a quiescent state, and subsequently secrete cytokines including TNF- ⁇ , IL-6, and IL-8.
  • Cytokine expression in turn, maintains the activated state of the endothelium, allowing extravasation of additional immunocytes, and the activated state of the keratinocytes which secrete TGF- ⁇ and IL-8.
  • Keratinocyte IL-8 recruits immunocytes from the dermis into the epidermis. During passage through the dermis, T-cells encounter the activated dermal dendrocytes which efficiently activate the T H -1 phenotype.
  • the activated T-cells continue to migrate into the epidermis, where they are stimulated by keratinocyte-expressed ICAM-1 and MHC class II.
  • IFN- ⁇ secreted by the T-cells synergizes with the TNF- ⁇ from dermal dendrocytes to increase keratinocyte proliferation and the levels of TGF- ⁇ , IL-8, and IL-6 production.
  • IFN- ⁇ also feeds back to the dermal dendrocyte, maintaining the activated phenotype and the inflammatory cycle.
  • Elevated serum titres of IL-6 increases synthesis of acute phase proteins including complement factors by the liver, and antibody production by plasma cells. Increased complement and antibody levels increases the probability of autoimmune reactions.
  • TN F- ⁇ expression by the dermal dendrocyte to maintain activated endothelium and keratinocytes and IFN- ⁇ expression by T-cells to maintain activated dermal dendrocytes.
  • corticosteroids There are 3 million patients in the United States afflicted with psoriasis.
  • the available treatments for psoriasis are corticosteroids.
  • the most widely prescribed are TEMOVATE (clobetasol propionate), LIDEX (fluocinonide), DIPROLENE (betamethasone propionate), PSORCON (diflorasone diacetate) and TRIAMCINOLONE formulated for topical application.
  • the mechanism of action of corticosteroids is multifactorial. This is a palliative therapy because the underlying cause of the disease remains, and upon discontinuation of the treatment the disease returns. Discontinuation of treatment is often prompted by the appearance of adverse effects such as atrophy, telangiectasias and purpura.
  • Corticosteroids are not recommended for prolonged treatments or when treatment of large and/or inflamed areas is required.
  • Alternative treatments include retinoids, such as etretinate, which has been approved for treatment of severe, refractory psoriasis.
  • Alternative retinoid-based treatments are in advanced clinical trials.
  • Retinoids act by converting keratinocytes to a differentiated state and restoration of normal skin development.
  • Immunosuppressive drugs such as cyclosporine are also in the advanced stages of clinical trials. Due to the nonspecific mechanism of action of corticosteroids, retinoids and immunosuppressives, these treatments exhibit severe side effects and should not be used for extended periods of time unless the condition is life-threatening or disabling. There is a need for a less toxic, effective therapeutic agent in psoriatic patients.
  • the human immunodeficiency virus causes several fundamental changes in the human immune system from the time of infection until the development of full-blown acquired immunodeficiency syndrome (AIDS). These changes include a shift in the ratio of CD4+ to AIDS.
  • CD8+ T-cells sustained elevation of IL-4 levels, episodic elevation of TNF- ⁇ and TNF- ⁇ levels, hypergammaglobulinemia, and lymphoma/leukemia (Rosenberg & Fauci, 1990 Immun. Today 11 , 176; Weiss 1993 Science
  • TNF- ⁇ and IL-6 are often found to be elevated in AIDS patients (Weiss, 1993 supra).
  • tissue culture HIV infection of monocytes isolated from healthy individuals stimulates secretion of both TNF- ⁇ and IL-6. This response has been reproduced using purified gp120, the viral coat protein responsible for binding to CD-4 (Buonaguro et al., 1992 J. Virol. 66, 7159). It has also been demonstrated that the viral gene regulator, Tat, can directly induce TNF transcription.
  • the ability of HIV to directly stimulate secretion of TNF- ⁇ and IL-6 may be an adaptive mechanism of the virus.
  • TNF- ⁇ has been shown to upregulate transcription of the LTR of HIV, increasing the number of HIV-specific transcripts in infected cells.
  • IL-6 enhances HIV production, but at a post-transcriptional level, apparently increasing the efficiency with which HIV transcripts are translated into protein.
  • stimulation of TNF- ⁇ secretion by the HIV virus may promote infection of neighboring CD4+ cells both by enhancing virus production from latently infected cells and by driving replication of the virus in newly infected cells.
  • the role of TNF- ⁇ in HIV replication has been well established in tissue culture models of infection (Sher et al., 1992 Immun. Rev. 127, 183), suggesting that the mutual induction of HIV replication and TNF- ⁇ replication may create positive feedback in vivo.
  • evidence for the presence of such positive feedback in infected patients is not abundant. TNF- ⁇ levels are found to be elevated in some, but not all patients tested.
  • TNF- ⁇ can stimulate the proliferation of spindle cells isolated from Kaposi's sarcoma lesions of AIDS patients (Barillari et al., 1992 J Immunol 149, 3727).
  • a therapeutic agent that inhibits cytokine gene expression, inhibits adhesion molecule expression, and mimics the anti-inflammatory effects of glucocorticoids (without inducing steroid-responsive genes) is ideal for the treatment of inflammatory and autoimmune disorders.
  • Disease targets for such a drug are numerous. Target indications and the delivery options each entails are summarized below. In all cases, because of the potential immunosuppressive properties of a ribozyme that cleaves the specified sites in TNF- ⁇ mRNA, uses are limited to local delivery, acute indications, or ex vivo treatment.
  • ribozymes Exogenous delivery of ribozymes to macrophages can be achieved by intraperitoneal or intravenous injections. Ribozymes will be delivered by incorporation into liposomes or by complexing with cationic lipids.
  • RA Raster rheumatoid arthritis
  • adenovirus adenovirus
  • retrovirus adeno-associated virus vectors
  • adenovirus vector can be administered by direct injection into the synovium: high efficiency of gene transfer and expression for several months would be expected (B.J. Roessler, E.P. Allen, J.M. Wilson, J.W. Hartman, B. L. Davidson, J. Clin. Invest. 92, 1085-1092 (1993)). It is unlikely that the course of the disease could be reversed by the transient, local administration of an anti-inflammatory agent. Multiple administrations may be necessary. Retrovirus and adeno-associated virus vectors would lead to permanent gene transfer and expression in the joint. However, permanent expression of a potent anti-inflammatory agent may lead to local immune deficiency.
  • the psoriatic plaque is a particularly good candidate for ribozyme or vector delivery.
  • the stratum corneum of the plaque is thinned, providing access to the proliferating keratinocytes. T-cells and dermal dendrocytes can be efficiently targeted by trans-epidermal diffusion .
  • ribozymes of the present invention that cleave TNF- ⁇ mRNA and thereby TNF- ⁇ activity have many potential therapeutic uses, and there are reasonable modes of delivering the ribozymes in a number of the possible indications. Development of an effective ribozyme that inhibits TNF- ⁇ function is described above; available cellular and activity assays are number, reproducible, and accurate. Animal models for TNF- ⁇ function and for each of the suggested disease targets exist and can be used to optimize activity.
  • Chronic myelogenous leukemia exhibits a characteristic disease course, presenting initially as a chronic granulocytic hyperplasia, and invariably evolving into an acute leukemia which is caused by the clonal expansion of a cell with a less differentiated phenotype (i.e.. the blast crisis stage of the disease).
  • CML is an unstable disease which ultimately progresses to a terminal stage which resembles acute leukemia. This lethal disease affects approximately 16,000 patients a year.
  • Chemotherapeutic agents such as hydroxyurea or busulfan can reduce the leukemic burden but do not impact the life expectancy of the patient (e.g. approximately 4 years). Consequently, CML patients are candidates for bone marrow transplantation (BMT) therapy. However, for those patients which survive BMT, disease recurrence remains a major obstacle
  • the Philadelphia (Ph) chromosome which results from the translocation of the abl oncogene from chromosome 9 to the bcr gene on chromosome 22 is found in greater than 95% of CML patients and in 10-25% of all cases of acute lymphoblastic leukemia [(ALL); Fourth International Workshop on Chromosomes in Leukemia 1982, Cancer Genet. Cytogenet. 11 , 316].
  • the leukemic cells express bcr-abl fusion mRNAs in which exon 2 (b2-a2 junction) or exon 3 (b3-a2 junction) from the major breakpoint cluster region of the bcr gene is spliced to exon 2 of the abl gene.
  • exon 2 b2-a2 junction
  • exon 3 b3-a2 junction
  • the first exon of the bcr gene is spliced to exon 2 of the abl gene (Hooberman et al., 1989 Proc. Nat. Acad. Sci. USA 86, 4259; Heisterkamp et al., 1988 Nucleic Acids Res. 16, 10069).
  • the b3-a2 and b2-a2 fusion mRNAs encode 210 kd bcr-abl fusion proteins which exhibit oncogenic activity (Daley et al., 1990 Science 247, 824; Heisterkamp et al., 1990 Nature 344, 251).
  • the importance of the bcr-abl fusion protein (p210 bcr-a bl ) in the evolution and maintenance of the leukemic phenotype in human disease has been demonstrated using antisense oligonucleotide inhibition of p210 bcr-a bl expression.
  • These inhibitory molecules have been shown to inhibit the in vitro proliferation of leukemic cells in bone marrow from CML patients.
  • ribozymes as therapeutic agents for leukemias, such as chronic myelogenous leukemia (CML) by targeting the specific junction region of bcr-abl fusion transcripts. It indicates causing cleavage by a ribozyme at or near the breakpoint of such a hybrid chromosome, specifically it includes cleavage at the sequence GUX, where X is A, U or G.
  • CML chronic myelogenous leukemia
  • GUX where X is A, U or G.
  • the one example presented is to cleave the sequence 5' AGC AG AGUU (cleavage site) CAA AAGCCCU-3'.
  • the invention features use of ribozymes to inhibit the development or expression of a transformed phenotype in man and other animals by modulating expression of a gene that contributes to the expression of CML. Cleavage of targeted mRNAs expressed in pre-neoplastic and transformed cells elicits inhibition of the transformed state.
  • the invention can be used to treat cancer or pre-neoplastic conditions.
  • Two preferred administration protocols can be used, either in vivo administration to reduce the tumor burden, or ex vivo treatment to eradicate transformed cells from tissues such as bone marrow prior to reimplantation.
  • This invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of CML.
  • the mRNA targets are present in the 425 nucleotides surrounding the fusion sites of the bcr and abl sequences in the b2-a2 and b3-a2 recombinant mRNAs.
  • Other sequences in the 5' portion of the bcr mRNA or the 3' portion of the abl mRNA may also be targeted for ribozyme cleavage. Cleavage at any of these sites in the fusion mRNA molecules will result in inhibition of translation of the fusion protein in treated cells.
  • the invention provides a class of chemical cleaving agents which exhibit a high degree of specificity for the mRNA causative of CML.
  • Such enzymatic RNA molecules can be delivered exogenously or endogenously to afflicted cells.
  • the small size (less than 40 nucleotides, preferably between 32 and 36 nucleotides in length) of the molecule allows the cost of treatment to be reduced.
  • the enzymatic RNA molecules of this invention can be used to treat human CML or precancerous conditions.
  • Affected animals can be treated at the time of cancer detection or in a prophylactic manner. This timing of treatment will reduce the number of affected cells and disable cellular replication. This is possible because the ribozymes are designed to disable those structures required for successful cellular proliferation.
  • Ribozymes of this invention block to some extent p210 bcr-abl expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to tissues in animal models of CML. Ribozyme cleavage of bcr/abl mRNA in these systems may prevent or alleviate disease symptoms or conditions.
  • the sequence of human bcr/abl mRNA can be screened for accessible sites using a computer folding algorithm. Regions of the mRNA that did not form secondary folding structures and that contain potential hammerhead or hairpin ribozyme cleavage sites can be identified. These sites are shown in Table 29 (All sequences are 5' to 3' in the tables). The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme. The sequences of the chemically synthesized ribozymes most useful in this study are shown in Table 30.
  • sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity.
  • stem-loop II sequence of hammerhead ribozymes listed in Table 30 (5'-GGCCGAAAGGCC-3') can be altered (substitution, deletion, and/or insertion) to contain any sequence provided, a minimum of two base-paired stem structure can form.
  • the sequences listed in Tables 30 may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables.
  • ribozyme motifs By engineering ribozyme motifs we have designed several ribozymes directed against bcr-abl mRNA sequences. These have been synthesized with modifications that improve their nuclease resistance as described above. These ribozymes cleave bcr-abl target sequences in vitro. The ribozymes are tested for function in vivo by exogenous delivery to cells expressing bcr-abl. Ribozymes are delivered by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA vectors. Expression of bcr-abl is monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis.
  • This invention relates to the use of ribozymes as inhibitors of respiratory syncytial virus (RSV) production, and in particular, the inhibition of RSV replication.
  • RSV respiratory syncytial virus
  • RSV is a member of the virus family paramyxoviridae and is classified under the genus Pneumovirus (for a review see McIntosh and Chanock, 1990 in Virology ed. B.N. Fields, pp. 1045, Raven Press Ltd. NY).
  • the infectious virus particle is composed of a nucleocapsid enclosed within an envelope.
  • the nucleocapsid is composed of a linear negative singlestranded non-segmented RNA associated with repeating subunits of capsid proteins to form a compact structure and thereby protect the RNA from nuclease degradation.
  • the entire nucleocapsid is enclosed by the envelope.
  • the size of the virus particle ranges from 150 - 300 nm in diameter.
  • the RSV genome encodes ten viral proteins essential for viral production.
  • RSV protein products include two structural glycoproteins (G and F) found in the envelope spikes, two matrix proteins [M and M2 (22K)] found in the inner membrane, three proteins localized in the nucleocapsid (N, P and L), one protein that is present on the surface of the infected cell (SH), and two nonstructural proteins [NS1 (1C) and NS2 (1 B)] found only in the infected cell.
  • G and F structural glycoproteins found in the envelope spikes
  • M and M2 (22K) found in the inner membrane
  • N, P and L three proteins localized in the nucleocapsid
  • SH protein that is present on the surface of the infected cell
  • NS1 (1C) and NS2 (1 B) found only in the infected cell.
  • the mRNAs for the 10 RSV proteins have similar 5' and 3' ends. UV-inactivation studies suggest that a single promoter is used with multiple transcription
  • the order of transcription corresponding to the protein assignment on the genomic RNA is 1C, 1B, N, P, M, SH, G, F, 22K and L genes (Huang et al., 1985 Virus Res. 2, 157) and transcript abundance corresponds to the order of gene assignment (for example the 1C and 1B mRNAs are much more abundant than the L mRNA. Synthesis of viral message begins immediately after RSV infection of cells and reaches a maximum at 14 hours post-infection (McIntosh and Chanock, supra).
  • Subgroup A There are two antigenic subgroups of RSV, A and B, which can circulate simultaneously in the community in varying proportions in different years (McIntosh and Chanock, supra).
  • Subgroup A usually predominates. Within the two subgroups there are numerous strains. By the limited sequence analysis available it seems that homology at the nucleotide level is more complete within than between subgroups, although sequence divergence has been noted within subgroups as well.
  • Antigenic determinates result primarily from both surface glycoproteins, F and G. For F, at least half of the neutralization epitopes have been stably maintained over a period of 30 years. For G however, A and B subgroups may be related antigenically by as little as a few percent.
  • Respiratory Syncytial Virus is the major cause of lower respiratory tract illness during infancy and childhood (Hall, supra) and as such is associated with an estimated 90,000 hospitalizations and 4500 deaths in the United States alone (Update: respiratory syncytial virus activity - United States, 1993, Mmwr Morb Mortal Wkly Rep, 42, 971). Infection with RSV generally outranks all other microbial agents leading to both pneumonia and bronchitis. While primarily affecting children under two years of age, immunity is not complete and reinfection of older children and adults, especially hospital care givers (McIntosh and Chanock, supra), is not uncommon. Immunocompromised patients are severely affected and RSV infection is a major complication for patients undergoing bone marrow transplantation .
  • Uneventftl RSV respiratory disease resembles a common cold and recovery is in 7 to 12 days.
  • Initial symptoms rhinorrhea, nasal congestion, slight fever, etc.
  • lower respiratory tract signs of infection that include a cough and wheezing.
  • these mild symptoms quickly progress to tachypnea, cyanosis, and listlessness and hospitalization is required.
  • infants with underlying cardiac or respiratory disease the progression of symptoms is especially rapid and can lead to respiratory failure by the second or third day of illness. With modem intensive care however, overall mortality is usually less than 5% of hospitalized patients (McIntosh and Chanock, supra).
  • Ribavirin therapy is associated with a decrease in the severity of the symptoms, improved arterial oxygen and a decrease in the amount of viral shedding at the end of the treatment period. It is not certain, however, whether ribavirin therapy actually shortens the patients' hospital stay or diminishes the need for supportive therapies (McIntosh and Chanock, supra).
  • ribavirin therapy is especially clear for high risk infants, those with the most serious symptoms or for patients with underlying bronchopulmonary or cardiac disease.
  • Inhibition of the viral polymerase complex is supported as the main mechanism for inhibition of RSV by ribavirin, since viral but not cellular polypeptide synthesis is inhibited by ribavirin in RSV-infected cells (Antiviral Agents and Viral Diseases of Man, 3rd edition. 1990. (eds. G.J. Galasso, R.J. Whitley, and T.C. Merigan) Raven Press Ltd., NY].
  • ribavirin is at least partially effective against RSV infection when delivered by aerosolization, it can be assumed that the target cells are at or near the epithelial surface.
  • RSV antigen had not spread any deeper than the superficial layers of the respiratory epithelium in autopsy studies of fatal pneumonia (McIntosh and Chanock, supra).
  • Jennings et al., WO 94/13688 indicates that targets for specific types of ribozymes include respiratory syncytical virus.
  • the invention features novel enzymatic RNA molecules, or ribozymes, and methods for their use for inhibiting production of respiratory syncytial virus (RSV).
  • RSV respiratory syncytial virus
  • the invention also features cleavage of the genomic RNA and mRNA of these viruses by use of ribozymes.
  • the ribozyme molecules described are targeted to the NS1 (1C), NS2 (1B) and N viral genes. These genes are known in the art (for a review see McIntosh and Chanock, 1990 supra ).
  • Ribozymes that cleave the specified sites in RSV mR ⁇ As represent a novel therapeutic approach to respiratory disorders. Applicant indicates that ribozymes are able to inhibit the activity of RSV and that the catalytic activity of the ribozymes is required for their inhibitory effect.
  • the ribozymes have binding arms which are complementary to the sequences in Tables 31 , 33, 35, 37 and 38. Examples of such ribozymes are shown in Tables 32, 34, 36-38. Examples of such ribozymes consist essentially of sequences defined in these Tables. By “consists essentially of” is meant that the active ribozyme contains an enzymatic center equivalent to those in the examples, and binding arms able to bind mR ⁇ A such that cleavage at the target site occurs. Other sequences may be present which do not interfere with such cleavage. Ribozymes of this invention block to some extent RSV production and can be used to treat disease or diagnose such disease.
  • Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of respiratory disorders. Ribozyme cleavage of RSV encoded mR ⁇ As or the genomic R ⁇ A in these systems may alleviate disease symptoms. While all ten RSV encoded proteins (1C, 1B, N, P, M, SH, 22K, F, G, and L) are essential for viral life cycle and are all potential targets for ribozyme cleavage, certain proteins (mRNAs) are more favorable for ribozyme targeting than the others. For example RSV encoded proteins 1C, 1 B, SH and 22K are not found in other members of the family paramyxoviridae and appear to be unique to RSV.
  • RSV encoded proteins 1C, 1 B, SH and 22K are not found in other members of the family paramyxoviridae and appear to be unique to RSV.
  • RSV proteins 1C, 1 B and N are highly conserved among various subtypes at both the nucleotide and amino acid levels. Also, 1C, 1B and N are the most abundant of all RSV proteins.
  • the sequence of human RSV mRNAs encoding 1C, 1 B and N proteins are screened for accessible sites using a computer folding algorithm.
  • Hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 31 , 33, 34, 37 and 38 (All sequences are 5' to 3' in the tables.)
  • the nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme.
  • Ribozymes of the hammerhead or hairpin motif are designed to anneal to various sites in the mRNA message.
  • the binding arms are complementary to the target site sequences described above.
  • the ribozymes are chemically synthesized.
  • Hairpin ribozymes are synthesized in two parts and annealed to reconstruct the active ribozyme (Chowrira and Burke, 1992 Nucleic Acids Res., 20, 2835-2840). Hairpin ribozymes are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbeck, 1989, Methods Enzymol. 180, 51). All ribozymes are modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-o-methyl, 2'-H (for a review see Usman and Cedergren, 1992 TIBS 17, 34).
  • Ribozymes are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography and are resuspended in water.
  • the sequences of the chemically synthesized ribozymes useful in this study are shown in Tables 32, 34, 36, 37 and 38. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity.
  • stem-loop II sequence of hammerhead ribozymes listed in Tables 32 and 34 can be altered (substitution, deletion, and/or insertion) to contain any sequences provided a minimum of two base-paired stem structure can form.
  • stem-loop IV sequence of hairpin ribozymes listed in Tables 37 and 38 (5'-CACGUUGUG-3') can be altered (substitution, deletion, and/or insertion) to contain any sequence, provided a minimum of two base-paired stem structure can form.
  • the sequences listed in Tables 32, 34, 36, 37 and 38 may be formed of ribonucleotides or other nucleotides or non-nucleotides.
  • ribozymes are equivalent to the ribozymes described specifically in the Tables.
  • ribozyme motifs we have designed several ribozymes directed against RSV encoded mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave target sequences in vitro is evaluated.
  • RSV Ribozymes that block the induction of RSV activity and/or 1C, 1 B and N protein encoding mRNAs by more than 90% will be identified. Optimizing Ribozyme Activity
  • Ribozyme activity can be optimized as described by Draper et al., PCT WO93/23569. The details will not be repeated here, but include altering the length of the ribozyme binding arms or chemically synthesizing ribozymes with modifications that prevent their degradation by serum ribonucleases (see e.g., Eckstein et al., International Publication No. WO 92/07065; Perrault et al., 1990 Nature 344, 565; Pieken et al., 1991 Science 253, 314; Usman and Cedergren, 1992 Trends in Biochem. Sci. 17, 334; Usman et al., International Publication No.
  • Ribozymes may be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
  • the RNA/vehicle combination is locally delivered by direct injection or by use of a catheter, infusion pump or stent.
  • Alternative routes of delivery include, but are not limited to, intravenous injection, intramuscular injection, subcutaneous injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. More detailed descriptions of ribozyme delivery and administration are provided in Sullivan, et al., supra and Draper, et al., supra which have been incorporated by reference herein.
  • Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990 Proc. Natl. Acad. Sci. U S A. 87, 6743-7: Gao and Huang 1993 Nucleic Acids Res., 21 , 2867-72; Lieber et al., 1993 Methods Enzymol., 217, 47-66; Zhou et al., 1990 Mol. Cell. Biol.. 10, 4529-37).
  • ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992 Antisense Res.
  • ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral, or alpha virus vectors).
  • plasmid DNA vectors such as adenovirus or adeno-associated virus vectors
  • viral RNA vectors such as retroviral, or alpha virus vectors
  • a transcription unit expressing a ribozyme that cleaves target RNA is inserted into a plasmid
  • DNA vector a retrovirus DNA viral vector, an adenovirus DNA viral vector or an adeno-associated virus vector or alpha virus vector. These and other vectors have been used to transfer genes to live animals (for a review see
  • the vectors are delivered as recombinant viral particles.
  • DNA may be delivered alone or complexed with vehicles (as described for RNA above).
  • the DNA, DNA/vehicle complexes, or the recombinant virus particles are locally administered to the site of treatment, e.g., through the use of a catheter, stent or infusion pump.
  • Ribozymes of this invention may be used as diagnostic tools to examine genetic drift and mutations within diseased cells.
  • the close relationship between ribozyme activity and the structure of the target RNA allows the detection of mutations in any region of the molecule which alters the base-pairing and three-dimensional structure of the target RNA.
  • multiple ribozymes described in this invention one may map nucleotide changes which are important to RNA structure and function in vitro, as well as in cells and tissues. Cleavage of target RNAs with ribozymes may be used to inhibit gene expression and define the role (essentially) of specified gene products in the progression of disease. In this manner, other genetic targets may be defined as important mediators of the disease.
  • ribozymes of this invention include detection of the presence of mRNA associated with ICAM-1 , relA, TNF- ⁇ , p210, bcr-abl or RSV related condition. Such RNA is detected by determining the presence of a cleavage product after treatment with a ribozyme using standard methodology.
  • ribozymes which can cleave only wild-type or mutant forms of the target RNA are used for the assay.
  • the first ribozyme is used to identify wild-type RNA present in the sample and the second ribozyme will be used to identify mutant RNA in the sample.
  • synthetic substrates of both wild-type and mutant RNA will be cleaved by both ribozymes to demonstrate the relative ribozyme efficiencies in the reactions and the absence of cleavage of the "non-targeted" RNA species.
  • the cleavage products from the synthetic substrates will also serve to generate size markers for the analysis of wild-type and mutant RNAs in the sample population.
  • each analysis will require two ribozymes, two substrates and one unknown sample which will be combined into six reactions.
  • the presence of cleavage products will be determined using an RNAse protection assay so that full-length and cleavage fragments of each RNA can be analyzed in one lane of a polyacrylamide gel. It is not absolutely required to quantify the results to gain insight into the expression of mutant RNAs and putative risk of the desired phenotypic changes in target cells.
  • the expression of mRNA whose protein product is implicated in the development of the phenotype i.e., ICAM-1 , rel A, TNF ⁇ , p210 bcr-abl or RSV is adequate to establish risk.
  • RNA levels will be adequate and will decrease the cost of the initial diagnosis. Higher mutant form to wild-type ratios will be correlated with higher risk whether RNA levels are compared qualitatively or quantitatively.
  • RNA, enzymatic RNA or modified RNA molecules in greater than milligram quantities with high biological activity.
  • Applicant has determined that the synthesis of enzymatically active RNA in high yield and quantity is dependent upon certain critical steps used during its preparation. Specifically, it is important that the RNA phosphoramidites are coupled efficiently in terms of both yield and time, that correct exocyclic amino protecting groups be used, that the appropriate conditions for the removal of the exocyclic amino protecting groups and the alkylsilyl protecting groups on the 2'-hydroxyl are used, and that the correct work-up and purification procedure of the resulting ribozyme be used.
  • RNA molecule i.e., about 30 to 40 nucleotide bases
  • the protection of the amino functions of the bases requires either amide or substituted amide protecting groups, which must be, on the one hand, stable enough to survive the conditions of synthesis, and on the other hand, removable at the end of the synthesis.
  • amide protecting groups shown in Figure 8 in particular, benzoyl for adenosine, isobutyryl or benzoyl for cytidine, and isobutyryl for guanosine, which may be removed at the end of the synthesis by incubating the RNA in NH 3 /EtOH (ethanolic ammonia) for 20 h at 65 °C.
  • NH 3 /EtOH ethanolic ammonia
  • an incubation in ethanolic ammonia for 4 h at 65 °C is used to obtain complete removal of these protecting groups.
  • Removal of the alkylsilyl 2'-hydroxyl protecting groups can be accomplished using a tetrahydrofuran solution of TBAF at room temperature for 8-24 h.
  • RNA molecule is by either ethanol precipitation, or an anion exchange cartridge desalting, as described in Scaringe et al. Nucleic Acids Res. 1990, 18, 5433-5341.
  • the purification of the long RNA sequences may be accomplished by a two-step chromatographic procedure in which the molecule is first purified on a reverse phase column with either the trityl group at the 5' position on or off. This purification is accomplished using an acetonitrile gradient with triethylammonium or bicarbonate salts as the aqueous phase.
  • the trityl on purification the trityl group may be removed by the addition of an acid and drying of the partially purified RNA molecule.
  • the final purification is carried out on an anion exchange column, using alkali metal perchlorate salt gradients to elute the fully purified RNA molecule as the appropriate metal salts, e.g. Na + , Li + etc.
  • a final de-salting step on a small reverse-phase cartridge completes the purification procedure. Applicant has found that such a procedure not only fails to adversely affect activity of a ribozyme, but may improve its activity to cleave target RNA molecules.
  • Alkyl refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups.
  • the alkyl group has 1 to 12 carbons. More preferably it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkenyl group has 1 to 12 carbons. More preferably it is a lower alkenyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • alkyl also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkynyl group has 1 to 12 carbons. More preferably it is a lower alkynyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • alkyl groups may also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups.
  • An "aryl” group refers to an aromatic group which has at least one ring having a conjugated ⁇ electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted.
  • the preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups.
  • alkylaryl refers to an alkyl group (as described above) covalently joined to an aryl group (as described above.
  • Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted.
  • Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms.
  • Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted.
  • An "amide” refers to an -C(O)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • An “ester” refers to an -C(O)-OR', where R is either alkyl, aryl, alkylaryl or hydrogen. 2.
  • alkylamine where alkyl is preferably methyl, ethyl, propyl or butyl
  • AMA NH 4 OH/alkylamine
  • Other alkylamines e.g. ethylamine, propylamine, butylamine etc. may also be used.
  • anion-exchange resins to purify and/or analyze the fully deprotected RNA.
  • These resins include, but are not limited to, quartenary or tertiary amino derivatized stationary phases such as silica or polystyrene. Specific examples include Dionex-NA100 ® , Mono-Q ® , Poros-Q ® .
  • the invention features an improved method for the coupling of RNA phosphoramidites; for the removal of amide or substituted amide protecting groups; and for the removal of 2'-hydroxyl alkylsilyl protecting groups.
  • Such methods enhance the production of RNA or analogs of the type described above (e.g., with substituted 2'-groups), and allow efficient synthesis of large amounts of such RNA.
  • RNA may also have enzymatic activity and be purified without loss of that activity. While specific examples are given herein, those in the art will recognize that equivalent chemical reactions can be performed with the alternative chemicals noted above, which can be optimized and selected by routine experimentation.
  • the invention features an improved method for the purification or analysis of RNA or enzymatic RNA molecules (e.g. 28-70 nucleotides in length) by passing said RNA or enzymatic RNA molecule over an HPLC, e.g., reverse phase and/or an anion exchange chromatography column.
  • HPLC e.g., reverse phase and/or an anion exchange chromatography column.
  • the method of purification improves the catalytic activity of enzymatic RNAs over the gel purification method (see Figure 10).
  • the purification of long RNA molecules may be accomplished using anion exchange chromatography, particularly in conjunction with alkali perchlorate salts.
  • This system may be used to purify very long RNA molecules.
  • a Dionex NucleoPak 100 ⁇ or a Pharmacia Mono Q ® anion exchange column for the purification of RNA by the anion exchange method.
  • This anion exchange purification may be used following a reverse-phase purification or prior to reverse phase purification. This method results in the formation of a sodium salt of the ribozyme during the chromatography. Replacement of the sodium alkali earth salt by other metal salts, e.g., lithium, magnesium or calcium perchlorate, yields the corresponding salt of the RNA molecule during the purification.
  • the reverse phase purification is best accomplished using polymeric, e.g. polystyrene based, reverse-phase media, using either a 5'-trityl-on or 5'-trityl-off method. Either molecule may be recovered using this reverse-phase method, and then, once detritylated, the two fractions may be pooled and then submitted to an anion exchange purification step as described above.
  • polymeric e.g. polystyrene based, reverse-phase media
  • Either molecule may be recovered using this reverse-phase method, and then, once detritylated, the two fractions may be pooled and then submitted to an anion exchange purification step as described above.
  • the method includes passing the enzymatically active RNA molecule over a reverse phase HPLC column; the enzymatically active RNA molecule is produced in a synthetic chemical method and not by an enzymatic process; and the enzymatic RNA molecule is partially blocked, and the partially blocked enzymatically active RNA molecule is passed over a reverse phase HPLC column to separate it from other RNA molecules.
  • the enzymatically active RNA molecule after passage over the reverse phase HPLC column, is deprotected and passed over a second reverse phase HPLC column (which may be the same as the reverse phase HPLC column), to remove the enzymatic RNA molecule from other components.
  • the column is a silica or organic polymer-based C4, C8 or C18 column having a porosity of at least 125 A, preferably 300 A, and a particle size of at least 2 ⁇ m, preferably 5 ⁇ m.
  • RNA molecules may be accomplished chemically or enzymatically.
  • chemical synthesis the use of tetrazole as an activator of RNA phosphoramidites is known (Usman et al. J. Am. Chem. Soc. 1987, 109, 7845-7854).
  • a 0.5 M solution of tetrazole is allowed to react with the RNA phosphoramidite and couple with the polymer bound 5'-hydroxyl group for 10 m.
  • Applicant has determined that using 0.25-0.5 M solutions of 5-S-alkyltetrazoies for only 5 min gives equivalent or better results. The following exemplifies the procedure.
  • the method of synthesis used follows the general procedure for RNA synthesis as described in Usman et al., 1987 supra and in Scaringe et al., Nucleic Acids Res. 1990, 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
  • the major difference used was the activating agent, 5-S-ethyl or -methyltetrazole @ 0.25 M concentration for 5 min.
  • oligonucleotide synthesis reagents for the 394 Detritylation solution was 2% TCA in methylene chloride; capping was performed with 16% N-Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I 2 , 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. S-Ethyl tetrazole solution (0.25 M in acetonitrile) was made up from the solid obtained from Applied Biosystems.
  • Oligonucleotide synthesis reagents for the 390Z Detritylation solution was 2% DCA in methylene chloride; capping was performed with 16% N-Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I 2 , 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. S-Ethyl tetrazole solution (0.25-0.5 M in acetonitrile) was made up from the solid obtained from Applied Biosystems.
  • the first step of the deprotection of R ⁇ A molecules may be accomplished by removal of the exocyclic amino protecting groups with either ⁇ H 4 OH/EtOH:3/1 (Usman et al. J. Am. Chem. Soc. 1987, 109, 7845-7854) or NH 3 /EtOH (Scaringe et al. Nucleic Acids Res. 1990, 18, 5433-5341) for ⁇ 20 h @ 55-65 °C. Applicant has determined that the use of methylamine or NH4OH/methylamine for 10-15 min @ 55-65 °C gives equivalent or better results. The following exemplifies the procedure.
  • the polymer-bound oligonucleotide either trityl-on or off, was suspended in a solution of methylamine (MA) or NH 4 OH/methylamine (AMA) @ 55-65 °C for 5-15 min to remove the exocyclic amino protecting groups.
  • MA methylamine
  • AMA NH 4 OH/methylamine
  • the polymer-bound oligoribonucleotide was transferred from the synthesis column to a 4 mL glass screw top vial. NH4OH and aqueous methylamine were pre-mixed in equal volumes. 4 mL of the resulting reagent was added to the vial, equilibrated for 5 m at RT and then heated at 55 or 65 °C for 5-15 min.
  • the supernatant was removed from the polymer support.
  • the support was washed with 1.0 mL of EtOH:MeCN:H 2 O/3:1 :1 , vortexed and the supernatant was then added to the first supernatant.
  • the combined supernatants, containing the oligoribonucleotide, were dried to a white powder. The same procedure was followed for the aqueous methylamine reagent.
  • Table 40 is a summary of the results obtained using the improvements outlined in this application for base deprotection.
  • the second step of the deprotection of RNA molecules may be accomplished by removal of the 2'-hydroxyl alkylsilyl protecting group using TBAF for 8-24 h (Usman et al. J. Am. Chem. Soc. 1987, 109, 7845-7854). Applicant has determined that the use of anhydrous TEA•HF in N-methylpyrrolidine ( ⁇ MP) for 0.5-1.5 h @ 55-65 °C gives equivalent or better results. The following exemplifies this procedure.
  • the ammonia-deprotected oligoribonucleotide was resuspended in 250 ⁇ L of 1.4 M anhydrous HF solution (1.5 mL N-methylpyrrolidine, 750 ⁇ L TEA and 1.0 mL TEA•3HF) and heated to 65 °C for 1.5 h. 9 mL of 50 mM TEAB was added to quench the reaction. The resulting solution was loaded onto a Qiagen 500 ® anion exchange cartridge (Qiagen Inc.) prewashed with 10 mL of 50 mM TEAB. After washing the cartridge with 10 mL of 50 mM TEAB, the R ⁇ A was eluted with 10 mL of 2 M TEAB and dried down to a white powder.
  • anhydrous HF solution 1.5 mL N-methylpyrrolidine, 750 ⁇ L TEA and 1.0 mL TEA•3HF
  • 9 mL of 50 mM TEAB was added to
  • Table 41 is a summary of the results obtained using the improvements outlined in this application for alkylsilyl deprotection.
  • the crude material was diluted to 5 mL with diethylpyrocarbonate treated water.
  • the sample was injected onto either a Pharmacia Mono Q ® 16/10 or Dionex ⁇ ucleoPac ® column with 100% buffer A (10 mM ⁇ aClO 4 ).
  • a gradient from 180-210 mM NaClO 4 at a rate of 0.85 mM/void volume for a Pharmacia Mono Q® anion-exchange column or 100-150 mM NaClO 4 at a rate of 1.7 mM/void volume for a Pionex NucleoPac ® anion-exchange column was used to elute the RNA.
  • Fractions were analyzed by a HP-1090 HPLC with a Dionex NucleoPac ® column. Fractions containing full length product at >80% by peak area were pooled.
  • the crude material was desalted by applying the solution that resulted from quenching of the desilylation reaction to a 53 mL Pharmacia HiLoad 26/10 Q-Sepharose® Fast Flow column. The column was thoroughly washed with 10 mM sodium perchlorate buffer. The oligonucleotide was eluted from the column with 300 mM sodium perchlorate.
  • the eluent was quantitated and an analytical HPLC was run to determine the percent full length material in the synthesis.
  • the eluent was diluted four fold in sterile H 2 O to lower the salt concentration and applied to a Pharmacia Mono Q ® 16/10 column.
  • a gradient from 10-185 mM sodium perchlorate was run over 4 column volumes to elute shorter sequences, the full length product was then eluted in a gradient from 185-214 mM sodium perchlorate in 30 column volumes.
  • the fractions of interest were analyzed on a HP-1090 HPLC with a Pionex NucleoPac ® column. Fractions containing over 85% full length material were pooled. The pool was applied to a Pharmacia RPC ® column for desalting.
  • the crude material was desalted by applying the solution that resulted from quenching of the desilylation reaction to a 53 mL Pharmacia HiLoad 26/10 Q-Sepharose ® Fast Flow column.
  • the column was thoroughly washed with 20 mM NH 4 CO 3 H/10% CH 3 CN buffer.
  • the oligonucleotide was eluted from the column with 1.5 M NH 4 CO 3 H/10% acetonitrile.
  • the eluent was quantitated and an analytical HPLC was run to determine the percent full length material present in the synthesis.
  • the oligonucleotide was then applied to a Pharmacia Resource RPC column.
  • RNA substrates 15-25-mers
  • purified 5'-end labeled ribozymes ⁇ 36-mers
  • Ribozyme stock solutions were 1 ⁇ M, 200 nM, 40 nM or 8 nM and the final substrate RNA concentrations were - 1 nM.
  • Total reaction volumes were 50 ⁇ L.
  • aqueous methyl amine is an efficient reagent to deprotect bases in an RNA molecule.
  • the RNA sample needs to be dried completely prior to the deprotection of the sugar 2'-hydroxyl groups.
  • deprotection of RNA synthesized on a large scale e.g., 100 ⁇ mol
  • applicant describes a one pot deprotection protocol (Fig. 12). According to this protocol, anhydrous methylamine is used in place of aqueous methyl amine.
  • Base deprotection is carried out at 65 °C for 15 min and the reaction is allowed to cool for 10 min. Deprotection of 2'-hydroxyl groups is then carried out in the same container for 90 min in a TEA•3HF reagent. The reaction is quenched with 16 mM TEAB solution.
  • hammerhead ribozyme targeted to site B is synthesized using RNA phosphoramadite chemistry and deprotected using either a two pot or a one pot protocol. Profiles of these ribozymes on an HPLC column are compared. The figure shows that RNAs deprotected by either the one pot or the two pot protocols yield similar full-length product profiles. Applicant has shown that using a one pot deprotection protocol, time required for RNA deprotection can be reduced considerably without compromising the quality or the yield of full length RNA.
  • ribozymes targeted to site B are tested for their ability to cleave RNA.
  • ribozymes that are deprotected using one pot protocol have catalytic activity comparable to ribozymes that are deprotected using a two pot protocol.
  • TETD tetraethylthiuram disulfide
  • Beaucage reagent Vu and Hirschbein, 1991 supra
  • TETD requires long sulfurization times (600 seconds for DNA and 3600 seconds for RNA). It has recently been shown that for sulfurization of DNA oligonucleotides, Beaucage reagent is more efficient than TETD (Wyrzykiewicz and Ravikumar, 1994 Bioorganic Med. Chem. 4, 1519).
  • Beaucage reagent has also been used to synthesize phosphorothioate oligonucleotides containing 2'-deoxy-2'-fluoro modifications wherein the wait time is 10 min (Kawasaki et al., 1992 J. Med. Chem).
  • the method of synthesis used follows the procedure for RNA synthesis as described herein and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
  • the sulfurization step for RNA described in the literature is a 8 second delivery and 10 min wait steps (Beaucage and Iyer, 1991 Tetrahedron 49, 6123).
  • a major improvement is the use of an activating agent, 5-S-ethyltetrazole or 5-S-methyltetrazole at a concentration of 0.25 M for 5 min. Additionally, for those linkages which are phosporothioate, the iodine solution is replaced with a 0.05 M solution of 3H-1 ,2-benzodithiole-3-one 1 ,1-dioxide (Beaucage reagent) in acetonitrile. The delivery time for the sulfurization step is reduced to 5 seconds and the wait time is reduced to 300 seconds.
  • RNA synthesis is conducted on a 394 (ABI) synthesizer using a modified 2.5 ⁇ mol scale protocol with a reduced 5 min coupling step for alkylsilyl protected RNA and 2.5 min coupling step for 2'-O-methylated
  • RNA RNA.
  • oligonucleotide synthesis reagents for the 394 synthesizer detritylation solution was 2% TCA in methylene chloride; capping was performed with 16% N-Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I 2 , 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. S-Ethyl tetrazole solution (0.25 M in acetonitrile) was made up from the solid obtained from Applied Biosystems. Sulfurizing reagent was obtained from Glen Research.
  • a 36 mer hammerhead ribozyme is synthesized which is targeted to site C.
  • the ribozyme is synthesized to contain phosphorothioate linkages at four positions towards the 5' end.
  • R ⁇ A cleavage activity of this ribozyme is shown in Fig. 16.
  • Activity of the phosphorothioate ribozyme is comparable to the activity of a ribozyme lacking any phosphorothioate linkages.
  • Example 13 Protocol for the synthesis of 2'- ⁇ -phtalimido-nucleoside phosphoramidite
  • the 2'-amino group of a 2'-deoxy-2'-amino nucleoside is normally protected with N-(9-flourenylmethoxycarbonyl) (Fmoc; Imazawa and Eckstein, 1979 supra; Pieken et al., 1991 Science 253, 314).
  • This protecting group is not stable in CH3CN solution or even in dry form during prolonged storage at -20 °C.
  • phosphoramidite 17 was synthesized starting from 2'-deoxy-2'-aminonucleoside (12) using transient protection with Markevich reagent (Markiewicz J. Chem. Res. 1979, S, 24). An intermediate 13 was obtained in 50% yield, however subsequent introduction of N-phtaloyl (Pht) group by Nefken's method (Nefkens, 1960 Nature 185, 306), desilylation (15), dimethoxytrytilation (16) and phosphitylation led to phosphoramidite 17. Since overall yield of this multi-step procedure was low (20%) applicant investigated some alternative approaches, concentrating on selective introduction of N-phtaloyl group without acylation of 5' and 3' hydroxyls.
  • N-Pht-derivative 15 When 2'-deoxy-2'-amino-nucleoside was reacted with 1.05 equivalents of Nefkens reagent in DMF overnight with subsequent treatment with Et 3 N (1 hour) only 10-15% of N and 5'(3')-bis-phtaloyl derivatives were formed with the major component being N-Pht-derivative 15.
  • the N,O-bis by-products could be selectively and quantitively converted to N-Pht derivative 15 by treatment of crude reaction mixture with cat. KCN/MeOH.
  • a convenient "one-pot" procedure for the synthesis of key intermediate 16 involves selective N-phthaloylation with subsequent dimethoxytrytilation by DMTCI/Et3N and resulting in the preparation of DMT derivative 16 in 85% overall yield as follows. Standard phosphytilation of 16 produced phosphoramidite 17 in 87% yield.
  • 2'-amino nucleoside for example 2'-amino uridine (US Biochemicals® part # 77140) was co-evaporated twice from dry dimethyl formamide (Dmf) and dried in vacuo overnight.
  • the (trimethylsilyl)ethoxymethyl ether (SEM) seems a suitable substitute. This protecting group is stable to base and all but the harshest acidic conditions. Therefore it is stable under the conditions required for oligonucleotide synthesis. It can be readily introduced and the oxygen carbon bond makes it unable to migrate. Finally, the SEM group can be removed with BF 3 •OEt 2 very quickly.
  • the method can involve use of standard RNA synthesis conditions as discussed below, or any other equivalent steps. Those in the art are familiar with such steps.
  • the nucleotide used can be any normal nucleotide or may be substituted in various positions by methods well known in the art, e.g., as described by
  • This invention also features a method for covalently linking a SEM group to the 2'-position of a nucleotide.
  • the method involves contacting a nucleoside with an SEM-containing molecule under SEM bonding conditions.
  • the conditions are dibutyltin oxide, tetrabutylammonium fluoride and SEM-Cl. Those in the art, however, will recognize that other equivalent conditions can also be used.
  • the invention features a method for removal of an SEM group from a nucleoside molecule or an oligonucleotide.
  • the method involves contacting the molecule or oligonucleotide with boron trifluoride etherate (BF 3 •OEt 2 ) under SEM removing conditions, e.g., in acetonitrile.
  • boron trifluoride etherate BF 3 •OEt 2
  • RNA synthesis there is shown the method for solid phase synthesis of RNA.
  • a 2',5'-protected nucleotide is contacted with a solid phase bound nucleotide under RNA synthesis conditions to form a dinucleotide.
  • the protecting group (R) at the 2'-position in prior art methods can be a silyl ether, as shown in the Figure.
  • an SEM group is used in place of the silyl ether. Otherwise RNA synthesis can be performed by standard methodology.
  • FIG. 19 there is shown the synthesis of 2'-O-SEM protected nucleosides and phosphoramadites.
  • a 5'-protected nucleoside (1 ) is protected at the 2'- or 3'-position by contacting with a derivative of SEM under appropriate conditions. Specifically, those conditions include contacting the nucleoside with dibutyltin oxide and SEM chloride.
  • the 2 regioisomers are separated by chromatography and the 2'-protected moiety is converted into a phosphoramidite by standard procedure.
  • the 3'-protected nucleoside is converted into a succinate derivative suitable for derivatization of a solid support.
  • RNA containing an SEM group is performed.
  • step 1 the base protecting groups and cyanoethyl groups are removed by standard procedure.
  • the SEM group is then removed as shown in the Figure.
  • Nucleoside 2 was detritylated following standard methods, as shown in Figure 19.
  • Example 16 Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl)-5',3'-O-Acetyl Uridine (5)
  • Nucleoside 3 was succinylated and coupled to the support following standard procedures, as shown in Figure 19.
  • Example 19 Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl)-5'-O- Dimethoxytrityl Uridine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (8)
  • RNA synthesis follows the general procedure for RNA synthesis as described in Usman, N.; Ogilvie.K.K.; Jiang, M.-Y.; Cedergren.R.J. J. Am. Chem. Soc. 1987, 109, 7845-7854 and in Scaringe, S.A.; Franklyn.C; Usman, N. Nucl. Acids Res. 1990, 18, 5433-5441.
  • the phosphoramidite 8 was coupled following standard RNA methods to provide a 10-mer of uridylic acid. Syntheses were conducted on a 394 (ABI) synthesizer using a modified 2.5 ⁇ mol scale protocol with a 10 m coupling step.
  • oligonucleotide synthesis reagents for the 394 Detritylation solution was 2% TCA in methylene chloride; capping was performed with 16% N- Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I 2 , 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. Referring to Figure 21. the homopolymer was base deprotected with
  • the invention features a method for preparing multi-copy cassettes encoding a defined ribozyme structure for production of a ribozyme at a decreased cost.
  • a vector is produced which encodes a plurality of ribozymes which are cleaved at their 3' and 5' ends from an RNA transcript producted from the vector by only one other ribozyme.
  • the system is useful for scaling up production of a ribozyme, which may be either modified or unmodified, in situ or in vitro.
  • Such vector systems can be used to express a desired ribozyme in a specific cell, or can be used in an in vitro system to allow productiuon of large amounts of a desired riboqyne,
  • the vectors of this invention allow a higher yield synthesis of a ribozyme in the form of an RNA transcript which is cleaved in situ or in vitro before or after transcript isolation.
  • this invention is distinct from the prior art in that a single ribozyme is used to process the 3' and 5' ends of each therapeutic, transacting or desired ribozyme instead of processing only one end, or only one ribozyme.
  • This allows smaller vectors to be derived with multiple transacting ribozymes released by only one other ribozyme from the mRNA transcript.
  • Applicant has also provided methods by which the activity of such ribozymes is increased compared to those in the art, by designing ribozyme-encoding vectors and the corresponding transcript such that folding of the mRNA does not interfere with processing by the releasing ribozyme.
  • the stability of the ribozyme produced in this method can be enhanced by provision of sequences at the termini of the ribozymes as described by Draper et al., PCT WO 93/23509, hereby incorporated by reference herein.
  • the method of this invention is advantageous since it provides high yield synthesis of ribozymes by use of low cost transcription-based protocols, compared to existing chemical ribozyme synthesis, and can use isolation techniques currently used to purify chemically synthesized oligonucleotides.
  • the method allows synthesis of ribozymes in high yield at low cost for analytical, diagnostic, or therapeutic applications.
  • the method is also useful for synthesis of ribozymes in vitro for ribozyme structural studies, enzymatic studies, target RNA accessibility studies, transcription inhibition studies and nuclease protection studies, much is described by Draper et al., PCT WO 93/23509 hereby incorporated by reference herein.
  • the method can also be used to produce ribozymes in situ either to increase the intracellular concentration of a desired therapeutic ribozyme, or to produce a concatameric transcript for subsequent in vitro isolation of unit length ribozyme.
  • the desired ribozyme can be used to inhibit gene expression in molecular genetic analyses or in infectious cell systems, and to test the efficacy of a therapeutic molecule or treat afflicted cells.
  • the invention features a vector which includes a bacterial, viral or eucaryotic promoter within a plasmid, cosmid, phagmid, virus, viroid, virusoid or phage vector.
  • Other vectors are equally suitable and include double-stranded, or partially double-stranded DNA, formed by an amplification method such as the polymerase chain reaction, or doublestranded, partially double-stranded or single-stranded RNA, formed by sitedirected homologous recombination into viral or viroid RNA genomes.
  • Such vectors need not be circular.
  • a first ribozyme-encoding region Transcriptionally linked to the promoter region is a first ribozyme-encoding region, and nucleotide sequences encoding a ribozyme cleavage sequence which is placed on either side of a region encoding a therapeutic or otherwise desired second ribozyme.
  • Suitable restriction endonuclease sites can be provided to ease construction of this vector in DNA vectors or in requisite DNA vectors of an RNA expression system.
  • the desired second ribozyme may be any desired type of ribozyme, such as a hammerhead, hairpin , hepatitis delta virus (HDV) or other catalytic center, and can include group I and group II introns, as discussed above.
  • HDV hepatitis delta virus
  • the first ribozyme is chosen to cleave the encoded cleavage sequence, and may also be any desired ribozyme, for example, a Tetrahymena derived ribozyme, which may, for example, include an imbedded restriction endonuclease site in the center of a self-recognition sequence to aid in vector construction. This endonuclease site is useful for construction of the vector, and subsequent analysis of the vector.
  • RNA transcript which includes the first and second ribozyme sequences.
  • the first ribozyme sequence is able to act, under appropriate conditions, to cause cleavage at the cleavage sites to release the second ribozyme sequences.
  • These second ribozyme sequences can then act at their target RNA sites, or can be isolated for later use or analysis.
  • the invention features a vector which includes a first nucleic acid sequence (encoding a first ribozyme having intramolecular cleaving activity), and a second nucleic acid sequence (encoding a second ribozyme having intermolecular cleaving enzymatic activity) flanked by nucleic acid sequences encoding RNA which is cleaved by the first ribozyme to release the second ribozyme from the RNA transcript encoded by the vector.
  • the second ribozyme may be flanked by the first ribozyme either on the 5' side or 3' side. If desired, the first ribozyme may be encoded on a separate vector and may have intermolecular cleaving activity.
  • the first ribozyme can be chosen to be any self-cleaving ribozyme, and the second ribozyme may be chosen to be any desired ribozyme.
  • the flanking sequences are chosen to include sequences recognized by the first ribozyme.
  • the vector is caused to express RNA from these nucleic acid sequences, that RNA has the ability under appropriate conditions to cleave each of the flanking regions and thereby release one or more copies of the second ribozyme.
  • several different second ribozymes can be produced by the same vector, or several different vectors can be placed in the same vessel or cell to produce different ribozymes.
  • the vector includes a plurality of the nucleic acid sequences encoding the second ribozyme, each flanked by nucleic acid sequences recognized by the first ribozyme. Most preferably, such a plurality includes at least six to nine or even between 60 - 100 nucleic acid sequences.
  • the vector includes a promoter which regulates expression of the nucleic acid encoding the ribozymes from the vector; and the vector is chosen from a plasmid, cosmid, phagmid, virus, viroid or phage.
  • the plurality of nucleic acid sequences are identical and are arranged in sequential order such that each has an identical end nearest to the promoter.
  • a poly(A) sequence adjacent to the sequence encoding the first or second ribozyme may be provided to increase stability of the RNA produced by the vector; and a restriction endonuclease site adjacent to the nucleic acid encoding the first ribozyme is provided to allow insertion of nucleic acid encoding the second ribozyme during construction of the vector.
  • the invention features a method for formation of a ribozyme expression vector by providing a vector including nucleic acid encoding a first ribozyme, as discussed above, and providing a single-stranded PNA encoding a second ribozyme, as discussed above.
  • the single-stranded PNA is then allowed to anneal to form a partial duplex PNA which can be filled in by a treatment with an appropriate enzyme, such as a PNA polymerase in the presence of dNTPs, to form a duplex DNA which can then be ligated to the vector.
  • the invention features a method for production of ribozymes by providing a vector as described above, expressing RNA from that vector, and allowing cleavage by the first ribozyme to release the second ribozyme.
  • three different ribozyme motifs are used as cis-cleaving ribozymes.
  • the hammerhead, hairpin, and hepatitis delta virus (HDV) ribozyme motifs consist of small, well-defined sequences that rapidly self-cleave in vitro (Symons, 1992 Annu. Rev. Biochem. 61 , 641). While structural and functional differences exist among the three ribozyme motifs, they self-process efficiently in vivo. All three ribozyme motifs self-process to 87-95% completion in the absence of 3' flanking sequences.
  • HDV hepatitis delta virus
  • the self-processing constructs described in this invention are significantly more active than those reported by Taira et al., 1990 supra: and Altffler et al., 1992 Gene 122, 85.
  • the present invention enables the use of cis-cleaving ribozymes to efficiently truncate RNA molecules at specific sites in vivo by ensuring lack of secondary structure which prevents processing.
  • the preferred method of isolating therapeutic ribozyme is by a chromatographic technique.
  • HPLC purification methods and reverse HPLC purification methods described by Draper et al., PCT WO 93/23509, hereby incorporated by reference herein, can be used.
  • the attachment of complementary oligonucleotides to cellulose or other chromatography columns allows isolation of the therapeutic second ribozyme, for example, by hybridization to the region between the flanking arms and the enzymatic RNA. This hybridization will select against the short flanking sequences without the desired enzymatic RNA, and against the releasing first ribozyme.
  • the hybridization can be accomplished in the presence of a chaotropic agent to prevent nuclease degradation.
  • the oligonucleotides on the matrix can be modified to minimize nuclease activity, for example, by provision of 2'-0-methyl RNA oligonucleotides.
  • modifications of the oligonucleotide attached to the column matrix will allow the multiple use of the column with minimal oligo degradation.
  • Many such modifications are known in the art, but a chemically stable non-reducible modification is preferred.
  • phosphorothioate modifications can also be used.
  • the expressed ribozyme RNA can be isolated from bacterial or eucaryotic cells by routine procedures such as lysis followed by guanidine isothiocyanate isolation.
  • the current known self-cleaving site of Tetrahymena can be used in an alternative vector of this invention. If desired, the full-length Tetrahymena sequence may be used, or a shorter sequence may be used. It is preferred that, in order to decrease the superfluous sequences in the self-cleaving site at the 5' cleavage end, the hairpin normally present in the Tetrahymena ribozyme should contain the therapeutic second ribozyme 3' sequence and its complement. That is, the first releasing ribozyme-encoding DNA is provided in two portions, separated by DNA encoding the desired second ribozyme.
  • CGAGGA is provided in the self-cleaving site loop such that it is in a stem structure recognized by the Tetrahymena ribozyme.
  • the loop of the stem may include a restriction endonuclease site into which the desired second ribozyme-encoding DNA is placed.
  • the vector may be used in a therapeutic protocol by use of the systems described by Lechner, PCT WO 92/13070, hereby incorporated by reference herein, to allow a timed expression of the therapeutic second ribozyme, as well as an appropriate shut off of cell or gene function.
  • the vector will include a promoter which appropriately expresses enzymatically active RNA only in the presence of an RNA or another molecule which indicates the presence of an undesired organism or state. Such enzymatically active RNA will then kill or harm the cell in which it exists, as described by Lechner, id ., or act to cause reduced expression of a desired protein product.
  • RNA vectors may also be used in this invention.
  • the vectors include plant viroids, plant viruses which contain single or double-stranded RNA genomes and animal viruses which contain RNA genomes, such as the picornaviruses, myxoviruses, paramyxoviruses, hepatitis A virus, reovirus and retroviruses. In many instances cited, use of these viral vectors also results in tissue specific delivery of the ribozymes.
  • Example 21 Design of self-processing cassettes
  • applicant compared the in vitro and in vivo cis-cleaving activity of three different ribozyme motifs-the hammerhead, the hairpin and the hepatitis delta virus ribozyme-in order to assess their potential to process the ends of transcripts in vivo.
  • all the ribozyme cassettes contained the same trans-acting hammerhead ribozyme followed immediately by one of the three cis-acting ribozymes ( Figure 23-25).
  • HH refers to the cis-cleaving cassette containing a hammerhead ribozyme
  • HP and HDV refer to the cassettes containing hairpin and hepatitis delta virus cis-cleaving ribozymes, respectively.
  • the general design of the ribozyme cassettes, as well as specific differences among the cassettes, are outlined below.
  • a sequence predicted to form a stable stem-loop structure is included at the 5' end of all the transcripts.
  • the hairpin stem contains the T7 RNA polymerase initiation sequence (Milligan & Uhlenbeck, 1989 Methods Enzymol.
  • the trans-acting ribozyme used in this study is targeted to a site B (5'...CUGGAGUC ⁇ GACCUUC...3').
  • all transcripts also contain a single nucleotide between the 5' stem-loop and the first nucleotide of the ribozyme.
  • the linker nucleotide was required to obtain the same activity in vitro that was measured with an identical ribozyme lacking the 5' hairpin.
  • the three cis-cleaving ribozymes have different requirements at the site of cleavage, slight differences were unavoidable at the 3' end of the processed transcript.
  • the junction between the trans- and cis-acting ribozyme is, however, designed so that there is minimal extraneous sequence left at the 3' end of the trans-cleaving ribozyme once cis-cleavage occurs.
  • the only differences between the constructs lie in the 3' binding arm of the ribozyme, where either 6 or 7 nucleotides, 5'-ACUCCA(+/-G)-3', complementary to the target sequence are present and where, after processing, two to five extra nucleotides remain.
  • the cis-cleaving hammerhead ribozyme used in the HH cassette is based on the design of Grosshans and Cech, 1991 supra. As shown in Figure 23, the 3' binding arm of the trans-acting ribozyme is included in the required base-pairing interactions of the cis-cleaving ribozyme to form stem I. Two extra nucleotides, UC, were included at the end of the 3' binding arm to form the self-processing hammerhead ribozyme site (Ruffner et al., 1990 supra) which remain on the 3' end of the trans-acting ribozyme following self-processing.
  • the hairpin ribozyme portion of the HP self-processing construct is based on the minimal wild-type sequence (Hampel & Tritz, 1989 supra).
  • a tetra-loop at the end of helix 1 (3' side of the cleavage site) serves to link the two portions and thus allows a minimal five nucleotides to remain at the end of the released trans-acting ribozyme following self-processing.
  • Two variants of HP were designed: HP(GU) and HP(GC).
  • the HP(GU) was constructed with a G ⁇ U wobble base pair in helix 2 (A 52 G substitution; Figure 24).
  • the HDV ribozyme self-processes efficiently when the nucleotide 5' to the cleavage site is a pyrimidine, and somewhat less so when adenosine is in that position. No other sequence requirements have been identified upstream of the cleavage site, however, we have observed some decrease in activity when a stem-loop structure was present within 2 nt of the cleavage site.
  • the HDV self-processing construct (Fig 25) was designed to generate the trans-acting hammerhead ribozyme with only two additional nucleotides at its 3' end after self-processing.
  • the HDV sequence used here is based on the anti-genomic sequence (Perrota & Been, 1992 supra) but includes the modifications of Been et al., 1992 (Biochemistry 31 , 11843) in which cis-cleavage activity of the ribozyme was improved by the substitution of a shortened helix 4 for a wild-type stem-loop ( Figure 25).
  • oligonucleotides 60-90 nucleotides
  • the singlestrand portions of annealed oligonucleotides were converted to doublestrands using Sequenase® (U.S. Biochemicals). Insert DNA was ligated into EcoR1/HindIII-digested puc18 and transformed into E.
  • each construct was transcribed and allowed to undergo self-processing under identical conditions at 37°C
  • equal amounts of linearized DNA templates bearing the various ribozyme cassettes were transcribed in the presence of [ ⁇ - 32 P]GTP to generate 5' end-labeled transcripts.
  • Mg 2+ was included at 10 mM above the nucleotide concentration so that cleavage by all the ribozyme cassettes would be supported.
  • Transcription templates were linearized at several positions by digestion with different restriction enzymes so that self-processing in the presence of increasing lengths of downstream sequence could be compared (see Fig. 26).
  • the resulting transcripts have either 4-5 non-ribozyme nucleotides at the 3' end (HindIII-digested template), 220 nucleotides (NdeI digested templates) or 454 nucleotides of downstream sequence (Real digested template).
  • HP(GC) is more efficient than the HP(GU) ribozyme, both in the presence and in the absence of extra downstream sequence.
  • the activity of HP(GU) falls off more dramatically when downstream sequence is present.
  • the stronger G:C base pair likely contributes to the HP(GC) construct's ability to fold correctly (and/or more quickly) into the productive structure, even when as much as 216 extra nucleotides are present downstream.
  • the HH ribozyme construct is also quite efficient at self-processing, and slightly better than the HP(GU) construct even when downstream sequence is present.
  • HindIII-digested template 250 ng was used in a standard transcription reaction mixture containing: 50 mM Tris ⁇ HCl pH 8.3; 1 mM ATP, GTP and UTP; 50 ⁇ M CTP; 40 ⁇ Ci [ ⁇ - 32 P]CTP; 12 mM MgCl 2 ; 10 mM DTT.
  • the transcription/self-processing reaction was initiated by the addition of T7 RNA polymerase (15 U/ ⁇ l). Aliquots of 5 ⁇ l were taken at regular time intervals and the reaction was stopped by adding an equal volume of 2x formamide loading buffer (95% formamide, 15 mM EDTA, & dyes) and freezing on dry ice.
  • Transcripts containing the trans ribozyme with or without 3' flanking sequences were assayed for their ability to cleave their target in trans.
  • transcripts from three templates were resolved on a preparative gel and bands corresponding both to processed trans-acting ribozymes from the HH transcription reaction, and to full-length HH(mutant) and ⁇ HDV transcripts were isolated.
  • the trans-acting ribozyme portion is identical-with the exception of sequences at their 3' ends.
  • the HH trans-acting ribozyme contains only an additional UC at its 3' end, while HH(mutant) and ⁇ HDV have 52 and 37 nucleotides, respectively, at their 3' ends.
  • a 622 nucleotide, internally-labeled target RNA was incubated, under ribozyme excess conditions, along with the three ribozyme transcripts in a standard reaction buffer.
  • RNA was transcribed in a standard transcription buffer in the presence of [ ⁇ - 32 P]CTP (Chowrira & Burke, 1991 supra).
  • the reaction mixture was treated with 15 units of ribonuclease-free DNasel, extracted with phenol followed chloroform:isoamyl alcohol (25:1), precipitated with isopropanol and washed with 70% ethanol.
  • the dried pellet was resuspended in 20 ⁇ l DEPC-treated water and stored at -20°C.
  • Unlabeled ribozyme (1 ⁇ M) and internally labeled 622 nt substrate RNA ( ⁇ 10 nM) were denatured and renatured separately in a standard cleavage buffer (containing 50 mM Tris ⁇ HCl pH 7.5 and 10 mM MgCl 2 ) by heating to 90°C for 2 min. and slow cooling to 37°C for 10 min. The reaction was initiated by mixing the ribozyme and substrate mixtures and incubating at 37°C Aliquots of 5 ⁇ l were taken at regular time intervals, quenched by adding an equal volume of 2X formamide gel loading buffer and frozen on dry ice.
  • a standard cleavage buffer containing 50 mM Tris ⁇ HCl pH 7.5 and 10 mM MgCl 2
  • the samples were resolved on 5% polyacrylamide sequencing gel and results were quantitatively analyzed by radioanalytic imaging of gels with a Phosphorlmager® (Molecular Dynamics, Sunnyvale, CA).
  • Phosphorlmager® Molecular Dynamics, Sunnyvale, CA.
  • the HH trans-acting ribozyme cleaves the target RNA approximately
  • ⁇ HDV sequences are predicted to form multiple structures involving the 3' target-binding arm of the trans ribozyme that have stabilities ranging from 1-2 kcal/mole.
  • the observed reductions in activity for the ⁇ HDV and HH(mutant) constructs are consistent with the predicted folded structures, and it reinforces the view that the flanking sequences can decrease the catalytic efficiency of a ribozyme through nonproductive interactions with either the ribozyme or the substrate or both.
  • Example 25 RNA self-processing in vivo
  • Monolayers of a mouse L9 fibroblast cell line (OST7-1 ; Elroy-Stein and Moss, 1990 supra) were grown in 6-well plates with ⁇ 5 ⁇ 10 5 cells/well. Cells were transfected with circular plasmids (5 ⁇ g/well) using the calcium phosphate-DNA precipitation method (Maniatis et al., 1982 supra). Cells were lysed (4 hours post-transfection) by the addition of standard lysis buffer (200 ⁇ l/well) containing 4M guanadinium isothiocyanate, 25 mM sodium citrate (pH 7.0), 0.5% sarkosyl (Chomczynski and Sacchi, 1987 Anal. Biochem.
  • standard lysis buffer 200 ⁇ l/well
  • 4M guanadinium isothiocyanate 25 mM sodium citrate (pH 7.0)
  • 0.5% sarkosyl (Chomczynski and Sacchi, 1987 Anal. Biochem.
  • RNA pellet was resuspended in 0.2 M ammonium acetate and reprecipitated with ethanol. The pellet was then washed with 70% ethanol and resuspended in DEPC-treated water.
  • RNA Purified cellular RNA (3 ⁇ g/reaction) was first denatured in the presence of a 5' end-labeled DNA primer (100 pmol) by heating to 90°C for 2 min. in the absence of Mg 2+ , and then snap-cooling on ice for at least 15 min. This protocol allows for efficient annealing of the primer to its complementary RNA sequence.
  • the primer was extended using Superscript II reverse transcriptase (8 U/ ⁇ l; BRL) in a buffer containing 50 mM Tris ⁇ HCl pH 8.3; 10 mM DTT; 75 mM KCl; 1 mM MgCl 2 ; 1 mM each dNTP. The extension reaction was carried out at 42°C for 10 min.
  • the reaction was terminated by adding an equal volume of 2x formamide gel loading buffer and freezing on crushed dry ice.
  • the samples were resolved on a 10% polyacrylamide sequencing gel.
  • the primer sequences are as follows: HH primer, 5'-CTCCAGTTTCGAGCTTT-3'; HDV primer, 5'-A A G T A G C C C A G G T C G G A C C - 3 ' ; H P p ri m e r , 5 ' - ACCAGGTAATATACCACAAC-3'.
  • RNA self-processing might occur during cell lysis, RNA isolation and /or the primer extension assay.
  • Two precautions were taken to exclude this possibility.
  • 50 mM EDTA was included in the lysis buffer.
  • EDTA is a strong chelator of divalent metal ions such as Mg 2+ and Ca 2+ that are necessary for ribozyme activity. Divalent metal ions are therefore unavailable to self-processing RNAs following cell lysis.
  • a second precaution involved using primers in the primer-extension assay that were designed to hybridize to essential regions of the processing ribozyme. Binding of these primers should prevent the 3' cis-acting ribozymes from folding into the conformation essential for catalytic activity.
  • the first experiment involves primer extension analysis on full-length precursor RNAs that were added to non-transfected OST7-1 lysates after cell lysis. Thus, only if self-processing is occurring at some point after lysis would cleavage products be detected.
  • Full-length precursor RNAs were prepared by transcribing under conditions of low Mg 2+ (5 mM) and high NTP concentration (total 12 mM) in an attempt to eliminate the free Mg 2+ required for the self-processing reaction (Michel et al. 1992 Genes & Dev. 6, 1373).
  • RNA samples were gel-purified, and a known amount was added to lysates of non-transfected OST7-1 cells. RNA was purified from these lysates and incubated for 1 hr in DEPC-treated water at 37° C prior to the standard primer extension analysis (Figure 29. in vitro "-MgCl 2 " control). The predominant RNA detected in all cases corresponds to the primer extension product of full-length precursor RNAs. If, instead, the purified RNA containing the full-length precursor is incubated in 10 mM MgCl 2 prior to the primer extension analysis, most or all of the RNA detected by primer extension analysis undergoes cleavage (Figure 29. in vitro "+MgCl 2 " control). These results indicate that the standard RNA isolation and primer extension protocols used here do not provide a favorable environment for RNA self-processing, even though the RNA in question is inherently able to undergo self-cleavage.
  • RNAs were prepared and added to non-transfected OST7-1 lysates as in the previous control.
  • the internally-labeled precursor RNAs were carried through the RNA purification and primer extension reactions (in the presence of unlabeled primers) and analyzed to determine the extent of self-processing.
  • the vast majority of the added full-length RNA remained intact during the entire process of RNA isolation and primer extension.
  • Sequences in figures 23 through 25 are meant to be non-limiting examples. Those in the art will recognize that other embodiments can be readily generated using techniques generally known in the art. In addition, those in the art will recognize that Applicant provides guidance through the above examples as to how to best design vectors of this invention so that secondary structure of the mRNA allows efficient cleavage by releasing ribozymes. Thus, the specific constructs are not limiting in this invention. Such constructs can be readily tested as described above for such secondary structure, either by computer folding algorithms or empirically. Such constructs will then allow at least 80% completion of release of ribozymes, which can be readily determined as described above or by methods known in the art. That is, any such secondary structure in the RNA does not reduce release of the ribozymes by more than 20%.
  • RNA polymerase III RNA polymerase III
  • This stem structure is formed by hydrogen bond interactions (either Watson-Crick or non-Watson-Crick) between nucleotides in the 3' region (at least 8 bases) and complementary nucleotides in the 5' terminus of the same RNA molecule.
  • pol III promoter systems can also be used, for example, tRNA (Hall et al., 1982 Cell 29, 3-5), 5S RNA (Nielsen et al., 1993, Nucleic Acids Res. 21 , 3631-3636), adenovirus VA RNA (Fowlkes and Shenk, 1980 Cell 22, 405-413), U6 snRNA (Gupta and Reddy, 1990 Nucleic Acids Res. 19, 2073-2075), vault RNA (Kickoefer et al., 1993 J. Biol. Chem. 268, 7868-7873), telomerase RNA (Romero and Blackburn, 1991 Cell 67, 343-353), and others.
  • the construct described in this invention is able to accumulate RNA to a significantly higher level than other constructs, even those in which 5' and 3' ends are involved in hairpin loops.
  • the level of expression of a foreign RNA can be increased to between 20,000 and 50,000 copies per cell.
  • the molecules can be used as agonist or antagonist RNAs (affinity RNAs).
  • affinity RNAs RNAs
  • the invention features a pol III promoter system (e.g., a type 2 system) used to synthesize a chimeric RNA molecule which includes tRNA sequences and a desired RNA (e.g., a tRNA-based molecule).
  • a pol III promoter system e.g., a type 2 system
  • the RNA molecule in the following example has an A box and a B box of the type 2 pol III promoter system and has a 5' terminus or region able to base-pair with at least 8 bases of a complementary 3' end or region of the same RNA molecule.
  • This is meant to be a specific example. Those in the art will recognize that this is but one example, and other embodiments can be readily generated using other pol III promoter systems and techniques generally known in the art.
  • terminal is meant the terminal bases of an RNA molecule, ending in a 3' hydroxyl or 5' phosphate or 5' cap moiety.
  • region is meant a stretch of bases 5' or 3' from the terminus that are involved in base-paired interactions. It need not be adjacent to the end of the RNA. Applicant has determined that base pairing of at least one end of the RNA molecule with a region not more than about 50 bases, and preferably only 20 bases, from the other end of the molecule provides a useful molecule able to be expressed at high levels.
  • 3' region is meant a stretch of bases 3' from the terminus that are involved in intramolecular bas-paired interaction with complementary nucleotides in the 5' terminus of the same molecule.
  • the 3' region can be designed to include the 3' terminus. The 3' region therefore is > 0 nucleotides from the 3' terminus.
  • the 3' region is one nucleotide from the 3' terminus.
  • the 3' region is ⁇ 43 nt from 3' terminus.
  • tRNA molecule is meant a type 2 pol III driven RNA molecule that is generally derived from any recognized tRNA gene.
  • DNA encoding such molecules is readily available and can be modified as desired to alter one or more bases within the DNA encoding the RNA molecule and/or the promoter system.
  • such molecules include an A box and a B box that consist of sequences which are well known in the art (and examples of which can be found throughout the literature). These A and B boxes have a certain consensus sequence which is essential for a optimal pol III transcription.
  • chimeric tRNA molecule is meant a RNA molecule that includes a pol III promoter (type 2) region.
  • a chimeric tRNA molecule might contain an intramolecular base-paired structure between the 3' region and complementary 5' terminus of the molecule, and includes a foreign RNA sequence at any location within the molecule which does not affect the activity of the type 2 pol III promoter boxes.
  • a foreign RNA may be provided at the 3' end of the B box, or may be provided in between the A and the B box, with the B box moved to an appropriate location either within the foreign RNA or another location such that it is effective to provide pol III transcription.
  • the RNA molecule may include a hammerhead ribozyme with the B box of a type 2 pol III promoter provided in stem II of the ribozyme.
  • the B box may be provided in stem IV region of a hairpin ribozyme.
  • RNA any foreign RNA molecule which is useful from a therapeutic, diagnostic, or other viewpoint.
  • Such molecules include antisense RNA molecules, decoy RNA molecules, enzymatic RNA, therapeutic editing RNA and agonist and antagonist RNA.
  • antisense RNA is meant a non-enzymatic RNA molecule that binds to another RNA (target RNA) by means of RNA-RNA interactions and alters the activity of the target RNA (Eguchi et al., 1991 Annu. Rev. Biochem. 60, 631-652).
  • enzymatic RNA is meant an RNA molecule with enzymatic activity (Cech, 1988 J. American. Med. Assoc. 260, 3030-3035).
  • Enzymatic nucleic acids ribozymes act by first binding to a target RNA.
  • Such binding occurs through the target binding portion of a enzymatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA.
  • the enzymatic nucleic acid first recognizes and then binds a target RNA through base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA.
  • decoy RNA is meant an RNA molecule that mimics the natural binding domain for a ligand. The decoy RNA therefore competes with natural binding target for the binding of a specific ligand.
  • TAR HIV trans-activation response
  • TAR HIV trans-activation response
  • RNA can act as a "decoy” and efficiently binds HIV tat protein, thereby preventing it from binding to TAR sequences encoded in the HIV RNA (Sullenger et al., 1990 Cell 63, 601-608). This is meant to be a specific example. Those in the art will recognize that this is but one example, and other embodiments can be readily generated using techniques generally known in the art.
  • RNA RNA or DNA
  • RNA or DNA cellular target
  • RNA By “agonist RNA” is meant an RNA molecule that can bind to protein receptors with high affinity and cause the stimulation of specific cellular pathways.
  • antagonist RNA By “antagonist RNA” is meant an RNA molecule that can bind to cellular proteins and prevent it from performing its normal biological function (for example, see Tsai et al., 1992 Proc. Natl. Acad. Sci. USA 89, 8864-8868).
  • the invention includes vectors encoding RNA molecules as described above, cells including such vectors, methods for producing the desired RNA, and use of the vectors and cells to produce this RNA.
  • the invention features a transcribed non-naturally occuring RNA molecule which includes a desired therapeutic RNA portion and an intramolecular stem formed by base-pairing interactions between a 3' region and complementary nucleotides at the 5' terminus in the RNA.
  • the stem preferably includes at least 8 base pairs, but may have more, for example, 15 or 16 base pairs.
  • the 5' terminus of the chimeric tRNA includes a portion of the precursor molecule of the primary tRNA molecule, of which ⁇ 8 nucleotides are involved in base-pairing interaction with the 3' region; the chimeric tRNA contains A and B boxes; natural sequences 3' of the B box are deleted, which prevents endogenous RNA processing; the desired RNA molecule is at the 3' end of the B box; the desired RNA molecule is between the A and the B box; the desired RNA molecule includes the B box; the desired RNA molecule is selected from the group consisting of antisense RNA, decoy RNA, therapeutic editing RNA, enzymatic RNA, agonist RNA and antagonist RNA; the molecule has an intramolecular stem resulting from a base-paired interaction between the 5' terminus of the RNA and a complementary 3' region within the same RNA, and includes at least 8 bases; and the 5' terminus is able to base pair with at least 15 bases of the 3' region
  • the molecule is transcribed by a RNA polymerase III based promoter system, e.g., a type 2 pol III promoter system; the molecule is a chimeric tRNA, and may have the A and B boxes of a type 2 pol III promoter separated by between 0 and 300 bases; DNA vector encoding the RNA molecule of claim 51.
  • a RNA polymerase III based promoter system e.g., a type 2 pol III promoter system
  • the molecule is a chimeric tRNA, and may have the A and B boxes of a type 2 pol III promoter separated by between 0 and 300 bases
  • DNA vector encoding the RNA molecule of claim 51 is transcribed by a RNA polymerase III based promoter system, e.g., a type 2 pol III promoter system
  • the molecule is a chimeric tRNA, and may have the A and B boxes of a type 2 pol III promoter separated by between 0 and
  • the invention features an RNA or DNA vector encoding the above RNA molecule, with the portions of the vector encoding the RNA functioning as a RNA pol III promoter; or a cell containing the vector ; or a method to provide a desired RNA molecule in a cell, by introducing the molecule into a cell with an RNA molecule as described above.
  • the cells can be derived from animals, plants or human beings.
  • RNA-based gene therapy approaches In order for RNA-based gene therapy approaches to be effective, sufficient amounts of the therapeutic RNA must accumulate in the appropriate intracellular compartment of the treated cells. Accumulation is a function of both promoter strength of the antiviral gene, and the intracellular stability of the antiviral RNA.
  • RNA polymerase II polymerase II
  • poly III RNA polymerase III
  • Pol II produces messenger R ⁇ As located exclusively in the cytoplasm
  • pol III produces functional R ⁇ As found in both the nucleus and the cytoplasm.
  • Pol II promoters tend to be more tissue restricted
  • pol III genes encode tR ⁇ As and other functional R ⁇ As necessary for basic "housekeeping" functions in all cell types. Therefore, pol III promoters are likely to be expressed in all tissue types.
  • pol III transcripts from a given gene accumulate to much greater levels in cells relative to pol II genes. Intracellular accumulation of therapeutic R ⁇ As is also dependent on the method of gene transfer used. For example, the retroviral vectors presently used to accomplish stable gene transfer, integrate randomly into the genome of target cells.
  • a pol III based genetic element derived from a human tRNA i met gene and termed ⁇ 3-5 has been adapted to express antiviral RNAs (Sullenger et al., 1990 Mol. Cell. Biol. 10, 6512-6523). This element was inserted into the DC retroviral vector (Sullenger et al., 1990 Mol. Cell. Biol. 10, 6512-6523) to accomplish stable gene transfer, and used to express antisense RNAs against moloney murine leukemia virus and anti-HIV decoy RNAs (Sullenger et al., 1990 Mol.
  • HHI hammerhead
  • RNA with enzymatic activity RNA with enzymatic activity
  • Fig. 34 ribozyme expression in human T cell lines using the ⁇ 3-5 vector system
  • ⁇ 3-5/HHI ribozyme expression in human T cell lines using the ⁇ 3-5 vector system
  • ribozymes were found to accumulate to less than 100 copies per cell in the bulk T cell populations.
  • the applicant made a series of modified ⁇ 3-5 gene units containing enhanced promoter elements to increase transcription rates, and inserted structural elements to improve the intracellular stability of the ribozyme transcripts (Fig. 34).
  • S35 One of these modified gene units, termed S35, gave rise to more than a 100-fold increase in ribozyme accumulation in bulk T cell populations relative to the original ⁇ 3-5/HHI vector system. Ribozyme accumulation in individual clonal lines from the pooled T cell populations ranged from 10 to greater than 100 fold more than those achieved with the original ⁇ 3-5/HHI version of this vector.
  • the S35 gene unit may be used to express other therapeutic RNAs including, but not limited to, ribozymes, antisense, decoy, therapeutic editing, agonist and antagonist RNAs. Application of the S35 gene unit would not be limited to antiviral therapies, but also to other diseases, such as cancer, in which therapeutic RNAs may be effective.
  • the S35 gene unit may be used in the context of other vector systems besides retroviral vectors, including but not limited to, other stable gene transfer systems such as adeno-associated virus (AAV; Carter, 1992 Curr. Opin. Genet. Dev. 3, 74), as well as transient vector systems such as plasmid delivery and adenoviral vectors (Berkner, 1988 BioTechniques 6, 616-629).
  • AAV adeno-associated virus
  • transient vector systems such as plasmid delivery and adenoviral vectors (Berkner, 1988 BioTechniques 6, 616-629).
  • the S35 vector encodes a truncated version of a tRNA wherein the 3' region of the RNA is base-paired to complementary nucleotides at the 5' terminus, which includes the 5' precursor portion that is normally processed off during tRNA maturation.
  • a tRNA truncated version of a tRNA wherein the 3' region of the RNA is base-paired to complementary nucleotides at the 5' terminus, which includes the 5' precursor portion that is normally processed off during tRNA maturation.
  • ⁇ 3-5/HHI ribozyme-encoding sequences
  • One strategy involved the incorporation of stem-loop structures into the termini of the transcript. Two such constructs were cloned, S3 which contains a stem-loop structure at the 3' end, and S5 which contains stem-loop structures at both ends of the transcript ( Figure 34).
  • the second strategy involved modification of the 3' terminal sequences such that the 5' terminus and the 3' end sequences can form a stable base-paired stem.
  • FIG 48 there is provided a general structure for a chimeric RNA molecule of this invention.
  • Each N independently represents none or a number of bases which may or may not be base paired.
  • the A and B boxes are optional and can be any known A or B box, or a consensus sequence as exemplified in the figure.
  • the desired nucleic acid to be expressed can be any location in the molecule, but preferably is on those places shown adjacent to or between the A and B boxes (designated by arrows).
  • Figure 49 shows one example of such a structure in which a desired RNA is provided 3' of the intramolecular stem. A specific example of such a construct is provided in Figures 50 and 51.
  • Oligonucleotides encoding the S35 insert that overlap by at least 15 nucleotides were designed (5' GATCCACTCTGCTGTTCTGTTTTTGA 3' and 5' CGCGTCAAAAACAGAACAGCAGAGTG 3').
  • the oligonucleotides (10 ⁇ M each) were denatured by boiling for 5 min in a buffer containing 40 mM Tris.HCl, pH8.0.
  • the oligonucleotides were allowed to anneal by snap cooling on ice for 10-15 min.
  • the annealed oligonucleotide mixture was converted into a double-stranded molecule using Sequenase® enzyme (US Biochemicals) in a buffer containing 40 mM Tris.HCI, pH7.5, 20 mM MgCl 2 , 50 mM NaCl, 0.5 mM each of the four deoxyribonucleotide triphosphates, 10 mM DTT.
  • the reaction was allowed to proceed at 37°C for 30 min.
  • the reaction was stopped by heating to 70°C for 15 min.
  • the double stranded DNA was digested with appropriate restriction endonucleases (BamHI and MluI) to generate ends that were suitable for cloning into the ⁇ 3-5 vector.
  • the double-stranded insert DNA was ligated to the ⁇ 3-5 vector DNA by incubating at room temperature (about 20°C) for 60 min in a buffer containing 66 mM Tris.HCl, pH 7.6, 6.6 mM MgCl 2 , 10 mM DTT, 0.066 ⁇ M ATP and 0.1 U/ ⁇ l T4 DNA Ligase (US Biochemicals).
  • Competent E. coli bacterial strain was transformed with the recombinant vector DNA by mixing the cells and DNA on ice for 60 min. The mixture was heat-shocked by heating to 37°C for 1 min. The reaction mixture was diluted with LB media and the cells were allowed to recover for 60 min at 37°C The cells were plated on LB agar plates and incubated at 37°C for ⁇ 18 h.
  • Plasmid DNA was isolated from an overnight culture of recombinant clones using standard protocols (Ausubel et al., Curr. Protocols Mol. Biology 1990, Wiley & Sons, NY).
  • the identity of the clones were determined by sequencing the plasmid DNA using the Sequenase DNA sequencing kit (US Biochemicals).
  • the resulting recombinant ⁇ 3-5 vector contains the S35 sequence.
  • the HHI encoding DNA was cloned into this ⁇ 3-5-S35 containing vector using SaclI and BamHI restriction sites.
  • RNA from the transduced MT2 cells were extracted and the presence of ⁇ 3-5/ribozyme chimeric transcripts were assayed by Northern analysis (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY).
  • Northern analysis of RNA extracted from MT2 transductants showed that ⁇ 3-5/ribozyme chimeras of appropriate sizes were expressed (Fig. 35.36).
  • Fig. 35.36 Northern analysis of RNA extracted from MT2 transductants showed that ⁇ 3-5/ribozyme chimeras of appropriate sizes were expressed (Fig. 35.36).
  • Fig. 35.36 The pattern of expression seen from the ⁇ 3-5/HHI ribozyme chimera was similar to 12 other ribozymes cloned into the ⁇ 3-5 vector (not shown).
  • ⁇ 3-5/HHI ribozyme chimeras accumulated, on average, to less than 100 copies per cell. Addition of a stem-loop onto the 3' end of ⁇ 3-5/HHI did not lead to increased ⁇ 3-5 levels (S3 in Fig. 35,36). The S5 construct containing both 5' and 3' stem-loop structures also did not lead to increased ribozyme levels (Fig. 35.36).
  • the S35 construct expression in MT2 cells was about 100-fold more abundant relative to the original ⁇ 3-5/HHI vector transcripts (Fig. 35,36). This may be due to increased stability of the S35 transcript.
  • RNA extracted from the transduced MT2 T cells were incubated with a labeled substrate containing the HHI cleavage site ( Figure 37). Ribozyme activity in all but the S35 constructs, was too low to detect. However, ribozyme activity was detectable in S35-transduced T cell RNA. Comparison of the activity observed in the S35-transduced MT2 RNA with that seen with MT2 RNA in which varying amounts of in vitro transcribed S5 ribozyme chimeras, indicated that between 1-3 nM of S35 ribozyme was present in S35-transduced MT2 RNA. This level of activity corresponds to an intracellular concentration of 5,000-15,000 ribozyme molecules per cell.
  • Example 29 Clonal variation Variation in the ribozyme expression levels among cells making up the bulk population was determined by generating several clonal cell lines from the bulk S35 transduced CEM line (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY) and the ribozyme expression and activity levels in the individual clones were measured ( Figure 38 and 39). All the individual clones were found to express active ribozyme. The ribozyme activity detected from each clone correlated well with the relative amounts of ribozyme observed by Northern analysis.
  • ribozyme levels among the clones ranged from approximately 1 ,000 molecules per cell in clone G to 11 ,000 molecules per cell in clone H (Fig. 38).
  • the fact that all 14 clones were found to express ribozyme indicate that the percentage of cells in the bulk population expressing ribozyme is also very high.
  • the lowest level of expression in the clones was still more than 10-fold that seen in bulk cells transduced with the original ⁇ 3-5 vector. Therefore, the S35 gene unit should be much more effective in a gene therapy setting in which bulk cells are removed, transduced and then reintroduced back into a patient.
  • TRZ A transcription unit, termed TRZ, is designed that contains the S35 motif ( Figure 52).
  • a desired RNA e.g. ribozyme
  • TRZ-A and TRZ-B are non-limiting examples of the TRZ-tRNA chimera.
  • a hammerhead ribozyme targeted to site I (HHITRZ-A; Fig. 53) and a hairpin ribozyme (HPITRZ-A; Fig. 54), also targeted to site I, is cloned individually into the indicated region of TRZ tRNA chimera.
  • the resulting ribozyme trancripts retain full RNA cleavage activity (see for example Fig. 55). Applicant has shown that efficient expression of these TRZ tRNA chimera can be achieved in mammalian cells.
  • RNAs like antisense, therapeutic editing RNAs, decoys, can be readily inserted into the indicated region of TRZ-tRNA chimera to achieve therapeutic levels of RNA expression in mammalian cells.
  • Ribozyme expression in T cell lines stably-transduced with either a retroviral-based or an Adeno-associated virus (AAV)-based ribozyme expression vector ( Figure 56).
  • the human T cell lines MT2 and CEM were transduced with either retroviral or AAV vectors encoding a neomycin slelctable marker and a ribozyme (S35/HHI) expressed from pol III met i tRNA-driven promoter.
  • Cells stably-transduced with the vectors were selectivelyt expanded medium containing the neomycin antibiotic derivative, G418 (0.7 mg/ml). Ribozyme expression in the stable cell lines was then alalyzed by Northern analysis.
  • the probe used to detect ribozyme transcripts also cross-hybridized with human met i tRNA sequences.
  • S35/HHI RNA accumulates to significant levels in MT2 and CEM cells when transduced with either the retrovirus or the AAV vector.
  • adenovirus vector Figure 57
  • plasmid DNA vector plasmid DNA vector
  • alpha virus vectors plasmid DNA vector
  • alpha virus vectors alpha virus vectors
  • the transcription units can be expressed individually or in multiples using pol II and/or pol III promoters.
  • ribozymes generated by ligation methods described in Praper et al., PCT WO 93/23569, to cleave target RNA was tested on either matched substrate RNA (Fig. 58) or long (622 nt) RNA (Fig. 59, 60 and 61).
  • Matched substrate RNAs were chemically synthesized using solid-phase RNA synthesis chemistry (Scaringe et al., 1990 Nucleic Acids Res. 18, 5433-5441). Substrate RNA was 5' end-labeled using [ ⁇ - 32 P] ATP and polynucleotide kinase (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). Ribozyme reactions were carried out under ribozyme excess conditions (kcat/KM; Herschlag and Cech, 1990 Biochemistry 29, 10159-10171).
  • ribozyme and substrate RNA were denatured and renatured separately by heating to 90°C and snap cooling on ice for 10 min in a buffer containing 50 mM Tris. HCl pH 7.5 and 10 mM MgCl 2 .
  • Cleavage reaction was initiated by mixing the ribozyme with the substrate at 37°C Aliquots of 5 ⁇ l were taken at regular intervals of time and the reaction was stopped by mixing with equal volume of formamide gel loading buffer (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). The samples were resolved on 20 % polyacrylamide-urea gel. Refering to Fig. 58.
  • RPI A is a HH ribozyme with 6/6 binding arms. This ribozyme was synthesized chemically either as a one piece ribozyme or was synthesized in two fragments followed by ligation to generate a one piece ribozyme. The kcat/KM values for the two ribozymes were comparable.
  • the target RNA (containing HH ribozyme cleavage sites B, C and D) was transcribed from this PCR amplified template using T7 RNA polymerase.
  • the transcript was internally labeled during transcription by including [ ⁇ - 32 P] CTP as one of the four ribonucleotide triphosphates.
  • the transcription mixture was treated with DNase-1 , following transcription at 37°C for 2 hours, to digest away the DNA template used in the transcription.
  • RNA was precipitated with Isopropanol and the pellet was washed two times with 70% ethanol to get rid of salt and nucleotides used in the transcription reaction.
  • RNA is resuspended in DEPC-treated water and stored at 4°C Ribozyme cleavage reactions were carried out under ribozyme excess (kcat/KM) conditions [Herschlag and Cech 1990 supra]. Briefly, 1000 nM ribozyme and 10 nM internally labeled target RNA were denatured separately by heating to 90°C for 2 min in the presence of 50 mM Tris.HCl, pH 7.5 and 10 mM MgCl 2. The RNAs were renatured by cooling to 37°C for 10-20 min.
  • RNA phosphoramidite chemistry To decrease the cost of chemical synthesis of RNA, applicant was interested in determining whether the length of stem II region of a typical hammerhead ribozyme ( ⁇ 4 bp stem II) can be shortened without decreasing the catalytic efficiency of the HH ribozyme.
  • the length of stem II was systematically shortened by one base-pair at a time.
  • HH ribozymes with three and two base-paired stem II were chemically synthesized using solid-phase RNA phosphoramidite chemistry (Scaringe et al., 1990 supra).
  • RNA molecules were chemically synthesized having the nucleotide base sequence shown in Fig. 65 for both the 5' and 3' fragments.
  • the 3' fragments are phosphorylated and ligated to the 5' fragment essentially as described in example 37.
  • the 3' and 5' fragments can hybridize together at helix 4 and are covalently linked via GAAA sequence.
  • helix 4 is shown as 3 base pairs it may be formed with only 1 or 2 base pairs.
  • Fig. 62 and 65 The target and the ribozyme sequences shown in Fig. 62 and 65 are meant to be non-limiting examples. Those in the art will recognize that other embodiments can be readily generated using other sequences and techniques generally known in the art.
  • a new helix i.e., a sequence able to form a double-stranded region with another single-stranded nucleic acid
  • This helix is provided at the 3' end of the ribozyme after helix 3 as shown in Figure 3.
  • at least two extra bases may be provided in helix 2 and a portion of the substrate corresponding to helix 2 may be either directly linked to the 5' portion able to hydrogen bond to the 3' end of the hairpin or may have a linker of atleast one base.
  • trans-cleaving is meant that the ribozyme is able to act in trans to cleave another RNA molecule which is not covalently linked to the ribozyme itself.
  • the ribozyme is not able to act on itself in an intramolecular cleavage reaction.
  • base-pair is meant a nucleic acid that can form hydrogen bond(s) with other RNA sequence by either traditional Watson-Crick or other non-traditional types (for example Hoogsteen type) of interactions.
  • the increase in length of helix 2 of a hairpin ribozyme has several advantages. These include improved stability of the ribozyme-target complex in vivo . In addition, an increase in the recognition sequence of the hairpin ribozyme improves the specificity of the ribozyme. This also makes possible the targeting of potential hairpin ribozyme sites that would otherwise be inaccessible due to neighboring secondary structure.
  • helix 2 of a hairpin ribozyme enhances trans-ligation reaction catalyzed by the ribozyme.
  • Trans-ligation reactions catalyzed by the regular hairpin ribozyme (4 bp helix 2) is very inefficient (Komatsu et al., 1993 Nucleic Acids Res. 21 , 185). This is attributed to weak base-pairing interactions between substrate RNAs and the ribozyme.
  • H2 can be 6 bp without significantly reducing the activity of the hairpin ribozyme.
  • the H2 arm length variation does not appear to be sequence dependent.
  • HP ribozymes with 6 bp H2 have been designed against five different target RNAs and all five ribozymes efficiently cleaved their cognate target RNA. Additionally, two of these ribozymes were able to successfully inhibit gene expression (e.g., TNF- ⁇ ) in mammalian cells. Results of these experiments are shown below.
  • HP ribozymes with 7 and 8 bp H2 are also capable of cleaving target RNA in a sequence-specific manner, however, the rate of the cleavage reaction is lower than those catalyzed by HP ribozymes with 6 bp H2.
  • a nucleoside or nucleotide sugar introduces an additional center of chirality into the sugar moiety.
  • Fig. 75 the general structures of 5'-C-alkylnucleotides belonging to the D-allose, 2, and L-talose, 3, sugar families are shown.
  • Useful specific D-allose and L-talose nucleotide derivatives are shown in Figure 76. 29-32 and Figure 77, 58-61 respectively.
  • This invention relates to the use of 5'-C-alkylnucleotides in oligonucleotides, which are particularly useful for enzymatic cleavage of RNA or single-stranded DNA, and also as antisense oligonucleotides.
  • 5'-C-alkylnucleotide-containing enzymatic nucleic acids are catalytic nucleic molecules that contain 5'-C-alkylnucleotide components replacing, but not limited to, double stranded stems, single stranded "catalytic core" sequences, single-stranded loops or single-stranded recognition sequences.
  • RNA or DNA molecules are able to cleave (preferably, repeatedly cleave) separate RNA or DNA molecules in a nucleotide base sequence specific manner.
  • Such catalytic nucleic acids can also act to cleave intramolecularly if that is desired.
  • enzymatic molecules can be targeted to virtually any RNA transcript.
  • 5'-C-alkylnucleotides which may be present in enzymatic nucleic acid or even in antisense oligonucleotides. Such nucleotides are useful since they enhance the stability of the antisense or enzymatic molecule, and can be used in locations which do not affect the desired activity of the molecule.
  • the enhanced stability may make the loss of activity of less consequence.
  • a 5'-C-alkyl-containing molecule has 10% the activity of the unmodified molecule, but has 10-fold higher stability in vivo then it has utility in the present invention.
  • the same analysis is true for antisense oligonucleotides containing such modifications.
  • the invention also relates to novel intermediates useful in the synthesis of such nucleotides and oligonucleotides (examples of which are shown in the Figures), and to methods for their synthesis.
  • the invention features 5'-C-alkylnucleosides, that is a nucleotide base having at the 5'-position on the sugar molecule an alkyl moiety.
  • the invention also features 5'-C-alkylnucleotides, and in preferred embodiments features those where the nucleotide is not uridine or thymidine. That is, the invention preferably includes all those nucleotides useful for making enzymatic nucleic acids or antisense molecules that are not described by the art discussed above.
  • the sugar of the nucleoside or nucleotide is in an optically pure form, as the talose or allose sugar. Examples of various alkyl groups useful in this invention are shown in
  • each R 1 group is any alkyl.
  • an "alkyl” group refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups.
  • the alkyl group has 1 to 12 carbons. More preferably it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkenyl group has 1 to 12 carbons. More preferably it is a lower alkenyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • alkyl also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkynyl group has 1 to 12 carbons. More preferably it is a lower alkynyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • alkyl groups may also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups.
  • An "aryl” group refers to an aromatic group which has at least one ring having a conjugated ⁇ electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted.
  • the preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups.
  • alkylaryl refers to an alkyl group (as described above) covalently joined to an aryl group (as described above.
  • Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted.
  • Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms.
  • Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted.
  • An "amide” refers to an -C(O)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • An “ester” refers to an -C(0)-OR', where R is either alkyl, aryl, alkylaryl or hydrogen.
  • the invention features oligonucleotides having one or more 5'-C-alkylnucleotides; e.g. enzymatic nucleic acids having a 5'-C-alkylnucleotide; and a method for producing an enzymatic nucleic acid molecule having enhanced activity to cleave an RNA or single-stranded DNA molecule, by forming the enzymatic molecule with at least one nucleotide having at its 5'-position an alkyl group.
  • the invention features 5'-C-alkylnucleotide triphosphates. These triphosphates can be used in standard protocols to form useful oligonucleotides of this invention.
  • the 5'-C-alkyl derivatives of this invention provide enhanced stability to the oligonulceotides containing them. While they may also reduce absolute activity in an in vitro assay they will provide enhanced overall activity in vivo. Below are provided assays to determine which such molecules are useful. Those in the art will recognize that equivalent assays can be readily devised.
  • the invention features a method for conversion of a protected allo sugar to a protected talo sugar. In the method, the protected allo sugar is contacted with triphenyl phosphine, diethylazodicarboxylate, and p-nitrobenzoic acid under inversion causing conditions to provide the protected talo sugar. While one example of such conditions is provided below, those in the art will recognize other such conditions. Applicant has found that such conversion allows for ready synthesis of all types of nucleotide bases as exemplified in the figures.
  • Figure 1 shows base numbering of a hammerhead motif in which the numbering of various nucleotides in a hammerhead ribozyme is provided. This is not to be taken as an indication that the Figure is prior art to the pending claims, or that the art discussed is prior art to those claims. Referring to Figure 1.
  • the preferred sequence of a hammerhead ribozyme in a 5'- to 3'-direction of the catalytic core is CUGANGAG [base paired with] CGAAA.
  • CUGANGAG base paired with
  • CGAAA CGAAA
  • 5'-C-alkyl substituted phosphoramidites may be incorporated not only into hammerhead ribozymes, but also into hairpin, hepatitis delta virus, Group 1 or Group 2 intron catalytic nucleic acids, or into antisense oligonucleotides. They are, therefore, of general use in any nucleic acid structure.
  • Example 38 Methyl-2,3-O-Isopropylidine-6-Deoxy- ⁇ -D-allofuranoside (4)
  • Example 39 Methyl-2,3-O-Isopropylidine-5-O-t-Butyldiphenylsilyl-6-Deoxy- ⁇ -D-Allofuranoside (5).
  • Example 40 Methyl-5-O-t-Butyldiphenylsilyl-6-Deoxy- ⁇ -D-Allofuranoside (6).
  • Methylfuranoside 6 (7.0 g, 17.5 mmol) was coevaporated with pyridine (2 ⁇ 100 mL) and dissolved in pyridine (100 mL). Benzoyl chloride (5.4 g, 38.5 mmol) was added and the reaction mixture was left at RT for 16 h. Dry EtOH (50 mL) was added and the reaction mixture was evaporated to dryness after 0.5 h. The residue was dissolved in CH 2 Cl 2 (300 mL), washed with sat. NaHCO 3 (2 ⁇ 75 mL), brine (2 ⁇ 75 mL) dried over MgSO 4 and evaporated to dryness. The product was purified by flash chromatography in CH 2 Cl 2 to yield 9.5 g (89%) of compound 7.
  • Example 42 1-O-Acetyl-2,3-di-O-benzoyl-5-O-t-Butyldiphenylsilyl-6-Deoxy- ⁇ -D-Allofuranose (8).
  • Dibenzoate 7 (4.7 g, 7.7 mmol) was dissolved in a mixture of AcOH (10.0 mL), AC2O (20.0 mL) and EtOAc (30 mL) and the reaction mixture was cooled 0 °C 98% H 2 SO 4 (0.15 mL) was then added. The reaction mixture was kept at 0 °C for 16 h, and then poured into a cold 1 :1 mixture of sat. NaHCO 3 and EtOAc (150 mL).
  • N 4 -benzoylcytosine (1.84 g, 8.56 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (3 h) occurred, and then for an additional hour.
  • the reaction mixture was cooled to RT evaporated to dryness and coevaporated with dry toluene (2 ⁇ 50 mL). To the residue was added a solution of of acetates 8 (3.6 g, 5.6 mmol) in dry CH 3 CN (100 mL), followed by CF 3 SO 3 SiMe 3 (4.76 g, 21.4 mmol).
  • N 6 -benzoyladenine (2.86 g, 11.86 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (7 h) occurred, and then for an additional hour.
  • the reaction mixture was cooled to RT evaporated to dryness and coevaporated with dry toluene (2 ⁇ 50 mL).
  • To the residue was added a solution of of acetates 8 (3.6 g, 5.6 mmol) in dry CH 3 CN (100 mL) followed by CF 3 SO 3 SiMe 3 (6.59 g, 29.7 mmol).
  • the reaction mixture was boiled under reflux for 8 h, cooled to RT, concentrated to 1/3 of its original volume, diluted with CH 2 Cl 2 (100 mL) and extracted with sat. NaHCO 3 (2 ⁇ 50 mL), brine (2 ⁇ 50 mL) dried over MgSO 4 and evaporated to dryness.
  • the product 11 was purified by flash chromatography using a gradient of 0-5% MeOH in CH 2 Cl 2 . Yield: 2.7 g (60%).
  • Example 46 N 2 -Isobutyryl-9-(2',3'-di-O-Benzoyl-5'-O-t-Butyldiphenylsilyl-6'-Deoxy- ⁇ -D-Allofuranosyl)guanine (12).
  • N 2 -lsobutyrylguanine (1.47 g , 11.2 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (6 h) occurred, and then for an additional hour. The reaction mixture was cooled to RT evaporated to dryness and coevaporated with dry toluene (2 ⁇ 50 mL).
  • Nucleoside 15 (0.55 g, 0.92 mmol) was dissolved in dry CH 2 Cl 2 (50 mL). AgNO 3 (0.34 g, 2.0 mmol), dimethoxytrityl chloride (0.68 g, 2.0 mmol) and sym-collidine (0.48 g) were added under Ar. The reaction mixture was stirred for 2h, diluted with CH 2 Cl 2 (100 mL), filtered, evaporated to dryness and coevaporated with toluene (2 ⁇ 50 mL). Purification by flash chromatography using a gradient of 0-5% MeOH in CH 2 Cl 2 yielded 0.8 g (97%) of compound 19.
  • Nucleoside 19 (1.8 g, 2 mmol) was dissolved in dioxane (50 mL), cooled to 0 °C and 2 M NaOH (50 mL) was added. The reaction mixture was kept at 0 °C for 45 m, neutralized with Dowex 50 (Pyr + form), filtered and the resin was washed with MeOH (2 x 50 mL). The filtrate was then evaporated to dryness. Purification by flash chromatography using a gradient of 0-10% MeOH in CH 2 Cl 2 yielded 1.1 g (80%) of 23.
  • Example 50 N 6 -Benzoyl-9-(-5'-O-Dimethoxytrityl-2'-O-t-butyldimethylsilyl-6'-Deoxy- ⁇ -D-Allofuranosyl)adenine (27).
  • Nucleoside 23 (1.2 g, 1.8 mmol) was dissolved in dry THF (50 mL). Pyridine (0.50 g, 8 mmol) and AgNO 3 (0.4 g, 2.3 mmol) were added. After the AgNO 3 dissolved (1.5 h), t-butyldimethylsilyl chloride (0.35 g , 2.3 mmol) was added and the reaction mixture was stirred at RT for 16 h. The reaction mixture was diluted with CH 2 Cl 2 (100 mL), filtered into sat. NaHCO 3 (50 mL), extracted, the organic layer washed with brine (2 ⁇ 50 mL), dried over MgSO 4 and evaporated to dryness. The product 27 was purified by flash chromatography using a hexanes:EtOAc / 7:3 gradient. Yield: 0.7 g (50%).
  • Example 51 N 6 -Benzoyl-9-(-5'-O-Dimethoxytrityl-2'-O-t-butyldimethylsilyl-6'-Deoxy- ⁇ -D-Allofuranosyl)adenine-3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (31).
  • the alkyl substituted nucleotides of this invention can be used to form stable oligonucleotides as discussed above for use in enzymatic cleavage or antisense situations.
  • Such oligonucleotides can be formed enzymatically using triphosphate forms by standard procedure. Administration of such oligonucleotides is by standard procedure. See Sullivan et al., PCT WO 94/ 02595.
  • the ribozymes and the target RNA containing site O were synthesized, deprotected and purified as described above. RNA cleavage assay was carried our at 37°C in the presence of 10 mM MgCl 2 as described above.
  • Applicant has substituted 5'-C-Me-L-talo nucleotides at positions A6, A9, A9 + G10, C11.1 and C11.1 + G10, as shown in Figure 78 (HH-01 to HH-05).
  • HH-0 1,2,4 and 5 showed almost wild type activity ( Figure 79).
  • HH-03 demonstrated low catalytic activity.
  • Ribozymes HH-01, 2, 3, 4 and 5 are also extremely resistant to degradation by human serum nucleases.
  • This invention uses 2'-deoxy-2'-alkylnucleotides in oligonucleotides, which are particularly useful for enzymatic cleavage of RNA or single-stranded DNA, and also as antisense oligonucleotides.
  • 2'-deoxy-2'-alkylnucleotide-containing enzymatic nucleic acids are catalytic nucleic molecules that contain 2'-deoxy-2'-alkylnucleotide components replacing, but not limited to, double stranded stems, single stranded "catalytic core" sequences, single-stranded loops or single-stranded recognition sequences.
  • RNA or DNA molecules are able to cleave (preferably, repeatedly cleave) separate RNA or DNA molecules in a nucleotide base sequence specific manner.
  • Such catalytic nucleic acids can also act to cleave intramolecularly if that is desired.
  • Such enzymatic molecules can be targeted to virtually any RNA transcript.
  • 2'-deoxy-2'-alkylnucleotides which may be present in enzymatic nucleic acid or even in antisense oligonucleotides. Contrary to the findings of De Mesmaeker et al. applicant has found that such nucleotides are useful since they enhance the stability of the antisense or enzymatic molecule, and can be used in locations which do not affect the desired activity of the molecule. That is, while the presence of the 2'-alkyl group may reduce binding affinity of the oligonucleotide containing this modification, if that moiety is not in an essential base pair forming region then the enhanced stability that it provides to the molecule is advantageous.
  • the enhanced stability may make the loss of activity of less consequence.
  • a 2'-deoxy-2'-alkyl-containing molecule has 10% the activity of the unmodified molecule, but has 10-fold higher stability in vivo then it has utility in the present invention.
  • antisense oligonucleotides containing such modifications The same analysis is true for antisense oligonucleotides containing such modifications.
  • the invention also relates to novel intermediates useful in the synthesis of such nucleotides and oligonucleotides (examples of which are shown in the Figures), and to methods for their synthesis.
  • the invention features 2'-deoxy-2'-alkylnucleotides, that is a nucleotide base having at the 2'-position on the sugar molecule an alkyl moiety and in preferred embodiments features those where the nucleotide is not uridine or thymidine. That is, the invention preferably includes all those nucleotides useful for making enzymatic nucleic acids or antisense molecules that are not described by the art discussed above.
  • each R group is any alkyl.
  • alkyl does not include alkoxy groups which have an "-O-alkyl” group, where "alkyl” is defined as described above, where the O is adjacent the 2'-position of the sugar molecule.
  • the invention features oligonucleotides having one or more 2'-deoxy-2'-alkylnucleotides (preferably not a 2'-alkyl- uridine or thymidine); e.g. enzymatic nucleic acids having a 2'-deoxy-2'-alkylnucleotide; and a method for producing an enzymatic nucleic acid molecule having enhanced activity to cleave an RNA or single-stranded DNA molecule, by forming the enzymatic molecule with at least one nucleotide having at its 2'-position an alkyl group.
  • the invention features 2'-deoxy-2'-alkylnucleotide triphosphates. These triphosphates can be used in standard protocols to form useful oligonucleotides of this invention.
  • the 2'-alkyl derivatives of this invention provide enhanced stability to the oligonulceotides containing them. While they may also reduce absolute activity in an in vitro assay they will provide enhanced overall activity in vivo. Below are provided assays to determine which such molecules are useful. Those in the art will recognize that equivalent assays can be readily devised.
  • the invention features hammerhead motifs having enzymatic activity having ribonucleotides at locations shown in Figure 80 at 5, 6, 8, 12, and 15.1 , and having substituted ribonucleotides at other positions in the core and in the substrate binding arms if desired.
  • core refers to positions between bases 3 and 14 in Figure 80, and the binding arms correspond to the bases from the 3'-end to base 15.1 , and from the 5'-end to base 2).
  • Applicant has found that use of ribonucleotides at these five locations in the core provide a molecule having sufficient enzymatic activity even when modified nucleotides are present at other sites in the motif.
  • Figure 80 shows base numbering of a hammerhead motif in which the numbering of various nucleotides in a hammerhead ribozyme is provided. This is not to be taken as an indication that the Figure is prior art to the pending claims, or that the art discussed is prior art to those claims.
  • the preferred sequence of a hammerhead ribozyme in a 5'- to 3'-direction of the catalytic core is CUGANGAG[base paired with]CGAAA.
  • Ribozymes from Figure 80 and Table 45 were synthesized and assayed for catalytic activity and nuclease resistance. With the exception of entries 8 and 17, all of the modified ribozymes retained at lease 1/10 of the wild-type catalytic activity. From Table 45, all 2'-modified ribozymes showed very large and significant increases in stability in human serum (shown) and in the other fluids described below (Example 55, data not shown). The order of most agressive nuclease activity was fetal bovine serum, > human serum >human plasma > human synovial fluid. As an overall measure of the effect of these 2'-substitutions on stability and activity, a ratio ⁇ was calculated (Table 45).
  • compound 37 may be used as a general intermediate to prepare derivatized 2'C-alkyl phosphoramidites, where X is CH 3 , or an alkyl, or other group described above.
  • X is CH 3 , or an alkyl, or other group described above.
  • the following are non-limiting examples showing the synthesis of nucleic acids using 2'-C-alkyl substituted phosphoramidites, the syntheses of the amidites, their testing for enzymatic activity and nuclease resistance.
  • the method of synthesis used generally follows the procedure for normal RNA synthesis as described in Usman, N.; Ogilvie.K.K.; Jiang, M.-Y.; Cedergren.R.J. J. Am. Chem. Soc. 1987, 109, 7845-7854 and in Scaringe, S.A.; Franklyn.C; Usman, N. Nucleic Acids Res. 1990, 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end (compounds 10, 12, 17, 22, 31 , 18, 26, 32, 36 and 38).
  • these 2'-alkyl substituted phosphoramidites may be incorporated not only into hammerhead ribozymes, but also into hairpin, hepatitis delta virus, Group I or Group II intron catalytic nucleic acids, or into antisense oligonucleotides. They are, therefore, of general use in any nucleic acid structure.
  • RNA substrates 15-25-mers
  • purified 5'-end labeled ribozymes ⁇ 36-mers
  • Ribozyme stock solutions were 1 mM, 200 nM, 40 nM or 8 nM and the final substrate RNA concentrations were - 1 nM.
  • Total reaction volumes were 50 mL.
  • Example 58 5'-O-Dimethoxytrityl-2'-C-Allyl-Uridine (9) A solution of 8 (1.25 g, 2.45 mmol) in 10 mL of dry tetrahydrofuran
  • Example 60 3',5'-O-(Tetraisopropyl-disiloxane-1 ,3-diyl)-2'-C-Allyl-N 4 -Acetyl-Cytidine (11) Triethylamine (6.35 mL, 45.55 mmol) was added dropwise to a stirred ice-cooled mixture of 1 ,2,4-triazole (5.66 g, 81.99 mmol) and phosphorous oxychloride (0.86 mL, 9.11 mmol) in 50 mL of anhydrous acetonitrile.
  • Example 66 2'-Deoxy-2'-Difluoromethylene-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-Uridine 2'-Keto-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)uridine 14 (1.92 g,
  • Example 67 2'-Deoxy-2'-Difluoromethylene-Uridine 2'-Deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine (3.1 g, 5.9 mmol) dissolved in THF (20 mL) was treated with 1 M TBAF in THF (10 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on silica gel column. 2'-Deoxy-2'-difluoromethylene-uridine (1.1 g, 4.0 mmol, 68%) was eluted with 20% MeOH in CH 2 Cl 2 .
  • Triethylamine (4.8 mL, 34 mmol) was added to a solution of POCl 3 (0.65 mL, 6.8 mmol) and 1 ,2,4-triazole (2.1 g, 30.6 mmol) in acetonitrile (20 mL) at 0 °C
  • a solution of 2'-deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl) uridine 19 (1.65 g, 3.4 mmol) in acetonitrile (20 mL) was added dropwise to the above reaction mixture and left to stir at room temperature for 4 h.
  • Example 72 1-(2'-Deoxy-2,-Methylene-5'-O-Dimethoxytrityl- ⁇ -D-ribofuranosyl)-4-N-Acetyl-Cytosine 3'-(2-Cyanoethyl-N,N-diisopropylphosphoramidite) (22) 1-(2'-Deoxy-2'-methylene-5'-O-dimethoxytrityl- ⁇ -D-ribofuranosyl)-4-N-acetyl-cytosine 21 (0.88 g, 1.5 mmol) dissolved in dry CH 2 Cl 2 (10 mL) was placed in a round-bottom flask under Ar.
  • Example 74 1-(2'-Deoxy-2'-Difluoromethylene-5'-O-Dimethoxytrityl- ⁇ -D-ribofuranosyl)-4-N-Acetyl-Cytosine (25)
  • Example 75 1-(2'-Deoxy-2'-Difluoromethylene-5'-O-Dimethoxytrityl- ⁇ -D-ribofuranosyl)-4- N-Acetylcytosine 3'-(2-cyanoethyl-N,N-diisopropylphosphoramidite) (26)
  • Acetic anhydride (4.6 mL) was added to a solution of 3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine (Brown, J.; Christodolou, C.; Jones.S.; Modak.A.; Reese, C; Sibanda,S.; Ubasawa A. J. Chem .Soc. Perkin Trans. I 1989, 1735) (6.2 g, 9.2 mmol) in DMSO (37 mL) and the resulting mixture was stirred at room temperature for 24 h. The mixture was concentrated in vacuo.
  • Example 77 2'-Deoxy-2'-methylene-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-Butylbenzoyl)-Adenosine (29)
  • sec-butyllithium in hexanes (11.2 mL, 14.6 mmol) was added to a suspension of triphenylmethylphosphonium iodide (7.07 g,17.5 mmol) in THF (25 mL) cooled at -78 °C
  • the homogeneous orange solution was allowed to warm to -30 °C and a solution of 2'-keto-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine 28 (4.87 g, 7.3 mmol) in THF
  • Methyl(triphenylphosphoranylidine)acetate (5.4 g, 16 mmol) was added to a solution of 2'-keto-3',5'-O-(tetraisopropyl disiloxane-1 ,3-diyl)-uridine 14 in CH 2 Cl 2 under argon. The mixture was left to stir at RT for 30 h. CH 2 Cl 2 (100 mL) and water were added (20 mL), and the solution was neutralized with a cooled solution of 2% HCl. The organic layer was washed with H 2 O (20 mL), 5% aq. ⁇ aHCO 3 (20 mL), H 2 O to neutrality, and brine (10 mL).
  • Example 87 5'-O-DMT-2'-Deoxy-2'-Methoxycarbonylmethylidine-Uridine 2'-Deoxy-2'-methoxycarbonylmethylidine-uridine 34 (1.2 g, 4.02 mmol) was dissolved in pyridine (20 mL). A solution of DMT-Cl (1.5 g, 4.42 mmol) in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH 2 Cl 2 (100 mL) and washed with sat. NaHCO 3 , water and brine.
  • alkyl substituted nucleotides of this invention can be used to form stable oligonucleotides as discussed above for use in enzymatic cleavage or antisense situations.
  • Such oligonucleotides can be formed enzymatically using triphosphate forms by standard procedure. Administration of such oligonucleotides is by standard procedure. See Sullivan et al. PCT WO 94/02595.
  • This invention synthesis and uses 3' and/or 5' dihalophosphonate-, e.g., 3' or 5'-CF 2 -phosphonate-, substituted nucleotides that maintain or enhance the catalytic activity and/or nuclease resistance of an enzymatic or antisense molecule.
  • catalytic nucleic molecules that contain 5'-and/or 3'-dihalophosphonate nucleotide components replacing, but not limited to, double-stranded stems, single-stranded "catalytic core" sequences, single-stranded loops or single-stranded recognition sequences. These molecules are able to cleave (preferably, repeatedly cleave) separate RNA or DNA molecules in a nucleotide base sequence specific manner. Such catalytic nucleic acids can also act to cleave intramolecularly if that is desired.
  • Such enzymatic molecules can be targeted to virtually any RNA or DNA transcript.
  • This invention concerns nucleic acids formed of standard nucleotides or modified nucleotides, which also contain at least one 5'-dihalophosphonate and/or one 3'-dihalophosphonate group.
  • intermediates may be incorporated into catalytic or antisense nucleic acids by either chemical (conversion of the nucleoside 5'-deoxy-5'-dihalomethylphosphonates into suitably protected phosphoramidites 12a or solid supports 12b, e.g., Figure 88) or enzymatic means (conversion of the nucleoside 5'-deoxy-5'-dihalomethylphosphonates into their triphosphates, e.g., 14 Figure 89, for T7 transcription).
  • chemical conversion of the nucleoside 5'-deoxy-5'-dihalomethylphosphonates into suitably protected phosphoramidites 12a or solid supports 12b, e.g., Figure 88
  • enzymatic means conversion of the nucleoside 5'-deoxy-5'-dihalomethylphosphonates into their triphosphates, e.g., 14 Figure 89, for T7 transcription.
  • the invention features 5' and/or 3'-dihalonucleotides and nucleic acids containing such 5' and/or 3'-dihalonucleotides.
  • the general structure of such molecules is shown below.
  • R 1 is H, OH, or R, where R is a hydroxyl protecting group, e.g., acyl, alkysilyl, or carbonate; each R 2 is separately H, OH, or R; each R 3 is separately a phosphate protecting group, e.g., methyl, ethyl, cyanoethyl, p-nitrophenyl, or chlorophenyl; each X is separately any halogen; and each B is any nucleotide base.
  • R 1 is H, OH, or R, where R is a hydroxyl protecting group, e.g., acyl, alkysilyl, or carbonate
  • each R 2 is separately H, OH, or R
  • each R 3 is separately a phosphate protecting group, e.g., methyl, ethyl, cyanoethyl, p-nitrophenyl, or chlorophenyl
  • each X is separately any halogen
  • each B is any
  • the invention in particular features nucleic acid molecules having such modified nucleotides and enzymatic activity.
  • the invention features a method for synthesis of such nucleoside 5'-deoxy-5'-dihalo and/or 3'-deoxy-3'-dihalophosphonates by condensing a dihalophosphonate-containing sugar with a pyrimidine or a purine under conditions suitable to form a nucleoside 5'-deoxy-5'-dihalophosphonate and/or a 3'-deoxy-3'-dihalophosphonate.
  • Phosphonic acids may exhibit important biological properties because of their similarity to phosphates (Engel, Chem. Rev. 1977, 77, 349-367). Blackburn and Kent (J. Chem. Soc, Perkin Trans. 1986, 913-917) indicate that based on electronic and steric considerations _-fluoro and _,_-difluoromethylphosphonates might mimic phosphate esters better than the corresponding phosphonates.
  • Analogues of pyro- and triphosphates 1 where the bridging oxygen atoms are replaced by a difluoromethylene group, have been employed as substrates in enzymatic processes (Blackburn et al., Nucleosides & Nucleotides 1985, 4, 165-167; Blackburn et al., Chem. Scr. 1986, 26, 21 -24).
  • 9-(5,5-Difluoro-5-phosphonopentyl)guanine (2) has been utilized as a multisubstrate analogue inhibitor of purine nucleoside phosphorylase (Halazy et al., J. Am. Chem. Soc. 1991 , 113, 315-317).
  • Oligonucleotides containing methylene groups in place of phosphodiester 5'-oxygens are resistant toward nucleases that cleave phosphodiester linkages between phosphorus and the 5'-oxygen (Breaker et al., Biochemistry 1993, 32, 9125-9128), but can still form stable complexes with complementary sequences.
  • Heinemann et al. (Nucleic Acids Res. 1991 , 19, 427-433) found that a single 3'-methylenephosphonate linkage had a minor influence on the conformation of a DNA octamer double helix.
  • Methyl 2,3-O - i s o p ro pyl i d e n e- ⁇ -D-ribofu ranose a, a-difluoromethylphosphonate (4) was synthesized from the 5-aldehyde according to the procedure of Martin et al. (Tetrahedron Lett. 1992, 33, 1839-1842) ( Figure 87). Removal of the isopropylidene group was accomplished under mild conditions (I 2 -MeOH, reflux, 18 h (Szarek et al., Tetrahedron Lett.
  • Nucleoside 5'-deoxy-5'-difluoromethylphosphonates 8 were finally purified on a DEAE Sephadex A-25 (HCO 3 -) column using a 0.01-0.25 M TEAB gradient for elution and obtained as their sodium salts (82% 8a; 87% 8b; 82% 8c).
  • nucleoside 5'-deoxy-5'-difluoromethylphosphonates may be incorporated not only into hammerhead ribozymes, but also into hairpin, hepatitis delta virus, Group 1 or Group 2 introns, or into antisense oligonucleotides. They are, therefore, of general use in any nucleic acid structure.
  • Example 92 Synthesis of Modified Triphosphate
  • nucleoside dihalophosphonates described herein are advantageous as modified nucleotides in any nucleic acid structure, e.g., catalytic or antisense, since they are resistant to exo- and endonucleases that normally degrade unmodified nucleic acids in vivo. They also do not perturb the normal structure of the nucleic acid in which they are incorporated thereby maintaining any activity associated with that structure. These compounds may also be of use as monomers as antiviral and/or antitumor drugs.
  • This invention replaces 2'-hydroxyl group of a ribonucleotide moiety with a 2'-amido or 2'-peptido moiety.
  • the 3' and 5' portions of the sugar of a nucleotide may be substituted, or the phosphate group may be substituted with amido or peptido moieties.
  • such a nucleotide has the general structure shown in Formula I below:
  • the base (B) is any one of the standard bases or is a modified nucleotide base known to those in the art, or can be a hydrogen group.
  • R 1 or R 2 is H or an alkyl, alkene or alkyne group containing between 2 and 10 carbon atoms, or hydrogen, an amine (primary, secondary or tertiary, e.g., R 3 NR 4 where each R 3 and R 4 independently is hydrogen or an alkyl, alkene or alkyne having between 2 and 10 carbon atoms, or is a residue of an amino acid, Le,., an amide), an alkyl group, or an amino acid (D or L forms) or peptide containing between 2 and 5 amino acids.
  • the zigzag lines represent hydrogen, or a bond to another base or other chemical moiety known in the art.
  • one of R 1 , R 2 and R 3 is an H, and the other is an amino acid or peptide.
  • RNA can assume a much more complex structural form than DNA because of the presence of the 2'-hydroxyl group in RNA. This group is able to provide additional hydrogen bonding with other hydrogen donors, acceptors and metal ions within the
  • RNA molecule RNA molecule.
  • Applicant now provides molecules which have a modified amine group at the 2' position, such that significantly more complex structures can be formed by the modified oligonucleotide.
  • modification with a 2'-amido or peptido group leads to expansion and enrichment of the side-chain hydrogen bonding network.
  • the amide and peptide moieties are responsible for complex structural formation of the oligonucleotide and can form strong complexes with other bases, and interfere with standard base pairing interactions. Such interference will allow the formation of a complex nucleic acid and protein conglomerate.
  • Oligonucleotides of this invention are significantly more stable than existing oligonucleotides and can potentially form biologically active bioconjugates not previously possible for oligonucleotides. They may also be used for in vitro selection of unique aptamers, that is, randomly generated oligonucleotides which can be folded into an effective ligand for a target protein, nucleic acid or
  • the invention features an oligonucleotide containing the modified base shown in Formula I, above.
  • the oligonucleotide may include a 3' or 5' nucleotide having a 3' or 5' located amino acid or aminoacyl group.
  • an "O" may be replaced with an S
  • the sugar may lack a base (i.e., abasic) and the phosphate moiety may be modified to include other substitutions (see Sproat, supra).
  • Example 93 General procedure for the preparation of 2'-aminoacyl-2'-deoxy-2'-aminonucleoside conjugates.
  • the scheme shows synthesis of conjugate of 2'-d-2'-aminouridine.
  • nucelotides e.g., adenosine, cytidine, guanosine
  • abasic moieties e.g., adenosine, cytidine, guanosine
  • Example 94 RNA cleavage bv hammerhead ribozymes containing 2'-aminoacyl modifications.
  • Hammerhead ribozymes targeted to site N are synthesized using solid-phase synthesis, as described above. U4 and U7 positions are modified, individually or in combination, with either 2'-NH-alanine or 2'-NH-lysine.
  • RNA cleavage assay in vitro Substrate RNA is 5' end-labeled using [ ⁇ - 32 P] ATP and T4 polynucleotide kinase (US Biochemicals). Cleavage reactions were carried out under ribozyme "excess" conditions. Trace amount ( ⁇ 1 nM) of 5' end-labeled substrate and 40 nM unlabeled ribozyme are denatured and renatured separately by heating to 90°C for 2 min and snap-cooling on ice for 10 -15 min. The ribozyme and substrate are incubated, separately, at 37°C for 10 min in a buffer containing 50 mM Tris-HCl and 10 mM MgCl 2 .
  • the reaction is initiated by mixing the ribozyme and substrate solutions and incubating at 37°C Aliquots of 5 ⁇ l are taken at regular intervals of time and the reaction is quenched by mixing with equal volume of 2X formamide stop mix. The samples are resolved on 20 % denaturing polyacrylamide gels. The results are quantified and percentage of target RNA cleaved is plotted as a function of time.
  • ribozymes containing 2'-NH-alanine or 2'-NH-lysine modifications at U4 and U7 positions cleave the target RNA efficiently.
  • Sequences listed in Figure 94 and the modifications described in Figure 95 are meant to be non-limiting examples. Those skilled in the art will recognize that variants (base-substitutions, deletions, insertions, mutations, chemical modifications) of the ribozyme and RNA containing other 2'-hydroxyl group modifications, including but not limited to amino acids, peptides and cholesterol, can be readily generated using techniques known in the art, and are within the scope of the present invention.
  • the modified solid support (has an OH group instead of the standard NH 2 end group) was prepared according to Haralambidis et al., Tetrahedron Lett. 1987, 28, 5199, (P denotes aminopropyl CPG or polystyrene type support).
  • O-DMT or NH-monomethoxytrityl (NH-MMT amino acid was attached to the above solid support using standard procedures for derivatization of the solid support (Gait, 1984, supra) creating a base-labile ester bond between amino acids and the support.
  • This support is suitable for the construction of RNA/DNA chain using suitably protected nucleoside phosphoramidites.
  • aminoacyl group(s) is attached to the phosphate group at the 5'-end of the RNA using standard procedures described above.
  • This invention uses a complementary oligonucleotide to position a nucleotide base changing activity at a particular site on a gene (RNA or genomic DNA), such that the nucleotide modifying activity will change (or revert) a mutation to wild-type, or its equivalent.
  • a revertant may be achieved by changing any one of the three codon positions.
  • creation of a stop codon in a deleterious gene is defined here as reverting a mutant phenotype to wild-type.
  • An example of this type of reversion is creating a stop codon in a critical HIV proviral gene in a human.
  • RNA is provided with a complementary (Watson-crick) oligonucleotide sequence to that in the target molecule.
  • sequence modifying oligonucleotide would (analogously to an antisense oligonucleotide or ribozyme) have to be continuously present to revert the RNA as it is made by the cell. Such a reversion would be transient and would potentially require continuous addition of more sequence modifying oligonucleotide.
  • a second approach targets DNA (Fig. 101) and has the advantage that changes may be permanently encoded in the target cell's genetic code.
  • a single course (or several courses) of treatment may lead to permanent reversion of the genetic disease. If inadvertent chromosomal mutations are introduced this may cause cancer, mutate other genes, or cause genetic changes in the germ-line (in patients of reproductive age).
  • the base changing activity is a specific methylation that may modulate gene expression it would not necessarily lead to germ-line transmission. See Lewin, Genes, 1983 John Wilely & Sons, Inc.
  • Complementary base pairing to single-stranded PNA or RNA is one method of directing an oligonucleotide to a particular site of PNA. This could occur by a strand displacement mechanism or by targeting PNA when it is single-stranded (such as during replication, or transcription).
  • Another method is using triple-strand binding (triplex formation) to double-stranded PNA, which is an established technique for binding polypyrimidine tracts, and can be extended to recognize all 4 nucleotides. See Povsic, T., Strobel, S., & Dervan, P. (1992). Sequence-specific double-strand alkylation and cleavage of DNA mediated by triple-helix formation. J. Am. Chem. Soc.
  • antisense oligonucleotides have been used to redirect an incorrect splice into order to obtain correct splicing of a splice mutant globin gene in vitro.
  • a complementary oligomer is used to correct an existiing mutant RNA, instead of the traditional approach of inhibiting that RNA by antisense.
  • the oligonucleotide will modify the nucleic acid sequence. This can be accomplished by activating an endogenous enzyme (see Figure 102). by appropriate positioning of an enzyme (or ribozyme) conjugated (or activated by the duplex) to the oligonucleotide, or by appropriate positioning of a chemical mutagen. Specific mutagens, such as nitrous acid which deaminates C to U, are most useful, but others can also be used if inactivation of a harmful RNA is desired.
  • RNA editing is an naturally occurring event in mammalian cells in which a sequence modifying activity edits a RNA to its proper sequence post-transcriptionally. Higuchi, M.,, Single, F., Kohler, M., Sommer, B., and Seeburg, P. (1993) RNA Editing of AMPA Receptor Subunit GluR-B: A base-paired intron-exon structure determines position and efficiency Cell 75:1361-1370. The machinery involved in RNA editing can be co-opted by a suitable oligonucleotide in order to promote chemical modification.
  • the changes in the base created by the methods of this invention cause a change in the nucleotide sequence, either directly, or after DNA repair by normal cellular mechanisms. These changes functionally correct a genetic defect or introduce a stop codon.
  • an active chemical group e.g., an alkylator
  • an antisense or triple strand oligonucleotide is attached to an antisense or triple strand oligonucleotide in order to chemically inactivate the target RNA or DNA.
  • this invention creates an alteration to an existing base in a nucleic acid molecule so that the base is read in vivo as a different base. This includes correcting a sequence instead of inactivating a gene but can also include inactivating a deleterious gene.
  • the invention features a method for altering in vivo the nucleotide base sequence of a naturally occurring mutant nucleic acid molecule.
  • the method includes contacting the nucleic acid molecule in vivo with an oligonucleotide or peptide nucleic acid or other sequence specific binding molecules able to form a duplex or triplex molecule with the nucleic acid molecule. After formation of the duplex or triplex molecule a base modifying activity chemically or enzymatically alters the targeted base directly, or after nucleic acid repair in vivo. This results in the functional alteration of the nucleic acid sequence.
  • alter is meant that one or more chemical moieties in a targeted base, or bases, is altered so that the mutant nucleic acid will be functionally different.
  • this is distinct from prior methods of correcting defects in DNA, such as homologous recombination, in which an entire segment of the targeted sequence is replaced with a segment of DNA from the transfected nucleic acid.
  • This is also distinct from other methods that use reactive groups to inactivate a RNA or DNA target, in that this method functionally corrects the sequence of the target, instead of merely damaging it, by causing it to be read by a polymerase as a different base from the original base.
  • the naturally occurring enzymes in a cell can be utilized to cause the chemical alteration, examples of which are provided below.
  • RNA molecule may be altered so that it can cause production of a desired protein, or a DNA molecule can be altered so that upon DNA repair, the DNA sequence is changed.
  • mutant it is meant a nucleic acid molecule which is altered in some way compared to equivalent molecules present in a normal individual.
  • Such mutants may be well known in the art, and include, molecules present in individuals with known genetic deficiencies, such as muscular dystrophy, or diabetes and the like. It also includes individuals with diseases or conditions characterized by abnormal expression of a gene, such as cancer, thalassemia's and sickle cell anemia, and cystic fibrosis. It allows modulation of lipid metabolism to reduce artery disease, treatment of integrated AIDS genomes, and AIDs RNA, and Alzeimer's disease.
  • this invention concerns alteration of a base in a mutant to provide a "wild type" phenotype and/or genotype.
  • the oligonucleotide is of a length (at least 12 bases, preferably 17 - 22) sufficient to activate dsRNA deaminase in vivo to cause conversion of an adenine base to inosine; the oligonucleotide is an enzymatic nucleic acid molecule that is active to chemically modify a base (see below); the nucleic acid molecule is DNA or RNA; the oligonucleotide includes a chemical mutagen, e.g., the mutagen is nitrous acid; and the oligonucleotide causes deamination of 5-methylcytosine to thymidine, cytosine to uracil, or adenine to inosine, or methtylation of cytosine to 5-methylcytosine.
  • the invention features correction of a mutation, rather than inactivation of a target by causing a mutation.
  • Sequence modifying activities can change one nucleotide to another
  • Sequence modifying activities could also delete or add one or more nucleotides to a sequence.
  • a specific embodiment of adding sequences is described by Sullenger and Cech, PCT/US94/12976 hereby incorporated by reference herein), in which entire exons with wild-type sequence are spliced into a mutant transcript.
  • the present invention features only the addition of a few bases (1 - 3).
  • the invention features ribozymes or enzymatic nucleic acid molecules active to change the chemical structure of an existing base in a separate nucleic acid molecule. Applicant is the first to determine that such molecules would be useful, and to provide a description of how such molecules might be isolated.
  • Molecules used to achieve in situ reversion can be delivered using the existing means employed for delivering antisense molecules and ribozymes, including liposomes and cationic lipid complexes. If the in situ reverting molecule is composed only of RNA, then expression vectors can be used in a gene therapy protocol to produce the reverting molecules endogenously, analogously to antisense or ribozymes expression vectors. There are several advantages of using such an expression vector, rather than simply replacing the gene through standard gene therapy. Firstly, this approach would limit the production of the corrected gene to cells that already express that gene. Furthermore, the corrected gene would be properly regulated by its natural transcriptional promoter.
  • mutant RNA creates a dominant gain of function protein (e.g., in sickle cell anemia), where correction of the mutant RNA is necessary to stop the production of the deleterious mutant protein, and allow production of the corrected protein.
  • function protein e.g., in sickle cell anemia
  • RNAs were different from the DNA sequence that encodes them.
  • RNA editing cellular RNA are post-transcriptionally modified to a) create a translation initiation and termination codons, b) enable tRNA and rRNA to fold into a functional conformation (for a review see Bass, B. L. (1993) In The RNA World. R. Gesteland, R. and Atkins, J. eds. (Cold Spring Harbor, New York; CSH Lab. Press) pp. 383-418).
  • the process of RNA editing includes base modification, deletion and insertion of nucleotides.
  • RNA editing process is widespread among lower eukaryotes, very few RNAs (four) have been reported to undergo editing in mammals (Bass, supra).
  • the predominant mode of RNA editing in mammalian system is base modification (C ⁇ U and A ⁇ G).
  • the mechanism of RNA editing in the mammalian system is postulated to be that C ⁇ U conversion is catalyzed by cytidine deaminase.
  • the mechanism of conversion of A ⁇ G has recently been reported for glutamate receptor B subunit (gluR-B) in rat PC12 cells (Higuchi, M. et al. (1993) Cell 75, 1361-1370).
  • Higuchi gluR-B mRNA precursor attains a structure such that intron 11 and exon 11 can form a stable stem-loop structure.
  • This stem-loop structure is a substrate for a nuclear double strand-specific adenosine deaminase enzyme. The deamination will result in the conversion of A ⁇ l. Reverse transcription followed by double strand synthesis will result in the incorporation of G in place of A.
  • the endogenous deaminase activity or other such activities can be utilized to achieve targeted base modification.
  • the following are examples of the invention to illustrate different methods by which in vivo conversion of a base can be achieved. These are provided only to clarify specific embodiments of the invention and are not limiting to the invention. Those in the art will recognize that equivalent methods can be readily devised within the scope of the claims.
  • Example 97 Exploiting cellular dsRNA dependent Adenine to Inosine converter:
  • This stop codon can be reverted by targeting an antisense RNA which is long enough to activate the dsRNA deaminase, which converts Adenines to Inosines.
  • the A to I transition will be read by the ribosome as an A to G transition in some cases and will thereby functionally revert the stop codon. While other A's in this region may be converted to I's and read as G, converting an A to I (G) cannot create a stop codon.
  • the A to I transitions in the region surrounding the target mutation will create some point mutations, however, the function of the dystrophin protein is rarely inactivated by point mutations.
  • the reverted mRNA was then translated in a cell lysate and assayed for luciferase activity.
  • the A to I transition was read by the ribosome as an A to G transition and the stop codon has successfully been reverted with the lysate treated complex.
  • an irrelevant non-complementary RNA oligonucleotide was added to the dystrophin/luciferase mRNA.
  • no translation luciferase activity
  • the hybrid was not treated with extract, and again no translation (Iuciferase activity) is observed ( Figure 103).
  • RNA oligonucleotides were synthesized on a 394 (ABI) synthesizer using phosphoramidite chemistry.
  • sequence of the synthetic complementary RNA that binds to the mutant dystrophin sequence is as follows (5' to 3'):
  • RNA oligonucleotides Fifty nanograms of mRNA pre-annealed to the RNA oligonucleotides was added to 7ul of nuclear extracts containing 1 mM ATP, 15mM EDTA, 1600un/ml RNasin and 12.5mM Tris pH 8 to a total volume of 12ul. Bass, B.L. & Weintraub, H. supra.
  • This mixture which contains the dsRNA deaminase activity, was incubated for 30 minutes at 25°C Next, 1.5ul of this mixture was added to a rabbit reticulocyte lysate in vitro translation mixture and translated for two hours according to the manufacturers protocol (Life Technologies, Gaithersberg, MP), except that an additional 1.3 mM magnesium acetate was added to compensate for the EPTA carried through from the nuclear extract mixture.
  • Luciferase assays were performed on 15ul of extract with the Promega luciferase assay system (Promega, Madison, WI), and luminescence was detected with a 96 well luminometer, and the results are displayed in the graph in figure 102.
  • Reactive groups such as alkylators that can modify nucleotide bases in targeted RNA or DNA have been conjugated to oligonucleotides. Additionally enzymes that modify nucleic acids have been conjugated to oligonucleotides. (Knorre, P.G., Valentin, V.V., Valentina, F.Z., Lebedev, AN. & Federova, O.S. Design and targeted reactions of oligonucleotide derivatives 1 -366 (CRC Press, Novosibirsk, 1993). In the past these conjugated chemical groups or enzymes have been used to inactivate DNA or RNA that is specifically targeted by antisense or triple-strand interactions. Below is a list of useful base changing activities that could be used to change the sequence of DNA or RNA targeted by antisense or triple strand interactions, in order to achieve in situ reversion of mutations, as described herein (see figure 100-104).
  • cytosine to create uracil (performed by the enzyme cytidine deaminase (Bass, B.L. in The RNA World (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1993) or by chemical groups similar to nitrous acid that promote oxidative deamination (Microbial Genetics, David Freifelder, Jones and Bartlett Publishers, Inc., Boston, 1987, PP.226-230.)
  • the following matrix shows that the chemical modifications noted can cause transversion reversions (pyrimidine to pyrimidine, or purine to purine) in RNA or DNA.
  • the transversions are not preferred because these are more difficult chemical transformations.
  • the footnotes refer to the specific desired chemical transformations.
  • the bold footnotes refer to the reaction on the opposite DNA strand. For example, if one desires to change an A to a G, this can be accomplished at the DNA level by using reaction #5 to change a T to a C in the opposing strand. In this example an A/T base pair goes to A/C , then when the DNA is replicated, or mismatch repair occurs this can become G/C, thus the original A has been converted to a G.
  • ISR matrix ISR matrix
  • RNA is designed that folds back on itself (this is similar to approaches already used to select for RNA ligases, Bartel, D. and Szostak, J. (1993)
  • RNA reverse transcribed into DNA (that is, using standard in vitro evolution protocol. Tuerk and Gold, 249 Science 505, 1990) , and then the DNA is selected for having a base change.
  • a restriction enzyme cleavage and size selection or its equivalent is used to isolate the fraction of DNAs with the appropriate base change. The cycle could then be repeated many times.
  • the in vitro selection (evolution) strategy is similar to approaches developed by Joyce (Beaudry, A. A. and Joyce, G.F. (1992) Science 257, 635-641 ; Joyce, G. F. (1992) Scientific American 267.
  • each member contains two domains: a) one domain consists of a region with defined (known) nucleotide sequence; b) the second domain consists of a region with degenerate (random) sequence.
  • the known nucleotide sequence domain enables: 1) the nucleic acid to bind to its target (the region flanking the mutant nucleotide), 2) complimentary DNA (cDNA) synthesis and PCR amplification of molecules selected for their base modifying activity, 3) introduction of restriction endonuclease site for the purpose of cloning.
  • the degenerate domain can be created to be completely random (each of the four nucleotides represented at every position within the random region) or the degeneracy can be partial (Beaudry, A. A. and Joyce, G.F. (1992) Science 257, 635-641).
  • the degenerate domain is flanked by regions containing known sequences (see Figure 105), such that the degenerate domain is placed across from the mutant base (the base that is targeted for modification).
  • This random library of nucleic acids is incubated under conditions that ensure folding of the nucleic acids into conformations that facilitate the catalysis of base modification (the reaction protocol may also include certain cofactors like ATP or GTP or an S-adenosyl-methionine (if methylation is desired) in order to make the selection more stringent).
  • the reaction protocol may also include certain cofactors like ATP or GTP or an S-adenosyl-methionine (if methylation is desired) in order to make the selection more stringent).
  • nucleic acids are converted into complimentary DNA (if the starting pool of nucleic acids is RNA).
  • Nucleic acids with base modification can be separated from rest of the population of nucleic acids by using a variety of methods.
  • a restriction endonuclease cleavage site can either be created or abolished as a result of base modification. If a restriction endonuclease site is created as a result of base modification, then the library can be digested with the restriction endonuclease (RE). The fraction of the population that is cleaved by the RE is the population that has been able to catalyze the base modification reaction (active pool).
  • a new piece of DNA (containing oligonucleotide primer binding sites for PCR and RE sites for cloning) is ligated to the termini of the active pool to facilitate PCR amplification and subsequent cycles (if necessary) of selection.
  • the final pool of nucleic acids with the best base modifying activity is cloned in to a plasmid vector and transformed into bacterial hosts. Recombinant plasmids can then be isolated from transformed bacteria and the identity of clones can be determined using DNA sequencing techniques.
  • Base modifying enzymatic nucleic acids (identified via in vitro selection) can be used to cause the chemical modification in vivo.
  • the ribozyme could be evolved to specifically bind a protein having an enzymatic base changing acitivity.
  • ribozymes can be used to cause the above chemical modifications in vivo.
  • the ribozymes or above noted antisense-type molecules can be administered by methods discussed in the above referenced art.
  • RNA portion of the R-loop can be used to target the whole R-loop complex to a desirable intracellular or cellular site, and aid in cellular uptake of the complex.
  • the invention features a method for introduction of enzymatic nucleic acid into a cell or tissue.
  • a complex of a first nucleic acid encoding the enzymatic nucleic acid and a second nucleic acid molecule is provided.
  • the second nucleic acid molecule has sufficient complementarity with the first nucleic acid to be able to form an R-loop base pair structure under physiological conditions.
  • the R-loop is formed in a region of the first nucleic acid molecule which promotes expression of RNA from the first nucleic acid under physiological conditions.
  • the method further includes contacting the complex with a cell or tissue under conditions in which the enzymatic nucleic acid is produced within the cell or tissue.
  • complex is simply meant that the two nucleic acid molecules interact by intermolecular bond formation (such as by hydrogen bonding) between two complementary base-paired sequences.
  • the complex will generally be stable under physiological condition such that it is able to cause initiation of transcription from the first nucleic acid molecule.
  • the first and second nucleic acid molecules may be formed from any desired nucleotide bases, either those naturally occurring (such as adenine, guanine, thymine and cytosine), or other bases well known in the art, or may have modifications at the sugar or phosphate moieties to allow greater stability or greater complex formation to be achieved.
  • such molecules may contain non-nucleotides in place of nucleotides.
  • modifications are well known in the art, see e.g., Eckstein et al., International Publication No. WO 92/07065; Perrault et al., 1990 Nature 344, 565; Pieken et al., 1991 Science.
  • sufficient complementarity is meant that sufficient base pairing occurs so that the R-loop base pair structure can be formed under the appropriate conditions to cause transcription of the enzymatic nucleic acid. Those in the art will recognize routine tests by which such sufficient base pairs can be determined. In general, between about 15 - 80 bases is sufficient in this invention.
  • physiological condition is meant the condition in the cell or tissue to be targeted by the first nucleic acid molecule, although the R-loop complex may be formed under many other conditions.
  • physiological condition is meant the condition in the cell or tissue to be targeted by the first nucleic acid molecule, although the R-loop complex may be formed under many other conditions.
  • One example is use of a standard physiological saline at 37°C, but it is simply desirable in this invention that the R-loop structure exists to some extent at the site of action so that the expression of the desired nucleic acid will be achieved at that site of action. While it is preferred that the R-loop structure be stable under those conditions, even a minimal amount of formation of the R-loop structure to cause expression will be sufficient.
  • the invention features a method for introduction of ribonucleic acid within a cell or tissue by forming an R-loop base-paired structure (as described above) with the first nucleic acid molecule lacking any promoter region or transcription termination signal such that once expression is initiated it will continue until the first nucleic acid is degraded.
  • the invention features a method in which the second nucleic acid is provided with a localization factor, such as a protein, e.g., an antibody, transferin, a nuclear localization peptide, or folate, or other such compounds well known in the art, which will aid in targeting the R-loop complex to a desired cell or tissue.
  • a localization factor such as a protein, e.g., an antibody, transferin, a nuclear localization peptide, or folate, or other such compounds well known in the art, which will aid in targeting the R-loop complex to a desired cell or tissue.
  • the first nucleic acid is a plasmid, e.g., one without a promoter or a transcription termination signal ;
  • the second nucleic acid is of length between about 40-200 bases and is formed of ribonucleotides at a majority of positions; and the second nucleic is covalently bonded with a ligand such as a nucleic acid, protein, peptide, lipid, carbohydrate, cellular receptor, nuclear localization factor, or is attached to maleimide or a thiol group:
  • the first nucleic acid is an expression plasmid lacking a promoter able to express a desired gene, e.g., it is a double-stranded molecule formed with a majority of deoxyribonucleic acids;
  • the R-loop complex is a RNA/DNA heteroduplex; no promoter or leader region is provided in the first nucleic acid; and the R-loop is adapted to prevent nucleosome assembly and is designed to aid recruitment of
  • the first nucleic acid encodes one or more enzymatic nucleic acids, e.g., it is formed with a plurality of intramolecular and intermolecular cleaving enzymatic nucleic acids to allow release of therapeutic enzymatic nucleic acid in vivo.
  • the invention features a complex of the above first nucleic acid molecules and second nucleic acid molecules.
  • R-loop complex
  • An R-loop complex is designed to provide a non-integrating plasmid so that, when an RNA polymerase binds to the plasmid, transcription is continuous until the plasmid is degraded. This is achieved by hybridizing an RNA molecule, 40 to 200 nucleotides in length, to a DNA expression plasmid resulting in an R-loop structure (see figure 106).
  • This RNA when conjugated with a ligand that binds to a cell surface receptor, triggers internalization of the plasmid/RNA-ligand complex. Formation of R-loops in general is described by DeWet, 1987 Methods in Enzymol. 145, 235; Neuwald et al., 1977 J. Virol. 21 ,1019; and Meyer et al., 1986 J. Ult. Mol. Str. Res. 96, 187. Thus, those in the art can readily design complexes of this invention following the teachings of the art.
  • Promoters placed in retroviral genomes have not always behaved as planned in that the additional promoter will serve as a stop signal or reverses the direction of the polymerase. Applicant was told that creation of an R-loop between the promoter and the reporter gene increased the transfection efficiency. Incubation of an RNA molecule with a double-stranded DNA molecule, containing a region of complementarity with the RNA will result in the formation of a stable RNA-DNA hetroduplex and the DNA strand that has a sequence identical to the RNA will be displaced into a loop-like structure called the R-loop. This displacement of DNA strand occurs because an RNA-DNA duplex is more stable compared to a DNA-DNA duplex.
  • RNA that hybridized to the leader sequence between the promoter and the gene, or directly to the first 80 nucleotides of the mRNA increased the expression levels 8-10 fold.
  • the proposed mechanism is that R-loop formation prevents nucleosome assembly, thus making the DNA more accessible for transcription.
  • the R-loop may resemble a RNA primer promoting either DNA replication or transcription (Daube and von Hippel, 1992, supra).
  • One of the salient features of this invention is to generate R-loops in expression vectors of choice and introduce them into cells to achieve enhanced expression from the expression vector. The presence of an R-loop may aid in the recruitment of cellular transcription machinery. Once an RNA polymerase binds to the plasmid and initiates transcription, the process will continue until a termination signal is reached, or the plasmid is degraded.
  • This invention will increase the expression of ribozymes inside a cell.
  • the idea is to construct a plasmid with no transcription termination signal, such that a transcript-containing multiple ribozyme units can be generated.
  • self-processing ribozymes can be cloned downstream of each therapeutic ribozyme (see figure 107) as described by Draper supra.
  • RNA used to generate R-loop structures can be covalently linked to a ligand (nucleic acid, proteins, peptides, lipids, carbohydrates, etc).
  • ligand nucleic acid, proteins, peptides, lipids, carbohydrates, etc.
  • Specific ligands can be chosen such that the ligand can bind selectively to a desired cell surface receptor. This ligand-receptor interaction will help internalize a plasmid containing an R-loop.
  • RNA is used to attach the ligand to the DNA such that localization of the gene to certain regions of the cell is achieved.
  • One of several methods can be used to attach a ligand to RNA.
  • RNA containing a 6 carbon spacer having a terminal primary amine is added into the RNA (see figure 108).
  • This amino group can be directly derivatized with the ligand, such as folate (Lee and Low, 1994 J. Biol. Chem. 269, 3198-3204).
  • the RNA containing a 6 carbon spacer with a terminal amine group is mixed with folate and the mixture is reacted with activators like 1 -(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC).
  • EDC 1 -(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • This reaction should be carried out in the presence of 1-Hydroxybenzotriazole hydrate (HOBT) to prevent any undesirable side reactions.
  • HOBT 1-Hydroxybenzotriazole hydrate
  • the RNA can also be derivatized with a heterobifuctional crosslinking agent (or linker) like succinimidyl 4-(p-maleimidophenyl)butyrate (SMPB).
  • SMPB succinimidyl 4-(p-maleimidophenyl)butyrate
  • the SMPB introduces a maleimide into the RNA.
  • This maleimide can then react with a thiol moiety either in a peptide or in a protein.
  • Thiols can also be introduced into proteins or peptides that lack naturally occurring thiols using succinylacetylthioacetate.
  • the amino linker can be attached at the 5' end or 3' end of the RNA.
  • the RNA can also contain a series of nucleotides that do not hybridize to the DNA and extend the linker away from the RNA/DNA complex, thus increasing the accessibility of the ligand for its receptor and not interfering with the hybridization.
  • These techniques can be used to link peptides such as nuclear localization signal (NLS) peptides (Lanford et al., 1984 Cell 37, 801-813; Kalderon et al., 1984 Cell 39, 499-509; Goldfarb et al., 1986 Nature 322, 641-644)and/or proteins like the transferrin (Curiel et al., 1991 Proc. Natl. Acad. Sci. USA 88, 8850-8854; Wagner et al., 1992 Proc.
  • NLS nuclear localization signal
  • an intrinsic thiol can be used to react with the maleimide or the thiols can be introduced into the protein itself using either iminothiolate or succinimidyl acetyl thioacetate (SATA; Duncan et al., 1983 Anal. Biochem 132, 68).
  • SATA requires an additional deprotection step using 0.5 M hydroxylamine.
  • liposomes can be used to cause an R-loop complex to be delivered to an appropriate intracellular cite by techniques well known in the art.
  • pH-sensitive liposomes Connor and Huang, 1986 Cancer Res. 46, 3431-3435
  • DNA transfection can be used to facilitate DNA transfection.
  • In vitro selection strategies can be used to select nucleic acids that a) can form stable R-loops b) selectively bind to specific cell surface receptors. These nucleic acids can then be covalently linked to each other.
  • the known nucleotide sequence domain enables: 1) the nucleic acid to bind to its target (a specific region of the double strand DNA), 2) complimentary DNA (cDNA) synthesis and PCR amplification of molecules selected for their affinity to form R-loop and/or their ability to bind to a specific receptor, 3) introduction of a restriction endonuclease site for the purpose of cloning.
  • the degenerate domain can be created to be completely random (each of the four nucleotides represented at every position within the random region) or the degeneracy can be partial (Beaudry and Joyce, 1992 Science 257, 635-641). In this invention, the degenerate domain is flanked by regions containing known sequences.
  • This random library of nucleic acids is incubated under conditions that ensure equilibrium binding to either double-stranded DNA or cell surface receptor. Following incubation, nucleic acids are converted into complementary DNA (if the starting pool of nucleic acids is RNA). Nucleic acids with desired characteristics can be separated from the rest of the population of nucleic acids by using a variety of methods (Joyce, 1992 supra). The desired pool of nucleic acids can then be carried through subsequent rounds of selection to enrich the population with the most desired traits. These molecules are then cloned in to appropriate vectors. Recombinant plasmids can then be isolated from transformed bacteria and the identity of clones can be determined using DNA sequencing techniques. Other embodiments are within the following claims. TABLE 1
  • RNAseP RNA M1 RNA
  • Size ⁇ 290 to 400 nucleotides.
  • RNA portion of a ribonucleoprotein enzyme Cleaves tRNA precursors to form mature tRNA.
  • RNA RNAs of the tobacco ringspot virus, ara bis mosaic virus and chicory yellow mottle virus
  • HDV Hepatitis Delta Virus

Abstract

Enzymatic RNA molecules which cleave ICAM-1 mRNA, IL-5 mRNA, rel A mRNA, TNF-α mRNA, RSV mRNA or RSV genomic RNA, or CML associated mRNA, and use of these molecules for the treatment of pathological conditions related to those mRNA-levels; ribonucleosides or nucleotides modified in 2', 3' or 5', methods for their synthesis, purification and deprotection; vectors containing multiple enzymatic nucleic acids, optionally in chimeric form with tRNAs; method for introducing enzymatic nucleic acids into cells by forming a complex with a second nucleic acid, where the complex is capable of taking an R-loop base-paired structure; method for altering a mutant nucleic acid in vivo by hybridization with an oligonucleotide capable of activating dsRNA deaminase, comprising an enzymatic activity or a chemical mutagen. Further are disclosed trans-cleaving or -ligating hairpin ribozymes lacking a substrate RNA moiety, as well as hammerhead ribozymes having an interconnecting loop between base pairs in stem II.

Description

METHOD AND REAGENT FOR INHIBITING THE EXPRESSION OF DISEASE RELATED GENES
Background of the Invention
This invention relates to reagents useful as inhibitors of gene expression relating to diseases such as inflammatory or autoimmune disorders, chronic myelogenous leukemia, or respiratory tract illness. Summary of the Invention
The invention features novel enzymatic RNA molecules, or ribozymes, and methods for their use for inhibiting the expression of disease related genes, e.g., ICAM-1 , IL-5, relA, TNF-α, p210 bcr-abl, and respiratory syncytial virus genes. Such ribozymes can be used in a method for treatment of diseases caused by the expression of these genes in man and other animals, including other primates.
Ribozymes are RNA molecules having an enzymatic activity which is able to repeatedly cleave other separate RNA molecules in a nucleotide base sequence specific manner. Such enzymatic RNA molecules can be targeted to virtually any RNA transcript, and efficient cleavage has been achieved in vitro. Kim et al., 84 Proc. Natl. Acad. Sci. USA 8788, 1987; Haseloff and Gerlach, 334 Nature 585, 1988; Cech, 260 JAMA 3030, 1988; and Jefferies et al., 17 Nucleic Acids Research 1371 , 1989.
Six basic varieties of naturally-occurring enzymatic RNAs are known presently. Each can catalyze the hydrolysis of RNA phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions. Table 1 summarizes some of the characteristics of these ribozymes.
Ribozymes act by first binding to a target RNA. Such binding occurs through the target RNA binding portion of a ribozyme which is held in close proximity to an enzymatic portion of the RNA which acts to cleave the target RNA. Thus, the ribozyme first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA. Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After a ribozyme has bound and cleaved its RNA target it is released from that RNA to search for another target and can repeatedly bind and cleave new targets. The enzymatic nature of a ribozyme is advantageous over other technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the effective concentration of ribozyme necessary to effect a therapeutic treatment is lower than that of an antisense oligonucleotide. The advantage reflects the ability of the ribozyme to act enzymatically. Thus, a single ribozyme molecule is able to cleave many molecules of target RNA. In addition, the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding, but also on the mechanism by which the molecule inhibits the expression of the RNA to which it binds. That is, the inhibition is caused by cleavage of the RNA target and so specificity is defined as the ration of the rate of cleavage of the targeted RNA over the rate of cleavage of non-targeted RNA. This cleavage mechanism is dependent upon factors additional to those involved in base pairing. Thus, it is thought that the specificity of action of a ribozyme is greater than that of antisense oligonucleotide binding the same RNA site. With their catalytic activity and increased site specificity, ribozymes represent more potent and safe therapeutic molecules than antisense oligonucleotides. Thus, in a first aspect, this invention relates to ribozymes, or enzymatic
RNA molecules, directed to cleave RNA species encoding ICAM-1 , IL-5, relA, TNF-α, p210bcr-abl, or RSV proteins. In particular, applicant describes the selection and function of ribozymes capable of cleaving these RNAs and their use to reduce levels of ICAM-1 , IL-5, relA, TNF-α, p210 bor-abl or RSV proteins in various tissues to treat the diseases discussed herein. Such ribozymes are also useful for diagnostic uses.
Applicant indicates that these ribozymes are able to inhibit expression of ICAM-1 , IL-5, rel A, TNF-α, p21θbcr-abl. or RSV genes and that the catalytic activity of the ribozymes is required for their inhibitory effect. Those of ordinary skill in the art, will find that it is clear from the examples described that other ribozymes that cleave target ICAM-1 , IL-5, rel A, TNF-α, p210b cr-abl, or RSV encoding mRNAs may be readily designed and are within the invention.
These chemically or enzymatically synthesized RNA molecules contain substrate binding domains that bind to accessible regions of their target mRNAs. The RNA molecules also contain domains that catalyze the cleavage of RNA. Upon binding, the ribozymes cleave the target encoding mRNAs, preventing translation and protein accumulation. In the absence of the expression of the target gene, a therapeutic effect may be observed.
By "gene" is meant to refer to either the protein coding regions of the cognate mRNA, or any regulatory regions in the RNA which regulate synthesis of the protein or stability of the mRNA; the term also refers to those regions of an mRNA which encode the ORF of a cognate polypeptide product, and the proviral genome.
By "enzymatic RNA molecule" it is meant an RNA molecule which has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity which is active to specifically cleave RNA in that target. That is, the enzymatic RNA molecule is able to intermolecularly cleave RNA and thereby inactivate a target RNA molecule. This complementarity functions to allow sufficient hybridization of the enzymatic RNA molecule to the target RNA to allow the cleavage to occur. One hundred percent complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention. By "equivalent" RNA to a virus is meant to include those naturally occurring viral encoded RNA molecules associated with viral caused diseases in various animals, including humans, cats, simians, and other primates. These viral or viral-encoded RNAs have similar structures and equivalent genes to each other.
By "complementarity" it is meant a nucleaic acid that can form hydrogen bond(s) with other RNA sequence by either traditional WatsonCrick or other non-traditional types (for examplke, Hoogsteen type) of base-paired interactions.
In preferred embodiments of this invention, the enzymatic nucleic acid molecule is formed in a hammerhead or hairpin motif, but may also be formed in the motif of a hepatitis delta virus, group I intron or RNaseP RNA (in associateion with an RNA guide sequence) or Neurospora VS RNA. Examples of such hammerhead motifs are described by Rossi et al., 1992, Aids Research and Human Retroviruses , 8,183, of hairpin motifs by Hampel and Tritz, 1989 Biochemistry, 28, 4929, EP 0360257 and Hampel et al., 1990, Nucleic Acids Res. 18,299 and an example of the hepatitis delta virus motif is described by Perotta and Been, 1992 Biochemistry, 31 16 of the RNaseP motif by Guerrier-Takada et al., 1983 Cell, 35 849, cleavage of RNA. Upon binding, the ribozymes cleave the target encoding mRNAs, preventing translation and protein accumulation. In the absence of the expression of the target gene, a therapeutic effect may be observed.
By "gene" is meant to refer to either the protein coding regions of the cognate mRNA, or any regulatory regions in the RNA which regulate synthesis of the protein or stability of the mRNA; the term also refers to those regions of an mRNA which encode the ORF of a cognate polypeptide product, and the proviral genome.
By "enzymatic RNA molecule" it is meant an RNA molecule which has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity which is active to specifically cleave RNA in that target. That is, the enzymatic RNA molecule is able to intermolecularly cleave RNA and thereby inactivate a target RNA molecule. This complementarity functions to allow sufficient hybridization of the enzymatic RNA molecule to the target RNA to allow the cleavage to occur. One hundred percent complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention. By "equivalent" RNA to a virus is meant to include those naturally occurring viral encoded RNA molecules associated with viral caused diseases in various animals, including humans, cats, simians, and other primates. These viral or viral-encoded RNAs have similar structures and equivalent genes to each other.
By "complementarity" it is meant a nucleaic acid that can form hydrogen bond(s) with other RNA sequence by either traditional Watson-Crick or other non-traditional types (for examplke, Hoogsteen type) of base-paired interactions.
In preferred embodiments of this invention, the enzymatic nucleic acid molecule is formed in a hammerhead or hairpin motif, but may also be formed in the motif of a hepatitis delta virus, group I intron or RNaseP RNA (in associateion with an RNA guide sequence) or Neurospora VS RNA. Examples of such hammerhead motifs are described by Rossi et al., 1992, Aids Research and Human Retroviruses , 8,183, of hairpin motifs by Hampel and Tritz, 1989 Biochemistry, 28, 4929, EP 0360257 and Hampel et al., 1990, Nucleic Acids Res. 18,299 and an example of the hepatitis delta virus motif is described by Perotta and Been, 1992 Biochemistry, 31 16 of the RNaseP motif by Guerrier-Takada et al., 1983 Cell, 35 849, expressed in eukaryotic cells from the appropriate DNA or RNA vector. The activity of such ribozymes can be augmented by their release from the primary transcript by a second ribozyme (Draper et al., PCT W093/23569, and Sullivan et al., PCT WO94/02595, both hereby incorporated in their totality by reference herein; Ohkawa, J., et al., 1992, Nucleic Acids Symp. Ser. 27, 15-6; Taira, K. et al., Nucleic Acids Res., 19, 5125-30; Ventura, M., et al., 1993, Nucleic Acids Res., 21 , 3249-55, Chowrira et al., 1994 J. Biol. Chem., 269. 25856 ).
By "inhibit" is meant that the activity or level of ICAM-1 , Rel A, IL-5, TNF-α, p210bcr-abl or RSV encoding mRNA is reduced below that observed in the absense of the ribozyme, and preferably is below that level observed in the presence of an inactive RNA molecule able to bind to the same site on the mRNA, but unable to cleave that RNA.
Such ribozymes are useful for the prevention of the diseases and conditions discussed above, and any other diseases or conditions that are related to the level of ICAM-1 , IL-5, Rel A, TNF-α, p210bcr-abl or RSV protein or activity in a cell or tissue. By "related" is meant that the inhibition of ICAM-1 , IL-5, Rel A, TNF-α, P210bcr-abl or RSV mRNA translation, and thus reduction in the level of, ICAM-1 , IL-5, Rel A, TNF-α, p210bcl-abl or RSV proteins will relieve to some extent the symptoms of the disease or condition.
Ribozymes are added directly, or can be complexed with cationic lipids, packaged within liposomes, or otherwise delivered to target cells. The RNA or RNA complexes can be locally administered to relevant tissues through the use of a catheter, infusion pump or stent, with or without their incorporation in biopolymers. In preferred embodiments, the ribozymes have binding arms which are complementary to the sequences in Tables 2,3,6-9, 11 , 13, 15-23, 27, 28, 31 , 33, 34, 36 and 37.
Examples of such ribozymes are shown in Tables 4-8, 10, 12, 14-16, 19-22, 24, 26-28, 30, 32, 34 and 36-38. Examples of such ribozymes consist essentially of sequences defined in these Tables. By "consists essentially of" is meant that the active ribozyme contains an enzymatic center equivalent to those in the examples, and binding arms able to bind mRNA such that cleavage at the target site occurs. Other sequences may be present which do not interfere with such cleavage. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity. For example, stem-loop II sequence of hammerhead ribozymes listed in the above identified Tables can be altered (substitution, deletion, and/or insertion) to contain any sequences provided a minimum of two base-paired stem structure can form. Similarly, stem-loop IV sequence of hairpin ribozymes listed in the above identified Tables can be altered (substitution, deletion, and/or insertion) to contain any sequence, provided a minimum of two base-paired stem structure can form. The sequence listed in the above identified Tables may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables.
In another aspect of the invention, ribozymes that cleave target molecules and inhibit ICAM-1 , IL-5, Rel A, TNF-α, p210bcr-abl or RSV gene expression are expressed from transcription units inserted into DNA, RNA, or viral vectors. Another means of accumulating high concentrations of a ribozyme(s) within cells is to incorporate the ribozyme-encoding sequences into a DNA or RNA expression vector. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III). Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990 Proc. Natl. Acad. Sci. USA, 87, 6743-7; Gao and Huang 1993 Nucleic Acids Res., 21 2867-72; Lieber et al., 1993 Methods Enzymol., 217, 47-66; Zhou et al., 1990 Mol. Cell. Biol., 10, 4529-37). Several investigators have demonstrated that ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992 Antisense Res. Dev., 2, 3-15; Ojwang et al., 1992 Proc. Natl. Acad. Sci. USA, 90, 6340-4; L'Huiller et al., 1992 EMBO J. 11 , 4411-8; Lisziewicz et al., 1993 Proc. Natl. Acad. Sci. U.S.A., 90 8000-4). The above ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors).
Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims.
Description Of The Preferred Embodiments The drawings will first briefly be described. Drawings:
Figure 1 is a diagrammatic representation of the hammerhead ribozyme domain known in the art. Stem II can be≥ 2 base-pair long.
Figure 2(a) is a diagrammatic representation of the hammerhead ribozyme domain known in the art; Figure 2(b) is a diagrammatic representation of the hammerhead ribozyme as divided by Uhlenbeck (1987, Nature, 327, 596-600) into a substrate and enzyme portion; Figure 2(c) is a similar diagram showing the hammerhead divided by Haseloff and Gerlach (1988, Nature, 334, 585-591) into two portions; and Figure 2(d) is a similar diagram showing the hammerhead divided by Jeffries and Symons (1989, Nυcl. Acids. Res., 17, 1371-1371) into two portions.
Figure 3 is a diagrammatic representation of the general structure of a hairpin ribozyme. Helix 2 (H2) is provided with a least 4 base pairs (i.e., n is 1 ,2,3 or 4) and helix 5 can be optionally provided of length 2 or more bases (preferably 3-20 bases, i.e., m is from 1-20 or more). Helix 2 and helix 5 may be covalently linked by one or more bases (i.e., r is≥ 1 base).
Helix 1 , 4 or 5 may also be extended by 2 or more base pairs (e.g., 4-20 base pairs) to stabilize the ribozyme structure, and preferably is a protein binding site. In each instance, each N and N' independently is any normal or modified base and each dash represents a potential base-pairing interaction. These nucleotides may be modified at the sugar, base or phosphate. Complete base-pairing is not required in the helices, but is preferred. Helix 1 and 4 can be of any size (i.e., o and p is each independently from 0 to any number, e.g. 20) as long as some base-pairing is maintained. Essential bases are shown as specific bases in the structure, but those in the art will recognize that one or more may be modified chemically (abasic, base, sugar and/or phosphate modifications) or replaced with another base without significant effect. Helix 4 can be formed from two separate molecules, i.e., without a connecting loop. The connecting loop when present may be a ribonucleotide with or without modifications to its base, sugar or phosphate, "q" is≥ 2 bases. The connecting loop can also be replaced with a non-nucleotide linker molecule. H refers to bases A, U, or C. Y refers to pyrimidine bases. " " refers to a covalent bond.
Figure 4 is a representation of the general structure of the hepatitis delta virus ribozyme domain known in the art.
Figure 5 is a representation of the general structure of the self-cleaving VS RNA ribozyme domain.
Figure 6 is a diagrammatic representation of the genetic map of RSV strain A2. Figure 7 is a diagrammatic representation of the solid-phase synthesis of RNA.
Figure 8 is a diagrammatic representation of exocyclic amino protecting groups for nucleic acid synthesis.
Figure 9 is a diagrammatic representation of the deprotection of RNA. Figure 10 is a graphical representation of the cleavage of an RNA substrate by ribozymes synthesized, deprotected and purified using the improved methods described herein.
Figure 11 is a schematic representation of a two pot deprotection protocol. Base deprotection is carried out with aqueous methyl amine at 65°C for 10 min. The sample is dried in a speed-vac for 2-24 hours depending on the scale of RNA synthesis. Silyl protecting group at the 2'-hydroxyl position is removed by treating the sample with 1.4 M anhydrous HF at 65°C for 1.5 hours.
Figure 12 is a schematic representation of a one pot deprotection of RNA synthesized using RNA phosphoramidite chemistry. Anhydrous methyl amine is used to deprotect bases at 65°C for 15 min. The sample is allowed to cool for 10 min before adding TEA•3HF reagent, to the same pot, to remove protecting groups at the 2'-hydroxyI position. The deprotection is carried out for 1.5 hours.
Figs. 13a - b is a HPLC profile of a 36 nt long ribozyme, targeted to site B. The RNA is deprotected using either the two pot or the one pot deprotection protocol. The peaks corresponding to full-length RNA is indicated. The sequence for site B is CCUGGGCCAGGGAUUA
AUGGAGAUGCCCACU.
Figure 14 is a graph comparing RNA cleavage activity of ribozymes deprotected by two pot vs one pot deprotection protocols. Figure 15 is a schematic representation of an improved method of synthesizing RNA containing phosphorothioate linkages.
Figure 16 shows RNA cleavage reaction catalyzed by ribozymes containing phosphorothioate linkages. Hammerhead ribozyme targeted to site C is synthesized such that 4 nts at the 5' end contain phosphorothioate linkages. P=O refers to ribozyme without phosphorothioate linkages. P=S refers to ribozyme with phosphorothioate linkages. The sequence for site C is UCAUUUUGGCCAUCUC UUCCUUCAGGCGUGG.
Figure 17 is a schematic representation of synthesis of 2'-N-phtalimido-nucleoside phosphoramidite. Figure 18 is a diagrammatic representation of a prior art method for the solid-phase synthesis of RNA using silyl ethers, and the method of this invention using SEM as a 2'-protecting group.
Figure 19 is a diagrammatic representation of the synthesis of 2'- SEM-protected nucleosides and phosphoramidites useful for the synthesis of RNA. B is any nucleotide base as exemplified in the Figure, P is purine and I is inosine. Standard abbreviations are used throughout this application, well known to those in the art.
Figure 20 is a diagrammatic representation of a prior art method for deprotection of RNA using TBDMS protection of the 2'-hydroxyl group. Figure 21 is a diagrammatic representation of the deprotection of RNA having SEM protection of the 2'-hydroxyl group. Figure 22 is a representation of an HPLC chromatogram of a fully deprotected 10-mer of uridylic acid.
Figs. 23 - 25 are diagrammatic representations of hammerhead, hairpin or hepatitis delta virus ribozyme containing self-processing RNA transcript. Solid arrows indicate self-processing sites. Boxes indicate the sites of nucleotide substitution. Solid lines are drawn to show the binding sites of primers used in a primer-extension assay. Lower case letters indicate vector sequence present in the RNA when transcribed from a HindIII-linearized plasmid. (23) HH Cassette, transcript containing the hammerhead trans-acting ribozyme linked to a 3' cis-acting hammerhead ribozyme. The structure of the hammerhead ribozyme is based on phylogenetic and mutational analysis (reviewed by Symons, 1992 supra). The trans ribozyme domain extends from nucleotide 1 through 49. After 3'-end processing, the trans-ribozyme contains 2 non-ribozyme nucleotides (UC at positions 50 and 51) at its 3' end. The 3' processing ribozyme is comprised of nucleotides 44 through 96. Roman numerals I, II and III, indicate the three helices that contribute to the structure of the 3' cis-acting hammerhead ribozyme (Hertel et al., 1992 Nucleic Acids Res. 20, 3252). Substitution of G70 and A71 to U and G respectively, inactivates the hammerhead ribozyme (Ruffner et al., 1990 Biochemistry 29, 10695) and generates the HH(mutant) construct. (24) HP Cassette, transcript containing the hammerhead trans-acting ribozyme linked to a 3' cis-acting hairpin ribozyme. The structure of the hairpin ribozyme is based on phylogenetic and mutational analysis (Berzal-Herranz et al., 1993 EMBO. J 12, 2567). The trans-ribozyme domain extends from nucleotide 1 through 49. After 3'-end processing, the trans-ribozyme contains 5 non-ribozyme nucleotides (UGGCA at positions 50 to 54) at its 3' end. The 3' cis-acting ribozyme is comprised of nucleotides 50 through 115. The transcript named HP(GU) was constructed with a potential wobble base pair between G52 and U77; HP(GC) has a Watson-Crick base pair between G52 and C77. A shortened helix 1 (5 base pairs) and a stable tetraloop (GAAA) at the end of helix 1 was used to connect the substrate with the catalytic domain of the hairpin ribozyme (Feldstein & Bruening, 1993 Nucleic Acids Res. 21 , 1991 ; Altschuler et al., 1992 supra). (25) HDV Cassette, transcript containing the trans-acting hammerhead ribozyme linked to a 3' cis-acting hepatitis delta virus (HDV) ribozyme. The secondary structure of the HDV ribozyme is as proposed by Been and coworkers (Been et al., 1992 Biochemistry 31 , 11843). The trans-ribozyme domain extends from nucleotides 1 through 48. After 3'-end processing, the trans-ribozyme contains 2 non-ribozyme nucleotides (AA at positions 49 to 50) at its 3' end. The 3' cis-acting HDV ribozyme is comprised of nucleotides 50 through 114. Roman numerals I, II, III & IV, indicate the location of four helices within the 3' cis-acting HDV ribozyme (Perrota & Been, 1991 Nature 350, 434). The ΔHDV transcript contains a 31 nucleotide deletion in the HDV portion of the transcript (nucleotides 84 through 115 deleted). Fig. 26 is a schematic representation of a plasmid containing the insert encoding self-processing cassette. The figure is not drawn to scale.
Fig. 27 demonstrates the effect of 3' flanking sequences on RNA self-processing in vitro. H, Plasmid templates linearized with HindIII restriction enzyme. Transcripts from H templates contain four non-ribozyme nucleotides at the 3' end. N, Plasmid templates linearized with Ndel restriction enzyme. Transcripts from Ν templates contain 220 non-ribozyme nucleotides at the 3' end. R, Plasmid templates linearized with Rcal restriction enzyme. Transcripts from R templates contain 450 non-ribozyme nucleotides at the 3' end. Fig. 28 shows the effect of 3' flanking sequences on the transcleavage reaction catalyzed by a hammerhead ribozyme. A 622 nt internally-labeled RΝA (<10 nM) was incubated with ribozyme (1000 nM) under single turn-over conditions (Herschlag and Cech, 1990 Biochemistry 29, 10159). HH+2, HH+37, and HH+52 are trans-acting ribozymes produced by transcription from the HH, ΔHDV, and HH(mutant) constructs, respectively, and that contain 2, 37 and 52 extra nucleotides on the 3' end. The plot of the fraction of uncleaved substrate versus time was fit to a double exponential curve using the KaleidaGraph graphing program (Synergy Software, Reading, PA). A double exponential curve fit was used because the data points did not fall on a single exponential curve, presumably due to varying conformers of ribozyme and/or substrate RΝA.
Fig. 29 shows RΝA self-processing in OST7-1 cells. In vitro lanes contain full-length, unprocessed transcripts that were added to cellular lysates prior to RΝA extraction. These RΝAs were either pre-incubated with MgCl2 (+) or with DEPC-treated water (-) prior to being hybridized with 5' end-labeled primers. Cellular lanes contain total cellular RNA from cells transfected with one of the four self-processing constructs. Cellular RNA are probed for ribozyme expression using a sequence specific primer-extension assay. Solid arrows indicate the location of primer extension bands corresponding to Full-Length RNA and 3' Cleavage Products.
Figs. 30,31 are diagrammatic representations of self-processing cassettes that will release trans-acting ribozymes with defined, stable stemloop structures at the 5' and the 3' end following self-processing. 30, shows various permutations of a hammerhead self-processing cassette. 31 , shows various permutations of a hairpin self-processing cassette.
Figs. 32a-b Schematic representation of RNA polymerse III promoter structure. Arrow indicates the transcription start site and the direction of coding region. A, B and C, refer to consensus A, B and C box promoter sequences. I, refers to intermediate cis-acting promoter sequence. PSE, refers to proximal sequence element. DSE, refers to distal sequence element. ATF, refers to activating transcription factor binding element. ?, refers to cis-acting sequence element that has not been fully characterized. EBER, Epstein-Barr-virus-encoded-RNA. TATA is a box well known in the art. Figs. 33a-e Sequence of the primary tRNAi met and Δ3-5 transcripts.
The A and B box are internal promoter regions necessary for pol III transcription. Arrows indicate the sites of endogenous tRNA processing. The Δ3-5 transcript is a truncated version of tRNA wherein the sequence 3' of B box has been deleted (Adeniyi-Jones et al., 1984 supra). This modification renders the Δ 3-5 RNA resistant to endogenous tRNA processing.
Figure 34. Schematic representation of RNA structural motifs inserted into the Δ3-5 RNA. Δ3-5/HHI- a hammerhead (HHI) ribozyme was cloned at the 3' region of Δ3-5 RNA; S3- a stable stem-loop structure was incorporated at the 3' end of the Δ3-5/HHI chimera; S5- stable stem-loop structures were incorporated at the 5' and the 3' ends of Δ3-5/HHI ribozyme chimera; S35- sequence at the 3' end of the Δ3-5/HHI ribozyme chimera was altered to enable duplex formation between the 5' end and a complementary 3' region of the same RNA; S35Plus- in addition to structural alterations of S35, sequences were altered to facilitate additional duplex formation within the non-ribozyme sequence of the Δ3-5/HHI chimera.
Figures 35 and 36. Northern analysis to quantitate ribozyme expression in T cell lines transduced with Δ3-5 vectors. 35) Δ3-5/HHI and its variants were cloned individually into the DC retroviral vector (Sullenger et al., 1990 supra). Northern analysis of ribozyme chimeras expressed in MT-2 cells was performed. Total RNA was isolated from cells (Chomczynski & Sacchi, 1987 Analytical Biochemistry 162, 156-159), and transduced with various constructs described in Fig. 34. Northern analysis was carried out using standard protocols (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). Nomenclature is same as in Figure 34. This assay measures the level of expression from the type 2 pol III promoter. 36) Expression of S35 constructs in MT2 cells. S35 (+ribozyme), S35 construct containing HHI ribozyme. S35 (-ribozyme), S35 construct containing no ribozyme.
Figure 37. Ribozyme activity in total RNA extracted from transduced MT-2 cells. Total RNA was isolated from cells transduced with Δ3-5 constructs described in Figs. 35 and 36 In a standard ribozyme cleavage reaction, 5 μg total RNA and trace amounts of 5' terminus-labeled ribozyme target RNA were denatured separately by heating to 90°C for 2 min in the presence of 50 mM Tris-HCI, pH 7.5 and 10 mM MgCl2. RNAs were renatured by cooling the reaction mixture to 37°C for 10-15 min. Cleavage reaction was initiated by mixing the labeled substrate RNA and total cellular RNA at 37°C. The reaction was allowed to proceed for ~ 18h, following which the samples were resolved on a 20 % urea-polyacrylamide gel. Bands were visualized by autoradiography.
Figures 38 and 39. Ribozyme expression and activity levels in S35-transduced clonal CEM cell lines. 38) Northern analysis of S35-transduced clonal CEM cell lines. Standard curve was generated by spiking known concentrations of in vitro transcribed S5 RNA into total cellular RNA isolated from non-transduced CEM cells. Pool, contains RNA from pooled cells transduced with S35 construct. Pool (-G418 for 3 Mo), contains RNA from pooled cells that were initially selected for resistance to G418 and then grown in the absence of G418 for 3 months. Lanes A through N contain RNA from individual clones that were generated from the pooled cells transduced with S35 construct. tRNAi met, refers to the endogenous tRNA. S35, refers to the position of the ribozyme band. M, marker lane. 39) Activity levels in S35-transduced clonal CEM cell lines. RNA isolation and cleavage reactions were as described in Fig.37. Nomenclature is same as in Figs. 35 and 36 except, S, 5' terminus-labeled substrate RNA. P, 8 nt 5' terminus-labeled ribozyme-mediated RNA cleavage product.
Figures 40 and 41 are proposed secondary structures of S35 and S35 containing a desired RNA (HHI), respectively. The position of HHI ribozyme is indicated in figure 41. Intramolecular stem refers to the stem structure formed due to an intramolecular base-paired interaction between the 3' sequence and the complementary 5' terminus. The length of the stem ranges from 15-16 base-pairs. Location of the A and the B boxes are shown.
Figures 42 and 43 are proposed secondary structures of S35 plus and S35 plus containing HHI ribozyme.
Figures 44, 45, 46 and 47 are the nucleotide base sequences of S35, HHIS35, S35 Plus, and HHIS35 Plus respectively.
Figs. 48a-b is a general formula for pol III RNA of this invention.
Figure 49 is a digrammatic representation of 5T construct. In this construct the desired RNA is located 3' of the intramolecular stem.
Figures 50 and 51 contain proposed secondary structures of 5T construct alone and 5T contruct containing a desired RNA (HHI ribozyme) respectively.
Figure 52 is a diagrammatic representation of TRZ-tRNA chimeras. The site of desired RNA insertion is indicated.
Figure 53 shows the general structure of HHITRZ-A ribozyme chimera. A hammerhead ribozyme targeted to site I is inserted into the stem II region of TRZ-tRNA chimera.
Figure 54 shows the general structure of HPITRZ-A ribozyme chimera. A hairpin ribozyme targeted to site I is cloned into the indicated region of TRZ-tRNA chimera. Figure 55 shows a comparison of RNA cleavage activity of HHITRZ-A, HHITRZ-B and a chemically synthesized HHI hammerhead ribozymes.
Figure 56 shows expression of ribozymes in T cell lines that are stably transduced with viral vectors. M, markers; lane 1 , non-transduced CEM cells; lanes 2 and 3, MT2 and CEM cells transduced with retroviral vectors; lanes 4 and 5, MT2 and CEM cells transduced with AAV vectors.
Figs. 57a-b Schematic diagram of adeno-associated virus and adenovirues vectors for ribozyme delivery. Both vectors utilize one or more ribozyme encoding transcription units (RZ) based on RNA polymerase II or RNA polymerase III promoters. A. Diagram of an AAV-based vector containing minimal AAV sequences comprising the inverted terminal repeats (ITR) at each end of the vector genome, an optional selectable marker (Neo) driven by an exogenous promoter (Pro), a ribozyme transcription unit, and sufficient additional sequences (stuffer) to maintain a vector length suitable for efficient packaging. B. Diagram of ribozyme expressing adenovirus vectors containing deletions of one or more wild type adenoviorus coding regions (cross-hatched boxes marked as E1 , pIX, E3, and E4), and insertion of the ribozyme transcription unit at any or several of those regions of deletions. Fig. 58 is a graph showing the effect of arm length variation on the activity of ligated hammerhead (HH) ribozymes. Nomenclature 5/5, 6/6, 7/7, 8/8 and so on refers to the number of base-pairs being formed between the ribozyme and the target. For example, 5/8 means that the HH ribozyme forms 5 bp on the 5' side and 8 bp on the 3' side of the cleavage site for a total of 13 bp. -ΔG refers to the free energy of binding calculated for base-paired interactions between the ribozyme and the substrate RNA (Turner and Sugimoto, 1988 Ann. Rev. Biophys. Chem. 17, 167). RPI A is a HH ribozyme with 6/6 binding arms.
Figs. 59 and 60 and 61 show cleavage of long substrate (622 nt) by ligated HH ribozymes.
Fig. 62 is a diagrammatic representation of a hammerhead ribozyme (HH-H) targeted against a site termed H. Variants of HH-H are also shown that contain either a 2 base-paired stem II (HH-H1 and HH-H2) or a 3 base- paired stem II (HH-H3 and HH-H4). Figs. 63 and 64 show RNA cleavage activity of HH-I and its variants (see Fig.62). 63) cleavage of matched substrate RNA (15 nt). 64) cleavage of long substrate RNA (613 nt).
Figs. 65a-b is a schematic representation of a method of this invention to synthesize a full length hairpin ribozyme. No splint strand is required for ligation but rather the two fragments hybridize together at helix 4 prior to ligation. The only prerequisite is that the 3' fragment is phosphorylated at its 5' end and that the 3' end of the 5' fragment have a hydroxyl group. The hairpin ribozyme is targeted against site J. H1 and H2 are intermolecular helices formed between the ribozyme and the substrate. H3 and H4 are intramolecular helices formed within the hairpin ribozyme motif. Arrow indicates the cleavage site.
Fig. 66 shows RNA cleavage activity of ligated hairpin ribozymes targeted against site J. Figs. 67a-b is a diagrammatic representation of a Site K Hairpin
Ribozyme (HP-K) showing the proposed secondary structure of the hairpin ribozyme•substrate complex as described in the art (Berzal-Herranz et al., 1993 EMBO. J.12, 2567). The ribozyme has been assembled from two fragments (bimolecular ribozyme; Chowrira and Burke, 1992 Nucleic Acids Res. 20, 2835); #H1 and H2 represent intermolecular helix formation between the ribozyme and the substrate. H3 and H4 represent intramolecular helix formation within the ribozyme (intermolecular helix in the case of bimolecular ribozyme). Left panel (HP-K1) indicates 4 base-paired helix 2 and the right panel (HP-K2) indicates 6 base-paired helix 2. Arrow indicates the site of RNA cleavage. All the ribozymes discussed herein were chemically synthesized by solid phase synthesis using RNA phosphoramadite chemistry, unless otherwise indicated. Those skilled in the art will recognize that these ribozymes could also be made transcriptionally in vitro and in vivo. Figure 68 is a graph showing RNA cleavage by hairpin ribozymes targeted to site K. A plot of fraction of the target RNA uncleaved (fraction uncleaved) as a function of time is shown. HP-K2 (6 bp helix 2) cleaves a 422 target RNA to a greater extent than the HP-K1 (4 bp helix 2). To make internally-labeled substrate RNA for trans-ribozyme cleavage reactions, a 422 nt region (containing hairpin site A) was synthesized by PCR using primers that place the T7 RNA promoter upstream of the amplified sequence. Target RNA was transcribed in a standard transcription buffer in the presence of [α-32P]CTP (Chowrira & Burke, 1991 supra). The reaction mixture was treated with 15 units of ribonuclease-free DNasel, extracted with phenol followed chloroform:isoamyl alcohol (25:1), precipitated with isopropanol and washed with 70% ethanol. The dried pellet was resuspended in 20 μl DEPC-treated water and stored at -20°C.
Unlabeled ribozyme (1μM) and internally labeled 422 nt substrate RNA (<10 nM) were denatured and renatured separately in a standard cleavage buffer (containing 50 mM Tris·HCl pH 7.5 and 10 mM MgCl2) by heating to 90°C for 2 min. and slow cooling to 37°C for 10 min. The reaction was initiated by mixing the ribozyme and substrate mixtures and incubating at 37°C. Aliquots of 5 μl were taken at regular time intervals, quenched by adding an equal volume of 2X formamide gel loading buffer and frozen on dry ice. The samples were resolved on 5% polyacrylamide sequencing gel and results were quantitatively analyzed by radioanalytic imaging of gels with a Phosphorlmager (Molecular Dynamics, Sunnyvale, CA).
Figs. 69a-b is the Site L Hairpin Ribozyme (HP-L) showing proposed secondary structure of the hairpin ribozyme•substrate complex. The ribozyme was assembled from two fragments as described above. The nomenclature is the same as above.
Figure 70 shows RNA cleavage by hairpin ribozymes targeted to site L. A. plot of fraction of the target RNA uncleaved (fraction uncleaved) as a function of time is shown. HP-L2 (6 bp helix 2) cleaves a 2 KB target RNA to a greater extent than the HP-L1 (4 bp helix 2). To make internally- labeled substrate RNA for trans- ribozyme cleavage reactions, a 2 kB region (containing hairpin site L) was synthesized by PCR using primers that place the T7 RNA promoter upstream of the amplified sequence. The cleavage reactions were carried out as described above. Figs. 71 a-b shows a Site M Hairpin Ribozyme (HP-M) with the proposed secondary structure of the hairpin ribozyme•substrate complex. The ribozyme was assembled from two fragments as described above.
Figure 72 is a graph showing RNA cleavage by hairpin ribozymes targeted to site M. The ribozymes were tested at both 20°C and at 26°C. To make internally-labeled substrate RNA for trans-ribozyme cleavage reactions, a 1.9 KB region (containing hairpin site M) was synthesized by PCR using primers that place the 77 RNA promoter upstream of the amplified sequence. Cleavage reactions were carried out as described above except that 20°C and at 26°C temperatures were used.
Figs. 73a-d shows various structural modifications of the present invention. A) Hairpin ribozyme lacking helix 5. Nomenclature is same as described under figure 3. B) Hairpin ribozyme lacking helix 4 and helix 5. Helix 4 is replaced by a nucleotide loop wherein q is ≥ 2 bases. Nomenclature is same as described under figure 3. C) Hairpin ribozyme lacking helix 5. Helix 4 loop is replaced by a linker 103"L", wherein L is a non-nucleotide linker molecule (Benseler et al., 1993 J. Am. Chem. Soc. 115, 8483; Jennings et al., WO 94/13688). Nomenclature is same as described under figure 3. D) Hairpin ribozyme lacking helix 4 and helix 5. Helix 4 is replaced by non-nucleotide linker molecule "L" (Benseler et al., 1993 supra; Jennings et al., supra). Nomenclature is same as described under figure 3.
Figs. 74a-b shows Hairpin ribozymes containing nucleotide spacer region "s" at the indicated location, wherein s is ≥ 1 base. Hairpin ribozymes containing spacer region, can be synthesized as one fragment or can be assembled from multiple fragments. Nomenclature is same as described under figure 3.
Figs. 75a-e shows the structures of the 5'-C-alkyl-modified nucleotides. R1 is as defined above. R is OH, H, O-protecting group, NH, or any group described by the publications discussed above, and those described below. B is as defined in the Figure or any other equivalent nucleotide base. CE is cyanoethyl, DMT is a standard blocking group. Other abbreviations are standard in the art. Figure 76 is a diagrammatic representation of the synthesis of 5'-C-alkyl-D-allose nucleosides and their phosphoramidites.
Figure 77 is a diagrammatic representation of the synthesis of 5'-C-alkyl-L-talose nucleosides and their phosphoramidites. Figure 78 is a diagrammatic representation of hammerhead ribozymes targeted to site O containing 5'-C-methyl-L-talo modifications at various positions.
Figure 79 shows RNA cleavage activity of HH-O ribozymes. Fraction of target RNA uncleaved as a function of time is shown. Figure 80 is a diagrammatic representation of a position numbered hammerhead ribozyme (according to Hertel et al. Nucleic Acids Res. 1 992, 20, 3252) showing specific substitutions.
Figs. 81 a-j shows the structures of various 2'-alkyl modified nucleotides which exemplify those of this invention. R groups are alkyl groups, Z is a protecting group.
Figure 82 is a diagrammatic representation of the synthesis of 2'-C-allyl uridine and cytidine.
Figure 83 is a diagrammatic representation of the synthesis of 2'-C-methylene and 2'-C-difluoromethylene uridine. Figure 84 is a diagrammatic representation of the synthesis of 2'-C-methylene and 2'-C-difluoromethylene cytidine.
Figure 85 is a diagrammatic representation of the synthesis of 2'-C-methylene and 2'-C-difluoromethylene adenosine.
Figure 86 is a diagrammatic representation of the synthesis of 2'-C-carboxymethylidine uridine, 2'-C-methoxycarboxymethylidine uridine and derivatized amidites thereof. X is CH3 or alkyl as discussed above, or another substituent.
Figure 87 is a diagrammatic representation of a synthesis of nucleoside 5'-deoxy-5'-difluoromethylphosphonates. Figure 88 is a diagrammatic representation of the synthesis of nucleoside 5'-deoxy-5'-difluoromethylphosphonate 3'-phosphoramidites, dimers and solid supported dimers.
Figure 89 is a diagrammatic representation of the synthesis of nucleoside 5'-deoxy-5'-difluoromethylene triphosphates.
Figures 90 and 91 are diagrammatic representations of the synthesis of 3'-deoxy-3'-difluoromethylphosphonates and dimers.
Figure 92 is a schematic representation of synthesizing RNA phosphoramidite of a nucleotide containing a 2'-hydroxyl group modification of the present invention.
Figs. 93a-b describes a method for deprotection of oligonucleotides containing a 2'-hydroxyl group modification of the present invention.
Figure 94 is a diagrammatic representation of a hammerhead ribozyme targeted to site N. Positions of 2'-hydroxyl group substitution is indicated.
Figure 95 shows RNA cleavage activity of ribozymes containing a 2'-hydroxyl group modification of the present invention. All RNA, represents hammerhead ribozyme (HHN) with no 2'-hydroxyl group modifications. U7-ala, represents HHN ribozyme containing 2'-NH-alanine modification at the U7 position. U4/U7-ala, represents HHA containing 2'-NH-alanine modifications at U4 and U7 positions. U4 lys, represents HHA containing 2'-NH-lysine modification at U4 position. U7 lys, represents HHA containing 2'-NH-lysine modification at U7 position. U4/U7-lys, represents HHN containing 2'-NH-lysine modification at U4 and U7 positions. Figures 96 and 97 are schematic representations of synthesizing
(solid-phase synthesis) 3' ends of RNA with modification of the present invention. B, refers to either a base, modified base or an H.
Figure 98 and 99 are schematic representations of synthesizing (solid-phase synthesis) 5' ends of RNA with modification of the present invention. B, refers to either a base, modified base or an H.
Figures 100 and 101 are general schematic representations of the invention. Fig. 102a-d is a schematic representation of a method of the invention.
Fig. 103 is a graph of the results of the experiment diagrammed in figure 104.
Figure 104 is a diagrammatic representation of a fusion mRNA used in the experiment diagrammed in Fig. 102.
Figure 105 is a diagrammatic representation of a method for selection of useful ribozymes of this invention.
Figure 106 generally shows R-loop formation, and an R-loop complex. In addition, it indicates the location at which ligands can be provided to target the R-loop complex to cells using at least three different procedures, such as ligand receptor interaction, lipid or calcium phosphate mediated delivery, or electroporation.
Figure 107 shows a method for use of self-processing ribozymes to generate therapeutic ribozymes of unit length. This method is essentially described by Draper et al., PCT WO 93/23509.
Figure 108 shows a method of linking ligands like folate, carbohydrate or peptides to R-loop forming RNA.
Ribozymes of this invention block to some extent ICAM-1 , IL-5, rel A, TNF-α, p210bcr-abI, or RSV genes expression and can be used to treat diseases or diagnose such diseases. Ribozymes will be delivered to cells in culture and to tissues in animal models. Ribozyme cleavage of ICAM-1 , II-5, rel A, TNF-α ,p210bcr-abl, or RSV mRNA in these systems may prevent or alleviate disease symptoms or conditions.
I. Target sites Targets for useful ribozymes can be determined as disclosed in
Draper et al PCT WO93/23509, Sullivan et al., PCT WO94/02595 as well as by Draper et al., PCT/US94/13129 and hereby incorporated by reference herein in totality. Rather than repeat the guidance provided in those documents here, below are provided specific examples of such methods, not limiting to those in the art. Ribozymes to such targets are designed as described in those applications and synthesized to be tested in vitro and in vivo, as also described. Such ribozymes can also be optimized and delivered as described therein. While specific examples to animal and human RNA are provided, those in the art will recognize that the equivalent human RNA targets described can be used as described below. Thus, the same target may be used, but binding arms suitable for targeting human RNA sequences are present in the ribozyme. Such targets may also be selected as described below.
It must be established that the sites predicted by the computer-based RNA folding algorithm correspond to potential cleavage sites. Hammerhead or hairpin ribozymes are designed that could bind and are individually analyzed by computer folding (Jaeger et al., 1989 Proc. Natl. Acad. Sci., USA, 86 7706-7710) to assess whether the ribozyme sequences fold into the appropriate secondary structure. Those ribozymes with unfavorable intramolecular interactions between the binding arms and the catalytic core are eliminated from consideration. Varying binding arm lengths can be chosen to optimize activity. Generally, at least 5 bases on each arm are able to bind to, or otherwise interact with, the target RNA. mRNA is screened for accessible cleavage sites by the method described generally in Draper et al., PCT WO93/23569 hereby incorporated by reference herein. Briefly, DNA oligonucleotides representing potential hammerhead or hairpin ribozyme cleavage sites are synthesized. A polymerase chain reaction is used to generate a substrate for T7 RNA polymerase transcription from cDNA clones. Labeled RNA transcripts are synthesized in vitro from DNA templates. The oligonucleotides and the labeled trascripts are annealed, RNaseH is added and the mixtures are incubated for the designated times at 37°C. Reactions are stopped and RNA separated on sequencing polyacrylamide gels. The percentage of the substrate cleaved is determined by autoradiographic quantitation using a phosphor imaging system. From these data, hammerhead or hairpin ribozynme sites are chosen as the most accessible.
Ribozymes of the hammerhead or hairpin motif are designed to anneal to various sites in the mRNA message. The binding arms are complementary to the target site sequences desribed above. The ribozymes are chemically synthesized. The method of synthesis used follows the procedure for normal RNA synthesis as described in Usman et al., 1987 J. Am. Chem. Soc., 109, 7845 and in Scaringe et al., 1990 Nucleic Acids Res., 18, 5433 and made use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, phosphoramidites at the 3'-end. The average stepwise coupling yeilds are >98%. Inactive ribozymes are synthesized by substituting a U for G5 and a U for A 14 (numbering from Hertel et al., 1992 Nucleic Acids Res., 20, 3252). Hairpin ribozymes are synthesized in two parts and annealed to reconstruct the active ribozyme (Chowrira and Burke, 1992 Nucleic Acids Res., 20, 2835-2840). Ribozymes are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbach, 1989, Methods Enzymol, 180, 51). All ribozymes are modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-O-methyl, 2'H (for a review see Usman and Cedergren, 1992 TIBS 17,34). Ribozymes are purified by gel electrophoresis using heneral methods or are purified by high pressure liquid chromatography and are resuspended in water.
Example 1 : ICAM-1
Ribozymes that cleave ICAM-1 mRNA represent a novel therapeutic approach to inflammatory or autoimmune disorders. ICAM-1 function can be blocked therapeutically using monoclonal antibodies. Ribozymes have the advantage of being generally immunologically inert, whereas significant neutralizing anti-IgG responses can be observed with some monoclonal antibody treatments.
The following is a brief description of the physiological role of ICAM-1.
The discussion is not meant to be complete and is provided only for understanding of the invention that follows. This summary is not an admission that any of the work described below is prior art to the claimed invention.
Intercellular adhesion molecule-1 (ICAM-1) is a cell surface protein whose expression is induced by inflammatory mediators. ICAM-1 is required for adhesion of leukocytes to endothelial cells and for several immunological functions including antigen presentation, immunoglobulin production and cytotoxic cell activity. Blocking ICAM-1 function prevents immune cell recognition and activity during transplant rejection and in animal models of rheumatoid arthritis, asthma and reperfusion injury. Cell-cell adhesion plays a pivotal role in inflammatory and immune responses (Springer et al., 1987 Ann. Rev. Immunol. 5, 223-252). Cell adhesion is required for leukocytes to bind to and migrate through vascular endothelial cells. In addition, cell-cell adhesion is required for antigen presentation to T cells, for B cell induction by T cells, as well as for the cytotoxicity activity of T cells, NK cells, monocytes or granulocytes. Intercellular adhesion molecule-1 (ICAM-1) is a 110 kilodalton member of the immunoglobulin superfamily that is involved in all of these cell-cell interactions (Simmons et al., 1988 Nature (London) 331, 624-627). ICAM-1 is expressed on only a limited number of cells and at low levels in the absence of stimulation (Dustin et al., 1986 J. Immunol. 137, 245-254). Upon treatment with a number of inflammatory mediators (lipopolysaccharide, γ-interferon, tumor necrosis factor-α, or interleukin-1), a variety of cell types (endothelial, epithelial, fibroblastic and hematopoietic cells) in a variety of tissues express high levels of ICAM-1 on their surface (Sringer et. al. supra; Dustin et al., supra; and Rothlein et al., 1988 J. Immunol. 141 , 1665-1669). Induction occurs via increased transcription of ICAM-1 mRNA (Simmons et al., supra). Elevated expression is detectable after 4 hours and peaks after 16 - 24 hours of induction. ICAM-1 induction is critical for a number of inflammatory and immune responses. In vitro, antibodies to ICAM-1 block adhesion of leukocytes to cytokine-activated endothelial cells (Boyd,1988 Proc. Natl. Acad. Sci. USA 85, 3095-3099; Dustin and Springer, 1988 J. Cell Biol. 107, 321-331 ). Thus, ICAM-1 expression may be required for the extravasation of immune cells to sites of inflammation. Antibodies to ICAM-1 also block T cell killing, mixed lymphocyte reactions, and T cell-mediated B cell differentiation, suggesting that ICAM-1 is required for these cognate cell interactions (Boyd et al., supra). The importance of ICAM-1 in antigen presentation is underscored by the inability of ICAM-1 defective murine B cell mutants to stimulate antigen-dependent T cell proliferation (Dang et al., 1990 J. Immunol. 144, 4082-4091). Conversely, murine L cells require transfection with human ICAM-1 in addition to HLA-DR in order to present antigen to human T cells (Altmann et al., 1989 Nature (London) 338, 512-514). In summary, evidence in vitro indicates that ICAM-1 is required for cell-cell interactions critical to inflammatory responses, cellular immune responses, and humoral antibody responses. By engineering ribozyme motifs we have designed several ribozymes directed against ICAM-1 mRNA sequences. These have been synthesized with modifications that improve their nuclease resistance. These ribozymes cleave ICAM-1 target sequences in vitro. The sequence of human, rat and mouse ICAM-1 mRNA can be screened for accessible sites using a compter folding algorithm. Regions of the mRNA that did not form secondary folding structures and that contain potential hammerhead or hairpin ribozyme cleavage sites can be identified. These sites are shown in Tables 2, 3, and 6-9. (All sequences are 5' to 3' in the tables) While rat, mouse and human sequences can be screened and ribozymes thereafter designed, the human targeted sequences are of most utility.
The sequences of the chemically synthesized ribozymes useful in this study are shown in Tables 4 - 8 and 10. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity and may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables. The ribozymes will be tested for function in vivo by exogenous delivery to human umbilical vein endothelial cells (HUVEC). Ribozymes will be delivered by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA or RNA vectors described above. Cytokine-induced ICAM-1 expression will be monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. ICAM-1 mRNA levels will be assessed by Northern, by RNAse protection, by primer extension or by quantitative RT-PCR analysis. Ribozymes that block the induction of ICAM-1 protein and mRNA by more than 90% will be identified. As disclosed by Sullivan et al., PCT WO94/02595, incorporated by reference herein, ribozymes and/or genes encoding them will be locally delivered to transplant tissue ex vivo in animal models. Expression of the ribozyme will be monitored by its ability to block ex vivo induction of ICAM-1 mRNA and protein. The effect of the anti-ICAM-1 ribozymes on graft rejection will then be assessed. Similarly, ribozymes will be introduced into joints of mice with collagen-induced arthritis or rabbits with Streptococcal cell wall-induced arthritis. Liposome delivery, cationic lipid delivery, or adeno-associated virus vector delivery can be used. One dose
(or a few infrequent doses) of a stable anti-ICAM-1 ribozyme or a gene construct that constitutively expresses the ribozyme may abrogate inflammatory and immune responses in these diseases.
Uses
ICAM-1 plays a central role in immune cell recognition and function.
Ribozyme inhibition of ICAM-1 expression can reduce transplant rejection and alleviate symptoms in patients with rheumatoid arthritis, asthma or other acute and chronic inflammatory disorders. We have engineered several ribozymes that cleave ICAM-1 mRNA. Ribozymes that efficiently inhibit ICAM-1 expression in cells can be readily found and their activity measured with regard to their ability to block transplant rejection and arthritis symptoms in animal models. These anti-ICAM-1 ribozymes represent a novel therapeutic for the treatment of immunological or inflammatory disorders.
The therapeutic utility of reduction of activity of ICAM-1 function is evident in the following disease targets. The noted references indicate the role of ICAM-1 and the therapeutic potential of ribozymes described herein.
Thus, these targets can be therapeutically treated with agents that reduce
ICAM-1 expression or function. These diseases and the studies that support a critical role for ICAM-1 in their pathology are listed below. This list is not meant to be complete and those in the art will recognize further conditions and diseases that can be effectively treated using ribozymes of the present invention.
• Transplant rejection
ICAM-1 is expressed on venules and capillaries of human cardiac biopsies with histological evidence of graft rejection (Briscoe et al., 1991 Transplantation 51 , 537-539).
Antibody to ICAM-1 blocks renal (Cosimi et al., 1990 J. Immunol. 144, 4604- 4612) and cardiac (Flavin et al., 1991 Transplant. Proc. 23, 533-534) graft rejection in primates. A Phase I clinical trial of a monoclonal anti-ICAM-1 antibody showed significant reduction in rejection and a significant increase in graft function in human kidney transplant patients (Haug, et al., 1993 Transplantation 55, 766-72).
• Rheumatoid arthritis ICAM-1 overexpression is seen on synovial fibroblasts, endothelial cells, macrophages, and some lymphocytes (Chin et al., 1990 Arthritis Rheum 33, 1776-86; Koch et al., 1991 Lab Invest 64, 313-20).
Soluble ICAM-1 levels correlate with disease severity (Mason et al., 1993 Arthritis Rheum 36, 519-27). Anti-ICAM antibody inhibits collagen-induced arthritis in mice (Kakimoto et al.,
1992 Cell Immunol 142, 326-37).
Anti-ICAM antibody inhibits adjuvant-induced arthritis in rats (ligo et al., 1991 J Immunol 147, 4167-71).
• Myocardial ischemia, stroke, and reperfusion injury Anti-ICAM-1 antibody blocks adherence of neutrophils to anoxic endothelial cells (Yoshida et al., 1992 Am J Physiol 262, H1891-8).
Anti-ICAM-1 antibody reduces neurological damage in a rabbit model of cerebral stroke (Bowes et al., 1993 Exp Neurol 119, 215-9).
Anti-ICAM-1 antibody protects against reperfusion injury in a cat model of myocardial ischemia (Ma et al., 1992 Circulation 86, 937-46).
• Asthma
Antibody to ICAM-1 partially blocks eosinophil adhesion to endothelial cells and is overexpressed on inflamed airway endothelium and epithelium in vivo (Wegner et al., 1990 Science 247, 456-9). In a primate model of asthma, anti-ICAM-1 antibody blocks airway eosinophilia
(Wegneret al., supra) and prevents the resurgence of airway inflammation and hyper-responsiveness after dexamethosone treatment (Gundel et al., 1992 Clin Exp Allergy 22, 569-75).
• Psoriasis Surface ICAM-1 and a clipped, soluble version of ICAM-1 is expressed in psoriatic lesions and expression correlates with inflammation (Kellner et al., 1991 Br J Dermatol 125, 211-6; Griffiths 1989 J Am Acad Dermatol 20, 617-29; Schopf et al., 1993 Br J Dermatol 128, 34-7). Anti-ICAM antibody blocks keratinocyte antigen presentation to T cells
(Nickoloff et al., 1993 J Immunol 150, 2148-59 ).
• Kawasaki disease
Surface ICAM-1 expression correlates with the disease and is reduced by effective immunoglobulin treatment (Leung, et al., 1989 Lancet 2, 1298-302). Soluble ICAM levels are elevated in Kawasaki disease patients; particularly high levels are observed in patients with coronary artery lesions (Furukawa et al., 1992 Arthritis Rheum 35, 672-7; Tsuji, 1992 Arerugi 41 , 1507-14).
Circulating LFA-1+ T cells are depleted (presumably due to ICAM-1 mediated extravasation) in Kawasaki disease patients (Furukawa et al., 1993 Scand J Immunol 37, 377-80).
Example 2: IL-5
Ribozymes that cleave IL-5 mRNA represent a novel therapeutic approach to inflammatory disorders like asthma. The invention features use of ribozymes to treat chronic asthma, e.g., by inhibiting the synthesis of IL-5 in lymphocytes and preventing the recruitment and activation of eosinophils.
A number of cytokines besides IL-5 may also be involved in the activation of inflammation in asthmatic patients, including platelet activating factor, IL-1 , IL-3, IL-4, GM-CSF, TNF-α, gamma interferon, VCAM, ILAM-1 , ELAM-1 and NF-κB. In addition to these molecules, it is appreciated that any cellular receptors which mediate the activities of the cytokines are also good targets for intervention in inflammatory diseases. These targets include, but are not limited to, the IL-1 R and TNF-αR on keratinocytes, epithelial and endothelial cells in airways. Recent data suggest that certain neuropeptides may play a role in asthmatic symptoms. These peptides include substance P, neurokinin A and calcitonin-gene-related peptides.
These target genes may have more general roles in inflammatory diseases, but are currently assumed to have a role only in asthma. Ribozymes of this invention block to some extent IL-5 expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of asthma (Clutterbuck et al., 1989 supra: Garssen et al., 1991 Am. Rev. Respir. Dis. 144, 931-938; Larsen et al., 1992 J. Clin. Invest. 89, 747-752; Mauser et al., 1993 supra). Ribozyme cleavage of IL-5 mRNA in these systems may prevent inflammatory cell function and alleviate disease symptoms.
The sequence of human and mouse IL-5 mRNA were screened for accessible sites using a computer folding algorithm. Potential hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 11 , 13, and 14, 15. (All sequences are 5' to 3' in the tables.) While mouse and human sequences can be screened and ribozymes thereafter designed, the human targeted sequences are of most utility. However, mouse targeted ribozymes are useful to test efficacy of action of the ribozyme prior to testing in humans. The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme. (In Table 12, lower case letters indicate positions that are not conserved between the Human and the Mouse IL-5 sequences.) The sequences of the chemically synthesized ribozymes useful in this study are shown in Tables 12, 14 - 16. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity. For example, stem loop II sequence of hammerhead ribozymes listed in Tables 12 and 14 (5'-GGCCGAAAGGCC-3") can be altered (substitution, deletion and/or insertion) to contain any sequence provided, a minimum of two base-paired stem structure can form. Similarly, stem-loop IV sequence of hairpin ribozymes listed in Tables 15 and 16 (5'-CACGUUGUG-3') can be altered (substitution, deletion and/or insertion) to contain any sequence provided, a minimum of two base-paired stem structure can form. The sequences listed in Tables 12, 14 - 16 may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables. By engineering ribozyme motifs we have designed several ribozymes directed against IL-5 mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave IL-5 target sequences in vitro is evaluated.
The ribozymes will be tested for function in vivo by analyzing IL-5 expression levels. Ribozymes will be delivered to cells by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA or RNA vectors. IL-5 expression will be monitored by biological assays, ELISA, by indirect immunofluoresence, and/or by FACS analysis. IL-5 mRNA levels will be assessed by Northern analysis, RNAse protection or primer extension analysis or quantitative RT-PCR. Ribozymes that block the induction of IL-5 activity and/or IL-5 mRNA by more than 90% will be identified.
Uses
Interleukin 5 (IL-5), a cytokine produced by CD4+ T helper cells and mast cells, was originally termed B cell growth factor II (reviewed by Takatsu et al., 1988 Immunol. Rev. 102, 107). It stimulates proliferation of activated B cells and induces production of IgM and IgA. IL-5 plays a major role in eosinophil function by promoting differentiation (Clutterbuck et al., 1989 Blood 73, 1504-12), vascular adhesion (Walsh et al., 1990 Immunology 71 , 258-65) and in vitro survival of eosinophils (Lopez et al., 1988 J. Exp. Med. 167, 219-24). This cytokine also enhances histamine release from basophils (Hirai et al., 1990 J. Exp. Med. 172, 1525-8). The following summaries of clinical results support the selection of IL-5 as a primary target for the treatment of asthma:
Several studies have shown a direct correlation between the number of activated T cells and the number of eosinophils from asthmatic patients vs. normal patients (Oehling et al., 1992 J. Investig. Allergol. Clin. Immunol. 2, 295-9). Patients with either allergic asthma or intrinsic asthma were treated with corticosteroids. The bronchoalveolar lavage was monitored for eosinophils, activated T helper cells and recovery of pulmonary function over a 28 to 30 day period. The number of eosinophils and activated T helper cells decreased progressively with subsequent improvement in pulmonary function compared to intrinsic asthma patients with no corticosteroid treatment.
Bronchoalveolar lavage cells were screened for production of cytokines using in situ hybridization for mRNA. In situ hybridization signals were detected for IL-2, IL-3, IL-4, IL-5 and GM-CSF. Upregulation of mRNA was observed for IL-4, IL-5 and GM-CSF (Robinson et al., 1993 J. Allergy Clin. Immunol. 92, 313-24). Another study showed that upregulation of IL-5 transcripts from allergen challenged vs. saline challenged asthmatic patients (Krishnaswamy et al., 1993 Am. J. Respir. Cell. Mol. Biol. 9, 279-86).
An 18 patient study was performed to determine a mechanism of action for corticosteroid improvement of asthma symptoms. Improvement was monitored by methacholine responsiveness. A correlation was observed between the methacholine responsiveness, a reduction in the number of eosinophils, a reduction in the number of cells expressing IL-4 and IL-5 mRNA and an increase in number of cells expressing interferon-gamma.
Bronchial biopsies from 15 patients were analyzed 24 hours after allergen challenge (Bentley et al., 1993 Am. J. Respir. Cell. Mol. Biol. 8, 35-42). Increased numbers of eosinophils and IL-2 receptor positive cells were found in the biopsies. No differences in the numbers of total leukocytes, T lymphocytes, elastase-positive neutrophils, macrophages or mast cell subtypes were observed. The number of cells expressing IL-5 and GM-CSF mRNA significantly increased.
In another patient study, the eosinophil phenotype was the same for asthmatic patients and normal individuals. However, eosinophils from asthmatic patients had greater leukotriene C4 producing capacity and migration capacity. There were elevated levels of IL-3, IL-5 and GM-CSF in the circulation of asthmatics but not in normal individuals (Bruijnzeel et al., 1992 Schweiz. Med. Wochenschr. 122, 298-301).
Efficacy of antibody to IL-5 was assessed in a guinea pig asthma model. The animals were challenged with ovalbumin and assayed for eosinophilia and the responsiveness to the bronchioconstriction substance P. A 30 mg/kg dose of antibody administered i.p. blocked ovalbumin- induced increased sensitivity to substance P and blocked increases in bronchoalveolar and lung tissue accumulation of eosinophils (Mauser et al., 1993 Am. Rev. Respir. Dis. 148, 1623-7). In a separate study guinea pigs challenged for eight days with ovalbumin were treated with monoclonal antibody to IL-5. Treatment produced a reduction in the number of eosinophils in bronchoalveolar lavage. No reduction was observed for unchallenged guinea pigs and guinea pigs treated with a control antibody. Antibody treatment completely inhibited the development of hyperreactivity to histamine and arecoline after ovalbumin challenge (van Oosterhout et al., 1993 Am. Rev. Respir. Dis. 147, 548-52)
Results obtained from human clinical analysis and animal studies indicate the role of activated T helper cells, cytokines and eosinophils in asthma. The role of IL-5 in eosinophil development and function makes IL-5 a good candidate for target selection. The antibody studies neutralized IL-5 in the circulation thus preventing eosinophilia. Inhibition of the production of IL-5 will achieve the same goal.
Asthma - a prominent feature of asthma is the infiltration of eosinophils and deposition of toxic eosinophil proteins (e.g. major basic protein, eosinophil-derived neurotoxin) in the lung. A number of T-cell-derived factors like IL-5 are responsible for the activation and maintainance of eosinophils (Kay, 1991 J. Allergy Clin. Immun. 87, 893). Inhibition of IL-5 expression in the lungs can decrease the activation of eosinophils and will help alleviate the symptoms of asthma.
Atopy - is characterized by the developement of type I hypersensitive reactions associated with exposure to certain environmental antigens. One of the common clinical manifestations of atopy is eosinophilia (accumulation of abnormally high levels of eosinophils in the blood). Antibodies against IL-5 have been shown to lower the levels of eosinophils in mice (Cook et al., 1993 in Immunopharmacol. Eosinophils ed. Smith and Cook, pp. 193-216, Academic, London, UK)
Parasitic infection-related eosinophilia- infections with parasites like helminths, can lead to severe eosinophilia (Cook et al., 1993 supra). Animal models for eosinophilia suggest that infection of mice, for example, can lead to blood, peritoneal and/or tissue eosinophilia, all of which seem to be lowered to varying degrees by antibodies directed against IL-5.
Pulmonary infiltration eosinophilia- is characterised by accumulation of high levels of eosinophils in pulmonary parenchyma (Gleich, 1990 J. Allerαv Clin. Immunol. 85, 422). L-Tryptophan-associated eosinophilia-myalgia syndrome
(EMS)- The EMS disease is closely linked to the consumption of L-tryptophan, an essential aminoacid used to treat conditions like insomnia (for review see Varga et al., 1993 J Invest. Dermatol. 100, 97s). Pathologic and histologic studies have demonstrated high levels of eosinophils and mononuclear inflammatory cells in patients with EMS. It appears that IL-5 and transforming growth factor play a significant role in the development of EMS (Varga et al., 1993 supra) by activating eosinophils and other inflammatory cells. Thus, ribozymes of the present invention that cleave IL-5 mRNA and thereby IL-5 activity have many potential therapeutic uses, and there are reasonable modes of delivering the ribozymes in a number of the possible indications. Development of an effective ribozyme that inhibits IL-5 function is described above; available cellular and activity assays are numerous, reproducible, and accurate. Animal models for IL-5 function and for each of the suggested disease targets exist (Cook et al., 1993 supra) and can be used to optimize activity.
Example 3: NF-κB
Ribozymes that cleave rel A mRNA represent a novel therapeutic approach to inflammatory or autoimmune disorders. Inflammatory mediators such as lipopolysaccharide (LPS), interleukin-1 (IL-1) or tumor necrosis factor-a (TNF-α) act on cells by inducing transcription of a number of secondary mediators, including other cytokines and adhesion molecules. In many cases, this gene activation is known to be mediated by the transcriptional regulator, NF-κB. One subunit of NF-κB, the relA gene product (termed RelA or p65) is implicated specifically in the induction of inflammatory responses. Ribozyme therapy, due to its exquisite specificity, is particularly well-suited to target intracellular factors that contribute to disease pathology. Thus, ribozymes that cleave mRNA encoded by rel A or TNF-α may represent novel therapeutics for the treatment of inflammatory and autoimmune disorders.
The nuclear DNA-binding activity, NF-κB, was first identified as a factor that binds and activates the immunoglobulin κ light chain enhancer in B cells. NF-κB now is known to activate transcription of a variety of other cellular genes (e.g., cytokines, adhesion proteins, oncogenes and viral proteins) in response to a variety of stimuli (e.g., phorbol esters, mitogens, cytokines and oxidative stress). In addition, molecular and biochemical characterization of NF-κB has shown that the activity is due to a homodimer or heterodimer of a family of DNA binding subunits. Each subunit bears a stretch of 300 amino acids that is homologous to the oncogene, v-rel. The activity first described as NF-κB is a heterodimer of p49 or p50 with p65. The p49 and p50 subunits of NF-κB (encoded by the nf-κB2 or nf-κB1 genes, respectively) are generated from the precursors NF-κB1 (p105) or NF-κB2 (p100). The p65 subunit of NF-κB (now termed Rel A ) is encoded by the rel A locus.
The roles of each specific transcription-activating complex now are being elucidated in cells (N.D. Perkins, et al., 1992 Proc. Natl Acad. Sci USA 89, 1529-1533). For instance, the heterodimer of NF-κB1 and Rel A (p50/p65) activates transcription of the promoter for the adhesion molecule, VCAM-1 , while NF-κB2/RelA heterodimers (p49/p65) actually inhibit transcription (H.B. Shu, et al., Mol. Cell. Biol. 13, 6283-6289 (1993)). Conversely, heterodimers of NF-κB2/RelA (p49/p65) act with Tat-I to activate transcription of the HIV genome, while NF-κB1/RelA (p50/p65) heterodimers have little effect (J. Liu, N.D. Perkins, R.M. Schmid, G.J. Nabel, J. Virol. 1992 66, 3883-3887). Similarly, blocking rel A gene expression with antisense oligonucleotides specifically blocks embryonic stem cell adhesion; blocking NF-κB1 gene expression with antisense oligonucleotides had no effect on cellular adhesion (Narayanan et al., 1993 Mol. Cell. Biol. 13, 3802-3810). Thus, the promiscuous role initially assigned to NF-κB in transcriptional activation (M.J. Lenardo, D. Baltimore, 1989 Cell 58, 227-229) represents the sum of the activities of the rel family of DNA-binding proteins. This conclusion is supported by recent transgenic "knock-out" mice of individual members of the rel family. Such "knock-outs" show few developmental defects, suggesting that essential transcriptional activation functions can be performed by more than one member of the rel family.
A number of specific inhibitors of NF-κB function in cells exist, including treatment with phosphorothioate antisense oliogonucleotide, treatment with double-stranded NF-κB binding sites, and over expression of the natural inhibitor MAD-3 (an 1κB family member). These agents have been used to show that NF-κB is required for induction of a number of molecules involved in inflammation, as described below.
•NF-κB is required for phorbol ester-mediated induction of IL-6 (I. Kitajima, et al., Science 258, 1792-5 (1992)) and IL-8 (Kunsch and Rosen, 1993 Mol. Cell. Biol. 13, 6137-46).
•NF-κB is required for induction of the adhesion molecules ICAM-1 (Eck, et al., 1993 Mol. Cell. Biol. 13, 6530-6536), VCAM-1 (Shu et al., supra), and E-selectin (Read, et al., 1994 J. Exp. Med. 179, 503-512) on endothelial cells. •NF-κB is involved in the induction of the integrin subunit, CD18, and other adhesive properties of leukocytes (Eck et al., 1993 supra).
The above studies suggest that NF-κB is integrally involved in the induction of cytokines and adhesion molecules by inflammatory mediators. Two recent papers point to another connection between NF-κB and inflammation: glucocorticoids may exert their anti-inflammatory effects by inhibiting NF-κB. The glucocorticoid receptor and p65 both act at NF-κB binding sites in the ICAM-1 promoter (van de Stolpe, et al., 1994 J. Biol. Chem. 269, 6185-6192). Glucocorticoid receptor inhibits NF-κB-mediated induction of IL-6 (Ray and Prefontaine, 1994 Proc. Natl Acad. Sci USA 91 , 752-756). Conversely, overexpression of p65 inhibits glucocorticoid induction of the mouse mammary tumor virus promoter. Finally, protein cross-linking and co-immunoprecipitation experiments demonstrated direct physical interaction between p65 and the glucocorticoid receptor (Id.).
Ribozymes of this invention block to some extent NF-κB expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of restenosis, transplant rejection and rheumatoid arthritis. Ribozyme cleavage of relA mRNA in these systems may prevent inflammatory cell function and alleviate disease symptoms. The sequence of human and mouse re/A mRNA can be screened for accessible sites using a computer folding algorithm. Potential hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 17, 18 and 21-22. (All sequences are 5' to 3' in the tables.) While mouse and human sequences can be screened and ribozymes thereafter designed, the human targetted sequences are of most utility.
The sequences of the chemically synthesized ribozymes useful in this study are shown in Tables 19 - 22. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity and may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables. By engineering ribozyme motifs we have designed several ribozymes directed against rel A mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave relA target sequences in vitro is evaluated.
The ribozymes will be tested for function in vivo by analyzing cytokine-induced VCAM-1 , ICAM-1 , IL-6 and IL-8 expression levels. Ribozymes will be delivered to cells by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA and RNA vectors. Cytokine-induced VCAM-1 , ICAM-1 , IL-6 and IL-8 expression will be monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. Rel A mRNA levels will be assessed by Northern analysis, RNAse protection or primer extension analysis or quantitative RT-PCR. Activity of NF-κB will be monitored by gel-retardation assays. Ribozymes that block the induction of NF-κB activity and/or rel A mRNA by more than 50% will be identified. RNA ribozymes and/or genes encoding them will be locally delivered to transplant tissue ex vivo in animal models. Expression of the ribozyme will be monitored by its ability to block ex vivo induction of VCAM-1 , ICAM-1 , IL-6 and IL-8 mRNA and protein. The effect of the anti-rel A ribozymes on graft rejection will then be assessed. Similarly, ribozymes will be introduced into joints of mice with collagen-induced arthritis or rabbits with Streptococcal cell wall-induced arthritis. Liposome delivery, cationic lipid delivery, or adeno-associated virus vector delivery can be used. One dose (or a few infrequent doses) of a stable anti-relA ribozyme or a gene construct that constitutively expresses the ribozyme may abrogate inflammatory and immune responses in these diseases. Uses
A therapeutic agent that inhibits cytokine gene expression, inhibits adhesion molecule expression, and mimics the anti-inflammatory effects of glucocorticoids (without inducing steroid-responsive genes) is ideal for the treatment of inflammatory and autoimmune disorders. Disease targets for such a drug are numerous. Target indications and the delivery options each entails are summarized below. In all cases, because of the potential immunosuppressive properties of a ribozyme that cleaves rel A mRNA, uses are limited to local delivery, acute indications, or ex vivo treatment. •Rheumatoid arthritis (RA).
Due to the chronic nature of RA, a gene therapy approach is logical. Delivery of a ribozyme to inflamed joints is mediated by adenovirus, retrovirus, or adeno-associated virus vectors. For instance, the appropriate adenovirus vector can be administered by direct injection into the synovium: high efficiency of gene transfer and expression for several months would be expected (B.J. Roessler, E.D. Allen, J.M. Wilson, J.W. Hartman, B. L. Davidson, J. Clin. Invest. 92, 1085-1092 (1993)). It is unlikely that the course of the disease could be reversed by the transient, local administration of an anti-inflammatory agent. Multiple administrations may be necessary. Retrovirus and adeno-associated virus vectors would lead to permanent gene transfer and expression in the joint. However, permanent expression of a potent anti-inflammatory agent may lead to local immune deficiency.
•Restenosis. Expression of NF-κB in the vessel wall of pigs causes a narrowing of the luminal space due to excessive deposition of extracellular matrix components. This phenotype is similar to matrix deposition that occurs subsequent to coronary angioplasty. In addition, NF-κB is required for the expression of the oncogene c-myb (F.A. La Rosa, J.W. Pierce, G.E. Soneneshein, Mol. Cell. Biol. 14, 1039-44 (1994)). Thus NF-κB induces smooth muscle proliferation and the expression of excess matrix components: both processes are thought to contribute to reocclusion of vessels after coronary angioplasty.
•Transplantation. NF-κB is required for the induction of adhesion molecules (Eck et al., supra, K. O'Brien, et al., J. Clin. Invest. 92, 945-951 (1993)) that function in immune recognition and inflammatory responses. At least two potential modes of treatment are possible. In the first, transplanted organs are treated ex vivo with ribozymes or ribozyme expression vectors. Transient inhibition of NF-κB in the transplanted endothelium may be sufficient to prevent transplant-associated vasculitis and may significantly modulate graft rejection. In the second, donor B cells are treated ex vivo with ribozymes or ribozyme expression vectors. Recipients would receive the treatment prior to transplant. Treatment of a recipient with B cells that do not express T cell co-stimulatory molecules (such as ICAM-1 , VCAM-1 , and/or B7 an B7-2) can induce antigen-specific anergy. Tolerance to the donor's histocompatibility antigens could result; potentially, any donor could be used for any transplantation procedure. •Asthma.
Granulocyte macrophage colony stimulating factor (GM-CSF) is thought to play a major role in recruitment of eosinophils and other inflammatory cells during the late phase reaction to asthmatic trauma. Again, blocking the local induction of GM-CSF and other inflammatory mediators is likely to reduce the persistent inflammation observed in chronic asthmatics. Aerosol delivery of ribozymes or adenovirus ribozyme expression vectors is a feasible treatment.
•Gene Therapy.
Immune responses limit the efficacy of many gene transfer techniques. Cells transfected with retrovirus vectors have short lifetimes in immune competent individuals. The length of expression of adenovirus vectors in terminally differentiated cells is longer in neonatal or immunecompromised animals. Insertion of a small ribozyme expression cassette that modulates inflammatory and immune responses into existing adenovirus or retrovirus constructs will greatly enhance their potential.
Thus, ribozymes of the present invention that cleave rel A mRNA and thereby NF-κB activity have many potential therapeutic uses, and there are reasonable modes of delivering the ribozymes in a number of the possible indications. Development of an effective ribozyme that inhibits NF-κB function is described above; available cellular and activity assays are number, reproducible, and accurate. Animal models for NF-κB function (Kitajima, et al., supra) and for each of the suggested disease targets exist and can be used to optimize activity. Example 4: TNF-α
Ribozymes that cleave the specific cites in TNF-α mRNA represent a novel therapeutic approach to inflammatory or autoimmune disorders.
Tumor necrosis factor-α (TNF-α) is a protein, secreted by activated leukocytes, that is a potent mediator of inflammatory reactions. Injection of TNF-α into experimental animals can simulate the symptoms of systemic and local inflammatory diseases such as septic shock or rheumatoid arthritis.
TNF-α was initially described as a factor secreted by activated macrophages which mediates the destruction of solid tumors in mice (Old, 1985 Science 230, 4225-4231). TNF-α subsequently was found to be identical to cachectin, an agent responsible for the weight loss and wasting syndrome associated with tumors and chronic infections (Beutler, et al., 1985 Nature 316, 552-554). The cDNA and the genomic locus for TNF-α have been cloned and found to be related to TNF-β (Shakhov et al., 1990 J. Exp. Med. 171 , 35-47). Both TNF-α and TNF-β bind to the same receptors and have nearly identical biological activities. The two TNF receptors have been found on most cell types examined (Smith, et al., 1990 Science 248, 1019-1023). TNF-α secretion has been detected from monocytes/macrophages, CD4+ and CD8+ T-cells, B-cells, lymphokine activated killer cells, neutrophils, astrocytes, endothelial cells, smooth muscle cells, as well as various non-hematopoietic tumor cell lines ( for a review see Turestskaya et al., 1991 in Tumor Necrosis Factor: Structure. Function, and Mechanism of Action B. B. Aggarwal, J. Vilcek, Eds. Marcel Dekker, Inc., pp. 35-60). TNF-α is regulated transcriptionally and translationally, and requires proteolytic processing at the plasma membrane in order to be secreted (Kriegler et al., 1988 Cell 53, 45-53). Once secreted, the serum half life of TNF-α is approximately 30 minutes. The tight regulation of TNF-α is important due to the extreme toxicity of this cytokine. Increasing evidence indicates that overproduction of TNF-α during infections can lead to severe systemic toxicity and death (Tracey & Cerami, 1992 Am. J. Trop. Med. Hvα. 47, 2-7).
Antisense RNA and Hammerhead ribozymes have been used in an attempt to lower the expression level of TNF-α by targeting specified cleavage sites [Sioud et al., 1992 J. Mol. Biol. 223; 831 ; Sioud WO 94/10301 ; Kisich and co-workers, 1990 abstract (FASEB J. 4. A1860; 1991 slide presentation (J. Leukocyte Biol. sup. 2, 70); December, 1992 poster presentation at Anti-HIV Therapeutics Conference in SanDiego, CA; and "Development of anti-TNF-α ribozymes for the control of TNF-α gene expression"- Kisich, Doctoral Dissertation, 1993 University of California, Davis] listing various TNFα targeted ribozymes.
Ribozymes of this invention block to some extent TNF-α expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of septic shock and rheumatoid arthritis. Ribozyme cleavage of TNF-α mRNA in these systems may prevent inflammatory cell function and alleviate disease symptoms.
The sequence of human and mouse TNF-α mRNA can be screened for accessible sites using a computer folding algorithm. Hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 23, 25, and 27 - 28. (All sequences are 5' to 3' in the tables.) While mouse and human sequences can be screened and ribozymes thereafter designed, the human targeted sequences are of most utility. However, mouse targeted ribozymes are useful to test efficacy of action of the ribozyme prior to testing in humans. The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme. (In Table 24, lower case letters indicate positions that are not conserved between the human and the mouse TNF-α sequences.)
The sequences of the chemically synthesized ribozymes useful in this study are shown in Tables 24, 26 - 28. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity. For example, stem-loop II sequence of hammerhead ribozymes listed in Tables 24 and 26 (5'-GGCCGAAAGGCC-3') can be altered (substitution, deletion, and/or insertion) to contain any sequences provided a minimum of two base-paired stem structure can form. Similarly, stem-loop IV sequence of hairpin ribozymes listed in Tables 27 and 28 (5'-CACGUUGUG-3') can be altered (substitution, deletion, and/or insertion) to contain any sequence, provided a minimum of two base-paired stem structure can form. The sequences listed in Tables 24, 26 - 28 may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables or AAV .
In a preferred embodiment of the invention, a transcription unit expressing a ribozyme that cleaves TNF-α RNA is inserted into a plasmid
DNA vector or an adenovirus DNA viral vector or AAV or alpha virus or retroviris vectors. Viral vectors have been used to transfer genes to the intact vasculature or to joints of live animals (Willard et al., 1992
Circulation. 86, I-473.; Nabel et al., 1990 Science. 249, 1285-1288) and both vectors lead to transient gene expression. The adenovirus vector is delivered as recombinant adenoviral particles. DNA may be delivered alone or complexed with vehicles (as described for RNA above). The DNA,
DNA/vehicle complexes, or the recombinant adenovirus particles are locally administered to the site of treatment, e.g., through the use of an injection catheter, stent or infusion pump or are directly added to cells or tissues ex vivo.
In another preferred embodiment of the invention, a transcription unit expressing a ribozyme that cleaves TNF-α RNA is inserted into a retrovirus vector for sustained expression of ribozyme(s). By engineering ribozyme motifs we have designed several ribozymes directed against TNF-α mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave TNF-α target sequences in vitro is evaluated.
The ribozymes will be tested for function in cells by analyzing bacterial lipopolysaccharide (LPS)-induced TNF-α expression levels. Ribozymes will be delivered to cells by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA vectors. TNF-α expression will be monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. TNF-α mRNA levels will be assessed by Northern analysis, RNAse protection, primer extension analysis or quantitative RT-PCR. Ribozymes that block the induction of TNF-α activity and/or TNF-α mRNA by more than 90% will be identified.
RNA ribozymes and/or genes encoding them will be locally delivered to macrophages by intraperitoneal injection. After a period of ribozyme uptake, the peritoneal macrophages are harvested and induced ex vivo with LPS. The ribozymes that significantly reduce TNF-α secretion are selected. The TNF-α can also be induced after ribozyme treatment with fixed Streptococcus in the peritoneal cavity instead of ex vivo. In this fashion the ability of TNF-α ribozymes to block TNF-α secretion in a localized inflammatory response are evaluated. In addition, we will determine if the ribozymes can block an ongoing inflammatory response by delivering the TNF-α ribozymes after induction by the injection of fixed Streptococcus.
To examine the effect of anti-TNF-α ribozymes on systemic inflammation, the ribozymes are delivered by intravenous injection. The ability of the ribozymes to inhibit TNF-α secretion and lethal shock caused by systemic LPS administration are assessed. Similarly, TNF-α ribozymes can be introduced into the joints of mice with collagen-induced arthritis.
Either free delivery, liposome delivery, cationic lipid delivery, adeno-associated virus vector delivery, adenovirus vector delivery, retrovirus vector delivery or plasmid vector delivery in these animal model experiments can be used to supply ribozymes. One dose (or a few infrequent doses) of a stable anti-TNF-α ribozyme or a gene construct that constitutively expresses the ribozyme may abrogate tissue damage in these inflammatory diseases.
Macrophage isolation.
To produce responsive macrophages 1 ml of sterile fluid thioglycollate broth (Difco, Detroit, MI.) was injected i.p. into 6 week old female C57bl/6NCR mice 3 days before peritoneal lavage. Mice were maintained as specific pathogen free in autoclaved cages in a laminar flow hood and given sterilized water to minimize "spontaneous" activation of macrophages. The resulting peritoneal exudate cells (PEC) were obtained by lavage using Hanks balanced salt solution (HBSS) and were plated at 2.5X105/well in 96 well plates (Costar, Cambridge, MA.) with Eagles minimal essential medium (EMEM) containing 10% heat inactivated fetal bovine serum. After adhering for 2 hours the wells were washed to remove non-adherent cells. The resulting cultures were 97% macrophages as determined by morphology and staining for non-specific esterase.
Transfection of ribozymes into macrophages: The ribozymes were diluted to 2X final concentration, mixed with an equal volume of 11 nM lipofectamine (Life Technologies, Gaithersburg, MD.), and vortexed. 100 ml of lipid:ribozyme complex was then added directly to the cells, followed immediately by 10 ml fetal bovine serum. Three hours after ribozyme addition 100 ml of 1 mg/ml bacterial lipopolysaccaride (LPS) was added to each well to stimulate TNF production.
Quantitation of TNF-α in mouse macrophages:
Supernatants were sampled at 0, 2, 4, 8, and 24 hours post LPS stimulation and stored at -70°C. Quantitation of TNF-α was done by a specific ELISA. ELISA plates were coated with rabbit anti-mouse TNF-α serum at 1 :1000 dilution (Genzyme) followed by blocking with milk proteins and incubation with TNF-α containing supernatants. TNF-α was then detected using a murine TNF-α specific hamster monoclonal antibody (Genzyme). The ELISA was developed with goat anti-hamster IgG coupled to alkaline phosphatase.
Assessment of reagent toxicity:
Following ribozyme/lipid treatment of macrophages and harvesting of supernatants viability of the cells was assessed by incubation of the cells with 5 mg/ml of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT). This compound is reduced by the mitochondrial dihydrogenases, the activity of which correlates well with cell viability. After 12 hours the absorbance of reduced MTT is measured at 585 nm.
Uses
The association between TNF-α and bacterial sepsis, rheumatoid arthritis, and autoimmune disease make TNF-α an attractive target for therapeutic intervention [Tracy & Cerami 1992 supra: Williams et al., 1992
Proc. Natl. Acad. Sci. USA 89, 9784-9788; Jacob, 1992 J. Autoimmun. 5
(Supp. A), 133-143], Septic Shock
Septic shock is a complication of major surgery, bacterial infection, and polytrauma characterized by high fever, increased cardiac output, reduced blood pressure and a neutrophilic infiltrate into the lungs and other major organs. Current treatment options are limited to antibiotics to reduce the bacterial load and non-steroidal anti-inflammatories to reduce fever. Despite these treatments in the best intensive care settings, mortality from septic shock averages 50%, due primarily to multiple organ failure and disseminated vascular coagulation. Septic shock, with an incidence of 200,000 cases per year in the United States, is the major cause of death in intensive care units. In septic shock syndrome, tissue injury or bacterial products initiate massive immune activation, resulting in the secretion of pro-inflammatory cytokines which are not normally detected in the serum, such as TNF-α, interleukin-1 β (IL-1β), γ-interferon (IFN-γ), interleukin-6 (IL-6), and interleukin-8 (IL-8). Other non-cytokine mediators such as leukotriene b4, prostaglandin E2, C3a and C3d also reach high levels (de Boer et al., 1992 Immunopharmacology 24, 135-148).
TNF-α is detected early in the course of septic shock in a large fraction of patients (de Boer et al., 1992 supra). In animal models, injection of TNF-α has been shown to induce shock-like symptoms similar to those induced by LPS injection (Beutler et al., 1985 Science 229, 869-871); in contrast, injection of IL-1β, IL-6, or IL-8 does not induce shock. Injection of TNF-α also causes an elevation of IL-1β, IL-6, IL-8, PgE2, acute phase proteins, and TxA2 in the serum of experimental animals (de Boer et al., 1992 supra). In animal models the lethal effects of LPS can be blocked by preadministration of anti-TNF-α antibodies. The cumulative evidence indicates that TNF-α is a key player in the pathogenesis of septic shock, and therefore a good candidate for therapeutic intervention.
Rheumatoid Arthritis Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the joints leading to bone destruction and loss of joint function. At the cellular level, autoreactive T- lymphocytes and monocytes are typically present, and the synoviocytes often have altered morphology and immunostaining patterns. RA joints have been shown to contain elevated levels of TNF-α, IL-1α and IL-1β, IL-6, GM-CSF, and TGF- β (Abney et al., 1991 Imm. Rev. 119, 105-123), some or all of which may contribute to the pathological course of the disease.
Cells cultured from RA joints spontaneously secrete all of the proinflammatory cytokines detected in vivo. Addition of antisera against TNF-α to these cultures has been shown to reduce IL-1α/β production by these cells to undetectable levels (Abney et al., 1991 Supra). Thus, TNF-α may directly induce the production of other cytokines in the RA joint. Addition of the anti-inflammatory cytokine, TGF-β, has no effect on cytokine secretion by RA cultures. Immunocytochemical studies of human RA surgical specimens clearly demonstrate the production of TNF-α, IL-1α/β, and IL-6 from macrophages near the cartilage/pannus junction when the pannus in invading and overgrowing the cartilage (Chu et al., 1992 Br. J. Rheumatology 31 , 653-661). GM-CSF was shown to be produced mainly by vascular endothelium in these samples. Both TNF-α and TGF-β have been shown to be fibroblast growth factors, and may contribute to the accumulation of scar tissue in the RA joint. TNF-α has also been shown to increase osteoclast activity and bone resorbtion, and may have a role in the bone erosion commonly found in the RA joint (Cooper et al., 1992 Clin. Exp. Immunol. 89, 244-250). Elimination of TNF-α from the rheumatic joint would be predicted to reduce overall inflammation by reducing induction of MHC class II, IL-1α/β, II-6, and GM-CSF, and reducing T-cell activation. Osteoclast activity might also fall, reducing the rate of bone erosion at the joint. Finally, elimination of TNF-α would be expected to reduce accumulation of scar tissue within the joint by removal of a fibroblast growth factor.
Treatment with an anti-TNF-α antibody reduces joint swelling and the histological severity of collagen-induced arthritis in mice (Williams et al., 1992 Proc. Natl. Acad. Sci. USA 89, 9784-9788). In addition, a study of RA patients who have received i.v. infusions of anti-TNF-α monoclonal antibody reports a reduction in the number and severity of inflamed joints after treatment. The benefit of monoclonal antibody treatment in the long term may be limited by the expense and immunogenicity of the antibody.
Psoriasis
Psoriasis is an inflammatory disorder of the skin characterized by keratinocyte hyperproliferation and immune cell infiltrate (Kupper, 1990 J. Clin. Invest. 86, 1783-1789). It is a fairly common condition, affecting 1.5-2.0% of the population. The disorder ranges in severity from mild, with small flaky patches of skin, to severe, involving inflammation of the entire epidermis. The cellular infiltrate of psoriasis includes T-lymphocytes, neutrophils, macrophages, and dermal dendrocytes. The majority of T-lymphocytes are activated CD4+ cells of the TH-1 phenotype, although some CD8+ and CD4-/CD8- are also present. B lymphocytes are typically not found in abundance in psoriatic plaques.
Numerous hypotheses have been offered as to the proximal cause of psoriasis including auto-antibodies and auto-reactive T-cells, overproduction of growth factors, and genetic predisposition. Although there is evidence to support the involvement of each of these factors in psoriasis, they are neither mutually exclusive nor are any of them necessary and sufficient for the pathogenesis of psoriasis (Reeves, 1991 Semin. Dermatol. 10, 217).
The role of cytokines in the pathogenesis of psoriasis has been investigated. Among those cytokines found to be abnormally expressed were TGF-α , IL-1α, IL-1 β, IL-1 ra, IL-6, IL-8, IFN-γ, and TNF-α . In addition to abnormal cytokine production, elevated expression of ICAM-1 , ELAM-1 , and VCAM has been observed (Reeves, 1991 supra). This cytokine profile is similar to that of normal wound healing, with the notable exception that cytokine levels subside upon healing. Keratinocytes themselves have recently been shown to be capable of secreting EGF, TGF-α, IL-6, and TNF-α, which could increase proliferation in an autocrine fashion (Oxholm et al., 1991 APMIS 99, 58-64).
Nickoloff et al., 1993 (J Dermatol Sci. 6, 127-33) have proposed the following model for the initiation and maintenance of the psoriatic plaque:
Tissue damage induces the wound healing response in the skin. Keratinocytes secrete IL-1 α, IL-1 β, IL-6, IL-8, TNF-α. These factors activate the endothelium of dermal capillaries, recruiting PMNs, macrophages, and T-cells into the wound site.
Dermal dendrocytes near the dermal/epidermal junction remain activated when they should return to a quiescent state, and subsequently secrete cytokines including TNF-α, IL-6, and IL-8. Cytokine expression, in turn, maintains the activated state of the endothelium, allowing extravasation of additional immunocytes, and the activated state of the keratinocytes which secrete TGF-α and IL-8. Keratinocyte IL-8 recruits immunocytes from the dermis into the epidermis. During passage through the dermis, T-cells encounter the activated dermal dendrocytes which efficiently activate the TH-1 phenotype. The activated T-cells continue to migrate into the epidermis, where they are stimulated by keratinocyte-expressed ICAM-1 and MHC class II. IFN-γ secreted by the T-cells synergizes with the TNF-α from dermal dendrocytes to increase keratinocyte proliferation and the levels of TGF-α, IL-8, and IL-6 production. IFN-γ also feeds back to the dermal dendrocyte, maintaining the activated phenotype and the inflammatory cycle.
Elevated serum titres of IL-6 increases synthesis of acute phase proteins including complement factors by the liver, and antibody production by plasma cells. Increased complement and antibody levels increases the probability of autoimmune reactions.
Maintenance of the psoriatic plaque requires continued expression of all of these processes, but attractive points of therapeutic intervention are
TN F-α expression by the dermal dendrocyte to maintain activated endothelium and keratinocytes, and IFN-γ expression by T-cells to maintain activated dermal dendrocytes.
There are 3 million patients in the United States afflicted with psoriasis. The available treatments for psoriasis are corticosteroids. The most widely prescribed are TEMOVATE (clobetasol propionate), LIDEX (fluocinonide), DIPROLENE (betamethasone propionate), PSORCON (diflorasone diacetate) and TRIAMCINOLONE formulated for topical application. The mechanism of action of corticosteroids is multifactorial. This is a palliative therapy because the underlying cause of the disease remains, and upon discontinuation of the treatment the disease returns. Discontinuation of treatment is often prompted by the appearance of adverse effects such as atrophy, telangiectasias and purpura. Corticosteroids are not recommended for prolonged treatments or when treatment of large and/or inflamed areas is required. Alternative treatments include retinoids, such as etretinate, which has been approved for treatment of severe, refractory psoriasis. Alternative retinoid-based treatments are in advanced clinical trials. Retinoids act by converting keratinocytes to a differentiated state and restoration of normal skin development. Immunosuppressive drugs such as cyclosporine are also in the advanced stages of clinical trials. Due to the nonspecific mechanism of action of corticosteroids, retinoids and immunosuppressives, these treatments exhibit severe side effects and should not be used for extended periods of time unless the condition is life-threatening or disabling. There is a need for a less toxic, effective therapeutic agent in psoriatic patients.
HIV and AIDS
The human immunodeficiency virus (HIV) causes several fundamental changes in the human immune system from the time of infection until the development of full-blown acquired immunodeficiency syndrome (AIDS). These changes include a shift in the ratio of CD4+ to
CD8+ T-cells, sustained elevation of IL-4 levels, episodic elevation of TNF-α and TNF-β levels, hypergammaglobulinemia, and lymphoma/leukemia (Rosenberg & Fauci, 1990 Immun. Today 11 , 176; Weiss 1993 Science
260, 1273). Many patients experience a unique tumor, Kaposi's sarcoma and/or unusual opportunistic infections (e.g. Pneumocystis carinii, cytomegalovirus, herpesviruses, hepatitis viruses, papilloma viruses, and tuberculosis). The immunological dysfunction of individuals with AIDS suggests that some of the pathology may be due to cytokine dysregulation.
Levels of serum TNF-α and IL-6 are often found to be elevated in AIDS patients (Weiss, 1993 supra). In tissue culture, HIV infection of monocytes isolated from healthy individuals stimulates secretion of both TNF-α and IL-6. This response has been reproduced using purified gp120, the viral coat protein responsible for binding to CD-4 (Buonaguro et al., 1992 J. Virol. 66, 7159). It has also been demonstrated that the viral gene regulator, Tat, can directly induce TNF transcription. The ability of HIV to directly stimulate secretion of TNF-α and IL-6 may be an adaptive mechanism of the virus. TNF-α has been shown to upregulate transcription of the LTR of HIV, increasing the number of HIV-specific transcripts in infected cells. IL-6 enhances HIV production, but at a post-transcriptional level, apparently increasing the efficiency with which HIV transcripts are translated into protein. Thus, stimulation of TNF-α secretion by the HIV virus may promote infection of neighboring CD4+ cells both by enhancing virus production from latently infected cells and by driving replication of the virus in newly infected cells. The role of TNF-α in HIV replication has been well established in tissue culture models of infection (Sher et al., 1992 Immun. Rev. 127, 183), suggesting that the mutual induction of HIV replication and TNF-α replication may create positive feedback in vivo. However, evidence for the presence of such positive feedback in infected patients is not abundant. TNF-α levels are found to be elevated in some, but not all patients tested. Children with AIDS who were given zidovudine had reduced levels of TNF-α compared to those not given zidovudine (Cremoni et al., 1993 AIPS 7, 128). This correlation lends support to the hypothesis that reduced viral replication is physiologically linked to TNF-α levels. Furthermore, recently it has been shown that the polyclonal B cell activation associated with HIV infection is due to membrane-bound TNF-α. Thus, levels of secreted TNF-α may not accurately reflect the contribution of this cytokine to AIDS pathogenesis. Chronic elevation of TNF-α has been shown to shown to result in cachexia (Tracey et al., 1992 Am. J. Trop. Med. Hyg. 47, 2-7), increased autoimmune disease (Jacob, 1992 supra), lethargy, and immune suppression in animal models (Aderka et al., 1992 Isr. J. Med. Sci. 28, 126-130). The cachexia associated with AIDS may be associated with chronically elevated TNF-α frequently observed in AIDS patients. Similarly, TNF-α can stimulate the proliferation of spindle cells isolated from Kaposi's sarcoma lesions of AIDS patients (Barillari et al., 1992 J Immunol 149, 3727).
A therapeutic agent that inhibits cytokine gene expression, inhibits adhesion molecule expression, and mimics the anti-inflammatory effects of glucocorticoids (without inducing steroid-responsive genes) is ideal for the treatment of inflammatory and autoimmune disorders. Disease targets for such a drug are numerous. Target indications and the delivery options each entails are summarized below. In all cases, because of the potential immunosuppressive properties of a ribozyme that cleaves the specified sites in TNF-α mRNA, uses are limited to local delivery, acute indications, or ex vivo treatment.
•Septic shock. Exogenous delivery of ribozymes to macrophages can be achieved by intraperitoneal or intravenous injections. Ribozymes will be delivered by incorporation into liposomes or by complexing with cationic lipids.
•Rheumatoid arthritis (RA). Pue to the chronic nature of RA, a gene therapy approach is logical.
Pelivery of a ribozyme to inflamed joints is mediated by adenovirus, retrovirus, or adeno-associated virus vectors. For instance, the appropriate adenovirus vector can be administered by direct injection into the synovium: high efficiency of gene transfer and expression for several months would be expected (B.J. Roessler, E.P. Allen, J.M. Wilson, J.W. Hartman, B. L. Davidson, J. Clin. Invest. 92, 1085-1092 (1993)). It is unlikely that the course of the disease could be reversed by the transient, local administration of an anti-inflammatory agent. Multiple administrations may be necessary. Retrovirus and adeno-associated virus vectors would lead to permanent gene transfer and expression in the joint. However, permanent expression of a potent anti-inflammatory agent may lead to local immune deficiency.
•Psoriasis
The psoriatic plaque is a particularly good candidate for ribozyme or vector delivery. The stratum corneum of the plaque is thinned, providing access to the proliferating keratinocytes. T-cells and dermal dendrocytes can be efficiently targeted by trans-epidermal diffusion .
Organ culture systems for biopsy specimens of psoriatic and normal skin are described in current literature (Nickoloff et al., 1993 Supra). Primary human keratinocytes are easily obtained and will be grown into epidermal sheets in tissue culture. In addition to these tissue culture models, the flaky skin mouse develops psoriatic skin in response to UV light. This model would allow demonstration of animal efficacy for ribozyme treatments of psoriasis. «Gene Therapy.
Immune responses limit the efficacy of many gene transfer techniques. Cells transfected with retrovirus vectors have short lifetimes in immune competent individuals. The length of expression of adenovirus vectors in terminally differentiated cells is longer in neonatal or immunecompromised animals. Insertion of a small ribozyme expression cassette that modulates inflammatory and immune responses into existing adenovirus or retrovirus constructs will greatly enhance their potential. Thus, ribozymes of the present invention that cleave TNF-α mRNA and thereby TNF-α activity have many potential therapeutic uses, and there are reasonable modes of delivering the ribozymes in a number of the possible indications. Development of an effective ribozyme that inhibits TNF-α function is described above; available cellular and activity assays are number, reproducible, and accurate. Animal models for TNF-α function and for each of the suggested disease targets exist and can be used to optimize activity.
Example 5: p210bcr-abl
Chronic myelogenous leukemia exhibits a characteristic disease course, presenting initially as a chronic granulocytic hyperplasia, and invariably evolving into an acute leukemia which is caused by the clonal expansion of a cell with a less differentiated phenotype (i.e.. the blast crisis stage of the disease). CML is an unstable disease which ultimately progresses to a terminal stage which resembles acute leukemia. This lethal disease affects approximately 16,000 patients a year.
Chemotherapeutic agents such as hydroxyurea or busulfan can reduce the leukemic burden but do not impact the life expectancy of the patient (e.g. approximately 4 years). Consequently, CML patients are candidates for bone marrow transplantation (BMT) therapy. However, for those patients which survive BMT, disease recurrence remains a major obstacle
(Apperley et al., 1988 Br. J. Haematol. 69, 239).
The Philadelphia (Ph) chromosome which results from the translocation of the abl oncogene from chromosome 9 to the bcr gene on chromosome 22 is found in greater than 95% of CML patients and in 10-25% of all cases of acute lymphoblastic leukemia [(ALL); Fourth International Workshop on Chromosomes in Leukemia 1982, Cancer Genet. Cytogenet. 11 , 316]. In virtually all Ph-positive CMLs and approximately 50% of the Ph-positive ALLs, the leukemic cells express bcr-abl fusion mRNAs in which exon 2 (b2-a2 junction) or exon 3 (b3-a2 junction) from the major breakpoint cluster region of the bcr gene is spliced to exon 2 of the abl gene. Heisterkamp et al., 1985 Nature 315, 758; Shtivelman et al., 1987, Blood 69, 971). In the remaining cases of Ph-positive ALL, the first exon of the bcr gene is spliced to exon 2 of the abl gene (Hooberman et al., 1989 Proc. Nat. Acad. Sci. USA 86, 4259; Heisterkamp et al., 1988 Nucleic Acids Res. 16, 10069).
The b3-a2 and b2-a2 fusion mRNAs encode 210 kd bcr-abl fusion proteins which exhibit oncogenic activity (Daley et al., 1990 Science 247, 824; Heisterkamp et al., 1990 Nature 344, 251). The importance of the bcr-abl fusion protein (p210bcr-a bl) in the evolution and maintenance of the leukemic phenotype in human disease has been demonstrated using antisense oligonucleotide inhibition of p210bcr-a bl expression. These inhibitory molecules have been shown to inhibit the in vitro proliferation of leukemic cells in bone marrow from CML patients. Szczylik et al., 1991 Science 253, 562). Reddy, U.S. Patent 5,246,921 (hereby incorporated by reference herein) describes use of ribozymes as therapeutic agents for leukemias, such as chronic myelogenous leukemia (CML) by targeting the specific junction region of bcr-abl fusion transcripts. It indicates causing cleavage by a ribozyme at or near the breakpoint of such a hybrid chromosome, specifically it includes cleavage at the sequence GUX, where X is A, U or G. The one example presented is to cleave the sequence 5' AGC AG AGUU (cleavage site) CAA AAGCCCU-3'.
Scanlon WO 91/18625, WO 91/18624, and WO 91/18913 and Snyder et al., WO93/03141 and WO94/13793 describe a ribozyme effective to cleave oncogenic variants of H-ras RNA. This ribozyme is said to inhibit H-ras expression in response to external stimuli.
The invention features use of ribozymes to inhibit the development or expression of a transformed phenotype in man and other animals by modulating expression of a gene that contributes to the expression of CML. Cleavage of targeted mRNAs expressed in pre-neoplastic and transformed cells elicits inhibition of the transformed state.
The invention can be used to treat cancer or pre-neoplastic conditions. Two preferred administration protocols can be used, either in vivo administration to reduce the tumor burden, or ex vivo treatment to eradicate transformed cells from tissues such as bone marrow prior to reimplantation.
This invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of CML. The mRNA targets are present in the 425 nucleotides surrounding the fusion sites of the bcr and abl sequences in the b2-a2 and b3-a2 recombinant mRNAs. Other sequences in the 5' portion of the bcr mRNA or the 3' portion of the abl mRNA may also be targeted for ribozyme cleavage. Cleavage at any of these sites in the fusion mRNA molecules will result in inhibition of translation of the fusion protein in treated cells.
The invention provides a class of chemical cleaving agents which exhibit a high degree of specificity for the mRNA causative of CML. Such enzymatic RNA molecules can be delivered exogenously or endogenously to afflicted cells. In the preferred hammerhead motif the small size (less than 40 nucleotides, preferably between 32 and 36 nucleotides in length) of the molecule allows the cost of treatment to be reduced.
The smallest ribozyme delivered for any type of treatment reported to date (by Rossi et al.. 1992 supra) is an |n yjtro transcript having a length of 142 nucleotides. Synthesis of ribozymes greater than 100 nucleotides in length is very difficult using automated methods, and the therapeutic cost of such molecules is prohibitive. Delivery of ribozymes by expression vectors is primarily feasible using only ex vivo treatments. This limits the utility of this approach. In this invention, an alternative approach uses smaller ribozyme motifs and exogenous delivery. The simple structure of these molecules also increases the ability of the ribozyme to invade targeted regions of the mRNA structure. Thus, unlike the situation when the hammerhead structure is included within longer transcripts, there are no non-ribozyme flanking sequences to interfere with correct folding of the ribozyme structure, as well as complementary binding of the ribozyme to the mRNA target.
The enzymatic RNA molecules of this invention can be used to treat human CML or precancerous conditions. Affected animals can be treated at the time of cancer detection or in a prophylactic manner. This timing of treatment will reduce the number of affected cells and disable cellular replication. This is possible because the ribozymes are designed to disable those structures required for successful cellular proliferation.
Ribozymes of this invention block to some extent p210bcr-abl expression and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to tissues in animal models of CML. Ribozyme cleavage of bcr/abl mRNA in these systems may prevent or alleviate disease symptoms or conditions.
The sequence of human bcr/abl mRNA can be screened for accessible sites using a computer folding algorithm. Regions of the mRNA that did not form secondary folding structures and that contain potential hammerhead or hairpin ribozyme cleavage sites can be identified. These sites are shown in Table 29 (All sequences are 5' to 3' in the tables). The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme. The sequences of the chemically synthesized ribozymes most useful in this study are shown in Table 30. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity. For example, stem-loop II sequence of hammerhead ribozymes listed in Table 30 (5'-GGCCGAAAGGCC-3') can be altered (substitution, deletion, and/or insertion) to contain any sequence provided, a minimum of two base-paired stem structure can form. The sequences listed in Tables 30 may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables.
By engineering ribozyme motifs we have designed several ribozymes directed against bcr-abl mRNA sequences. These have been synthesized with modifications that improve their nuclease resistance as described above. These ribozymes cleave bcr-abl target sequences in vitro. The ribozymes are tested for function in vivo by exogenous delivery to cells expressing bcr-abl. Ribozymes are delivered by incorporation into liposomes, by complexing with cationic lipids, by microinjection, or by expression from DNA vectors. Expression of bcr-abl is monitored by ELISA, by indirect immunofluoresence, and/or by FACS analysis. Levels of bcr-abl mRNA are assessed by Northern analysis, RNase protection, by primer extension analysis or by quantitative RT-PCR techniques. Ribozymes that block the induction of p210bcr-abl) protein and mRNA by more than 20% are identified. Example 6: RSV
This invention relates to the use of ribozymes as inhibitors of respiratory syncytial virus (RSV) production, and in particular, the inhibition of RSV replication.
RSV is a member of the virus family paramyxoviridae and is classified under the genus Pneumovirus (for a review see McIntosh and Chanock, 1990 in Virology ed. B.N. Fields, pp. 1045, Raven Press Ltd. NY). The infectious virus particle is composed of a nucleocapsid enclosed within an envelope. The nucleocapsid is composed of a linear negative singlestranded non-segmented RNA associated with repeating subunits of capsid proteins to form a compact structure and thereby protect the RNA from nuclease degradation. The entire nucleocapsid is enclosed by the envelope. The size of the virus particle ranges from 150 - 300 nm in diameter. The complete life cycle of RSV takes place in the cytoplasm of infected cells and the nucleocapsid never reaches the nuclear compartment (Hall, 1990 in Principles and Practice of Infectious Diseases ed. Mandell et al., Churchill Livingstone, NY).
The RSV genome encodes ten viral proteins essential for viral production. RSV protein products include two structural glycoproteins (G and F) found in the envelope spikes, two matrix proteins [M and M2 (22K)] found in the inner membrane, three proteins localized in the nucleocapsid (N, P and L), one protein that is present on the surface of the infected cell (SH), and two nonstructural proteins [NS1 (1C) and NS2 (1 B)] found only in the infected cell. The mRNAs for the 10 RSV proteins have similar 5' and 3' ends. UV-inactivation studies suggest that a single promoter is used with multiple transcription initiation sites (Barik et al., 1992 J. Virol. 66, 6813). The order of transcription corresponding to the protein assignment on the genomic RNA is 1C, 1B, N, P, M, SH, G, F, 22K and L genes (Huang et al., 1985 Virus Res. 2, 157) and transcript abundance corresponds to the order of gene assignment (for example the 1C and 1B mRNAs are much more abundant than the L mRNA. Synthesis of viral message begins immediately after RSV infection of cells and reaches a maximum at 14 hours post-infection (McIntosh and Chanock, supra).
There are two antigenic subgroups of RSV, A and B, which can circulate simultaneously in the community in varying proportions in different years (McIntosh and Chanock, supra). Subgroup A usually predominates. Within the two subgroups there are numerous strains. By the limited sequence analysis available it seems that homology at the nucleotide level is more complete within than between subgroups, although sequence divergence has been noted within subgroups as well. Antigenic determinates result primarily from both surface glycoproteins, F and G. For F, at least half of the neutralization epitopes have been stably maintained over a period of 30 years. For G however, A and B subgroups may be related antigenically by as little as a few percent. On the nucleotide level, however, the majority of the divergence in the coding region of G is found in the sequence for the extracellular domain (Johnson et al., 1987, Proc. Natl. Acad. Sci. USA 84, 5625).
Respiratory Syncytial Virus (RSV) is the major cause of lower respiratory tract illness during infancy and childhood (Hall, supra) and as such is associated with an estimated 90,000 hospitalizations and 4500 deaths in the United States alone (Update: respiratory syncytial virus activity - United States, 1993, Mmwr Morb Mortal Wkly Rep, 42, 971). Infection with RSV generally outranks all other microbial agents leading to both pneumonia and bronchitis. While primarily affecting children under two years of age, immunity is not complete and reinfection of older children and adults, especially hospital care givers (McIntosh and Chanock, supra), is not uncommon. Immunocompromised patients are severely affected and RSV infection is a major complication for patients undergoing bone marrow transplantation .
Uneventftl RSV respiratory disease resembles a common cold and recovery is in 7 to 12 days. Initial symptoms (rhinorrhea, nasal congestion, slight fever, etc.) are followed in 1 to 3 days by lower respiratory tract signs of infection that include a cough and wheezing. In severe cases, these mild symptoms quickly progress to tachypnea, cyanosis, and listlessness and hospitalization is required. In infants with underlying cardiac or respiratory disease, the progression of symptoms is especially rapid and can lead to respiratory failure by the second or third day of illness. With modem intensive care however, overall mortality is usually less than 5% of hospitalized patients (McIntosh and Chanock, supra).
At present, neither an efficient vaccine nor a specific antiviral agent is available. An immune response to the viral surface glycoproteins can provide resistance to RSV in a number of experimental animals, and a subunit vaccine has been shown to be effective for up to 6 months in children previously hospitalized with an RSV infection (Tristam et al., 1993, J. Infect. Dis. 167, 191). An attenuated bovine RSV vaccine has also been shown to be effective in calves for a similar length of time (Kubota et al., 1992 J. Vet. Med. Sci. 54, 957). Previously however, a formalin-inactivated RSV vaccine was implicated in greater frequency of severe disease in subsequent natural infections with RSV (Connors et al., 1992 J. Virol. 66, 7444).
The current treatment for RSV infection requiring hospitalization is the use of aerosolized ribavirin, a guanosine analog [Antiviral Agents and Viral Diseases of Man, 3rd edition. 1990. (eds. G.J. Galasso, R.J. Whitley, and T.C. Merigan) Raven Press Ltd., NY.]. Ribavirin therapy is associated with a decrease in the severity of the symptoms, improved arterial oxygen and a decrease in the amount of viral shedding at the end of the treatment period. It is not certain, however, whether ribavirin therapy actually shortens the patients' hospital stay or diminishes the need for supportive therapies (McIntosh and Chanock, supra). The benefits of ribavirin therapy are especially clear for high risk infants, those with the most serious symptoms or for patients with underlying bronchopulmonary or cardiac disease. Inhibition of the viral polymerase complex is supported as the main mechanism for inhibition of RSV by ribavirin, since viral but not cellular polypeptide synthesis is inhibited by ribavirin in RSV-infected cells (Antiviral Agents and Viral Diseases of Man, 3rd edition. 1990. (eds. G.J. Galasso, R.J. Whitley, and T.C. Merigan) Raven Press Ltd., NY]. Since ribavirin is at least partially effective against RSV infection when delivered by aerosolization, it can be assumed that the target cells are at or near the epithelial surface. In this regard, RSV antigen had not spread any deeper than the superficial layers of the respiratory epithelium in autopsy studies of fatal pneumonia (McIntosh and Chanock, supra). Jennings et al., WO 94/13688 indicates that targets for specific types of ribozymes include respiratory syncytical virus. The invention features novel enzymatic RNA molecules, or ribozymes, and methods for their use for inhibiting production of respiratory syncytial virus (RSV). Such ribozymes can be used in a method for treatment of diseases caused by these related viruses in man and other animals. The invention also features cleavage of the genomic RNA and mRNA of these viruses by use of ribozymes. In particular, the ribozyme molecules described are targeted to the NS1 (1C), NS2 (1B) and N viral genes. These genes are known in the art (for a review see McIntosh and Chanock, 1990 supra ). Ribozymes that cleave the specified sites in RSV mRΝAs represent a novel therapeutic approach to respiratory disorders. Applicant indicates that ribozymes are able to inhibit the activity of RSV and that the catalytic activity of the ribozymes is required for their inhibitory effect. Those of ordinary skill in the art, will find that it is clear from the examples described that other ribozymes that cleave these sites in RSV mRΝAs encoding 1C, 1 B and Ν proteins may be readily designed and are within the invention. Also, those of ordinary skill in the art, will find that it is clear from the examples described that ribozymes cleaving other mRΝAs encoded by RSV (P, M, SH, G, F, 22K and L) and the genomic RΝA may be readily designed and are within the invention.
In preferred embodiments, the ribozymes have binding arms which are complementary to the sequences in Tables 31 , 33, 35, 37 and 38. Examples of such ribozymes are shown in Tables 32, 34, 36-38. Examples of such ribozymes consist essentially of sequences defined in these Tables. By "consists essentially of" is meant that the active ribozyme contains an enzymatic center equivalent to those in the examples, and binding arms able to bind mRΝA such that cleavage at the target site occurs. Other sequences may be present which do not interfere with such cleavage. Ribozymes of this invention block to some extent RSV production and can be used to treat disease or diagnose such disease. Ribozymes will be delivered to cells in culture and to cells or tissues in animal models of respiratory disorders. Ribozyme cleavage of RSV encoded mRΝAs or the genomic RΝA in these systems may alleviate disease symptoms. While all ten RSV encoded proteins (1C, 1B, N, P, M, SH, 22K, F, G, and L) are essential for viral life cycle and are all potential targets for ribozyme cleavage, certain proteins (mRNAs) are more favorable for ribozyme targeting than the others. For example RSV encoded proteins 1C, 1 B, SH and 22K are not found in other members of the family paramyxoviridae and appear to be unique to RSV. In contrast the ectodomain of the G protein and the signal sequence of the F protein show significant sequence divergence at the nucleotide level among various RSV sub-groups (Johnson et al., 1987 supra). RSV proteins 1C, 1 B and N are highly conserved among various subtypes at both the nucleotide and amino acid levels. Also, 1C, 1B and N are the most abundant of all RSV proteins.
The sequence of human RSV mRNAs encoding 1C, 1 B and N proteins are screened for accessible sites using a computer folding algorithm. Hammerhead or hairpin ribozyme cleavage sites were identified. These sites are shown in Tables 31 , 33, 34, 37 and 38 (All sequences are 5' to 3' in the tables.) The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of ribozyme. Ribozymes of the hammerhead or hairpin motif are designed to anneal to various sites in the mRNA message. The binding arms are complementary to the target site sequences described above. The ribozymes are chemically synthesized. The method of synthesis used follows the procedure for normal RNA synthesis as described in Usman et al., 1987 J. Am. Chem. Soc, 109, 7845-7854 and in Scaringe et al., 1990 Nucleic Acids Res., 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end. The average stepwise coupling yields were >98%. Inactive ribozymes were synthesized by substituting a U for G5 and a U for A14 (numbering from Hertel et al., 1992 Nucleic Acids Res., 20, 3252). Hairpin ribozymes are synthesized in two parts and annealed to reconstruct the active ribozyme (Chowrira and Burke, 1992 Nucleic Acids Res., 20, 2835-2840). Hairpin ribozymes are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbeck, 1989, Methods Enzymol. 180, 51). All ribozymes are modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-o-methyl, 2'-H (for a review see Usman and Cedergren, 1992 TIBS 17, 34). Ribozymes are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography and are resuspended in water. The sequences of the chemically synthesized ribozymes useful in this study are shown in Tables 32, 34, 36, 37 and 38. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the ribozyme (all but the binding arms) is altered to affect activity. For example, stem-loop II sequence of hammerhead ribozymes listed in Tables 32 and 34(5'-GGCCGAAAGGCC-3') can be altered (substitution, deletion, and/or insertion) to contain any sequences provided a minimum of two base-paired stem structure can form. Similarly, stem-loop IV sequence of hairpin ribozymes listed in Tables 37 and 38 (5'-CACGUUGUG-3') can be altered (substitution, deletion, and/or insertion) to contain any sequence, provided a minimum of two base-paired stem structure can form. The sequences listed in Tables 32, 34, 36, 37 and 38 may be formed of ribonucleotides or other nucleotides or non-nucleotides. Such ribozymes are equivalent to the ribozymes described specifically in the Tables. By engineering ribozyme motifs we have designed several ribozymes directed against RSV encoded mRNA sequences. These ribozymes are synthesized with modifications that improve their nuclease resistance. The ability of ribozymes to cleave target sequences in vitro is evaluated.
Numerous, common cell lines can be infected with RSV for experimental purposes. These include HeLa, Vero and several primary epithelial cell lines. A cotton rat animal model of experimental human RSV infection is also available, and the bovine RSV is quite homologous to the human viruses. Rapid clinical diagnosis is through the use of kits designed for the immunofluorescence staining of RSV-infected cells or an ELISA assay, both of which are adaptable for experimental study. RSV encoded mRNA levels will be assessed by Northern analysis, RNAse protection, primer extension analysis or quantitative RT-PCR. Ribozymes that block the induction of RSV activity and/or 1C, 1 B and N protein encoding mRNAs by more than 90% will be identified. Optimizing Ribozyme Activity
Ribozyme activity can be optimized as described by Draper et al., PCT WO93/23569. The details will not be repeated here, but include altering the length of the ribozyme binding arms or chemically synthesizing ribozymes with modifications that prevent their degradation by serum ribonucleases (see e.g., Eckstein et al., International Publication No. WO 92/07065; Perrault et al., 1990 Nature 344, 565; Pieken et al., 1991 Science 253, 314; Usman and Cedergren, 1992 Trends in Biochem. Sci. 17, 334; Usman et al., International Publication No. WO 93/15187; and Rossi et al., International Publication No. WO 91/03162, as well as Jennings et al., WO 94/13688, which describe various chemical modifications that can be made to the sugar moieties of enzymatic RNA molecules. All these publications are hereby incorporated by reference herein.), modifications which enhance their efficacy in cells, and removal of stem II bases to shorten RNA synthesis times and reduce chemical requirements.
Sullivan, et al., PCT WO94/02595, incorporated by reference herein, describes the general methods for delivery of enzymatic RNA molecules . Ribozymes may be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres. The RNA/vehicle combination is locally delivered by direct injection or by use of a catheter, infusion pump or stent. Alternative routes of delivery include, but are not limited to, intravenous injection, intramuscular injection, subcutaneous injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. More detailed descriptions of ribozyme delivery and administration are provided in Sullivan, et al., supra and Draper, et al., supra which have been incorporated by reference herein.
Another means of accumulating high concentrations of a ribozyme(s) within cells is to incorporate the ribozyme-encoding sequences into a DNA expression vector. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III). Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990 Proc. Natl. Acad. Sci. U S A. 87, 6743-7: Gao and Huang 1993 Nucleic Acids Res., 21 , 2867-72; Lieber et al., 1993 Methods Enzymol., 217, 47-66; Zhou et al., 1990 Mol. Cell. Biol.. 10, 4529-37). Several investigators have demonstrated that ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992 Antisense Res. Dev., 2, 3-15; Ojwang et al.. 1992 Proc. Natl. Acad. Sci. U S A. 89, 10802-6; Chen et al., 1992 Nucleic Acids Res., 20, 4581-9; Yu et al., 1993 Proc. Natl. Acad. Sci. U S A. 90, 6340-4; L'Huillier et al., 1992 EMBO J. 11. 4411-8; Lisziewicz et al., 1993 Proc. Natl. Acad. Sci. U. S. A., 90, 8000-4). The above ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral, or alpha virus vectors).
In a preferred embodiment of the invention, a transcription unit expressing a ribozyme that cleaves target RNA is inserted into a plasmid
DNA vector, a retrovirus DNA viral vector, an adenovirus DNA viral vector or an adeno-associated virus vector or alpha virus vector. These and other vectors have been used to transfer genes to live animals (for a review see
Friedman, 1989 Science 244, 1275-1281 ; Roemer and Friedman, 1992 Eur. J. Biochem. 208, 211-225) and leads to transient or stable gene expression. The vectors are delivered as recombinant viral particles. DNA may be delivered alone or complexed with vehicles (as described for RNA above). The DNA, DNA/vehicle complexes, or the recombinant virus particles are locally administered to the site of treatment, e.g., through the use of a catheter, stent or infusion pump.
Diagnostic uses
Ribozymes of this invention may be used as diagnostic tools to examine genetic drift and mutations within diseased cells. The close relationship between ribozyme activity and the structure of the target RNA allows the detection of mutations in any region of the molecule which alters the base-pairing and three-dimensional structure of the target RNA. By using multiple ribozymes described in this invention, one may map nucleotide changes which are important to RNA structure and function in vitro, as well as in cells and tissues. Cleavage of target RNAs with ribozymes may be used to inhibit gene expression and define the role (essentially) of specified gene products in the progression of disease. In this manner, other genetic targets may be defined as important mediators of the disease. These experiments will lead to better treatment of the disease progression by affording the possibility of combinational therapies (e.g.. multiple ribozymes targeted to different genes, ribozymes coupled with known small molecule inhibitors, or intermittent treatment with combinations of ribozymes and/or other chemical or biological molecules). Other in vitro uses of ribozymes of this invention are well known in the art, and include detection of the presence of mRNA associated with ICAM-1 , relA, TNF-α, p210, bcr-abl or RSV related condition. Such RNA is detected by determining the presence of a cleavage product after treatment with a ribozyme using standard methodology.
In a specific example, ribozymes which can cleave only wild-type or mutant forms of the target RNA are used for the assay. The first ribozyme is used to identify wild-type RNA present in the sample and the second ribozyme will be used to identify mutant RNA in the sample. As reaction controls, synthetic substrates of both wild-type and mutant RNA will be cleaved by both ribozymes to demonstrate the relative ribozyme efficiencies in the reactions and the absence of cleavage of the "non-targeted" RNA species. The cleavage products from the synthetic substrates will also serve to generate size markers for the analysis of wild-type and mutant RNAs in the sample population. Thus each analysis will require two ribozymes, two substrates and one unknown sample which will be combined into six reactions. The presence of cleavage products will be determined using an RNAse protection assay so that full-length and cleavage fragments of each RNA can be analyzed in one lane of a polyacrylamide gel. It is not absolutely required to quantify the results to gain insight into the expression of mutant RNAs and putative risk of the desired phenotypic changes in target cells. The expression of mRNA whose protein product is implicated in the development of the phenotype (i.e., ICAM-1 , rel A, TNFα, p210bcr-abl or RSV) is adequate to establish risk. If probes of comparable specific activity are used for both transcripts, then a qualitative comparison of RNA levels will be adequate and will decrease the cost of the initial diagnosis. Higher mutant form to wild-type ratios will be correlated with higher risk whether RNA levels are compared qualitatively or quantitatively.
II. Chemical Synthesis Of Ribozymes
There follows the chemical synthesis, deprotection, and purification of
RNA, enzymatic RNA or modified RNA molecules in greater than milligram quantities with high biological activity. Applicant has determined that the synthesis of enzymatically active RNA in high yield and quantity is dependent upon certain critical steps used during its preparation. Specifically, it is important that the RNA phosphoramidites are coupled efficiently in terms of both yield and time, that correct exocyclic amino protecting groups be used, that the appropriate conditions for the removal of the exocyclic amino protecting groups and the alkylsilyl protecting groups on the 2'-hydroxyl are used, and that the correct work-up and purification procedure of the resulting ribozyme be used.
To obtain a correct synthesis in terms of yield and biological activity of a large RNA molecule (i.e., about 30 to 40 nucleotide bases), the protection of the amino functions of the bases requires either amide or substituted amide protecting groups, which must be, on the one hand, stable enough to survive the conditions of synthesis, and on the other hand, removable at the end of the synthesis. These requirements are met by the amide protecting groups shown in Figure 8, in particular, benzoyl for adenosine, isobutyryl or benzoyl for cytidine, and isobutyryl for guanosine, which may be removed at the end of the synthesis by incubating the RNA in NH3/EtOH (ethanolic ammonia) for 20 h at 65 °C. In the case of the phenoxyacetyl type protecting groups shown in Figure 8 on guanosine and adenosine and acetyl protecting groups on cytidine, an incubation in ethanolic ammonia for 4 h at 65 °C is used to obtain complete removal of these protecting groups. Removal of the alkylsilyl 2'-hydroxyl protecting groups can be accomplished using a tetrahydrofuran solution of TBAF at room temperature for 8-24 h.
The most quantitative procedure for recovering the fully deprotected
RNA molecule is by either ethanol precipitation, or an anion exchange cartridge desalting, as described in Scaringe et al. Nucleic Acids Res. 1990, 18, 5433-5341. The purification of the long RNA sequences may be accomplished by a two-step chromatographic procedure in which the molecule is first purified on a reverse phase column with either the trityl group at the 5' position on or off. This purification is accomplished using an acetonitrile gradient with triethylammonium or bicarbonate salts as the aqueous phase. In the case of the trityl on purification, the trityl group may be removed by the addition of an acid and drying of the partially purified RNA molecule. The final purification is carried out on an anion exchange column, using alkali metal perchlorate salt gradients to elute the fully purified RNA molecule as the appropriate metal salts, e.g. Na+, Li+ etc. A final de-salting step on a small reverse-phase cartridge completes the purification procedure. Applicant has found that such a procedure not only fails to adversely affect activity of a ribozyme, but may improve its activity to cleave target RNA molecules.
Applicant has also determined that significant (see Tables 39-41) improvements in the yield of desired full length product (FLP) can be obtained by:
1. Using 5-S-alkyltetrazole at a delivered or effective concentration of 0.25-0.5 M or 0.15-0.35 M for the activation of the RNA (or analogue) amidite during the coupling step. (By delivered is meant that the actual amount of chemical in the reaction mix is known. This is possible for large scale synthesis since the reaction vessel is of size sufficient to allow such manipulations. The term effective means that available amount of chemical actually provided to the reaction mixture that is able to react with the other reagents present in the mixture. Those skilled in the art will recognize the meaning of these terms from the examples provided herein.) The time for this step is shortened from 10-15 m, vide supra, to 5-10 m. Alkyl, as used herein, refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, =O, =S, NO2 or N(CH3)2, amino, or SH. The term also includes alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkenyl group has 1 to 12 carbons. More preferably it is a lower alkenyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkenyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, =O, =S, NO2, halogen, N(CH3)2, amino, or SH. The term "alkyl" also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkynyl group has 1 to 12 carbons. More preferably it is a lower alkynyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkynyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, =O, =S, NO2 or N(CH3)2, amino or SH.
Such alkyl groups may also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups. An "aryl" group refers to an aromatic group which has at least one ring having a conjugated π electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted. The preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups. An "alkylaryl" group refers to an alkyl group (as described above) covalently joined to an aryl group (as described above. Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted. Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted. An "amide" refers to an -C(O)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen. An "ester" refers to an -C(O)-OR', where R is either alkyl, aryl, alkylaryl or hydrogen. 2. Using 5-S-alkyltetrazole at an effective, or final, concentration of 0.1-0.35 M for the activation of the RNA (or analogue) amidite during the coupling step. The time for this step is shortened from 10-15 m, vide supra, to 5-10 m.
3. Using alkylamine (MA, where alkyl is preferably methyl, ethyl, propyl or butyl) or NH4OH/alkylamine (AMA, with the same preferred alkyl groups as noted for MA) @ 65 °C for 10-15 m to remove the exocyclic amino protecting groups (vs 4-20 h @ 55-65 °C using NH4OH/EtOH or NH3/EtOH, vide supra). Other alkylamines, e.g. ethylamine, propylamine, butylamine etc. may also be used.
4. Using anhydrous triethylamine•hydrogen fluoride (aHF•TEA) @ 65 °C for 0.5-1.5 h to remove the 2'-hydroxyl alkylsilyl protecting group (vs 8 - 24 h using TBAF, vide supra or TEA•3HF for 24 h (Gasparutto et al. Nucleic Acids Res. 1992 , 20, 5159-5166). Other alkylamine•HF complexes may also be used, e.g. trimethylamine or diisopropylethylamine.
5. The use of anion-exchange resins to purify and/or analyze the fully deprotected RNA. These resins include, but are not limited to, quartenary or tertiary amino derivatized stationary phases such as silica or polystyrene. Specific examples include Dionex-NA100®, Mono-Q®, Poros-Q®.
Thus, the invention features an improved method for the coupling of RNA phosphoramidites; for the removal of amide or substituted amide protecting groups; and for the removal of 2'-hydroxyl alkylsilyl protecting groups. Such methods enhance the production of RNA or analogs of the type described above (e.g., with substituted 2'-groups), and allow efficient synthesis of large amounts of such RNA. Such RNA may also have enzymatic activity and be purified without loss of that activity. While specific examples are given herein, those in the art will recognize that equivalent chemical reactions can be performed with the alternative chemicals noted above, which can be optimized and selected by routine experimentation.
In another aspect, the invention features an improved method for the purification or analysis of RNA or enzymatic RNA molecules (e.g. 28-70 nucleotides in length) by passing said RNA or enzymatic RNA molecule over an HPLC, e.g., reverse phase and/or an anion exchange chromatography column. The method of purification improves the catalytic activity of enzymatic RNAs over the gel purification method (see Figure 10). Draper et al., PCT WO93/23569, incorporated by reference herein, disclosed reverse phase HPLC purification. The purification of long RNA molecules may be accomplished using anion exchange chromatography, particularly in conjunction with alkali perchlorate salts. This system may be used to purify very long RNA molecules. In particular, it is advantageous to use a Dionex NucleoPak 100© or a Pharmacia Mono Q® anion exchange column for the purification of RNA by the anion exchange method. This anion exchange purification may be used following a reverse-phase purification or prior to reverse phase purification. This method results in the formation of a sodium salt of the ribozyme during the chromatography. Replacement of the sodium alkali earth salt by other metal salts, e.g., lithium, magnesium or calcium perchlorate, yields the corresponding salt of the RNA molecule during the purification.
In the case of the 2-step purification procedure, in which the first step is a reverse phase purification followed by an anion exchange step, the reverse phase purification is best accomplished using polymeric, e.g. polystyrene based, reverse-phase media, using either a 5'-trityl-on or 5'-trityl-off method. Either molecule may be recovered using this reverse-phase method, and then, once detritylated, the two fractions may be pooled and then submitted to an anion exchange purification step as described above.
The method includes passing the enzymatically active RNA molecule over a reverse phase HPLC column; the enzymatically active RNA molecule is produced in a synthetic chemical method and not by an enzymatic process; and the enzymatic RNA molecule is partially blocked, and the partially blocked enzymatically active RNA molecule is passed over a reverse phase HPLC column to separate it from other RNA molecules.
In more preferred embodiments, the enzymatically active RNA molecule, after passage over the reverse phase HPLC column, is deprotected and passed over a second reverse phase HPLC column (which may be the same as the reverse phase HPLC column), to remove the enzymatic RNA molecule from other components. In addition, the column is a silica or organic polymer-based C4, C8 or C18 column having a porosity of at least 125 A, preferably 300 A, and a particle size of at least 2 μm, preferably 5 μm.
Activation
The synthesis of RNA molecules may be accomplished chemically or enzymatically. In the case of chemical synthesis the use of tetrazole as an activator of RNA phosphoramidites is known (Usman et al. J. Am. Chem. Soc. 1987, 109, 7845-7854). In this, and subsequent reports, a 0.5 M solution of tetrazole is allowed to react with the RNA phosphoramidite and couple with the polymer bound 5'-hydroxyl group for 10 m. Applicant has determined that using 0.25-0.5 M solutions of 5-S-alkyltetrazoies for only 5 min gives equivalent or better results. The following exemplifies the procedure.
Example 7: Synthesis of RNA and Ribozymes Using 5-S-Alkyltetrazoles as Activating Agent
The method of synthesis used follows the general procedure for RNA synthesis as described in Usman et al., 1987 supra and in Scaringe et al., Nucleic Acids Res. 1990, 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end. The major difference used was the activating agent, 5-S-ethyl or -methyltetrazole @ 0.25 M concentration for 5 min.
All small scale syntheses were conducted on a 394 (ABI) synthesizer using a modified 2.5 μmol scale protocol with a reduced 5 min coupling step for alkylsilyl protected RNA and 2.5 m coupling step for 2'-O-methylated RNA. A 6.5-fold excess (162.5 μL of 0.1 M = 32.5 μmol) of phosphoramidite and a 40-fold excess of S-ethyl tetrazole (400 μL of 0.25 M = 100 μmol) relative to polymer-bound 5'-hydroxyl was used in each coupling cycle. Average coupling yields on the 394, determined by colorimetric quantitation of the trityl fractions, was 97.5-99%. Other oligonucleotide synthesis reagents for the 394: Detritylation solution was 2% TCA in methylene chloride; capping was performed with 16% N-Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I2, 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. S-Ethyl tetrazole solution (0.25 M in acetonitrile) was made up from the solid obtained from Applied Biosystems.
All large scale syntheses were conducted on a modified (eight amidite port capacity) 390Z (ABI) synthesizer using a 25 μmol scale protocol with a
5-15 min coupling step for alkylsilyl protected RΝA and 7.5 m coupling step for 2'-O-methylated RΝA. A six-fold excess (1.5 mL of 0.1 M = 150 μmol) of phosphoramidite and a forty-five-fold excess of S-ethyl tetrazole (4.5 mL of 0.25 M = 1125 μmol) relative to polymer-bound 5'-hydroxyl was used in each coupling cycle. Average coupling yields on the 390Z, determined by colorimetric quantitation of the trityl fractions, was 95.0-96.7%. Oligonucleotide synthesis reagents for the 390Z: Detritylation solution was 2% DCA in methylene chloride; capping was performed with 16% N-Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I2, 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. S-Ethyl tetrazole solution (0.25-0.5 M in acetonitrile) was made up from the solid obtained from Applied Biosystems.
Deprotection
The first step of the deprotection of RΝA molecules may be accomplished by removal of the exocyclic amino protecting groups with either ΝH4OH/EtOH:3/1 (Usman et al. J. Am. Chem. Soc. 1987, 109, 7845-7854) or NH3/EtOH (Scaringe et al. Nucleic Acids Res. 1990, 18, 5433-5341) for ~20 h @ 55-65 °C. Applicant has determined that the use of methylamine or NH4OH/methylamine for 10-15 min @ 55-65 °C gives equivalent or better results. The following exemplifies the procedure.
Example 8: RNA and Ribozyme Deprotection of Exocyclic Amino
Protecting Groups Using Methylamine (MA) or NH4OH/Methylamine (AMA)
The polymer-bound oligonucleotide, either trityl-on or off, was suspended in a solution of methylamine (MA) or NH4OH/methylamine (AMA) @ 55-65 °C for 5-15 min to remove the exocyclic amino protecting groups. The polymer-bound oligoribonucleotide was transferred from the synthesis column to a 4 mL glass screw top vial. NH4OH and aqueous methylamine were pre-mixed in equal volumes. 4 mL of the resulting reagent was added to the vial, equilibrated for 5 m at RT and then heated at 55 or 65 °C for 5-15 min. After cooling to -20 °C, the supernatant was removed from the polymer support. The support was washed with 1.0 mL of EtOH:MeCN:H2O/3:1 :1 , vortexed and the supernatant was then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, were dried to a white powder. The same procedure was followed for the aqueous methylamine reagent.
Table 40 is a summary of the results obtained using the improvements outlined in this application for base deprotection. The second step of the deprotection of RNA molecules may be accomplished by removal of the 2'-hydroxyl alkylsilyl protecting group using TBAF for 8-24 h (Usman et al. J. Am. Chem. Soc. 1987, 109, 7845-7854). Applicant has determined that the use of anhydrous TEA•HF in N-methylpyrrolidine (ΝMP) for 0.5-1.5 h @ 55-65 °C gives equivalent or better results. The following exemplifies this procedure.
Example 9: RΝA and Ribozyme Deprotection of 2'-Hydroxyl Alkylsilyl Protecting Groups Using Anhydrous TEA•HF
To remove the alkylsilyl protecting groups, the ammonia-deprotected oligoribonucleotide was resuspended in 250 μL of 1.4 M anhydrous HF solution (1.5 mL N-methylpyrrolidine, 750 μL TEA and 1.0 mL TEA•3HF) and heated to 65 °C for 1.5 h. 9 mL of 50 mM TEAB was added to quench the reaction. The resulting solution was loaded onto a Qiagen 500® anion exchange cartridge (Qiagen Inc.) prewashed with 10 mL of 50 mM TEAB. After washing the cartridge with 10 mL of 50 mM TEAB, the RΝA was eluted with 10 mL of 2 M TEAB and dried down to a white powder.
Table 41 is a summary of the results obtained using the improvements outlined in this application for alkylsilyl deprotection.
Example 10: HPLC Purification, Anion Exchange column
For a small scale synthesis, the crude material was diluted to 5 mL with diethylpyrocarbonate treated water. The sample was injected onto either a Pharmacia Mono Q® 16/10 or Dionex ΝucleoPac® column with 100% buffer A (10 mM ΝaClO4). A gradient from 180-210 mM NaClO4 at a rate of 0.85 mM/void volume for a Pharmacia Mono Q® anion-exchange column or 100-150 mM NaClO4 at a rate of 1.7 mM/void volume for a Pionex NucleoPac® anion-exchange column was used to elute the RNA. Fractions were analyzed by a HP-1090 HPLC with a Dionex NucleoPac® column. Fractions containing full length product at >80% by peak area were pooled. For a trityl-off large scale synthesis, the crude material was desalted by applying the solution that resulted from quenching of the desilylation reaction to a 53 mL Pharmacia HiLoad 26/10 Q-Sepharose® Fast Flow column. The column was thoroughly washed with 10 mM sodium perchlorate buffer. The oligonucleotide was eluted from the column with 300 mM sodium perchlorate. The eluent was quantitated and an analytical HPLC was run to determine the percent full length material in the synthesis. The eluent was diluted four fold in sterile H2O to lower the salt concentration and applied to a Pharmacia Mono Q® 16/10 column. A gradient from 10-185 mM sodium perchlorate was run over 4 column volumes to elute shorter sequences, the full length product was then eluted in a gradient from 185-214 mM sodium perchlorate in 30 column volumes. The fractions of interest were analyzed on a HP-1090 HPLC with a Pionex NucleoPac® column. Fractions containing over 85% full length material were pooled. The pool was applied to a Pharmacia RPC® column for desalting.
For a trityl-on large scale synthesis, the crude material was desalted by applying the solution that resulted from quenching of the desilylation reaction to a 53 mL Pharmacia HiLoad 26/10 Q-Sepharose® Fast Flow column. The column was thoroughly washed with 20 mM NH4CO3H/10% CH3CN buffer. The oligonucleotide was eluted from the column with 1.5 M NH4CO3H/10% acetonitrile. The eluent was quantitated and an analytical HPLC was run to determine the percent full length material present in the synthesis. The oligonucleotide was then applied to a Pharmacia Resource RPC column. A gradient from 20-55% B (20 mM NH4CO3H/25% CH3CN, buffer A = 20 mM NH4CO3H/10% CH3CN) was run over 35 column volumes. The fractions of interest were analyzed on a HP-1090 HPLC with a Dionex NucleoPac® column. Fractions containing over 60% full length material were pooled. The pooled fractions were then submitted to manual detritylation with 80% acetic acid, dried down immediately, resuspended in sterile H2O, dried down and resuspended in H2O again. This material was analyzed on a HP 1090-HPLC with a Dionex NucleoPac® column. The material was purified by anion exchange chromatography as in the trityl-off scheme (vide supra). Example 11 Ribozyme Activity Assay
Purified 5'-end labeled RNA substrates (15-25-mers) and purified 5'-end labeled ribozymes (~36-mers) were both heated to 95 °C, quenched on ice and equilibrated at 37 °C, separately. Ribozyme stock solutions were 1 μM, 200 nM, 40 nM or 8 nM and the final substrate RNA concentrations were - 1 nM. Total reaction volumes were 50 μL. The assay buffer was 50 mM Tris-Cl, pH 7.5 and 10 mM MgCl2. Reactions were initiated by mixing substrate and ribozyme solutions at t = 0. Aliquots of 5 μL were removed at time points of 1 , 5, 15, 30, 60 and 120 m. Each aliquot was quenched in formamide loading buffer and loaded onto a 15% denaturing polyacrylamide gel for analysis. Quantitative analyses were performed using a phosphorimager (Molecular Dynamics).
Example 12: One pot deprotection of RNA
Applicant has shown that aqueous methyl amine is an efficient reagent to deprotect bases in an RNA molecule. However, in a time consuming step (2-24 hrs), the RNA sample needs to be dried completely prior to the deprotection of the sugar 2'-hydroxyl groups. Additionally, deprotection of RNA synthesized on a large scale (e.g., 100 μmol) becomes challenging since the volume of solid support used is quite large. In an attempt to minimize the time required for deprotection and to simplify the process of deprotection of RNA synthesized on a large scale, applicant describes a one pot deprotection protocol (Fig. 12). According to this protocol, anhydrous methylamine is used in place of aqueous methyl amine. Base deprotection is carried out at 65 °C for 15 min and the reaction is allowed to cool for 10 min. Deprotection of 2'-hydroxyl groups is then carried out in the same container for 90 min in a TEA•3HF reagent. The reaction is quenched with 16 mM TEAB solution.
Referring to Fig. 13. hammerhead ribozyme targeted to site B is synthesized using RNA phosphoramadite chemistry and deprotected using either a two pot or a one pot protocol. Profiles of these ribozymes on an HPLC column are compared. The figure shows that RNAs deprotected by either the one pot or the two pot protocols yield similar full-length product profiles. Applicant has shown that using a one pot deprotection protocol, time required for RNA deprotection can be reduced considerably without compromising the quality or the yield of full length RNA.
Referring to Fig. 14, hammerhead ribozymes targeted to site B (from Fig. 13) are tested for their ability to cleave RNA. As shown in the figure 14. ribozymes that are deprotected using one pot protocol have catalytic activity comparable to ribozymes that are deprotected using a two pot protocol. Example 12a. Improved protocol for the synthesis of phosphorothioate containing RNA and ribozymes using 5-S-Alkyltetrazoles as Activating Agent
The two sulfurizing reagents that have been used to synthesize ribophosphorothioates are tetraethylthiuram disulfide (TETD; Vu and Hirschbein, 1991 Tetrahedron Letter 31 , 3005), and 3H-1 ,2-benzodithiol-3-one 1 ,1-dioxide (Beaucage reagent; Vu and Hirschbein, 1991 supra). TETD requires long sulfurization times (600 seconds for DNA and 3600 seconds for RNA). It has recently been shown that for sulfurization of DNA oligonucleotides, Beaucage reagent is more efficient than TETD (Wyrzykiewicz and Ravikumar, 1994 Bioorganic Med. Chem. 4, 1519). Beaucage reagent has also been used to synthesize phosphorothioate oligonucleotides containing 2'-deoxy-2'-fluoro modifications wherein the wait time is 10 min (Kawasaki et al., 1992 J. Med. Chem). The method of synthesis used follows the procedure for RNA synthesis as described herein and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end. The sulfurization step for RNA described in the literature is a 8 second delivery and 10 min wait steps (Beaucage and Iyer, 1991 Tetrahedron 49, 6123). These conditions produced about 95% sulfurization as measured by HPLC analysis (Morvan et al., 1990 Tetrahedron Letter 31 , 7149). This 5% contaminating oxidation could arise from the presence of oxygen dissolved in solvents and/or slow release of traces of iodine adsorbed on the inner surface of delivery lines during previous synthesis.
A major improvement is the use of an activating agent, 5-S-ethyltetrazole or 5-S-methyltetrazole at a concentration of 0.25 M for 5 min. Additionally, for those linkages which are phosporothioate, the iodine solution is replaced with a 0.05 M solution of 3H-1 ,2-benzodithiole-3-one 1 ,1-dioxide (Beaucage reagent) in acetonitrile. The delivery time for the sulfurization step is reduced to 5 seconds and the wait time is reduced to 300 seconds.
RNA synthesis is conducted on a 394 (ABI) synthesizer using a modified 2.5 μmol scale protocol with a reduced 5 min coupling step for alkylsilyl protected RNA and 2.5 min coupling step for 2'-O-methylated
RNA. A 6.5-fold excess (162.5 μL of 0.1 M = 32.5 μmol) of phosphoramidite and a 40-fold excess of S-ethyl tetrazole (400 μL of 0.25 M = 100 μmol) relative to polymer-bound 5'-hydroxyl was used in each coupling cycle. Average coupling yields on the 394 synthesizer, determined by colorimetric quantitation of the trityl fractions, was 97.5-99%. Other oligonucleotide synthesis reagents for the 394 synthesizer: detritylation solution was 2% TCA in methylene chloride; capping was performed with 16% N-Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I2, 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. S-Ethyl tetrazole solution (0.25 M in acetonitrile) was made up from the solid obtained from Applied Biosystems. Sulfurizing reagent was obtained from Glen Research.
Average sulfurization efficiency (ASE) is determined using the formula: ASE = (PS/Total) 1/n-1 where, PS = integrated 31 P ΝMR values of the P=S diester
Total = integration value of all peaks n = length of oligo
Referring to tables 42 and 43, effects of varying the delivery and the wait time for sulfurization with Beaucage's reagent is described. These data suggest that 5 second wait time and 300 second delivery time is the condition under which ASE is maximum.
Using the above conditions a 36 mer hammerhead ribozyme is synthesized which is targeted to site C. The ribozyme is synthesized to contain phosphorothioate linkages at four positions towards the 5' end. RΝA cleavage activity of this ribozyme is shown in Fig. 16. Activity of the phosphorothioate ribozyme is comparable to the activity of a ribozyme lacking any phosphorothioate linkages.
Example 13: Protocol for the synthesis of 2'-Ν-phtalimido-nucleoside phosphoramidite The 2'-amino group of a 2'-deoxy-2'-amino nucleoside is normally protected with N-(9-flourenylmethoxycarbonyl) (Fmoc; Imazawa and Eckstein, 1979 supra; Pieken et al., 1991 Science 253, 314). This protecting group is not stable in CH3CN solution or even in dry form during prolonged storage at -20 °C. These problems need to be overcome in order to achieve large scale synthesis of RNA.
Applicant describes the use of alternative protecting groups for the 2'-amino group of 2'-deoxy-2'-amino nucleoside. Referring to Figure 17, phosphoramidite 17 was synthesized starting from 2'-deoxy-2'-aminonucleoside (12) using transient protection with Markevich reagent (Markiewicz J. Chem. Res. 1979, S, 24). An intermediate 13 was obtained in 50% yield, however subsequent introduction of N-phtaloyl (Pht) group by Nefken's method (Nefkens, 1960 Nature 185, 306), desilylation (15), dimethoxytrytilation (16) and phosphitylation led to phosphoramidite 17. Since overall yield of this multi-step procedure was low (20%) applicant investigated some alternative approaches, concentrating on selective introduction of N-phtaloyl group without acylation of 5' and 3' hydroxyls.
When 2'-deoxy-2'-amino-nucleoside was reacted with 1.05 equivalents of Nefkens reagent in DMF overnight with subsequent treatment with Et3N (1 hour) only 10-15% of N and 5'(3')-bis-phtaloyl derivatives were formed with the major component being N-Pht-derivative 15. The N,O-bis by-products could be selectively and quantitively converted to N-Pht derivative 15 by treatment of crude reaction mixture with cat. KCN/MeOH.
A convenient "one-pot" procedure for the synthesis of key intermediate 16 involves selective N-phthaloylation with subsequent dimethoxytrytilation by DMTCI/Et3N and resulting in the preparation of DMT derivative 16 in 85% overall yield as follows. Standard phosphytilation of 16 produced phosphoramidite 17 in 87% yield. One gram of 2'-amino nucleoside, for example 2'-amino uridine (US Biochemicals® part # 77140) was co-evaporated twice from dry dimethyl formamide (Dmf) and dried in vacuo overnight. 50 mis of Aldrich sure-seal Dmf was added to the dry 2'-amino uridine via syringe and the mixture was stirred for 10 minutes to produce a clear solution. 1 .0 grams (1.05 eq.) of N-carbethoxyphthalimide (Nefken's reagent, 98% Jannsen Chimica) was added and the solution was stirred overnight. Thin layer chromatography (TLC) showed 90% conversion to a faster moving products (10% ETOH in CHCl3) and 57 μl of TEA (0.1 eq.) was added to effect closure of the phthalimide ring. After 1 hour an additional 855 μl (1.5 eq.) of TEA was added followed by the addition of 1.53 grams (1.1 eq.) of DMT-Cl (Lancaster Synthesis®, 98%). The reaction mixture was left to stir overnight and quenched with ETOH after TLC showed greater than 90% desired product. Pmf was removed under vacuum and the mixture was washed with sodium bicarbonate solution (5% aq., 500 mls) and extracted with ethyl acetate (2× 200 mls). A 25mm × 300mm flash column (75 grams Merck flash silica) was used for purification. Compound eluted at 80 to 85% ethyl acetate in hexanes (yield: 80% purity: >95% by 1 HNMR). Phosphoramidites were then prepared using standard protocols described above. With phosphoramidite 17 in hand applicant synthesized several ribozymes with 2'-deoxy-2'-amino modifications. Analysis of the synthesis demonstrated coupling efficiency in 97-98% range. RNA cleavage activity of ribozymes containing 2'-deoxy-2'-amino-U modifications at U4 and/or U7 positions (see Figure 1), wherein the 2'-amino positions were either protected with Fmoc or Pht, was identical. Additionally, complete deprotection of 2'-deoxy-2'-amino-Uridine was confirmed by base-composition analysis. The coupling efficiency of phosphoramidite 17 was not effected over prolonged storage (1-2 months) at low temperatures.
Protecting 2' Position with a SEM Group There follows a method using the 2'-(trimethylsilyl)ethoxymethyl protecting group (SEM) in the synthesis of oligoribonucleotides, and in particular those enzymatic molecules described above. For the synthesis of RNA it is important that the 2'-hydroxyl protecting group be stable throughout the various steps of the synthesis and base deprotection. At the same time, this group should also be readily removed when desired. To that end the t-butyldimethylsilyl group has been efficacious (Usman, N.; Ogilvie.K.K.; Jiang, M.-Y.; Cedergren.R.J. J. Am. Chem. Soc. 1987, 109, 7845-7854 and Scaringe, S.A.; Franklyn.C.; Usman, N. Nucl. Acids Res. 1990, 18, 5433-5441 ). However, long exposure times to tetra-n-butylammonium fluoride (TBAF) are generally required to fully remove this protecting group from the 2'-hydroxyl. In addition, the bulky alkyl substituents can prove to be a hindrance to coupling thereby necessitating longer coupling times. Finally, it has been shown that the TBDMS group is base labile and is partially deprotected during treatment with ethanolic ammonia (Scaringe, S.A.; Franklyn.C.; Usman, N. Nucl. Acids Res. 1990, 18, 5433-5441 and Stawinski,J.; Stromberg.R.; Thelin.M.; Westman.E. Nucleic Acids Res. 1988, 16, 9285-9298).
The (trimethylsilyl)ethoxymethyl ether (SEM) seems a suitable substitute. This protecting group is stable to base and all but the harshest acidic conditions. Therefore it is stable under the conditions required for oligonucleotide synthesis. It can be readily introduced and the oxygen carbon bond makes it unable to migrate. Finally, the SEM group can be removed with BF3•OEt2 very quickly.
There follows a method for synthesis of RNA by protecting the 2'-position of a nucleotide during RNA synthesis with a
(trimethylsilyl)ethoxymethyl (SEM) group. The method can involve use of standard RNA synthesis conditions as discussed below, or any other equivalent steps. Those in the art are familiar with such steps. The nucleotide used can be any normal nucleotide or may be substituted in various positions by methods well known in the art, e.g., as described by
Eckstein et al., International Publication No. WO 92/07065, Perrault et al.,
Nature 1990, 344, 565-568, Pieken et al., Science 1991 , 253, 314-317,
Usman, N.; Cedergren.R.J. Trends in Biochem. Sci. 1992, 17, 334-339,
Usman et al., PCT W093/15187, and Sproat.B. European Patent Application 92110298.4 .
This invention also features a method for covalently linking a SEM group to the 2'-position of a nucleotide. The method involves contacting a nucleoside with an SEM-containing molecule under SEM bonding conditions. In a preferred embodiment, the conditions are dibutyltin oxide, tetrabutylammonium fluoride and SEM-Cl. Those in the art, however, will recognize that other equivalent conditions can also be used.
In another aspect, the invention features a method for removal of an SEM group from a nucleoside molecule or an oligonucleotide. The method involves contacting the molecule or oligonucleotide with boron trifluoride etherate (BF3•OEt2) under SEM removing conditions, e.g., in acetonitrile.
Referring to Figure 18. there is shown the method for solid phase synthesis of RNA. A 2',5'-protected nucleotide is contacted with a solid phase bound nucleotide under RNA synthesis conditions to form a dinucleotide. The protecting group (R) at the 2'-position in prior art methods can be a silyl ether, as shown in the Figure. In the method of the present invention, an SEM group is used in place of the silyl ether. Otherwise RNA synthesis can be performed by standard methodology.
Referring to Figure 19. there is shown the synthesis of 2'-O-SEM protected nucleosides and phosphoramadites. Briefly, a 5'-protected nucleoside (1 ) is protected at the 2'- or 3'-position by contacting with a derivative of SEM under appropriate conditions. Specifically, those conditions include contacting the nucleoside with dibutyltin oxide and SEM chloride. The 2 regioisomers are separated by chromatography and the 2'-protected moiety is converted into a phosphoramidite by standard procedure. The 3'-protected nucleoside is converted into a succinate derivative suitable for derivatization of a solid support.
Referring to Figure 20. a prior art method for deprotection of RNA using silyl ethers is shown. This contrasts with the method shown in Figure 21 in which deprotection of RNA containing an SEM group is performed. In step 1 , the base protecting groups and cyanoethyl groups are removed by standard procedure. The SEM group is then removed as shown in the Figure. The details of the synthesis of phosphoramidites and SEM protected nucleosides and their use in synthesis of oligonucleotides and subsequent deprotection of
Example 14: Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl)-5'-O- Dimethoxytrityl Uridine (2)
Referring to Figure 19. 5'-O-dimethoxytrityl uridine 1 (1.0 g, 1.83 mmol) in CH3CN (18 mL) was added dibutyltin oxide (1.0 g, 4.03 mmol) and TBAF (1 M, 2.38 mL, 2.38 mmol). The mixture was stirred for 2 h at RT (about 20-25°C) at which time (trimethylsilyl)ethoxymethyl chloride (SEM- Cl) (487 μL, 2.75 mmol) was added. The reaction mixture was stirred overnight and then filtered and evaporated. Flash chromatography (30% hexanes in ethyl acetate) yielded 347 mg (28.0%) of 2'-hydroxyl protected nucleoside 2 and 314 mg (25.3%) of 3'-hydroxyl protected nucleoside 3.
Example 15: Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl) Uridine (4)
Nucleoside 2 was detritylated following standard methods, as shown in Figure 19. Example 16: Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl)-5',3'-O-Acetyl Uridine (5)
Nucleoside 4 was acetylated following standard methods, as shown in Figure 19. Example 17: Synthesis of 5',3'-O-Acetyl Uridine (6)
Referring to Figure 19. the fully protected uridine 5 (32 mg, 0.07 mmol) was dissolved in CH3CN (700 μL) and BF3•OEt2 (17.5 μL, 0.14 mmol) was added. The reaction was stirred 15 m and MeOH was added to quench the reaction. Flash chromatography (5% MeOH in CH2Cl2) gave 20 mg (88%) of SEM deprotected nucleoside 6.
Example 18: Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl)-3'-O-Succinyl-5'-O- Dimethoxytrityl Uridine (2)
Nucleoside 3 was succinylated and coupled to the support following standard procedures, as shown in Figure 19. Example 19: Synthesis of 2'-O-((trimethylsilyl)ethoxymethyl)-5'-O- Dimethoxytrityl Uridine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (8)
Nucleoside 3 was phosphitylated following standard methods, as shown in Figure 19. Example 20: Synthesis of RNA Using 2'-O-SEM Protection
Referring to Figure 18. the method of synthesis used follows the general procedure for RNA synthesis as described in Usman, N.; Ogilvie.K.K.; Jiang, M.-Y.; Cedergren.R.J. J. Am. Chem. Soc. 1987, 109, 7845-7854 and in Scaringe, S.A.; Franklyn.C; Usman, N. Nucl. Acids Res. 1990, 18, 5433-5441. The phosphoramidite 8 was coupled following standard RNA methods to provide a 10-mer of uridylic acid. Syntheses were conducted on a 394 (ABI) synthesizer using a modified 2.5 μmol scale protocol with a 10 m coupling step. A thirteen-fold excess (325 μL of 0.1 M = 32.5 μmol) of phosphoramidite and a 80-fold excess of tetrazole (400 μL of 0.5 M = 200 μmol) relative to polymer-bound 5'-hydroxyl was used in each coupling cycle. Average coupling yields on the 394, determined by colorimetric quantitation of the trityl fractions, were 98-99%. Other oligonucleotide synthesis reagents for the 394: Detritylation solution was 2% TCA in methylene chloride; capping was performed with 16% N- Methyl imidazole in THF and 10% acetic anhydride/10% 2,6-lutidine in THF; oxidation solution was 16.9 mM I2, 49 mM pyridine, 9% water in THF. Fisher Synthesis Grade acetonitrile was used directly from the reagent bottle. Referring to Figure 21. the homopolymer was base deprotected with
N H3/EtOH at 65 °C The solution was decanted and the support was washed twice with a solution of 1 :1 :1 H2O:CH3CN:MeOH. The combined solutions were dried down and then diluted with CH3CN (1 mL). BF3•OEt2 (2.5 μL, 30 μmol) was added to the solution and aliquots were removed at ten time points. The results indicate that after 30 min deprotection is complete, as shown in Figure 22.
IIII Vectors Expressing Ribozymes
There follows a method for expression of a ribozyme in a bacterial or eucaryotic cell, and for production of large amounts of such a ribozyme. In general, the invention features a method for preparing multi-copy cassettes encoding a defined ribozyme structure for production of a ribozyme at a decreased cost. A vector is produced which encodes a plurality of ribozymes which are cleaved at their 3' and 5' ends from an RNA transcript producted from the vector by only one other ribozyme. The system is useful for scaling up production of a ribozyme, which may be either modified or unmodified, in situ or in vitro. Such vector systems can be used to express a desired ribozyme in a specific cell, or can be used in an in vitro system to allow productiuon of large amounts of a desired riboqyne, The vectors of this invention allow a higher yield synthesis of a ribozyme in the form of an RNA transcript which is cleaved in situ or in vitro before or after transcript isolation.
Thus, this invention is distinct from the prior art in that a single ribozyme is used to process the 3' and 5' ends of each therapeutic, transacting or desired ribozyme instead of processing only one end, or only one ribozyme. This allows smaller vectors to be derived with multiple transacting ribozymes released by only one other ribozyme from the mRNA transcript. Applicant has also provided methods by which the activity of such ribozymes is increased compared to those in the art, by designing ribozyme-encoding vectors and the corresponding transcript such that folding of the mRNA does not interfere with processing by the releasing ribozyme.
The stability of the ribozyme produced in this method can be enhanced by provision of sequences at the termini of the ribozymes as described by Draper et al., PCT WO 93/23509, hereby incorporated by reference herein.
The method of this invention is advantageous since it provides high yield synthesis of ribozymes by use of low cost transcription-based protocols, compared to existing chemical ribozyme synthesis, and can use isolation techniques currently used to purify chemically synthesized oligonucleotides. Thus, the method allows synthesis of ribozymes in high yield at low cost for analytical, diagnostic, or therapeutic applications.
The method is also useful for synthesis of ribozymes in vitro for ribozyme structural studies, enzymatic studies, target RNA accessibility studies, transcription inhibition studies and nuclease protection studies, much is described by Draper et al., PCT WO 93/23509 hereby incorporated by reference herein.
The method can also be used to produce ribozymes in situ either to increase the intracellular concentration of a desired therapeutic ribozyme, or to produce a concatameric transcript for subsequent in vitro isolation of unit length ribozyme. The desired ribozyme can be used to inhibit gene expression in molecular genetic analyses or in infectious cell systems, and to test the efficacy of a therapeutic molecule or treat afflicted cells.
Thus, in general, the invention features a vector which includes a bacterial, viral or eucaryotic promoter within a plasmid, cosmid, phagmid, virus, viroid, virusoid or phage vector. Other vectors are equally suitable and include double-stranded, or partially double-stranded DNA, formed by an amplification method such as the polymerase chain reaction, or doublestranded, partially double-stranded or single-stranded RNA, formed by sitedirected homologous recombination into viral or viroid RNA genomes. Such vectors need not be circular. Transcriptionally linked to the promoter region is a first ribozyme-encoding region, and nucleotide sequences encoding a ribozyme cleavage sequence which is placed on either side of a region encoding a therapeutic or otherwise desired second ribozyme. Suitable restriction endonuclease sites can be provided to ease construction of this vector in DNA vectors or in requisite DNA vectors of an RNA expression system. The desired second ribozyme may be any desired type of ribozyme, such as a hammerhead, hairpin , hepatitis delta virus (HDV) or other catalytic center, and can include group I and group II introns, as discussed above. The first ribozyme is chosen to cleave the encoded cleavage sequence, and may also be any desired ribozyme, for example, a Tetrahymena derived ribozyme, which may, for example, include an imbedded restriction endonuclease site in the center of a self-recognition sequence to aid in vector construction. This endonuclease site is useful for construction of the vector, and subsequent analysis of the vector.
When the promoter of such a vector is activated an RNA transcript is produced which includes the first and second ribozyme sequences. The first ribozyme sequence is able to act, under appropriate conditions, to cause cleavage at the cleavage sites to release the second ribozyme sequences. These second ribozyme sequences can then act at their target RNA sites, or can be isolated for later use or analysis.
Thus, in one aspect the invention features a vector which includes a first nucleic acid sequence (encoding a first ribozyme having intramolecular cleaving activity), and a second nucleic acid sequence (encoding a second ribozyme having intermolecular cleaving enzymatic activity) flanked by nucleic acid sequences encoding RNA which is cleaved by the first ribozyme to release the second ribozyme from the RNA transcript encoded by the vector. The second ribozyme may be flanked by the first ribozyme either on the 5' side or 3' side. If desired, the first ribozyme may be encoded on a separate vector and may have intermolecular cleaving activity.
As discussed above, the first ribozyme can be chosen to be any self-cleaving ribozyme, and the second ribozyme may be chosen to be any desired ribozyme. The flanking sequences are chosen to include sequences recognized by the first ribozyme. When the vector is caused to express RNA from these nucleic acid sequences, that RNA has the ability under appropriate conditions to cleave each of the flanking regions and thereby release one or more copies of the second ribozyme. If desired, several different second ribozymes can be produced by the same vector, or several different vectors can be placed in the same vessel or cell to produce different ribozymes.
In preferred embodiments, the vector includes a plurality of the nucleic acid sequences encoding the second ribozyme, each flanked by nucleic acid sequences recognized by the first ribozyme. Most preferably, such a plurality includes at least six to nine or even between 60 - 100 nucleic acid sequences. In other preferred embodiments, the vector includes a promoter which regulates expression of the nucleic acid encoding the ribozymes from the vector; and the vector is chosen from a plasmid, cosmid, phagmid, virus, viroid or phage. In a most preferred embodiment, the plurality of nucleic acid sequences are identical and are arranged in sequential order such that each has an identical end nearest to the promoter. If desired, a poly(A) sequence adjacent to the sequence encoding the first or second ribozyme may be provided to increase stability of the RNA produced by the vector; and a restriction endonuclease site adjacent to the nucleic acid encoding the first ribozyme is provided to allow insertion of nucleic acid encoding the second ribozyme during construction of the vector.
In a second aspect, the invention features a method for formation of a ribozyme expression vector by providing a vector including nucleic acid encoding a first ribozyme, as discussed above, and providing a single-stranded PNA encoding a second ribozyme, as discussed above. The single-stranded PNA is then allowed to anneal to form a partial duplex PNA which can be filled in by a treatment with an appropriate enzyme, such as a PNA polymerase in the presence of dNTPs, to form a duplex DNA which can then be ligated to the vector. Large vectors resulting from this method can then be selected to insure that a high copy number of the single-stranded DNA encoding the second ribozyme is incorporated into the vector. In a further aspect, the invention features a method for production of ribozymes by providing a vector as described above, expressing RNA from that vector, and allowing cleavage by the first ribozyme to release the second ribozyme.
In preferred embodiments, three different ribozyme motifs are used as cis-cleaving ribozymes. The hammerhead, hairpin, and hepatitis delta virus (HDV) ribozyme motifs consist of small, well-defined sequences that rapidly self-cleave in vitro (Symons, 1992 Annu. Rev. Biochem. 61 , 641). While structural and functional differences exist among the three ribozyme motifs, they self-process efficiently in vivo. All three ribozyme motifs self-process to 87-95% completion in the absence of 3' flanking sequences. In vitro, the self-processing constructs described in this invention are significantly more active than those reported by Taira et al., 1990 supra: and Altschuler et al., 1992 Gene 122, 85. The present invention enables the use of cis-cleaving ribozymes to efficiently truncate RNA molecules at specific sites in vivo by ensuring lack of secondary structure which prevents processing.
Isolation of Therapeutic Ribozyme
The preferred method of isolating therapeutic ribozyme is by a chromatographic technique. The HPLC purification methods and reverse HPLC purification methods described by Draper et al., PCT WO 93/23509, hereby incorporated by reference herein, can be used. Alternatively, the attachment of complementary oligonucleotides to cellulose or other chromatography columns allows isolation of the therapeutic second ribozyme, for example, by hybridization to the region between the flanking arms and the enzymatic RNA. This hybridization will select against the short flanking sequences without the desired enzymatic RNA, and against the releasing first ribozyme. The hybridization can be accomplished in the presence of a chaotropic agent to prevent nuclease degradation. The oligonucleotides on the matrix can be modified to minimize nuclease activity, for example, by provision of 2'-0-methyl RNA oligonucleotides. Such modifications of the oligonucleotide attached to the column matrix will allow the multiple use of the column with minimal oligo degradation. Many such modifications are known in the art, but a chemically stable non-reducible modification is preferred. For example, phosphorothioate modifications can also be used.
The expressed ribozyme RNA can be isolated from bacterial or eucaryotic cells by routine procedures such as lysis followed by guanidine isothiocyanate isolation.
The current known self-cleaving site of Tetrahymena can be used in an alternative vector of this invention. If desired, the full-length Tetrahymena sequence may be used, or a shorter sequence may be used. It is preferred that, in order to decrease the superfluous sequences in the self-cleaving site at the 5' cleavage end, the hairpin normally present in the Tetrahymena ribozyme should contain the therapeutic second ribozyme 3' sequence and its complement. That is, the first releasing ribozyme-encoding DNA is provided in two portions, separated by DNA encoding the desired second ribozyme. For example, if the therapeutic second ribozyme recognition sequence is CGGACGA/CGAGGA, then CGAGGA is provided in the self-cleaving site loop such that it is in a stem structure recognized by the Tetrahymena ribozyme. The loop of the stem may include a restriction endonuclease site into which the desired second ribozyme-encoding DNA is placed.
If desired, the vector may be used in a therapeutic protocol by use of the systems described by Lechner, PCT WO 92/13070, hereby incorporated by reference herein, to allow a timed expression of the therapeutic second ribozyme, as well as an appropriate shut off of cell or gene function. Thus, the vector will include a promoter which appropriately expresses enzymatically active RNA only in the presence of an RNA or another molecule which indicates the presence of an undesired organism or state. Such enzymatically active RNA will then kill or harm the cell in which it exists, as described by Lechner, id ., or act to cause reduced expression of a desired protein product.
A number of suitable RNA vectors may also be used in this invention. The vectors include plant viroids, plant viruses which contain single or double-stranded RNA genomes and animal viruses which contain RNA genomes, such as the picornaviruses, myxoviruses, paramyxoviruses, hepatitis A virus, reovirus and retroviruses. In many instances cited, use of these viral vectors also results in tissue specific delivery of the ribozymes.
Example 21 : Design of self-processing cassettes In a preferred embodiment, applicant compared the in vitro and in vivo cis-cleaving activity of three different ribozyme motifs-the hammerhead, the hairpin and the hepatitis delta virus ribozyme-in order to assess their potential to process the ends of transcripts in vivo. To make a direct comparison among the three, however, it is important to design the ribozyme-containing transcripts to be as similar as possible. To this end, all the ribozyme cassettes contained the same trans-acting hammerhead ribozyme followed immediately by one of the three cis-acting ribozymes (Figure 23-25). For simplicity, applicant refers to each cassette by an abbreviation that indicates the downstream cis-cleaving ribozyme only. Thus HH refers to the cis-cleaving cassette containing a hammerhead ribozyme, while HP and HDV refer to the cassettes containing hairpin and hepatitis delta virus cis-cleaving ribozymes, respectively. The general design of the ribozyme cassettes, as well as specific differences among the cassettes, are outlined below. A sequence predicted to form a stable stem-loop structure is included at the 5' end of all the transcripts. The hairpin stem contains the T7 RNA polymerase initiation sequence (Milligan & Uhlenbeck, 1989 Methods Enzymol. 180, 51) and its complement, separated be a stable tetra-loop (Antao et al., 1991 Nucleic Acids Res. 19, 5901). By incorporating the T7 initiation sequence into a stem-loop structure, applicant hoped to avoid nonproductive base pairing interactions with either the trans-acting ribozyme or with the cis-acting ribozyme. The presence of a hairpin at the end of a transcript may also contribute to the stability of the transcript in vivo. These are non-limiting examples. Those in the art will recognize that other embodiments can be readily generated using a variety of promoters, initiator sequences and stem-loop structure combinations generally known in the art.
The trans-acting ribozyme used in this study is targeted to a site B (5'...CUGGAGUCGACCUUC...3'). The 5' binding arm of the ribozyme, 5'-GAAGG UC-3' , and the core of the ribozyme, 5'-CUGAUGAGGCCGAAAGGCCGAA-3', remain constant in all cases. In addition, all transcripts also contain a single nucleotide between the 5' stem-loop and the first nucleotide of the ribozyme. The linker nucleotide was required to obtain the same activity in vitro that was measured with an identical ribozyme lacking the 5' hairpin. Because the three cis-cleaving ribozymes have different requirements at the site of cleavage, slight differences were unavoidable at the 3' end of the processed transcript. The junction between the trans- and cis-acting ribozyme is, however, designed so that there is minimal extraneous sequence left at the 3' end of the trans-cleaving ribozyme once cis-cleavage occurs. The only differences between the constructs lie in the 3' binding arm of the ribozyme, where either 6 or 7 nucleotides, 5'-ACUCCA(+/-G)-3', complementary to the target sequence are present and where, after processing, two to five extra nucleotides remain.
The cis-cleaving hammerhead ribozyme used in the HH cassette is based on the design of Grosshans and Cech, 1991 supra. As shown in Figure 23, the 3' binding arm of the trans-acting ribozyme is included in the required base-pairing interactions of the cis-cleaving ribozyme to form stem I. Two extra nucleotides, UC, were included at the end of the 3' binding arm to form the self-processing hammerhead ribozyme site (Ruffner et al., 1990 supra) which remain on the 3' end of the trans-acting ribozyme following self-processing.
The hairpin ribozyme portion of the HP self-processing construct is based on the minimal wild-type sequence (Hampel & Tritz, 1989 supra). A tetra-loop at the end of helix 1 (3' side of the cleavage site) serves to link the two portions and thus allows a minimal five nucleotides to remain at the end of the released trans-acting ribozyme following self-processing. Two variants of HP were designed: HP(GU) and HP(GC). The HP(GU) was constructed with a G·U wobble base pair in helix 2 (A52G substitution; Figure 24). This slight destabilization of helix 2 was intended to improve self-processing activity by promoting product release and preventing the reverse reaction (Berzal-Herranz et al., 1992 Genes & Dev. 6, 129; Chowrira et al., 1993 Biochemistry 32, 1088). The HP(GC) cassette was constructed as a control for strong base-pairing interactions in helix 2 (U77C and A52G substitution; Figure 24). Another modification to discourage the reverse ligation reaction of the hairpin ribozyme was to shorten helix 1 (Figure 24) by one base pair relative to the wild-type sequence (Chowrira & Burke, 1991 Biochemistry 30, 8518).
The HDV ribozyme self-processes efficiently when the nucleotide 5' to the cleavage site is a pyrimidine, and somewhat less so when adenosine is in that position. No other sequence requirements have been identified upstream of the cleavage site, however, we have observed some decrease in activity when a stem-loop structure was present within 2 nt of the cleavage site. The HDV self-processing construct (Fig 25) was designed to generate the trans-acting hammerhead ribozyme with only two additional nucleotides at its 3' end after self-processing. The HDV sequence used here is based on the anti-genomic sequence (Perrota & Been, 1992 supra) but includes the modifications of Been et al., 1992 (Biochemistry 31 , 11843) in which cis-cleavage activity of the ribozyme was improved by the substitution of a shortened helix 4 for a wild-type stem-loop (Figure 25).
To prepare DNA inserts that encode self-processing ribozyme cassettes, partially overlapping top- and bottom-strand oligonucleotides (60-90 nucleotides) were designed to include sequences for the T7 promoter, the trans-acting ribozyme, the cis-cleaving ribozyme and appropriate restriction sites for use in cloning (see Fig. 26). The singlestrand portions of annealed oligonucleotides were converted to doublestrands using Sequenase® (U.S. Biochemicals). Insert DNA was ligated into EcoR1/HindIII-digested puc18 and transformed into E. coli strain DH5α using standard protocols (Maniatis et al., 1982 in Molecular Cloning Cold Spring Harbor Press). The identity of positive clones was confirmed by sequencing small-scale plasmid preparations. Larger scale preparations of plasmid DNA for use as in vitro transcription templates and in transactions were prepared using the protocol and columns from QIAGEN Inc. (Studio City, CA) except that an additional ethanol precipitation was included as the final step.
Example 22: RNA Processing in vitro
Transcription reactions containing linear plasmid templates were carried out essentially as described (Milligan & Uhlenbeck, 1989 Supra: Chowrira & Burke, 1991 Supra). In order to prepare 5' end-labeled transcripts, standard transcription reactions were carried out in the presence of 10-20 μCi [γ-32P]GTP, 200 μM each NTP and 0.5 to 1 μg of linearized plasmid template. The concentration of MgCl2 was maintained at 10 mM above the total nucleotide concentration.
To compare the ability of the different ribozyme cassettes to self-process in vitro, each construct was transcribed and allowed to undergo self-processing under identical conditions at 37°C For these comparisons, equal amounts of linearized DNA templates bearing the various ribozyme cassettes were transcribed in the presence of [γ-32P]GTP to generate 5' end-labeled transcripts. In this manner only the full-length, unprocessed transcripts and the released trans-ribozymes are visualized by autoradiography. In all reactions, Mg2+ was included at 10 mM above the nucleotide concentration so that cleavage by all the ribozyme cassettes would be supported. Transcription templates were linearized at several positions by digestion with different restriction enzymes so that self-processing in the presence of increasing lengths of downstream sequence could be compared (see Fig. 26). The resulting transcripts have either 4-5 non-ribozyme nucleotides at the 3' end (HindIII-digested template), 220 nucleotides (NdeI digested templates) or 454 nucleotides of downstream sequence (Real digested template).
As shown in Figure 27, all four ribozyme cassettes are capable of self-processing and yield RΝA products of expected sizes. Two nucleotides essential for hammerhead ribozyme activity (Ruffner et al., 1990 supra) have been changed in the HH(mutant) core sequence (see Figure 23) and so this transcript is unable to undergo self-processing (Fig. 27). This is evidenced by the lack of a released 5' RΝA in the HH(mutant), although the full-length RΝAs are present . Comparison of the amounts of released trans-ribozyme (Fig. 27) indicate that there are differences in the ability of these ribozymes to self-process in vitro, especially with respect to the presence of downstream sequence. For the two HP constructs, it is clear that HP(GC) is more efficient than the HP(GU) ribozyme, both in the presence and in the absence of extra downstream sequence. In addition, the activity of HP(GU) falls off more dramatically when downstream sequence is present. The stronger G:C base pair likely contributes to the HP(GC) construct's ability to fold correctly (and/or more quickly) into the productive structure, even when as much as 216 extra nucleotides are present downstream. The HH ribozyme construct is also quite efficient at self-processing, and slightly better than the HP(GU) construct even when downstream sequence is present.
Of the three ribozyme motifs, the presence of extra downstream sequence seems to most affect the efficiency of HPV. When no extra sequence is present downstream, HPV is quite efficient and self-processes to approximately the same level as the HH and HP(GC) cassettes. However, when extra downstream sequence is present, the self-processing activity seems to decrease almost as dramatically as is seen with the (sub-optimal) HP(GU) cassette. Example 23: Kinetics of self-processing reaction
HindIII-digested template (250 ng) was used in a standard transcription reaction mixture containing: 50 mM Tris·HCl pH 8.3; 1 mM ATP, GTP and UTP; 50 μM CTP; 40 μCi [α-32P]CTP; 12 mM MgCl2; 10 mM DTT. The transcription/self-processing reaction was initiated by the addition of T7 RNA polymerase (15 U/μl). Aliquots of 5 μl were taken at regular time intervals and the reaction was stopped by adding an equal volume of 2x formamide loading buffer (95% formamide, 15 mM EDTA, & dyes) and freezing on dry ice. The samples were resolved on a 10% polyacrylamide sequencing gel and results were quantitated by PhosphorImager (Molecular Dynamics, Sunnyvale, CA). Ribozyme self-cleavage rates were determined from non-linear, least-squares fits (KaleidaGraph, Synergy Software, Reeding, PA) of the data to the equation:
Figure imgf000093_0001
where t represents time and k represents the unimolecular rate constant for cleavage (Long & Uhlenbeck, 1994 Proc. Natl. Acad. Sci. USA 91 , 6977).
Linear templates were prepared by digesting the plasmids with HindIII so that transcripts will contain only four to five vector-derived nucleotides at the 3' end (see Figure 23-25). By comparison of the unimolecular rate constant (k) determined for each construct, it is clear that HH is the most efficient at self-processing (Table 44). The HH transcript self-processes 2-fold faster than HDV and 3-fold faster than HP(GC) transcripts. Although the HP(GU) RNA undergoes self-processing, it is at least 6-fold slower than the HP(GC) construct. This is consistent with previous observations that the stability of helix 2 is essential for self-processing and trans-cleavage activity of the hairpin ribozyme (Hampel et al., 1990 supra: Chowrira & Burke, 1991 supra). The rate of HH self-cleavage during transcription measured here (1.2 min-1 ) is similar to the rate measured by Long and Uhlenbeck 1994 supra using a HH that has a different stem I and stem III. Self-processing rates during transcription for HP and HDV have not been previously reported. However, self-processing of the HDV ribozyme-as measured here during transcription-is significantly slower than when tested after isolation from a denaturing gel (Been et al., 1992 supra). This decrease likely reflects the difference in protocol as well as the presence of 5' flanking sequence in the HDV construct used here. Example 24: Effect of downstream sequences on trans-cleavage in vitro
Transcripts containing the trans ribozyme with or without 3' flanking sequences were assayed for their ability to cleave their target in trans. To this end, transcripts from three templates were resolved on a preparative gel and bands corresponding both to processed trans-acting ribozymes from the HH transcription reaction, and to full-length HH(mutant) and ΔHDV transcripts were isolated. In all three transcripts the trans-acting ribozyme portion is identical-with the exception of sequences at their 3' ends. The HH trans-acting ribozyme contains only an additional UC at its 3' end, while HH(mutant) and ΔHDV have 52 and 37 nucleotides, respectively, at their 3' ends. A 622 nucleotide, internally-labeled target RNA was incubated, under ribozyme excess conditions, along with the three ribozyme transcripts in a standard reaction buffer.
To make internally-labeled substrate RNA for trans-ribozyme cleavage reactions, a 622 nt region (containing hammerhead site P) was synthesized by PCR using primers that place the T7 RNA promoter upstream of the amplified sequence. Target RNA was transcribed in a standard transcription buffer in the presence of [α-32P]CTP (Chowrira & Burke, 1991 supra). The reaction mixture was treated with 15 units of ribonuclease-free DNasel, extracted with phenol followed chloroform:isoamyl alcohol (25:1), precipitated with isopropanol and washed with 70% ethanol. The dried pellet was resuspended in 20 μl DEPC-treated water and stored at -20°C.
Unlabeled ribozyme (1μM) and internally labeled 622 nt substrate RNA (<10 nM) were denatured and renatured separately in a standard cleavage buffer (containing 50 mM Tris·HCl pH 7.5 and 10 mM MgCl2) by heating to 90°C for 2 min. and slow cooling to 37°C for 10 min. The reaction was initiated by mixing the ribozyme and substrate mixtures and incubating at 37°C Aliquots of 5 μl were taken at regular time intervals, quenched by adding an equal volume of 2X formamide gel loading buffer and frozen on dry ice. The samples were resolved on 5% polyacrylamide sequencing gel and results were quantitatively analyzed by radioanalytic imaging of gels with a Phosphorlmager® (Molecular Dynamics, Sunnyvale, CA). The HH trans-acting ribozyme cleaves the target RNA approximately
10-fold faster than the ΔHDV transcript and greater than 20-fold faster than the HH(mutant) transcript (Figure 28). The additional nucleotides at the end of HH(mutant) form 7 base-pairs with the 3' target-binding arm of the trans-acting ribozyme (Figure 23). This interaction must be disrupted (at a cost of 6 kcal/mole) to make the trans-acting ribozyme available for binding the target sequence. In contrast, the additional nucleotides at the end of ΔHDV were not designed to form any strong, alternative base-pairing with the trans-ribozyme. Nevertheless, the ΔHDV sequences are predicted to form multiple structures involving the 3' target-binding arm of the trans ribozyme that have stabilities ranging from 1-2 kcal/mole. Thus, the observed reductions in activity for the ΔHDV and HH(mutant) constructs are consistent with the predicted folded structures, and it reinforces the view that the flanking sequences can decrease the catalytic efficiency of a ribozyme through nonproductive interactions with either the ribozyme or the substrate or both. Example 25: RNA self-processing in vivo
Since three of the constructs (HH, HDV and HP(GC)) self-process efficiently in solution, the affect of the mammalian cellular milieu on ribozyme self-processing was next explored by applicant. A transient expression system was employed to investigate ribozyme activity in vivo. A mouse cell line (OST7-1) that constitutively expresses T7 RNA polymerase in the cytoplasm was chosen for this study (Elroy-Stein and Moss, 1990 Proc. Natl. Acad. Sci. USA 87, 6743). In these cells plasmids containing a ribozyme cassette downstream of the T7 promoter will be transcribed efficiently in the cytoplasm (Elroy-Stein & Moss, 1990 supra). Monolayers of a mouse L9 fibroblast cell line (OST7-1 ; Elroy-Stein and Moss, 1990 supra) were grown in 6-well plates with ~ 5×105 cells/well. Cells were transfected with circular plasmids (5 μg/well) using the calcium phosphate-DNA precipitation method (Maniatis et al., 1982 supra). Cells were lysed (4 hours post-transfection) by the addition of standard lysis buffer (200 μl/well) containing 4M guanadinium isothiocyanate, 25 mM sodium citrate (pH 7.0), 0.5% sarkosyl (Chomczynski and Sacchi, 1987 Anal. Biochem. 162, 156), and 50 mM EDTA pH 8.0. The lysate was extracted once with water-saturated phenol followed by one extraction with chloroform:isoamyl alcohol (25:1). Total cellular RNA was precipitated with an equal volume of isopropanol. The RNA pellet was resuspended in 0.2 M ammonium acetate and reprecipitated with ethanol. The pellet was then washed with 70% ethanol and resuspended in DEPC-treated water.
Purified cellular RNA (3 μg/reaction) was first denatured in the presence of a 5' end-labeled DNA primer (100 pmol) by heating to 90°C for 2 min. in the absence of Mg2+, and then snap-cooling on ice for at least 15 min. This protocol allows for efficient annealing of the primer to its complementary RNA sequence. The primer was extended using Superscript II reverse transcriptase (8 U/μl; BRL) in a buffer containing 50 mM Tris·HCl pH 8.3; 10 mM DTT; 75 mM KCl; 1 mM MgCl2; 1 mM each dNTP. The extension reaction was carried out at 42°C for 10 min. The reaction was terminated by adding an equal volume of 2x formamide gel loading buffer and freezing on crushed dry ice. The samples were resolved on a 10% polyacrylamide sequencing gel. The primer sequences are as follows: HH primer, 5'-CTCCAGTTTCGAGCTTT-3'; HDV primer, 5'-A A G T A G C C C A G G T C G G A C C - 3 ' ; H P p ri m e r , 5 ' - ACCAGGTAATATACCACAAC-3'.
As shown in Figure 29. specific bands corresponding to full-length precursor RNA and 3' cleavage products were detected from cells transfected with the self-processing cassettes. All three constructs, in addition to being transcriptionally active, appear to self-process efficiently in the cytoplasm of OST7-1 cells. In particular, the HH and HP(GC) constructs self-process to greater than 95%. The overall extent of self-processing in OST7-1 cells appears to be strikingly similar to the extent of self-processing in vitro (Figure 29 "In Vitro +MgCl2" vs. "Cellular"). Consistent with the in vitro self-processing results, the HP(GU) cassette self-processed to approximately 50% in OST7-1 cells. As expected, transfection with plasmids containing the HH(mutant) cassette yielded a primer-extension product corresponding to the full-length RNA with no detectable cleavage products (Figure 29). The latter result strongly suggests that the primer extension band corresponding to the 3' cleavage product is not an artifact of reverse transcription.
Applicant was concerned with the possibility that RNA self-processing might occur during cell lysis, RNA isolation and /or the primer extension assay. Two precautions were taken to exclude this possibility. First, 50 mM EDTA was included in the lysis buffer. EDTA is a strong chelator of divalent metal ions such as Mg2+ and Ca2+ that are necessary for ribozyme activity. Divalent metal ions are therefore unavailable to self-processing RNAs following cell lysis. A second precaution involved using primers in the primer-extension assay that were designed to hybridize to essential regions of the processing ribozyme. Binding of these primers should prevent the 3' cis-acting ribozymes from folding into the conformation essential for catalytic activity.
Two experiments were carried out to further eliminate the possibility that self-processing is occurring either during RNA preparations or during the primer extension analysis. The first experiment involves primer extension analysis on full-length precursor RNAs that were added to non-transfected OST7-1 lysates after cell lysis. Thus, only if self-processing is occurring at some point after lysis would cleavage products be detected. Full-length precursor RNAs were prepared by transcribing under conditions of low Mg2+ (5 mM) and high NTP concentration (total 12 mM) in an attempt to eliminate the free Mg2+ required for the self-processing reaction (Michel et al. 1992 Genes & Dev. 6, 1373). The full-length precursor RNAs were gel-purified, and a known amount was added to lysates of non-transfected OST7-1 cells. RNA was purified from these lysates and incubated for 1 hr in DEPC-treated water at 37° C prior to the standard primer extension analysis (Figure 29. in vitro "-MgCl2" control). The predominant RNA detected in all cases corresponds to the primer extension product of full-length precursor RNAs. If, instead, the purified RNA containing the full-length precursor is incubated in 10 mM MgCl2 prior to the primer extension analysis, most or all of the RNA detected by primer extension analysis undergoes cleavage (Figure 29. in vitro "+MgCl2" control). These results indicate that the standard RNA isolation and primer extension protocols used here do not provide a favorable environment for RNA self-processing, even though the RNA in question is inherently able to undergo self-cleavage.
In a second experiment to demonstrate lack of self-processing during work up, internally-labeled precursor RNAs were prepared and added to non-transfected OST7-1 lysates as in the previous control. The internally-labeled precursor RNAs were carried through the RNA purification and primer extension reactions (in the presence of unlabeled primers) and analyzed to determine the extent of self-processing. By this analysis, the vast majority of the added full-length RNA remained intact during the entire process of RNA isolation and primer extension.
These two control experiments validate the protocols used and support applicant's conclusion that the self-processing reactions catalyzed by HH, HDV and HP(GC) cassettes are occurring in the cytoplasm of OST7-1 cells.
Sequences in figures 23 through 25 are meant to be non-limiting examples. Those in the art will recognize that other embodiments can be readily generated using techniques generally known in the art. In addition, those in the art will recognize that Applicant provides guidance through the above examples as to how to best design vectors of this invention so that secondary structure of the mRNA allows efficient cleavage by releasing ribozymes. Thus, the specific constructs are not limiting in this invention. Such constructs can be readily tested as described above for such secondary structure, either by computer folding algorithms or empirically. Such constructs will then allow at least 80% completion of release of ribozymes, which can be readily determined as described above or by methods known in the art. That is, any such secondary structure in the RNA does not reduce release of the ribozymes by more than 20%.
IV. Ribozymes Expressed by RNA Polymerase III
Applicant has determined that the level of production of a foreign RNA, using a RNA polymerase III (pol III) based system, can be significantly enhanced by ensuring that the RNA is produced with the 5' terminus and a 3' region of the RNA molecule base-paired together to form a stable intramolecular stem structure. This stem structure is formed by hydrogen bond interactions (either Watson-Crick or non-Watson-Crick) between nucleotides in the 3' region (at least 8 bases) and complementary nucleotides in the 5' terminus of the same RNA molecule. Although the example provided below involves a type 2 pol III gene unit, a number of other pol III promoter systems can also be used, for example, tRNA (Hall et al., 1982 Cell 29, 3-5), 5S RNA (Nielsen et al., 1993, Nucleic Acids Res. 21 , 3631-3636), adenovirus VA RNA (Fowlkes and Shenk, 1980 Cell 22, 405-413), U6 snRNA (Gupta and Reddy, 1990 Nucleic Acids Res. 19, 2073-2075), vault RNA (Kickoefer et al., 1993 J. Biol. Chem. 268, 7868-7873), telomerase RNA (Romero and Blackburn, 1991 Cell 67, 343-353), and others.
The construct described in this invention is able to accumulate RNA to a significantly higher level than other constructs, even those in which 5' and 3' ends are involved in hairpin loops. Using such a construct the level of expression of a foreign RNA can be increased to between 20,000 and 50,000 copies per cell. This makes such constructs, and the vectors encoding such constructs, excellent for use in decoy, therapeutic editing and antisense protocols as well as for ribozyme formation. In addition, the molecules can be used as agonist or antagonist RNAs (affinity RNAs). Generally, applicant believes that the intramolecular base-paired interaction between the 5' terminus and the 3' region of the RNA should be in a double-stranded structure in order to achieve enhanced RNA accumulation.
Thus, in one preferred embodiment the invention features a pol III promoter system (e.g., a type 2 system) used to synthesize a chimeric RNA molecule which includes tRNA sequences and a desired RNA (e.g., a tRNA-based molecule). The following exemplifies this invention with a type 2 pol III promoter and a tRNA gene. Specifically to illustrate the broad invention, the RNA molecule in the following example has an A box and a B box of the type 2 pol III promoter system and has a 5' terminus or region able to base-pair with at least 8 bases of a complementary 3' end or region of the same RNA molecule. This is meant to be a specific example. Those in the art will recognize that this is but one example, and other embodiments can be readily generated using other pol III promoter systems and techniques generally known in the art.
By "terminus" is meant the terminal bases of an RNA molecule, ending in a 3' hydroxyl or 5' phosphate or 5' cap moiety. By "region" is meant a stretch of bases 5' or 3' from the terminus that are involved in base-paired interactions. It need not be adjacent to the end of the RNA. Applicant has determined that base pairing of at least one end of the RNA molecule with a region not more than about 50 bases, and preferably only 20 bases, from the other end of the molecule provides a useful molecule able to be expressed at high levels.
By "3' region" is meant a stretch of bases 3' from the terminus that are involved in intramolecular bas-paired interaction with complementary nucleotides in the 5' terminus of the same molecule. The 3' region can be designed to include the 3' terminus. The 3' region therefore is > 0 nucleotides from the 3' terminus. For example, in the S35 construct described in the present invention (Fig. 40) the 3' region is one nucleotide from the 3' terminus. In another example, the 3' region is ~ 43 nt from 3' terminus. These examples are not meant to be limiting. Those in the art will recognize that other embodiments can be readily generated using techniques generally known in the art. Generally, it is preferred to have the 3' region within 100 bases of the 3' terminus.
By "tRNA molecule" is meant a type 2 pol III driven RNA molecule that is generally derived from any recognized tRNA gene. Those in the art will recognize that DNA encoding such molecules is readily available and can be modified as desired to alter one or more bases within the DNA encoding the RNA molecule and/or the promoter system. Generally, but not always, such molecules include an A box and a B box that consist of sequences which are well known in the art (and examples of which can be found throughout the literature). These A and B boxes have a certain consensus sequence which is essential for a optimal pol III transcription.
By "chimeric tRNA molecule" is meant a RNA molecule that includes a pol III promoter (type 2) region. A chimeric tRNA molecule, for example, might contain an intramolecular base-paired structure between the 3' region and complementary 5' terminus of the molecule, and includes a foreign RNA sequence at any location within the molecule which does not affect the activity of the type 2 pol III promoter boxes. Thus, such a foreign RNA may be provided at the 3' end of the B box, or may be provided in between the A and the B box, with the B box moved to an appropriate location either within the foreign RNA or another location such that it is effective to provide pol III transcription. In one example, the RNA molecule may include a hammerhead ribozyme with the B box of a type 2 pol III promoter provided in stem II of the ribozyme. In a second example, the B box may be provided in stem IV region of a hairpin ribozyme. A specific example of such RNA molecules is provided below. Those in the art will recognize that this is but one example, and other embodiments can be readily generated using techniques generally known in the art.
By "desired RNA" molecule is meant any foreign RNA molecule which is useful from a therapeutic, diagnostic, or other viewpoint. Such molecules include antisense RNA molecules, decoy RNA molecules, enzymatic RNA, therapeutic editing RNA and agonist and antagonist RNA.
By "antisense RNA" is meant a non-enzymatic RNA molecule that binds to another RNA (target RNA) by means of RNA-RNA interactions and alters the activity of the target RNA (Eguchi et al., 1991 Annu. Rev. Biochem. 60, 631-652). By "enzymatic RNA" is meant an RNA molecule with enzymatic activity (Cech, 1988 J. American. Med. Assoc. 260, 3030-3035). Enzymatic nucleic acids (ribozymes) act by first binding to a target RNA. Such binding occurs through the target binding portion of a enzymatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes and then binds a target RNA through base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA.
By "decoy RNA" is meant an RNA molecule that mimics the natural binding domain for a ligand. The decoy RNA therefore competes with natural binding target for the binding of a specific ligand. For example, it has been shown that over-expression of HIV trans-activation response (TAR) RNA can act as a "decoy" and efficiently binds HIV tat protein, thereby preventing it from binding to TAR sequences encoded in the HIV RNA (Sullenger et al., 1990 Cell 63, 601-608). This is meant to be a specific example. Those in the art will recognize that this is but one example, and other embodiments can be readily generated using techniques generally known in the art.
By "therapeutic editing RNA" is meant an antisense RNA that can bind to its cellular target (RNA or DNA) and mediate the modification of a specific base.
By "agonist RNA" is meant an RNA molecule that can bind to protein receptors with high affinity and cause the stimulation of specific cellular pathways. By "antagonist RNA" is meant an RNA molecule that can bind to cellular proteins and prevent it from performing its normal biological function (for example, see Tsai et al., 1992 Proc. Natl. Acad. Sci. USA 89, 8864-8868). In other aspects, the invention includes vectors encoding RNA molecules as described above, cells including such vectors, methods for producing the desired RNA, and use of the vectors and cells to produce this RNA.
Thus, the invention features a transcribed non-naturally occuring RNA molecule which includes a desired therapeutic RNA portion and an intramolecular stem formed by base-pairing interactions between a 3' region and complementary nucleotides at the 5' terminus in the RNA. The stem preferably includes at least 8 base pairs, but may have more, for example, 15 or 16 base pairs. In preferred embodiments, the 5' terminus of the chimeric tRNA includes a portion of the precursor molecule of the primary tRNA molecule, of which≥ 8 nucleotides are involved in base-pairing interaction with the 3' region; the chimeric tRNA contains A and B boxes; natural sequences 3' of the B box are deleted, which prevents endogenous RNA processing; the desired RNA molecule is at the 3' end of the B box; the desired RNA molecule is between the A and the B box; the desired RNA molecule includes the B box; the desired RNA molecule is selected from the group consisting of antisense RNA, decoy RNA, therapeutic editing RNA, enzymatic RNA, agonist RNA and antagonist RNA; the molecule has an intramolecular stem resulting from a base-paired interaction between the 5' terminus of the RNA and a complementary 3' region within the same RNA, and includes at least 8 bases; and the 5' terminus is able to base pair with at least 15 bases of the 3' region.
In most preferred embodiments, the molecule is transcribed by a RNA polymerase III based promoter system, e.g., a type 2 pol III promoter system; the molecule is a chimeric tRNA, and may have the A and B boxes of a type 2 pol III promoter separated by between 0 and 300 bases; DNA vector encoding the RNA molecule of claim 51. In other related aspects, the invention features an RNA or DNA vector encoding the above RNA molecule, with the portions of the vector encoding the RNA functioning as a RNA pol III promoter; or a cell containing the vector ; or a method to provide a desired RNA molecule in a cell, by introducing the molecule into a cell with an RNA molecule as described above. The cells can be derived from animals, plants or human beings.
In order for RNA-based gene therapy approaches to be effective, sufficient amounts of the therapeutic RNA must accumulate in the appropriate intracellular compartment of the treated cells. Accumulation is a function of both promoter strength of the antiviral gene, and the intracellular stability of the antiviral RNA. Both RNA polymerase II (pol II) and RNA polymerase III (pol III) based expression systems have been used to produce therapeutic RNAs in cells (Sarver & Rossi, 1993 AIDS Res. & Human Retroviruses 9, 483-487; Yu et al., 1993 P.N.A.S. (USA) 90, 6340-6344). However, pol III based expression cassettes are theoretically more attractive for use in expressing antiviral RΝAs for the following reasons. Pol II produces messenger RΝAs located exclusively in the cytoplasm, whereas pol III produces functional RΝAs found in both the nucleus and the cytoplasm. Pol II promoters tend to be more tissue restricted, whereas pol III genes encode tRΝAs and other functional RΝAs necessary for basic "housekeeping" functions in all cell types. Therefore, pol III promoters are likely to be expressed in all tissue types. Finally, pol III transcripts from a given gene accumulate to much greater levels in cells relative to pol II genes. Intracellular accumulation of therapeutic RΝAs is also dependent on the method of gene transfer used. For example, the retroviral vectors presently used to accomplish stable gene transfer, integrate randomly into the genome of target cells. This random integration leads to varied expression of the transferred gene in individual cells comprising the bulk treated cell population. Therefore, for maximum effectiveness, the transferred gene must have the capacity to express therapeutic amounts of the antiviral RΝA in the entire treated cell population, regardless of the integration site. Pol III System
The following is just one non-limiting example of the invention. A pol III based genetic element derived from a human tRNAi met gene and termed Δ3-5 (Fig. 33: Adeniyi-Jones et al., 1984 supra), has been adapted to express antiviral RNAs (Sullenger et al., 1990 Mol. Cell. Biol. 10, 6512-6523). This element was inserted into the DC retroviral vector (Sullenger et al., 1990 Mol. Cell. Biol. 10, 6512-6523) to accomplish stable gene transfer, and used to express antisense RNAs against moloney murine leukemia virus and anti-HIV decoy RNAs (Sullenger et al., 1990 Mol. Cell. Biol. 10, 6512-6523; Sullenger et al., 1990 Cell 63, 601-608; Sullenger et al., 1991 J. Virol. 65, 6811-6816; Lee et al., 1992 The New Biologist 4, 66-74). Clonal lines are expanded from individual cells present in the bulk population, and therefore express similar amounts of the therapeutic RNA in all cells. Development of a vector system that generates therapeutic levels of therapeutic RNA in all treated cells would represent a significant advancement in RNA based gene therapy modalities.
Applicant examined hammerhead (HHI) ribozyme (RNA with enzymatic activity) expression in human T cell lines using the Δ3-5 vector system (These constructs are termed "Δ3-5/HHI"; Fig. 34). On average, ribozymes were found to accumulate to less than 100 copies per cell in the bulk T cell populations. In an attempt to improve expression levels of the Δ3-5 chimera, the applicant made a series of modified Δ3-5 gene units containing enhanced promoter elements to increase transcription rates, and inserted structural elements to improve the intracellular stability of the ribozyme transcripts (Fig. 34). One of these modified gene units, termed S35, gave rise to more than a 100-fold increase in ribozyme accumulation in bulk T cell populations relative to the original Δ3-5/HHI vector system. Ribozyme accumulation in individual clonal lines from the pooled T cell populations ranged from 10 to greater than 100 fold more than those achieved with the original Δ3-5/HHI version of this vector.
The S35 gene unit may be used to express other therapeutic RNAs including, but not limited to, ribozymes, antisense, decoy, therapeutic editing, agonist and antagonist RNAs. Application of the S35 gene unit would not be limited to antiviral therapies, but also to other diseases, such as cancer, in which therapeutic RNAs may be effective. The S35 gene unit may be used in the context of other vector systems besides retroviral vectors, including but not limited to, other stable gene transfer systems such as adeno-associated virus (AAV; Carter, 1992 Curr. Opin. Genet. Dev. 3, 74), as well as transient vector systems such as plasmid delivery and adenoviral vectors (Berkner, 1988 BioTechniques 6, 616-629). As described below, the S35 vector encodes a truncated version of a tRNA wherein the 3' region of the RNA is base-paired to complementary nucleotides at the 5' terminus, which includes the 5' precursor portion that is normally processed off during tRNA maturation. Without being bound by any theory, Applicant believes this feature is important in the level of expression observed. Thus, those in the art can now design equivalent RNA molecules with such high expression levels. Below are provided examples of the methodology by which such vectors and tRNA molecules can be made.
Δ3-5 Vectors The use of a truncated human tRNAi met gene, termed Δ3-5 (Fig. 33:
Adeniyi-Jones et al., 1984 supra), to drive expression of antisense RNAs, and subsequently decoy RNAs (Sullenger et al., 1990 supra) has recently been reported. Because tRNA genes utilize internal pol III promoters, the antisense and decoy RNA sequences were expressed as chimeras containing tRNAi met sequences. The truncated tRNA genes were placed into the U3 region of the 3' moloney murine leukemia virus vector LTR (Sullenger et al., 1990 supra).
Base-Paired Structures
Since the Δ3-5 vector combination has been successfully used to express inhibitory levels of both antisense and decoy RNAs, applicant cloned ribozyme-encoding sequences (termed as "Δ3-5/HHI") into this vector to explore its utility for expressing therapeutic ribozymes. However, low ribozyme accumulation in human T cell lines stably transduced with this vector was observed (Fig. 35). To try and improve accumulation of the ribozyme, applicant incorporated various RNA structural elements (Fig. 34) into one of the ribozyme chimeras (Δ3-5/HHI).
Two strategies were used to try and protect the termini of the chimeric transcripts from exonucleolytic degredation. One strategy involved the incorporation of stem-loop structures into the termini of the transcript. Two such constructs were cloned, S3 which contains a stem-loop structure at the 3' end, and S5 which contains stem-loop structures at both ends of the transcript (Figure 34). The second strategy involved modification of the 3' terminal sequences such that the 5' terminus and the 3' end sequences can form a stable base-paired stem. Two such constructs were made: S35 in which the 3' end was altered to hybridize to the 5' leader and acceptor stem of the tRNAi met domain, and S35Plus which was identical to S35 but included more extensive structure formation within the non-ribozyme portion of the Δ3-5 chimeras (Figure 34). These stem-loop structures are also intended to sequester non-ribozyme sequences in structures that will prevent them from interfering with the catalytic activity of the ribozyme. These constructs were cloned, producer cell lines were generated, and stably-transduced human MT2 (Harada et al., 1985 supra) and CEM (Nara & Fischinger, 1988 supra) cell lines were established (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). The RNA sequences and structure of S35 and S35 Plus are provided in Figures 40-47.
Referring to Figure 48. there is provided a general structure for a chimeric RNA molecule of this invention. Each N independently represents none or a number of bases which may or may not be base paired. The A and B boxes are optional and can be any known A or B box, or a consensus sequence as exemplified in the figure. The desired nucleic acid to be expressed can be any location in the molecule, but preferably is on those places shown adjacent to or between the A and B boxes (designated by arrows). Figure 49 shows one example of such a structure in which a desired RNA is provided 3' of the intramolecular stem. A specific example of such a construct is provided in Figures 50 and 51.
Example 26: Cloning of Δ3-5-Ribozyme Chimera
Oligonucleotides encoding the S35 insert that overlap by at least 15 nucleotides were designed (5' GATCCACTCTGCTGTTCTGTTTTTGA 3' and 5' CGCGTCAAAAACAGAACAGCAGAGTG 3'). The oligonucleotides (10 μM each) were denatured by boiling for 5 min in a buffer containing 40 mM Tris.HCl, pH8.0. The oligonucleotides were allowed to anneal by snap cooling on ice for 10-15 min.
The annealed oligonucleotide mixture was converted into a double-stranded molecule using Sequenase® enzyme (US Biochemicals) in a buffer containing 40 mM Tris.HCI, pH7.5, 20 mM MgCl2, 50 mM NaCl, 0.5 mM each of the four deoxyribonucleotide triphosphates, 10 mM DTT. The reaction was allowed to proceed at 37°C for 30 min. The reaction was stopped by heating to 70°C for 15 min. The double stranded DNA was digested with appropriate restriction endonucleases (BamHI and MluI) to generate ends that were suitable for cloning into the Δ3-5 vector.
The double-stranded insert DNA was ligated to the Δ3-5 vector DNA by incubating at room temperature (about 20°C) for 60 min in a buffer containing 66 mM Tris.HCl, pH 7.6, 6.6 mM MgCl2, 10 mM DTT, 0.066 μM ATP and 0.1 U/μl T4 DNA Ligase (US Biochemicals).
Competent E. coli bacterial strain was transformed with the recombinant vector DNA by mixing the cells and DNA on ice for 60 min. The mixture was heat-shocked by heating to 37°C for 1 min. The reaction mixture was diluted with LB media and the cells were allowed to recover for 60 min at 37°C The cells were plated on LB agar plates and incubated at 37°C for ~ 18 h.
Plasmid DNA was isolated from an overnight culture of recombinant clones using standard protocols (Ausubel et al., Curr. Protocols Mol. Biology 1990, Wiley & Sons, NY).
The identity of the clones were determined by sequencing the plasmid DNA using the Sequenase DNA sequencing kit (US Biochemicals).
The resulting recombinant Δ3-5 vector contains the S35 sequence. The HHI encoding DNA was cloned into this Δ3-5-S35 containing vector using SaclI and BamHI restriction sites.
Example 27: Northern analysis
RNA from the transduced MT2 cells were extracted and the presence of Δ3-5/ribozyme chimeric transcripts were assayed by Northern analysis (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). Northern analysis of RNA extracted from MT2 transductants showed that Δ3-5/ribozyme chimeras of appropriate sizes were expressed (Fig. 35.36). In addition, these results demonstrated the relative differences in accumulation among the different constructs (Figure 35.36). The pattern of expression seen from the Δ3-5/HHI ribozyme chimera was similar to 12 other ribozymes cloned into the Δ3-5 vector (not shown). In MT-2 cell line, Δ3-5/HHI ribozyme chimeras accumulated, on average, to less than 100 copies per cell. Addition of a stem-loop onto the 3' end of Δ3-5/HHI did not lead to increased Δ3-5 levels (S3 in Fig. 35,36). The S5 construct containing both 5' and 3' stem-loop structures also did not lead to increased ribozyme levels (Fig. 35.36).
Interestingly, the S35 construct expression in MT2 cells was about 100-fold more abundant relative to the original Δ3-5/HHI vector transcripts (Fig. 35,36). This may be due to increased stability of the S35 transcript.
Example 28: Cleavage activity
To assay whether ribozymes transcribed in the transduced cells contained cleavage activity, total RNA extracted from the transduced MT2 T cells were incubated with a labeled substrate containing the HHI cleavage site (Figure 37). Ribozyme activity in all but the S35 constructs, was too low to detect. However, ribozyme activity was detectable in S35-transduced T cell RNA. Comparison of the activity observed in the S35-transduced MT2 RNA with that seen with MT2 RNA in which varying amounts of in vitro transcribed S5 ribozyme chimeras, indicated that between 1-3 nM of S35 ribozyme was present in S35-transduced MT2 RNA. This level of activity corresponds to an intracellular concentration of 5,000-15,000 ribozyme molecules per cell.
Example 29: Clonal variation Variation in the ribozyme expression levels among cells making up the bulk population was determined by generating several clonal cell lines from the bulk S35 transduced CEM line (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY) and the ribozyme expression and activity levels in the individual clones were measured (Figure 38 and 39). All the individual clones were found to express active ribozyme. The ribozyme activity detected from each clone correlated well with the relative amounts of ribozyme observed by Northern analysis. Steady state ribozyme levels among the clones ranged from approximately 1 ,000 molecules per cell in clone G to 11 ,000 molecules per cell in clone H (Fig. 38). The mean accumulation among the clones, calculated by averaging the ribozyme levels of the clones, exactly equaled the level measured in the parent bulk population. This suggests that the individual clones are representative of the variation present in the bulk population. The fact that all 14 clones were found to express ribozyme indicate that the percentage of cells in the bulk population expressing ribozyme is also very high. In addition, the lowest level of expression in the clones was still more than 10-fold that seen in bulk cells transduced with the original Δ3-5 vector. Therefore, the S35 gene unit should be much more effective in a gene therapy setting in which bulk cells are removed, transduced and then reintroduced back into a patient.
Example 30: Stability
Finally, the bulk S35-transduced line, resistant to G418, was propogated for a period of 3 months (in the absence of G418) to determine if ribozyme expression was stable over extended periods of time. This situation mimicks that found in the clinic in which bulk cells are transduced and then reintroduced into the patient and allowed to propogate. There was a modest 30% reduction of ribozyme expression after 3 months. This difference probably arose from cells with varying amount of ribozyme expression and exhibiting different growth rates in the culture becoming slightly more prevalent in the culture. However, ribozyme expression is apparently stable for at least this period of time.
Example 31 : Design and construction of TRZ-tRNA Chimera
A transcription unit, termed TRZ, is designed that contains the S35 motif (Figure 52). A desired RNA (e.g. ribozyme) can be inserted into the indicated region of TRZ tRNA chimera. This construct might provide additional stability to the desired RNA. TRZ-A and TRZ-B are non-limiting examples of the TRZ-tRNA chimera.
Referring to Fig. 53-54. a hammerhead ribozyme targeted to site I (HHITRZ-A; Fig. 53) and a hairpin ribozyme (HPITRZ-A; Fig. 54), also targeted to site I, is cloned individually into the indicated region of TRZ tRNA chimera. The resulting ribozyme trancripts retain full RNA cleavage activity (see for example Fig. 55). Applicant has shown that efficient expression of these TRZ tRNA chimera can be achieved in mammalian cells.
Besides ribozymes, desired RNAs like antisense, therapeutic editing RNAs, decoys, can be readily inserted into the indicated region of TRZ-tRNA chimera to achieve therapeutic levels of RNA expression in mammalian cells.
Sequences listed in Figures 40-47 and 50 - 54 are meant to be non-limiting examples. Those skilled in the art will recognize that variants (mutations, insertions and deletions) of the above examples can be readily generated using techniques known in the art, are within the scope of the present invention.
Example 32: Ribozyme expression in T cell lines
Ribozyme expression in T cell lines stably-transduced with either a retroviral-based or an Adeno-associated virus (AAV)-based ribozyme expression vector (Figure 56). The human T cell lines MT2 and CEM were transduced with either retroviral or AAV vectors encoding a neomycin slelctable marker and a ribozyme (S35/HHI) expressed from pol III metitRNA-driven promoter. Cells stably-transduced with the vectors were selectivelyt expanded medium containing the neomycin antibiotic derivative, G418 (0.7 mg/ml). Ribozyme expression in the stable cell lines was then alalyzed by Northern analysis. The probe used to detect ribozyme transcripts also cross-hybridized with human meti tRNA sequences. Refering to Figure 56, S35/HHI RNA accumulates to significant levels in MT2 and CEM cells when transduced with either the retrovirus or the AAV vector.
These are meant to be non-limiting examples, those skilled in the art will recognize that other vectors such as adenovirus vector (Figure 57), plasmid DNA vector, alpha virus vectors and the other derivatives there of, can be readily generated to deliver the desired RNA, using techniques known in the art and are within the scope of this invention. Additionally, the transcription units can be expressed individually or in multiples using pol II and/or pol III promoters.
References cited herein, as well as Draper WO 93/23569, 94/02495, 94/06331 , Sullenger WO 93/12657, Thompson WO 93/04573, and Sullivan WO 94/04609, and 93/11253 describe methods for use of vectors decribed herein, and are incorporated by reference herein. In particular these vectors are useful for administration of antisense and decoy RNA molecules. Example 33: Ligated Ribozymes are catalytically active
The ability of ribozymes generated by ligation methods, described in Praper et al., PCT WO 93/23569, to cleave target RNA was tested on either matched substrate RNA (Fig. 58) or long (622 nt) RNA (Fig. 59, 60 and 61).
Matched substrate RNAs were chemically synthesized using solid-phase RNA synthesis chemistry (Scaringe et al., 1990 Nucleic Acids Res. 18, 5433-5441). Substrate RNA was 5' end-labeled using [γ-32P] ATP and polynucleotide kinase (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). Ribozyme reactions were carried out under ribozyme excess conditions (kcat/KM; Herschlag and Cech, 1990 Biochemistry 29, 10159-10171). Briefly, ribozyme and substrate RNA were denatured and renatured separately by heating to 90°C and snap cooling on ice for 10 min in a buffer containing 50 mM Tris. HCl pH 7.5 and 10 mM MgCl2. Cleavage reaction was initiated by mixing the ribozyme with the substrate at 37°C Aliquots of 5 μl were taken at regular intervals of time and the reaction was stopped by mixing with equal volume of formamide gel loading buffer (Curr. Protocols Mol. Biol. 1992, ed. Ausubel et al., Wiley & Sons, NY). The samples were resolved on 20 % polyacrylamide-urea gel. Refering to Fig. 58. -ΔG refers to the free energy of binding calculated for base-paired interactions between the ribozyme and the substrate RNA (Turner and Sugimoto, 1988 Supra). RPI A is a HH ribozyme with 6/6 binding arms. This ribozyme was synthesized chemically either as a one piece ribozyme or was synthesized in two fragments followed by ligation to generate a one piece ribozyme. The kcat/KM values for the two ribozymes were comparable. A template containing T7 RNA polymerase promoter upstream of 622 nt long target sequence, was PCR amplified from a DNA clone. The target RNA (containing HH ribozyme cleavage sites B, C and D) was transcribed from this PCR amplified template using T7 RNA polymerase. The transcript was internally labeled during transcription by including [α-32P] CTP as one of the four ribonucleotide triphosphates. The transcription mixture was treated with DNase-1 , following transcription at 37°C for 2 hours, to digest away the DNA template used in the transcription. RNA was precipitated with Isopropanol and the pellet was washed two times with 70% ethanol to get rid of salt and nucleotides used in the transcription reaction. RNA is resuspended in DEPC-treated water and stored at 4°C Ribozyme cleavage reactions were carried out under ribozyme excess (kcat/KM) conditions [Herschlag and Cech 1990 supra]. Briefly, 1000 nM ribozyme and 10 nM internally labeled target RNA were denatured separately by heating to 90°C for 2 min in the presence of 50 mM Tris.HCl, pH 7.5 and 10 mM MgCl2. The RNAs were renatured by cooling to 37°C for 10-20 min. Cleavage reaction was initiated by mixing the ribozyme and target RNA at 37°C Aliquots of 5 μl were taken at regular intervals of time and the reaction was quenched by adding equal volume of stop buffer. The samples were resolved on a sequencing gel. Example 34: Hammerhead ribozymes with≥ 2 base-paired stem II are catalytically active
To decrease the cost of chemical synthesis of RNA, applicant was interested in determining whether the length of stem II region of a typical hammerhead ribozyme (≥ 4 bp stem II) can be shortened without decreasing the catalytic efficiency of the HH ribozyme. The length of stem II was systematically shortened by one base-pair at a time. HH ribozymes with three and two base-paired stem II were chemically synthesized using solid-phase RNA phosphoramidite chemistry (Scaringe et al., 1990 supra).
Matched and long substrate RNAs were synthesized and ribozyme assays were carried out as described in example 33. Referring to figures , 62, 63 and 64. data shows that shortening stem II of a hammerhead ribozyme does not significantly alter the catalytic efficiency. It is applicant's opinion that hammerhead ribozymes with≥ 2 base-paired stem II region are catalytically active. Example 35: Synthesis of catalytically active hairpin ribozymes
RNA molecules were chemically synthesized having the nucleotide base sequence shown in Fig. 65 for both the 5' and 3' fragments. The 3' fragments are phosphorylated and ligated to the 5' fragment essentially as described in example 37. As is evident from the Figure 65, the 3' and 5' fragments can hybridize together at helix 4 and are covalently linked via GAAA sequence. When this structure hybridizes to a substrate, a ribozyme•substrate complex structure is formed. While helix 4 is shown as 3 base pairs it may be formed with only 1 or 2 base pairs.
40 nM mixtures of ligated ribozymes were incubated with 1-5 nM 5' end-labeled matched substrates (chemically synthesized by solid-phase synthesis using RNA phosphoramidite chemistry) for different times in 50 mM Tris/HCl pH 7.5, 10 mM MgCl2 and shown to cleave the substrate efficiently (Fig.66).
The target and the ribozyme sequences shown in Fig. 62 and 65 are meant to be non-limiting examples. Those in the art will recognize that other embodiments can be readily generated using other sequences and techniques generally known in the art.
V. Constructs of Hairpin Ribozymes
There follows an improved trans-cleaving hairpin ribozyme in which a new helix (i.e., a sequence able to form a double-stranded region with another single-stranded nucleic acid) is provided in the ribozyme to base-pair with a 5' region of a separate substrate nucleic acid. This helix is provided at the 3' end of the ribozyme after helix 3 as shown in Figure 3. In addition, at least two extra bases may be provided in helix 2 and a portion of the substrate corresponding to helix 2 may be either directly linked to the 5' portion able to hydrogen bond to the 3' end of the hairpin or may have a linker of atleast one base. By trans-cleaving is meant that the ribozyme is able to act in trans to cleave another RNA molecule which is not covalently linked to the ribozyme itself. Thus, the ribozyme is not able to act on itself in an intramolecular cleavage reaction.
By "base-pair" is meant a nucleic acid that can form hydrogen bond(s) with other RNA sequence by either traditional Watson-Crick or other non-traditional types (for example Hoogsteen type) of interactions.
The increase in length of helix 2 of a hairpin ribozyme (with or without helix 5) has several advantages. These include improved stability of the ribozyme-target complex in vivo . In addition, an increase in the recognition sequence of the hairpin ribozyme improves the specificity of the ribozyme. This also makes possible the targeting of potential hairpin ribozyme sites that would otherwise be inaccessible due to neighboring secondary structure.
The increase in length of helix 2 of a hairpin ribozyme (with or without helix 5) enhances trans-ligation reaction catalyzed by the ribozyme. Trans-ligation reactions catalyzed by the regular hairpin ribozyme (4 bp helix 2) is very inefficient (Komatsu et al., 1993 Nucleic Acids Res. 21 , 185). This is attributed to weak base-pairing interactions between substrate RNAs and the ribozyme. By increasing the length of helix 2 (with or without helix 5) the rate of ligation (in vitro and in vivo) can be enhanced several fold. Results of experiments suggest that the length of H2 can be 6 bp without significantly reducing the activity of the hairpin ribozyme. The H2 arm length variation does not appear to be sequence dependent. HP ribozymes with 6 bp H2 have been designed against five different target RNAs and all five ribozymes efficiently cleaved their cognate target RNA. Additionally, two of these ribozymes were able to successfully inhibit gene expression (e.g., TNF-α) in mammalian cells. Results of these experiments are shown below.
HP ribozymes with 7 and 8 bp H2 are also capable of cleaving target RNA in a sequence-specific manner, however, the rate of the cleavage reaction is lower than those catalyzed by HP ribozymes with 6 bp H2.
Example 36: 4 and 6 base pair H2
Referring to Figures 67-72. HP ribozymes were synthesized as described above and tested for activity. Surprisingly, those with 6 base pairs in H2 were still as active as those with 4 base pairs. VI. Chemical Modification
Oligonucleotides with 5'-C-alkyl Group
The introduction of an alkyl group at the 5'-position of a nucleoside or nucleotide sugar introduces an additional center of chirality into the sugar moiety. Referring to Fig. 75. the general structures of 5'-C-alkylnucleotides belonging to the D-allose, 2, and L-talose, 3, sugar families are shown. The family names are derived from the known sugars D-allose and L-talose (R 1 = CH3 in 2 and 3 in Figure 75). Useful specific D-allose and L-talose nucleotide derivatives are shown in Figure 76. 29-32 and Figure 77, 58-61 respectively.
This invention relates to the use of 5'-C-alkylnucleotides in oligonucleotides, which are particularly useful for enzymatic cleavage of RNA or single-stranded DNA, and also as antisense oligonucleotides. As the term is used in this application, 5'-C-alkylnucleotide-containing enzymatic nucleic acids are catalytic nucleic molecules that contain 5'-C-alkylnucleotide components replacing, but not limited to, double stranded stems, single stranded "catalytic core" sequences, single-stranded loops or single-stranded recognition sequences. These molecules are able to cleave (preferably, repeatedly cleave) separate RNA or DNA molecules in a nucleotide base sequence specific manner. Such catalytic nucleic acids can also act to cleave intramolecularly if that is desired. Such enzymatic molecules can be targeted to virtually any RNA transcript. Also within the invention are 5'-C-alkylnucleotides which may be present in enzymatic nucleic acid or even in antisense oligonucleotides. Such nucleotides are useful since they enhance the stability of the antisense or enzymatic molecule, and can be used in locations which do not affect the desired activity of the molecule. That is, while the presence of the 5'-C-alkyl group may reduce binding affinity of the oligonucleotide containing this modification, if that moiety is not in an essential base pair forming region then the enhanced stability that it provides to the molecule is advantageous. In addition, while the reduced binding may reduce enzymatic activity, the enhanced stability may make the loss of activity of less consequence. Thus, for example, if a 5'-C-alkyl-containing molecule has 10% the activity of the unmodified molecule, but has 10-fold higher stability in vivo then it has utility in the present invention. The same analysis is true for antisense oligonucleotides containing such modifications. The invention also relates to novel intermediates useful in the synthesis of such nucleotides and oligonucleotides (examples of which are shown in the Figures), and to methods for their synthesis.
Thus, in one aspect, the invention features 5'-C-alkylnucleosides, that is a nucleotide base having at the 5'-position on the sugar molecule an alkyl moiety. In a related aspect, the invention also features 5'-C-alkylnucleotides, and in preferred embodiments features those where the nucleotide is not uridine or thymidine. That is, the invention preferably includes all those nucleotides useful for making enzymatic nucleic acids or antisense molecules that are not described by the art discussed above. In preferred embodiments, the sugar of the nucleoside or nucleotide is in an optically pure form, as the talose or allose sugar. Examples of various alkyl groups useful in this invention are shown in
Figure 75. where each R1 group is any alkyl. These examples are not limiting in the invention. Specifically, an "alkyl" group refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, =O, =S, NO2 or N(CH3)2, amino, or SH. The term also includes alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkenyl group has 1 to 12 carbons. More preferably it is a lower alkenyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkenyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, =O, =S, NO2, halogen, N(CH3)2, amino, or SH. The term "alkyl" also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkynyl group has 1 to 12 carbons. More preferably it is a lower alkynyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkynyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, =O, =S, NO2 or N(CH3)2, amino or SH.
Such alkyl groups may also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups. An "aryl" group refers to an aromatic group which has at least one ring having a conjugated π electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted. The preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups. An "alkylaryl" group refers to an alkyl group (as described above) covalently joined to an aryl group (as described above. Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted. Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted. An "amide" refers to an -C(O)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen. An "ester" refers to an -C(0)-OR', where R is either alkyl, aryl, alkylaryl or hydrogen. In other aspects, also related to those discussed above, the invention features oligonucleotides having one or more 5'-C-alkylnucleotides; e.g. enzymatic nucleic acids having a 5'-C-alkylnucleotide; and a method for producing an enzymatic nucleic acid molecule having enhanced activity to cleave an RNA or single-stranded DNA molecule, by forming the enzymatic molecule with at least one nucleotide having at its 5'-position an alkyl group. In other related aspects, the invention features 5'-C-alkylnucleotide triphosphates. These triphosphates can be used in standard protocols to form useful oligonucleotides of this invention.
The 5'-C-alkyl derivatives of this invention provide enhanced stability to the oligonulceotides containing them. While they may also reduce absolute activity in an in vitro assay they will provide enhanced overall activity in vivo. Below are provided assays to determine which such molecules are useful. Those in the art will recognize that equivalent assays can be readily devised. In another aspect, the invention features a method for conversion of a protected allo sugar to a protected talo sugar. In the method, the protected allo sugar is contacted with triphenyl phosphine, diethylazodicarboxylate, and p-nitrobenzoic acid under inversion causing conditions to provide the protected talo sugar. While one example of such conditions is provided below, those in the art will recognize other such conditions. Applicant has found that such conversion allows for ready synthesis of all types of nucleotide bases as exemplified in the figures.
While this invention is applicable to all oligonucleotides, applicant has found that the modified molecules of this invention are particulary useful for enzymatic RNA molecules. Thus, below is provided examples of such molecules. Those in the art will recognize that equivalent procedures can be used to make other molecules without such enzymatic activity. Specifically, Figure 1 shows base numbering of a hammerhead motif in which the numbering of various nucleotides in a hammerhead ribozyme is provided. This is not to be taken as an indication that the Figure is prior art to the pending claims, or that the art discussed is prior art to those claims. Referring to Figure 1. the preferred sequence of a hammerhead ribozyme in a 5'- to 3'-direction of the catalytic core is CUGANGAG [base paired with] CGAAA. In this invention, the use of 5'-C-alkyl substituted nucleotides that maintain or enhance the catalytic activity and or nuclease resistance of the hammerhead ribozyme is described. Substitutions of any nucleotide with any of the modified nucleotides shown in Figure 75 are possible.
The following are non-limiting examples showing the synthesis of nucleic acids using 5'-C-alkyl-substituted phosphoramidites and the syntheses of the amidites.
Example 37: Synthesis of Hammerhead Ribozymes Containing 5'-C-Alkylnucleotides & Other Modified Nucleotides
The method of synthesis would follow the procedure for normal RNA synthesis as described in Usman, N.; Ogilvie.K.K.; Jiang, M.-Y.; Cedergren.R.J. J. Am. Chem. Soc. 1987, 109, 7845-7854 and in Scaringe, S.A.; Franklyn,C.; Usman, N. Nucleic Acids Res. 1990, 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end (compounds 26-29 and 56-59). These 5'-C-alkyl substituted phosphoramidites may be incorporated not only into hammerhead ribozymes, but also into hairpin, hepatitis delta virus, Group 1 or Group 2 intron catalytic nucleic acids, or into antisense oligonucleotides. They are, therefore, of general use in any nucleic acid structure.
Example 38: Methyl-2,3-O-Isopropylidine-6-Deoxy-β-D-allofuranoside (4) A suspension of L-rhamnose (100 g, 0.55 mol), CuSO4 (120 g) and cone. H2SO4 (4.0 mL) in 1.0 L of dry acetone was mixed for 24 h at RT, then filtered. Cone. NH4OH (5 mL) was added to the filtrate and the newly formed precipitate was filtered. The residue was concentrated in vacuo, coevaporated with pyridine (2 × 300 mL), dissolved in pyridine (500 mL) and cooled to 0 °C A solution of p-toluenesufonylchloride (107 g , 0.56 mmol) in dry DCE (500 mL) was added dropwise over 0.5 h. The reaction mixture was left for 16 h at RT. The reaction was quenched by adding ice-water (0.5 L) and, after mixing for 0.5 h, was extracted with chloroform (0.75 L). The organic layer was washed with H2O (2 × 500 mL), 10% H2SO4 (2 × 300 mL), water (2 × 300 mL), sat. NaHCO3 (2 × 300 mL), brine (2 × 300 mL), dried over MgSO4 and evaporated to dryness. The residue (115 g) was dissolved in dry MeOH (1 L) and treated with NaOMe (23.2 g, 0.42 mmol) in MeOH. The reaction mixture was left for 16 h at 20 °C, neutralized with dry CO2 and evaporated to dryness. The residue was suspended in chloroform (750 mL), filtered , concentrated to 100 mL and purified by flash chromatography in CHCl3 to yield 45 g (37%) of compound 4.
Example 39: Methyl-2,3-O-Isopropylidine-5-O-t-Butyldiphenylsilyl-6-Deoxy-β-D-Allofuranoside (5).
To solution of methylfuranoside 4 (12.5 g 62.2 mmol) and AgNO3 (21.25 g, 125.0 mmol) in dry DMF (300 mL) t-butyldiphenylsilyl chloride (22.2 g , 81 mmol) was added dropwise under Ar over 0.5 h. The reaction mixture was stirred for 4 h at RT, diluted with CHCl3 (200 mL), filtered and evaporated to dryness (below 40 °C using a high vacuum oil pump). The residue was dissolved in CH2Cl2 (300 mL) washed with sat. NaHCO3 (2 × 50 mL), brine (2 × 50 mL), dried over MgSO4 and evaporated to dryness. The residue was purified by flash chromatography in CH2Cl2 to yield 20.0 g (75%) of compound 5.
Example 40: Methyl-5-O-t-Butyldiphenylsilyl-6-Deoxy-β-D-Allofuranoside (6).
Methylfuranoside 5 (13.5 g, 30.6 mmol) was dissolved in
CF3COOH:dioxane:H2O / 2:1 :1 (v/v/v, 200 mL) and stirred at 24 °C for 45 m. The reaction mixture was cooled to -10 °C, neutralized with conc. NH4OH (140 mL) and extracted with CH2Cl2 (500 mL). The organic layer was separated, washed with sat. NaHCO3 (2 × 75 mL), brine (2 × 75 mL), dried over MgSO4 and evaporated to dryness. The product 6 was purified by flash chromatography using a 0-10% MeOH gradient in CH2Cl2. Yield 9.0 g (76%). Example 41 : Methyl-2,3-di-O-Benzoyl-5-O-t-Butyldiphenylsilyl-6-Deoxy-β-D-Allofuranoside (7).
Methylfuranoside 6 (7.0 g, 17.5 mmol) was coevaporated with pyridine (2 × 100 mL) and dissolved in pyridine (100 mL). Benzoyl chloride (5.4 g, 38.5 mmol) was added and the reaction mixture was left at RT for 16 h. Dry EtOH (50 mL) was added and the reaction mixture was evaporated to dryness after 0.5 h. The residue was dissolved in CH2Cl2 (300 mL), washed with sat. NaHCO3 (2 × 75 mL), brine (2 × 75 mL) dried over MgSO4 and evaporated to dryness. The product was purified by flash chromatography in CH2Cl2 to yield 9.5 g (89%) of compound 7.
Example 42: 1-O-Acetyl-2,3-di-O-benzoyl-5-O-t-Butyldiphenylsilyl-6-Deoxy-β-D-Allofuranose (8). Dibenzoate 7 (4.7 g, 7.7 mmol) was dissolved in a mixture of AcOH (10.0 mL), AC2O (20.0 mL) and EtOAc (30 mL) and the reaction mixture was cooled 0 °C 98% H2SO4 (0.15 mL) was then added. The reaction mixture was kept at 0 °C for 16 h, and then poured into a cold 1 :1 mixture of sat. NaHCO3 and EtOAc (150 mL). After 0.5 h of vigorous stirring the organic phase was separated, washed with brine (2 × 75 mL), dried over MgSO4, evaporated to dryness and coevaporated with toluene (2 × 50 mL). The product was purified by flash chromatography using a gradient of 0-5% MeOH in CH2Cl2. Yield: 4.0 g (82% as a mixture of α and β isomers).
Example 43: 1-(2',3'-di-O-Benzoyl-5'-O-t-Butyldiphenylsilyl-6'-Deoxy-β-D-Allofuranosyl)uracil (9).
Uracil (1.44 g, 11.5 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (3 h) occurred, and then for an additional hour. The reaction mixture was cooled to RT, evaporated to dryness and coevaporated with dry toluene (2 × 50 mL). To the residue was added a solution of acetates 8 (6.36 g, 10.0 mmol) in dry CH3CN (100 mL), followed by CF3SO3SiMe3 (2.8 g, 12.6 mmol). The reaction mixture was kept at 24 °C for 16 h, concentrated to 1/3 of its original volume, diluted with 100 mL of CH2Cl2 and extracted with sat. NaHCO3 (2 × 50 mL), brine (2 × 50 mL) dried over MgS04, and evaporated to dryness. The product 9 was purified by flash chromatography using a gradient of 0-5% MeOH in CH2Cl2. Yield: 5.7 g (80%). Example 44: N4-Benzoyl-1-(2',3'-Di-O-Benzoyl-5'-O-t-Butyldiphenylsilyl-6'-Deoxy-β-D-Allofuranosyl)Cytosine (10). N4-benzoylcytosine (1.84 g, 8.56 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (3 h) occurred, and then for an additional hour. The reaction mixture was cooled to RT evaporated to dryness and coevaporated with dry toluene (2 × 50 mL). To the residue was added a solution of of acetates 8 (3.6 g, 5.6 mmol) in dry CH3CN (100 mL), followed by CF3SO3SiMe3 (4.76 g, 21.4 mmol). The reaction mixture was boiled under reflux for 5 h, cooled to RT, concentrated to 1/3 of its original volume, diluted with CH2Cl2 (100 mL) and extracted with sat. NaHCO3 (2 × 50 mL), brine (2 × 50 mL) dried over MgSO4 and evaporated to dryness. Purification by flash chromatography using a gradient of 0-5% MeOH in CH2Cl2 yielded 1.8 g (55%) of compound 10. Example 45: N6-Benzoyl-9-(2',3'-di-O-Benzoyl-5'-O-t-Butyldiphenylsilyl-6'-Deoxy-β-D-Allofuranosyl)adenine (11). N6-benzoyladenine (2.86 g, 11.86 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (7 h) occurred, and then for an additional hour. The reaction mixture was cooled to RT evaporated to dryness and coevaporated with dry toluene (2 × 50 mL). To the residue was added a solution of of acetates 8 (3.6 g, 5.6 mmol) in dry CH3CN (100 mL) followed by CF3SO3SiMe3 (6.59 g, 29.7 mmol). The reaction mixture was boiled under reflux for 8 h, cooled to RT, concentrated to 1/3 of its original volume, diluted with CH2Cl2 (100 mL) and extracted with sat. NaHCO3 (2 × 50 mL), brine (2 × 50 mL) dried over MgSO4 and evaporated to dryness. The product 11 was purified by flash chromatography using a gradient of 0-5% MeOH in CH2Cl2. Yield: 2.7 g (60%).
Example 46: N2-Isobutyryl-9-(2',3'-di-O-Benzoyl-5'-O-t-Butyldiphenylsilyl-6'-Deoxy-β-D-Allofuranosyl)guanine (12). N2-lsobutyrylguanine (1.47 g , 11.2 mmol) was suspended in mixture of hexamethyldisilazane (100 mL) and pyridine (50 mL) and boiled under reflux until complete dissolution (6 h) occurred, and then for an additional hour. The reaction mixture was cooled to RT evaporated to dryness and coevaporated with dry toluene (2 × 50 mL). To the residue was added a solution of of acetates 8 (3.4 g, 5.3 mmol) in dry CH3CN (100 mL) followed by CF3SO3SiMe3 (6.22 g, 28.0 mmol). The reaction mixture was boiled under reflux for 8 h, cooled to RT, concentrated to 1/3 of its original volume, diluted with CH2Cl2 (100 mL) and extracted with sat. NaHCO3 (2 x 50 mL), brine (2 × 50 mL) dried over MgSO4 and evaporated to dryness. The product 12 was purified by flash chromatography using a gradient of 0-2% MeOH in CH2Cl2. Yield: 2.1g (54%).
Example 47: N6--Benzoyl-9-(2',3'-di-O-benzoyl-6'-Deoxy-β-D-Allofuranosyl)adenine (15).
Nucleoside 11 (1.65 g, 2.0 mmol) was dissolved in THF (50 mL) and a 1 M solution of TBAF in THF (4 mL) was added. The reaction mixture was kept at RT for 4 h, evaporated to dryness and the product purified by flash chromatography using a gradient of 0-5% MeOH in CH2Cl2 to yield 1.0 g (85%) of compound 15. Example 48: N6-Benzoyl-9-(2',3'-di-O-Benzoyl-5'-O-Dimethoxytrityl-6'-Deoxy-β-D-Allofuranosyl)-adenine (19).
Nucleoside 15 (0.55 g, 0.92 mmol) was dissolved in dry CH2Cl2 (50 mL). AgNO3 (0.34 g, 2.0 mmol), dimethoxytrityl chloride (0.68 g, 2.0 mmol) and sym-collidine (0.48 g) were added under Ar. The reaction mixture was stirred for 2h, diluted with CH2Cl2 (100 mL), filtered, evaporated to dryness and coevaporated with toluene (2 × 50 mL). Purification by flash chromatography using a gradient of 0-5% MeOH in CH2Cl2 yielded 0.8 g (97%) of compound 19.
Example 49: N6-Benzoyl-9-(-5'-O-Dimethoxytrityl-6'-Deoxy-β-D-Allofuranosyl)adenine (23).
Nucleoside 19 (1.8 g, 2 mmol) was dissolved in dioxane (50 mL), cooled to 0 °C and 2 M NaOH (50 mL) was added. The reaction mixture was kept at 0 °C for 45 m, neutralized with Dowex 50 (Pyr+ form), filtered and the resin was washed with MeOH (2 x 50 mL). The filtrate was then evaporated to dryness. Purification by flash chromatography using a gradient of 0-10% MeOH in CH2Cl2 yielded 1.1 g (80%) of 23. Example 50: N6-Benzoyl-9-(-5'-O-Dimethoxytrityl-2'-O-t-butyldimethylsilyl-6'-Deoxy-β-D-Allofuranosyl)adenine (27).
Nucleoside 23 (1.2 g, 1.8 mmol) was dissolved in dry THF (50 mL). Pyridine (0.50 g, 8 mmol) and AgNO3 (0.4 g, 2.3 mmol) were added. After the AgNO3 dissolved (1.5 h), t-butyldimethylsilyl chloride (0.35 g , 2.3 mmol) was added and the reaction mixture was stirred at RT for 16 h. The reaction mixture was diluted with CH2Cl2 (100 mL), filtered into sat. NaHCO3 (50 mL), extracted, the organic layer washed with brine (2 × 50 mL), dried over MgSO4 and evaporated to dryness. The product 27 was purified by flash chromatography using a hexanes:EtOAc / 7:3 gradient. Yield: 0.7 g (50%).
Example 51 : N6-Benzoyl-9-(-5'-O-Dimethoxytrityl-2'-O-t-butyldimethylsilyl-6'-Deoxy-β-D-Allofuranosyl)adenine-3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (31).
Standard phosphitylation of 27 according to Scaringe, S. A.;
Franklyn.C; Usman, Ν. Nucleic Acids Res. 1990, 18, 5433-5441 yielded phosphoramidite 31 in 73% yield.
Example 52: Methyl-5-O-p-Νitrobenzoyl-2,3-O-Isopropylidine-6-deoxy-β-L-Tallofuranoside (5)
Methylfuranoside 4 (3.1 g 14.2 mmol) was dissolved in dry dioxane
(200 mL), p-nitrobenzoic acid (10.0 g, 60 mmol) and triphenylphosphine (15.74 g, 60.0 mmol) were added followed by DEAD (10.45 g, 60.0 mmol). The reaction mixture was left at RT for 16 h, EtOH (5 mL) was added, and after 0.5 h the reaction mixture was evaporated to dryness. The residue was dissolved in CH2Cl2 (300 mL) washed with sat. NaHCO3 (2 × 75 mL), brine (2 × 75 mL) dried over MgSO4 and evaporated to dryness. Purification by flash chromatography using a hexanes:EtOAc / 9:1 gradient yielded 4.1 g (78%) of compound 33. Subsequent debenzoylation (NaOMe/MeOH) and silylation (see preparation of 5) led to L-talofuranoside 34 which was converted to phosphoramidites 58-61 using the same methodology as described above for the preparation of the phosphoramidites of the D-allo-isomers 29-32.
The alkyl substituted nucleotides of this invention can be used to form stable oligonucleotides as discussed above for use in enzymatic cleavage or antisense situations. Such oligonucleotides can be formed enzymatically using triphosphate forms by standard procedure. Administration of such oligonucleotides is by standard procedure. See Sullivan et al., PCT WO 94/ 02595. The ribozymes and the target RNA containing site O were synthesized, deprotected and purified as described above. RNA cleavage assay was carried our at 37°C in the presence of 10 mM MgCl2 as described above.
Applicant has substituted 5'-C-Me-L-talo nucleotides at positions A6, A9, A9 + G10, C11.1 and C11.1 + G10, as shown in Figure 78 (HH-01 to HH-05). HH-0 1,2,4 and 5 showed almost wild type activity (Figure 79). However, HH-03 demonstrated low catalytic activity. Ribozymes HH-01, 2, 3, 4 and 5 are also extremely resistant to degradation by human serum nucleases. Oligonucleotides with 2'-Deoxy-2'-Alkylnucleotide
This invention uses 2'-deoxy-2'-alkylnucleotides in oligonucleotides, which are particularly useful for enzymatic cleavage of RNA or single-stranded DNA, and also as antisense oligonucleotides. As the term is used in this application, 2'-deoxy-2'-alkylnucleotide-containing enzymatic nucleic acids are catalytic nucleic molecules that contain 2'-deoxy-2'-alkylnucleotide components replacing, but not limited to, double stranded stems, single stranded "catalytic core" sequences, single-stranded loops or single-stranded recognition sequences. These molecules are able to cleave (preferably, repeatedly cleave) separate RNA or DNA molecules in a nucleotide base sequence specific manner. Such catalytic nucleic acids can also act to cleave intramolecularly if that is desired. Such enzymatic molecules can be targeted to virtually any RNA transcript.
Also within the invention are 2'-deoxy-2'-alkylnucleotides which may be present in enzymatic nucleic acid or even in antisense oligonucleotides. Contrary to the findings of De Mesmaeker et al. applicant has found that such nucleotides are useful since they enhance the stability of the antisense or enzymatic molecule, and can be used in locations which do not affect the desired activity of the molecule. That is, while the presence of the 2'-alkyl group may reduce binding affinity of the oligonucleotide containing this modification, if that moiety is not in an essential base pair forming region then the enhanced stability that it provides to the molecule is advantageous. In addition, while the reduced binding may reduce enzymatic activity, the enhanced stability may make the loss of activity of less consequence. Thus, for example, if a 2'-deoxy-2'-alkyl-containing molecule has 10% the activity of the unmodified molecule, but has 10-fold higher stability in vivo then it has utility in the present invention. The same analysis is true for antisense oligonucleotides containing such modifications. The invention also relates to novel intermediates useful in the synthesis of such nucleotides and oligonucleotides (examples of which are shown in the Figures), and to methods for their synthesis.
Thus, in one aspect, the invention features 2'-deoxy-2'-alkylnucleotides, that is a nucleotide base having at the 2'-position on the sugar molecule an alkyl moiety and in preferred embodiments features those where the nucleotide is not uridine or thymidine. That is, the invention preferably includes all those nucleotides useful for making enzymatic nucleic acids or antisense molecules that are not described by the art discussed above.
Examples of various alkyl groups useful in this invention are shown in
Figure 81 , where each R group is any alkyl. The term "alkyl" does not include alkoxy groups which have an "-O-alkyl" group, where "alkyl" is defined as described above, where the O is adjacent the 2'-position of the sugar molecule.
In other aspects, also related to those discussed above, the invention features oligonucleotides having one or more 2'-deoxy-2'-alkylnucleotides (preferably not a 2'-alkyl- uridine or thymidine); e.g. enzymatic nucleic acids having a 2'-deoxy-2'-alkylnucleotide; and a method for producing an enzymatic nucleic acid molecule having enhanced activity to cleave an RNA or single-stranded DNA molecule, by forming the enzymatic molecule with at least one nucleotide having at its 2'-position an alkyl group. In other related aspects, the invention features 2'-deoxy-2'-alkylnucleotide triphosphates. These triphosphates can be used in standard protocols to form useful oligonucleotides of this invention.
The 2'-alkyl derivatives of this invention provide enhanced stability to the oligonulceotides containing them. While they may also reduce absolute activity in an in vitro assay they will provide enhanced overall activity in vivo. Below are provided assays to determine which such molecules are useful. Those in the art will recognize that equivalent assays can be readily devised.
In another aspect, the invention features hammerhead motifs having enzymatic activity having ribonucleotides at locations shown in Figure 80 at 5, 6, 8, 12, and 15.1 , and having substituted ribonucleotides at other positions in the core and in the substrate binding arms if desired. (The term "core" refers to positions between bases 3 and 14 in Figure 80, and the binding arms correspond to the bases from the 3'-end to base 15.1 , and from the 5'-end to base 2). Applicant has found that use of ribonucleotides at these five locations in the core provide a molecule having sufficient enzymatic activity even when modified nucleotides are present at other sites in the motif. Other such combinations of useful ribonucleotides can be determined as described by Usman et al. supra. Figure 80 shows base numbering of a hammerhead motif in which the numbering of various nucleotides in a hammerhead ribozyme is provided. This is not to be taken as an indication that the Figure is prior art to the pending claims, or that the art discussed is prior art to those claims. Referring to Figure 80 the preferred sequence of a hammerhead ribozyme in a 5'- to 3'-direction of the catalytic core is CUGANGAG[base paired with]CGAAA. In this invention, the use of 2'-C-alkyl substituted nucleotides that maintain or enhance the catalytic activity and or nuclease resistance of the hammerhead ribozyme is described. Although substitutions of any nucleotide with any of the modified nucleotides shown in Figure 81 are possible, and were indeed synthesized, the basic structure composed of promarily 2'-O-Me nucleotides weth selected substitutions was chosen to maintain maximal catalytic activity (Yang et al. Biochemistry 1992, 31 , 5005-5009 and Paolella et al. , EMBO J. 1992, 11 , 1913-1919) and ease of synthesis, but is not limiting to this invention. Ribozymes from Figure 80 and Table 45 were synthesized and assayed for catalytic activity and nuclease resistance. With the exception of entries 8 and 17, all of the modified ribozymes retained at lease 1/10 of the wild-type catalytic activity. From Table 45, all 2'-modified ribozymes showed very large and significant increases in stability in human serum (shown) and in the other fluids described below (Example 55, data not shown). The order of most agressive nuclease activity was fetal bovine serum, > human serum >human plasma > human synovial fluid. As an overall measure of the effect of these 2'-substitutions on stability and activity, a ratio β was calculated (Table 45). This β value indicated that all modified ribozymes tested had significant, >100 - >1700 fold, increases in overall stability and activity. These increases in β indicate that the lifetime of these modified ribozymes in vivo are significantly increased which should lead to a more pronounced biological effect.
More general substitutions of the 2'-modified nucleotides from Figure 81 also increased the t 1/2 of the resulting modified ribozymes. However the catalytic activity of these ribozymes was decreased > 10-fold.
In Figure 86 compound 37 may be used as a general intermediate to prepare derivatized 2'C-alkyl phosphoramidites, where X is CH3, or an alkyl, or other group described above. The following are non-limiting examples showing the synthesis of nucleic acids using 2'-C-alkyl substituted phosphoramidites, the syntheses of the amidites, their testing for enzymatic activity and nuclease resistance.
Example 53: Synthesis of Hammerhead Ribozymes Containing 2'-Deoxy-2'-Alkylnucleotides & Other 2'-Modified Nucleotides
The method of synthesis used generally follows the procedure for normal RNA synthesis as described in Usman, N.; Ogilvie.K.K.; Jiang, M.-Y.; Cedergren.R.J. J. Am. Chem. Soc. 1987, 109, 7845-7854 and in Scaringe, S.A.; Franklyn.C; Usman, N. Nucleic Acids Res. 1990, 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end (compounds 10, 12, 17, 22, 31 , 18, 26, 32, 36 and 38). Other 2'-modified phosphoramidites were prepared according to: 3 & 4, Eckstein et al. International Publication No. WO 92/07065; and 5 Kois et al. Nucleosides & Nucleotides 1993, 12, 1093-1109. The average stepwise coupling yields were -98%. The 2'-substituted phosphoramidites were incorporated into hammerhead ribozymes as shown in Figure 80. However, these 2'-alkyl substituted phosphoramidites may be incorporated not only into hammerhead ribozymes, but also into hairpin, hepatitis delta virus, Group I or Group II intron catalytic nucleic acids, or into antisense oligonucleotides. They are, therefore, of general use in any nucleic acid structure.
Example 54: Ribozyme Activity Assay
Purified 5'-end labeled RNA substrates (15-25-mers) and purified 5'-end labeled ribozymes (~36-mers) were both heated to 95 °C, quenched on ice and equilibrated at 37 °C, separately. Ribozyme stock solutions were 1 mM, 200 nM, 40 nM or 8 nM and the final substrate RNA concentrations were - 1 nM. Total reaction volumes were 50 mL. The assay buffer was 50 mM Tris-Cl, pH 7.5 and 10 mM MgCl2. Reactions were initiated by mixing substrate and ribozyme solutions at t = 0. Aliquots of 5 mL were removed at time points of 1 , 5, 15, 30, 60 and 120 m. Each time point was quenched in formamide loading buffer and loaded onto a 15% denaturing polyacrylamide gel for analysis. Quantitative analyses were performed using a phosphorimager (Molecular Dynamics). Example 55: Stability Assay
500 pmol of gel-purified 5'-end-labeled ribozymes were precipitated in ethanol and pelleted by centrifugation. Each pellet was resuspended in 20 mL of appropriate fluid (human serum, human plasma, human synovial fluid or fetal bovine serum) by vortexing for 20 s at room temperature. The samples were placed into a 37 °C incubator and 2 mL aliquots were withdrawn after incubation for 0, 15, 30, 45, 60, 120, 240 and 480 m. Aliquots were added to 20 mL of a solution containing 95% formamide and 0.5X TBE (50 mM Tris, 50 mM borate, 1 mM EDTA) to quench further nuclease activity and the samples were frozen until loading onto gels. Ribozymes were size-fractionated by electrophoresis in 20% acrylamide/8M urea gels. The amount of intact ribozyme at each time point was quantified by scanning the bands with a phosphorimager (Molecular Dynamics) and the half-life of each ribozyme in the fluids was determined by plotting the percent intact ribozyme vs the time of incubation and extrapolation from the graph.
Example 56: 3',5'-O-(Tetraisopropyl-disiloxane-1 ,3-diyl)-2'-O-Phenoxythiocarbonyl-Uridine (7)
To a stirred solution of 3',5'-O-(tetraisopropyl-disiloxane-1 ,3-diyl)-uridine, 6, (15.1 g, 31 mmol, synthesized according to Nucleic Acid Chemistry, ed. Leroy Townsend, 1986 pp. 229-231) and dimethylaminopyridine (7.57 g, 62 mmol) a solution of phenylchlorothionoformate (5.15 mL, 37.2 mmol) in 50 mL of acetonitrile was added dropwise and the reaction stirred for 8 h. TLC (EtOAc:hexanes / 1 :1) showed disappearance of the starting material. The reaction mixture was evaporated, the residue dissolved in chloroform, washed with water and brine, the organic layer was dried over sodium sulfate, filtered and evaporated to dryness. The residue was purified by flash chromatography on silica gel with EtOAc:hexanes / 2:1 as eluent to give 16.44 g (85%) of 7. Example 57: 3'.5'-O-(Tetraisopropyl-disiloxane-1 ,3-diyl)-2'-C-Allyl -Uridine (8)
To a refluxing, under argon, solution of 3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-2'-O-phenoxythiocarbonyl-uridine, 7, (5 g, 8.03 mmol) and allyltributyltin (12.3 mL, 40.15 mmol) in dry toluene, benzoyl peroxide (0.5 g) was added portionwise during 1 h. The resulting mixture was allowed to reflux under argon for an additional 7-8 h. The reaction was then evaporated and the product 8 purified by flash chromatography on silica gel with EtOAc:hexanes / 1 :3 as eluent. Yield 2.82 g (68.7%).
Example 58: 5'-O-Dimethoxytrityl-2'-C-Allyl-Uridine (9) A solution of 8 (1.25 g, 2.45 mmol) in 10 mL of dry tetrahydrofuran
(THF) was treated with a 1 M solution of tetrabutylammoniumfluoride in THF (3.7 mL) for 10 m at room temperature. The resulting mixture was evaporated, the residue was loaded onto a silica gel column, washed with 1 L of chloroform, and the desired deprotected compound was eluted with chloroform:methanol / 9:1. Appropriate fractions were combined, solvents removed by evaporation, and the residue was dried by coevaporation with dry pyridine. The oily residue was redissolved in dry pyridine, dimethoxytritylchioride (1.2 eq) was added and the reaction mixture was left under anhydrous conditions overnight. The reaction was quenched with methanol (20 mL), evaporated, dissolved in chloroform, washed with 5% aq. sodium bicarbonate and brine. The organic layer was dried over sodium sulfate and evaporated. The residue was purified by flash chromatography on silica gel, EtOAc:hexanes / 1 :1 as eluent, to give 0.85 g (57%) of 9 as a white foam. Example 59: 5'-O-Dimethoxytrityl-2'-C-Allyl-Uridine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (10)
5'-O-Dimethoxytrityl-2'-C-allyl-uridine (0.64 g, 1.12 mmol) was dissolved in dry dichloromethane under dry argon. N,N-Diisopropylethylamine (0.39 mL, 2.24 mmol) was added and the solution was ice-cooled. 2-Cyanoethyl N,N-diisopropylchlorophosphoramidite (0.35 mL, 1.57 mmol) was added dropwise to the stirred reaction solution and stirring was continued for 2 h at RT. The reaction mixture was then ice-cooled and quenched with 12 mL of dry methanol. After stirring for 5 m, the mixture was concentrated in vacuo (40 °C) and purified by flash chromatography on silica gel using a gradient of 10-60% EtOAc in hexanes containing 1% triethylamine mixture as eluent. Yield: 0.78 g (90%), white foam.
Example 60: 3',5'-O-(Tetraisopropyl-disiloxane-1 ,3-diyl)-2'-C-Allyl-N4-Acetyl-Cytidine (11) Triethylamine (6.35 mL, 45.55 mmol) was added dropwise to a stirred ice-cooled mixture of 1 ,2,4-triazole (5.66 g, 81.99 mmol) and phosphorous oxychloride (0.86 mL, 9.11 mmol) in 50 mL of anhydrous acetonitrile. To the resulting suspension a solution of 3',5'-O-(tetraisopropyl-disiloxane-1 ,3-diyl)-2'-C-allyl uridine (2.32 g, 4.55 mmol) in 30 mL of acetonitrile was added dropwise and the reaction mixture was stirred for 4 h at room temperature. The reaction was concentrated in vacuo to a minimal volume (not to dryness). The residue was dissolved in chloroform and washed with water, saturated aq. sodium bicarbonate and brine. The organic layer was dried over sodium sulfate and the solvent was removed in vacuo. The resulting foam was dissolved in 50 mL of 1 ,4-dioxane and treated with 29% aq. ΝH4OH overnight at room temperature. TLC (chloroform:methanol /9:1) showed complete conversion of the starting material. The solution was evaporated, dried by coevaporation with anhydrous pyridine and acetylated with acetic anhydride (0.52 mL, 5.46 mmol) in pyridine overnight. The reaction mixture was quenched with methanol, evaporated, the residue was dissolved in chloroform, washed with sodium bicarbonate and brine. The organic layer was dried over sodium sulfate, evaporated to dryness and purified by flash chromatography on silica gel (3% MeOH in chloroform). Yield 2.3 g (90%) as a white foam. Example 61 : 5'-O-Dimethoxytrityl-2'-C-Allyl-N4-Acetyl-Cytidine
This compound was obtained analogously to the uridine derivative 9 in 55% yield.
Example 62: 5'-O-Dimethoxytrityl-2'-C-allyl-N4-Acetyl-Cytidine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (12)
2'-O-Dimethoxytrityl-2'-C-allyl-N4-acetyl cytidine (0.8 g, 1.31 mmol) was dissolved in dry dichloromethane under argon. N,N-Diisopropylethylamine (0.46 mL, 2.62 mmol) was added and the solution was ice-cooled. 2-Cyanoethyl N,N-diisopropylchlorophosphoramidite (0.38 mL, 1.7 mmol) was added dropwise to a stirred reaction solution and stirring was continued for 2 h at room temperature. The reaction mixture was then ice-cooled and quenched with 12 mL of dry methanol. After stirring for 5 m, the mixture was concentrated in vacuo (40 °C) and purified by flash chromatography on silica gel using chloroform:ethanol / 98:2 with 2% triethylamine mixture as eluent. Yield: 0.91 g (85%), white foam.
Example 63: 2'-Deoxy-2'-Methylene-Uridine
2'-Deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine 14 (Hansske.F.; Madej.D.; Robins.M. J. Tetrahedron 1984, 40, 125 and Matsuda.A.; Takenuki.K.; Tanaka.S.; Sasaki.T.; Ueda.T. J. Med. Chem. 1991 , 34, 812) (2.2 g, 4.55 mmol ) dissolved in THF (20 mL) was treated with 1 M TBAF in THF (10 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on a silica gel column. 2'-Deoxy-2'-methylene-uridine (1.0 g, 3.3 mmol, 72.5%) was eluted with 20% MeOH in CH2Cl2. Example 64: 5'-O-DMT-2'-Deoxy-2'-Methylene-Uridine (15)
2'-Deoxy-2'-methylene-uridine (0.91 g, 3.79 mmol) was dissolved in pyridine (10 mL) and a solution of DMT-Cl in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. ΝaHCO3, water and brine. The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using EtOAc:hexanes as eluant to yield 15 (0.43 g, 0.79 mmol, 22%). Example 65: 5'-O-DMT-2'-Deoxy-2'-Methylene-Uridine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (17)
1-(2'-Deoxy-2'-methylene-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-uracil (0.43 g, 0.8 mmol) dissolved in dry CH2Cl2 (15 mL) was placed in a round-bottom flask under Ar. Diisopropylethylamine (0.28 mL, 1.6 mmol) was added, followed by the dropwise addition of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (0.25 mL, 1.12 mmol). The reaction mixture was stirred 2 h at RT and quenched with ethanol (1 mL). After 10 m the mixture evaporated to a syrup in vacuo (40 °C). The product (0.3 g, 0.4 mmol, 50%) was purified by flash column chromatography over silica gel using a 25-70% EtOAc gradient in hexanes, containing 1% triethylamine, as eluant. Rf 0.42 (CH2Cl2: MeOH / 15:1)
Example 66: 2'-Deoxy-2'-Difluoromethylene-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-Uridine 2'-Keto-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)uridine 14 (1.92 g,
12.6 mmol) and triphenylphosphine (2.5 g, 9.25 mmol) were dissolved in diglyme (20 mL), and heated to a bath temperature of 160 °C A warm (60°C) solution of sodium chlorodifluoroacetate in diglyme (50 mL) was added (dropwise from an equilibrating dropping funnel) over a period of -1 h. The resulting mixture was further stirred for 2 h and concentrated in vacuo. The residue was dissolved in CH2Cl2 and chromatographed over silica gel. 2'-Deoxy-2'-difluoromethylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine (3.1 g, 5.9 mmol, 70%) eluted with 25% hexanes in EtOAc.
Example 67: 2'-Deoxy-2'-Difluoromethylene-Uridine 2'-Deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine (3.1 g, 5.9 mmol) dissolved in THF (20 mL) was treated with 1 M TBAF in THF (10 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on silica gel column. 2'-Deoxy-2'-difluoromethylene-uridine (1.1 g, 4.0 mmol, 68%) was eluted with 20% MeOH in CH2Cl2.
Example 68: 5'-O-DMT-2'-Deoxy-2'-Difluoromethylene-Uridine (16)
2'-Deoxy-2'-difluoromethylene-uridine (1.1 g, 4.0 mmol) was dissolved in pyridine (10 mL) and a solution of DMT-Cl (1.42 g, 4.18 mmol) in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. NaHCO3, water and brine. The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using 40% EtOAc:hexanes as eluant to yield 5'-O-DMT-2'-deoxy-2'-difluoromethylene-uridine 16 (1.05 g, 1.8 mmol, 45%).
Example 69: 5'-O-DMT-2'-Deoxy-2'-Difluoromethylene-Uridine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (18)
1-(2'-Deoxy-2'-difluoromethylene-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-uracil (0.577 g, 1 mmol) dissolved in dry CH2Cl2 (15 mL) was placed in a round-bottom flask under Ar. Diisopropylethylamine (0.36 mL, 2 mmol) was added, followed by the dropwise addition of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (0.44 mL, 1.4 mmol). The reaction mixture was stirred for 2 h at RT and quenched with ethanol (1 mL). After 10 m the mixture evaporated to a syrup in vacuo (40 °C). The product (0.404 g, 0.52 mmol, 52%) was purified by flash chromatography over silica gel using 20-50% EtOAc gradient in hexanes, containing 1% triethylamine, as eluant. Rf 0.48 (CH2Cl2: MeOH / 15:1). Example 70: 2'-Deoxy-2'-Methylene-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-4-N-Acetyl-Cytidine 20
Triethylamine (4.8 mL, 34 mmol) was added to a solution of POCl3 (0.65 mL, 6.8 mmol) and 1 ,2,4-triazole (2.1 g, 30.6 mmol) in acetonitrile (20 mL) at 0 °C A solution of 2'-deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl) uridine 19 (1.65 g, 3.4 mmol) in acetonitrile (20 mL) was added dropwise to the above reaction mixture and left to stir at room temperature for 4 h. The mixture was concentrated in vacuo, dissolved in CH2Cl2 (2 × 100 mL) and washed with 5% ΝaHCO3 (1 × 100 mL). The organic extracts were dried over Na2SO4 concentrated in vacuo, dissolved in dioxane (10 mL) and aq. ammonia (20 mL). The mixture was stirred for 12 h and concentrated in vacuo. The residue was azeotroped with anhydrous pyridine (2 × 20 mL). Acetic anhydride (3 mL) was added to the residue dissolved in pyridine, stirred at RT for 4 h and quenched with sat. NaHCO3 (5 mL). The mixture was concentrated in vacuo, dissolved in CH2Cl2 (2 × 100 mL) and washed with 5% NaHCO3 (1 × 100 mL). The organic extracts were dried over Na2SO4, concentrated in vacuo and the residue chromatographed over silica gel. 2'-Deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-4-N-acetyl-cytidine 20 (1.3 g, 2.5 mmol, 73%) was eluted with 20% EtOAc in hexanes. Example 71 : 1-(2'-Deoxy-2'-Methylene-5'-O-Dimethoxytrityl-β-D-ribofuranosyl)-4-N-Acetyl-Cytosine 21
2'-Deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-4-N-acetyl-cytidine 20 (1.3 g, 2.5 mmol) dissolved in THF (20 mL) was treated with 1 M TBAF in THF (3 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on silica gel column. 2'-Deoxy-2'-methylene-4-N-acetyl-cytidine (0.56 g, 1.99 mmol, 80%) was eluted with 10% MeOH in CH2Cl2. 2'-Deoxy-2'-methylene-4-N-acetyl-cytidine (0.56 g, 1.99 mmol) was dissolved in pyridine (10 mL) and a solution of DMT-Cl (0.81 g, 2.4 mmol) in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. ΝaHCO3 (50 mL), water (50 mL) and brine (50 mL). The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using EtOAc:hexanes / 60:40 as eluant to yield 21 (0.88 g, 1.5 mmol, 75%).
Example 72: 1-(2'-Deoxy-2,-Methylene-5'-O-Dimethoxytrityl-β-D-ribofuranosyl)-4-N-Acetyl-Cytosine 3'-(2-Cyanoethyl-N,N-diisopropylphosphoramidite) (22) 1-(2'-Deoxy-2'-methylene-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-4-N-acetyl-cytosine 21 (0.88 g, 1.5 mmol) dissolved in dry CH2Cl2 (10 mL) was placed in a round-bottom flask under Ar. Diisopropylethylamine (0.8 mL, 4.5 mmol) was added, followed by the dropwise addition of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (0.4 mL, 1.8 mmol). The reaction mixture was stirred 2 h at room temperature and quenched with ethanol (1 mL). After 10 m the mixture evaporated to a syrup in vacuo (40 °C). The product 22 (0.82 g, 1.04 mmol, 69%) was purified by flash chromatography over silica gel using 50-70% EtOAc gradient in hexanes, containing 1% triethylamine, as eluant. Rf 0.36 (CH2Cl2:MeOH / 20:1). Example 73: 2'-Deoxy-2'-Difluoromethylene-3',5'-O-(Tetraisopropyl disiloxane-1 ,3-diyl)-4-N-Acetyl-Cytidine (24)
Et3Ν (6.9 mL, 50 mmol) was added to a solution of POCl3 (0.94 mL, 10 mmol) and 1 ,2,4-triazole (3.1 g, 45 mmol) in acetonitrile (20 mL) at 0 °C A solution of 2'-deoxy-2'-difluoromethylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)uridine 23 ([described in example 14] 2.6 g, 5 mmol) in acetonitrile (20 mL) was added dropwise to the above reaction mixture and left to stir at RT for 4 h. The mixture was concentrated in vacuo, dissolved in CH2Cl2 (2 × 100 mL) and washed with 5% NaHCO3 (1 × 100 mL). The organic extracts were dried over Na2SO4 concentrated in vacuo, dissolved in dioxane (20 mL) and aq. ammonia (30 mL). The mixture was stirred for 12 h and concentrated in vacuo. The residue was azeotroped with anhydrous pyridine (2 × 20 mL). Acetic anhydride (5 mL) was added to the residue dissolved in pyridine, stirred at RT for 4 h and quenched with sat. NaHCO3 (5mL). The mixture was concentrated in vacuo, dissolved in CH2Cl2 (2 × 100 mL) and washed with 5% NaHCO3 (1 × 100 mL). The organic extracts were dried over Na2SO4, concentrated in vacuo and the residue chromatographed over silica gel. 2'-Deoxy-2'-difluoromethylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-4-N-acetyl-cytidine 24 (2.2 g, 3.9 mmol, 78%) was eluted with 20% EtOAc in hexanes.
Example 74: 1-(2'-Deoxy-2'-Difluoromethylene-5'-O-Dimethoxytrityl-β-D-ribofuranosyl)-4-N-Acetyl-Cytosine (25)
2'-Deoxy-2'-difluoromethylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-4-N-acetyl-cytidine 24 (2.2 g, 3.9 mmol) dissolved in THF (20 mL) was treated with 1 M TBAF in THF (3 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on a silica gel column. 2'-Deoxy-2'-difluoromethylene-4-N-acetyl-cytidine (0.89 g, 2.8 mmol, 72%) was eluted with 10% MeOH in CH2Cl2. 2'-Deoxy-2'-difluoromethylene-4-N-acetyl-cytidine (0.89 g, 2.8 mmol) was dissolved in pyridine (10 mL) and a solution of DMT-Cl (1.03 g, 3.1 mmol) in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. ΝaHCO3 (50 mL), water (50 mL) and brine (50 mL). The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using EtOAc:hexanes / 60:40 as eluant to yield 25 (1.2 g, 1.9 mmol, 68%).
Example 75: 1-(2'-Deoxy-2'-Difluoromethylene-5'-O-Dimethoxytrityl-β-D-ribofuranosyl)-4- N-Acetylcytosine 3'-(2-cyanoethyl-N,N-diisopropylphosphoramidite) (26)
1-(2'-Deoxy-2'-difluoromethylene-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-4-N-acetylcytosine 25 (0.6 g, 0.97 mmol) dissolved in dry CH2Cl2 (10 mL) was placed in a round-bottom flask under Ar. Diisopropylethylamine (0.5 mL, 2.9 mmol) was added, followed by the dropwise addition of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (0.4 mL, 1.8 mmol). The reaction mixture was stirred 2 h at RT and quenched with ethanol (1 mL). After 10 m the mixture was evaporated to a syrup in vacuo (40 °C). The product 26, a white foam (0.52 g, 0.63 mmol, 65%) was purified by flash chromatography over silica gel using 30-70% EtOAc gradient in hexanes, containing 1% triethylamine, as eluant. Rf 0.48 (CH2Cl2:MeOH / 20:1).
Example 76: 2'-Keto-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-Butylbenzoyl)-Adenosine (28)
Acetic anhydride (4.6 mL) was added to a solution of 3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine (Brown, J.; Christodolou, C.; Jones.S.; Modak.A.; Reese, C; Sibanda,S.; Ubasawa A. J. Chem .Soc. Perkin Trans. I 1989, 1735) (6.2 g, 9.2 mmol) in DMSO (37 mL) and the resulting mixture was stirred at room temperature for 24 h. The mixture was concentrated in vacuo. The residue was taken up in EtOAc and washed with water. The organic layer was dried over MgSO4 and concentrated in vacuo. The residue was purified on a silica gel column to yield 2'-keto-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine 28 (4.8 g, 7.2 mmol, 78%).
Example 77: 2'-Deoxy-2'-methylene-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-Butylbenzoyl)-Adenosine (29) Under a pressure of argon, sec-butyllithium in hexanes (11.2 mL, 14.6 mmol) was added to a suspension of triphenylmethylphosphonium iodide (7.07 g,17.5 mmol) in THF (25 mL) cooled at -78 °C The homogeneous orange solution was allowed to warm to -30 °C and a solution of 2'-keto-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine 28 (4.87 g, 7.3 mmol) in THF (25 mL) was transferred to this mixture under argon pressure. After warming to RT, stirring was continued for 24 h. THF was evaporated and replaced by CH2Cl2 (250 mL), water was added (20 mL), and the solution was neutralized with a cooled solution of 2% HCl. The organic layer was washed with H2O (20 mL), 5% aqueous NaHCO3 (20 mL), H2O to neutrality, and brine (10 mL). After drying (Na2SO4), the solvent was evaporated in vacuo to give the crude compound, which was chromatographed on a silica gel column. Elution with light petroleum ether:EtOAc / 7:3 afforded pure 2'-deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine 29 (3.86 g, 5.8 mmol, 79%).
Example 78: 2'-Deoxy-2'-Methylene-6-N-(4-t-Butylbenzoyl)-Adenosine
2'-Deoxy-2'-methylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine (3.86 g, 5.8 mmol) dissolved in THF (30 mL) was treated with 1 M TBAF in THF (15 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on a silica gel column. 2'-Deoxy-2'-methylene-6-N-(4-t-butylbenzoyl)-adenosine (1.8 g, 4.3 mmol, 74%) was eluted with 10% MeOH in CH2Cl2. Example 79: 5'-O-DMT-2'-Deoxy-2'-Methylene-6-N-(4-t-Butylbenzoyl)-Adenosine (29)
2'-Deoxy-2'-methylene-6-N-(4-t-butylbenzoyl)-adenosine (0.75 g, 1.77 mmol) was dissolved in pyridine (10 mL) and a solution of DMT-Cl (0.66 g, 1.98 mmol) in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. ΝaHCO3, water and brine. The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using 50% EtOAc:hexanes as an eluant to yield 29 (0.81 g, 1.1 mmol, 62%).
Example 80: 5'-O-DMT-2'-Deoxy-2'-Methylene-6-N-(4-t-Butylbenzoyl)- Adenosine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (31)
1-(2'-Deoxy-2'-methylene-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-6-N- (4-t-butylbenzoyl)-adenine 29 dissolved in dry CH2Cl2 (15 mL) was placed in a round bottom flask under Ar. Diisopropylethylamine was added, followed by the dropwise addition of 2-cyanoethyl N, N-diisopropylchlorophosphoramidite. The reaction mixture was stirred 2 h at RT and quenched with ethanol (1 mL). After 10 m the mixture was evaporated to a syrup in vacuo (40 °C). The product was purified by flash chromatography over silica gel using 30-50% EtOAc gradient in hexanes, containing 1 % triethylamine, as eluant (0.7 g, 0.76 mmol, 68%). Rf 0.45 (CH2Cl2: MeOH / 20:1)
Example 81 : 2'-Deoxy-2'-Difluoromethylene-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-Butylbenzoyl)-Adenosine
2'-Keto-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine 28 (6.7 g, 10 mmol) and triphenylphosphine (2.9 g, 11 mmol ) were dissolved in diglyme (20 mL), and heated to a bath temperature of 160 °C A warm (60 °C) solution of sodium chlorodifluoroacetate (2.3 g, 15 mmol) in diglyme (50 mL) was added (dropwise from an equilibrating dropping funnel) over a period of ~1 h. The resulting mixture was further stirred for 2 h and concentrated in vacuo. The residue was dissolved in CH2Cl2 and chromatographed over silica gel. 2'-Deoxy-2'-difluoromethylene-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine (4.1 g, 6.4 mmol, 64%) eluted with 15% hexanes in EtOAc.
Example 82: 2'-Deoxy-2'-Difluoromethylene-6-N-(4-f-Butylbenzoyl)-Adenosine
2'-Deoxy-2'-difluoromethylene-3' 5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-6-N-(4-t-butylbenzoyl)-adenosine (4.1 g, 6.4 mmol) dissolved in THF (20 mL) was treated with 1 M TBAF in THF (10 mL) for 20 m and concentrated in vacuo. The residue was triturated with petroleum ether and chromatographed on a silica gel column. 2'-Deoxy-2'-difluoromethylene-6-N-(4-t-butylbenzoyl)-adenosine (2.3 g, 4.9 mmol, 77%) was eluted with 20% MeOH in CH2Cl2.
Example 83: 5'-O-DMT-2'-Deoxy-2'-Difluoromethylene-6-N-(4-t-Butylbenzoyl)-Adenosine (30)
2'-Deoxy-2'-difluoromethylene-6-N-(4-t-butylbenzoyl)-adenosine (2.3 g, 4.9 mmol) was dissolved in pyridine (10 mL) and a solution of DMT-Cl in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. NaHCO3, water and brine. The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using 50% EtOAc:hexanes as eluant to yield 30 (2.6 g, 3.41 mmol, 69%).
Example 84: 5'-O-DMT-2'-Deoxy-2'-Difluoromethylene-6-N-(4-t-Butylbenzoyl)-Adenosine 3'-(2-Cyanoethyl N,N-diisopropylphosphoramidite) (32)
1-(2'-Deoxy-2'-difluoromethylene-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-6-N-(4-t-butylbenzoyl)-adenine 30 (2.6 g, 3.4 mmol) dissolved in dry C H 2Cl2 (25 mL) was placed in a round bottom flask under Ar. Diisopropylethylamine (1.2 mL, 6.8 mmol) was added, followed by the dropwise addition of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (1.06 mL, 4.76 mmol). The reaction mixture was stirred 2 h at RT and quenched with ethanol (1 mL). After 10 m the mixture evaporated to a syrup in vacuo (40 °C). 32 (2.3 g, 2.4 mmol, 70%) was purified by flash column chromatography over silica gel using 20-50% EtOAc gradient in hexanes, containing 1% triethylamine, as eluant. Rf 0.52 (CH2Cl2: MeOH /15:1).
Example 85: 2'-Deoxy-2'-Methoxycarbonylmethylidine-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-Uridine (33)
Methyl(triphenylphosphoranylidine)acetate (5.4 g, 16 mmol) was added to a solution of 2'-keto-3',5'-O-(tetraisopropyl disiloxane-1 ,3-diyl)-uridine 14 in CH2Cl2 under argon. The mixture was left to stir at RT for 30 h. CH2Cl2 (100 mL) and water were added (20 mL), and the solution was neutralized with a cooled solution of 2% HCl. The organic layer was washed with H2O (20 mL), 5% aq. ΝaHCO3 (20 mL), H2O to neutrality, and brine (10 mL). After drying (Na2SO4), the solvent was evaporated in vacuo to give crude product, that was chromatographed on a silica gel column. Elution with light petroleum ether:EtOAc / 7:3 afforded pure 2'-deoxy-2'-methoxycarbonylmethylidine-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine 33 (5.8 g, 10.8 mmol, 67.5%). Example 86: 2'-Deoxy-2'-Methoxycarbonylmethylidine-Uridine (34)
Et3N•3 HF (3 mL) was added to a solution of 2'-deoxy-2'-methoxycarboxylmethylidine-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine 33 (5 g, 9.3 mmol) dissolved in CH2CI2 (20 mL) and Et3N (15 mL). The resulting mixture was evaporated in vacuo after 1 h and chromatographed on a silica gel column eluting 2'-deoxy-2'-methoxycarbonylmethylidine-uridine 34 (2.4 g, 8 mmol, 86%) with THF:CH2Cl2 / 4:1.
Example 87: 5'-O-DMT-2'-Deoxy-2'-Methoxycarbonylmethylidine-Uridine 2'-Deoxy-2'-methoxycarbonylmethylidine-uridine 34 (1.2 g, 4.02 mmol) was dissolved in pyridine (20 mL). A solution of DMT-Cl (1.5 g, 4.42 mmol) in pyridine (10 mL) was added dropwise over 15 m. The resulting mixture was stirred at RT for 12 h and MeOH (2 mL) was added to quench the reaction. The mixture was concentrated in vacuo and the residue taken up in CH2Cl2 (100 mL) and washed with sat. NaHCO3, water and brine. The organic extracts were dried over MgSO4, concentrated in vacuo and purified over a silica gel column using 2-5% MeOH in CH2Cl2 as an eluant to yield 5'-O-DMT-2'-deoxy-2'-methoxycarbonylmethylidine-uridine 35 (2.03 g, 3.46 mmol, 86%). Example 88: 5'-O-DMT-2'-Deoxy-2'-Methoxycarbonylmethylidine-Uridine 3'-(2-cyanoethyl-N,N-diisopropylphosphoramidite) (36)
1-(2'-Deoxy-2'-2'-methoxycarbonylmethylidine-5'-O-dimethoxytrityl-β-D-ribofuranosyl)-uridine 35 (2.0 g, 3.4 mmol) dissolved in dry CH2Cl2 (10 mL) was placed in a round-bottom flask under Ar. Diisopropylethylamine (1.2 mL, 6.8 mmol) was added, followed by the dropwise addition of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (0.91 mL, 4.08 mmol). The reaction mixture was stirred 2 h at RT and quenched with ethanol (1 mL). After 10 m the mixture was evaporated to a syrup in vacuo (40 °C). 5'-O-DMT-2'-deoxy-2'-methoxycarbonylmethylidine-uridine 3'-(2-cyanoethyl-N,N-diisopropylphosphoramidite) 36 (1.8 g, 2.3 mmol, 67%) was purified by flash column chromatography over silica gel using a 30-60% EtOAc gradient in hexanes, containing 1% triethylamine, as eluant. Rf 0.44 (CH2Cl2:MeOH / 9.5:0.5). Example 89: 2'-Deoxy-2'-Carboxymethylidine-3',5'-O-(Tetraisopropyldisiloxane-1 ,3-diyl)-Uridine 37
2'-Peoxy-2'-methoxycarbonylmethylidine-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine 33 (5.0 g, 10.8 mmol) was dissolved in MeOH (50 mL) and 1 N NaOH solution (50 mL) was added to the stirred solution at RT. The mixture was stirred for 2 h and MeOH removed in vacuo. The pH of the aqueous layer was adjusted to 4.5 with 1 N HCl solution, extracted with EtOAc (2 × 100 mL), washed with brine, dried over MgSO4 and concentrated in vacuo to yield the crude acid. 2'-Deoxy-2'-carboxymethylidine-3',5'-O-(tetraisopropyldisiloxane-1 ,3-diyl)-uridine 37 (4.2 g, 7.8 mmol, 73%) was purified on a silica gel column using a gradient of 10-15% MeOH in CH2Cl2.
The alkyl substituted nucleotides of this invention can be used to form stable oligonucleotides as discussed above for use in enzymatic cleavage or antisense situations. Such oligonucleotides can be formed enzymatically using triphosphate forms by standard procedure. Administration of such oligonucleotides is by standard procedure. See Sullivan et al. PCT WO 94/02595.
Oligonucleotides with 3' and/or 5' Dihalophosphonate
This invention synthesis and uses 3' and/or 5' dihalophosphonate-, e.g., 3' or 5'-CF2-phosphonate-, substituted nucleotides that maintain or enhance the catalytic activity and/or nuclease resistance of an enzymatic or antisense molecule.
As the term is used in this application, 5'- and/or 3'-dihalophosphonate nucleotide containing ribozymes, deoxyribozymes (see
Usman et al., PCT/US94/11649, incorporated by reference herein), and chimeras of nucleotides, are catalytic nucleic molecules that contain 5'-and/or 3'-dihalophosphonate nucleotide components replacing, but not limited to, double-stranded stems, single-stranded "catalytic core" sequences, single-stranded loops or single-stranded recognition sequences. These molecules are able to cleave (preferably, repeatedly cleave) separate RNA or DNA molecules in a nucleotide base sequence specific manner. Such catalytic nucleic acids can also act to cleave intramolecularly if that is desired. Such enzymatic molecules can be targeted to virtually any RNA or DNA transcript. This invention concerns nucleic acids formed of standard nucleotides or modified nucleotides, which also contain at least one 5'-dihalophosphonate and/or one 3'-dihalophosphonate group.
The synthesis of 1 -O- Ac-2,3-di-O- Bz-D-ribofuranose 5-d-5+dihalomethylphosphonate in three steps from 1-O-methyl-2,3-O-isopropylidene-β-D-ribofuranose 5-deoxy-5-dihalomethylphosphonate is described (e.g., for the difluoro, in Figure 87). Condensation of this suitably derivatized sugar with silylated pyrimidines and purines affords novel nucleoside 5'-deoxy-5'-dihalomethylphosphonates. These intermediates may be incorporated into catalytic or antisense nucleic acids by either chemical (conversion of the nucleoside 5'-deoxy-5'-dihalomethylphosphonates into suitably protected phosphoramidites 12a or solid supports 12b, e.g., Figure 88) or enzymatic means (conversion of the nucleoside 5'-deoxy-5'-dihalomethylphosphonates into their triphosphates, e.g., 14 Figure 89, for T7 transcription).
Thus, in one aspect the invention features 5' and/or 3'-dihalonucleotides and nucleic acids containing such 5' and/or 3'-dihalonucleotides. The general structure of such molecules is shown below.
Figure imgf000142_0001
Figure imgf000142_0002
Figure imgf000142_0003
where R 1 is H, OH, or R, where R is a hydroxyl protecting group, e.g., acyl, alkysilyl, or carbonate; each R2 is separately H, OH, or R; each R3 is separately a phosphate protecting group, e.g., methyl, ethyl, cyanoethyl, p-nitrophenyl, or chlorophenyl; each X is separately any halogen; and each B is any nucleotide base.
The invention in particular features nucleic acid molecules having such modified nucleotides and enzymatic activity. In a related aspect the invention features a method for synthesis of such nucleoside 5'-deoxy-5'-dihalo and/or 3'-deoxy-3'-dihalophosphonates by condensing a dihalophosphonate-containing sugar with a pyrimidine or a purine under conditions suitable to form a nucleoside 5'-deoxy-5'-dihalophosphonate and/or a 3'-deoxy-3'-dihalophosphonate.
Phosphonic acids may exhibit important biological properties because of their similarity to phosphates (Engel, Chem. Rev. 1977, 77, 349-367). Blackburn and Kent (J. Chem. Soc, Perkin Trans. 1986, 913-917) indicate that based on electronic and steric considerations _-fluoro and _,_-difluoromethylphosphonates might mimic phosphate esters better than the corresponding phosphonates. Analogues of pyro- and triphosphates 1 , where the bridging oxygen atoms are replaced by a difluoromethylene group, have been employed as substrates in enzymatic processes (Blackburn et al., Nucleosides & Nucleotides 1985, 4, 165-167; Blackburn et al., Chem. Scr. 1986, 26, 21 -24). 9-(5,5-Difluoro-5-phosphonopentyl)guanine (2) has been utilized as a multisubstrate analogue inhibitor of purine nucleoside phosphorylase (Halazy et al., J. Am. Chem. Soc. 1991 , 113, 315-317). Oligonucleotides containing methylene groups in place of phosphodiester 5'-oxygens are resistant toward nucleases that cleave phosphodiester linkages between phosphorus and the 5'-oxygen (Breaker et al., Biochemistry 1993, 32, 9125-9128), but can still form stable complexes with complementary sequences. Heinemann et al. (Nucleic Acids Res. 1991 , 19, 427-433) found that a single 3'-methylenephosphonate linkage had a minor influence on the conformation of a DNA octamer double helix.
Figure imgf000144_0001
One common synthetic approach to α,α-difluoro-alkylphosphonates features the displacement of a leaving group from a suitable reactive substrate by diethyl (lithiodifluoromethyl)phosphonate (3) (Obayashi et al., Tetrahedron Lett. 1982, 23, 2323-2326). However, our attempts to synthesize nucleoside 5'-deoxy-5'-difluoro-methylphosphonates from 5'-deoxy-5'-iodonucleosides using 3 were unsuccessful, i.e. starting compounds were quantitatively recovered. The reaction of nucleoside 5'-aldehydes with 3, according to the procedure of Martin et al. (Martin et al., Tetrahedron Lett. 1992, 33, 1839-1842), led to a complex mixture of products. Recently, the synthesis of sugar α,α-difluoroalkylphosphonates from primary sugar triflates using 3 was described (Berkowitz et al., J. Org. Chem. 1993, 58, 6174-6176). Unfortunately, our experience is that nucleoside 5'-triflates are too unstable to be used in these syntheses. The following are non-limiting examples showing the synthesis of nucleoside 5'-deoxy-5'-difluoromethyl-phosphonates. Those in the art will recognize that equivalent methods can be readily devised based upon these examples. These examples demonstrate that it is possible to achieve synthesis of 5'-deoxy-5'-difluoro derivatives in good yield and thus guide those in the art to such equivalent methods. The examples also indicate utility of such synthesis to provide useful oligonucleotides as described above.
Those in the art will recognize that useful modified enzymatic nucleic acids can now be designed, much as described by Draper et al., PCT/US94/13129 hereby incorporated by reference herein (including drawings). Example 90: Synthesis of Nucleoside 5'-Peoxy-5'-difluoromethylphosphonates
Referring to Fig. 87, we synthesized a suitable glycosylating agent from the known D-ribose α,α-difluoromethylphosphonate (4) (Martin et al.,
Tetrahedron Lett. 1992, 33, 1839-1842) which served as a key intermediate for the synthesis of nucleoside 5'-difluoromethylphosphonates.
Methyl 2,3-O - i s o p ro pyl i d e n e- β -D-ribofu ranose a, a-difluoromethylphosphonate (4) was synthesized from the 5-aldehyde according to the procedure of Martin et al. (Tetrahedron Lett. 1992, 33, 1839-1842) (Figure 87). Removal of the isopropylidene group was accomplished under mild conditions (I2-MeOH, reflux, 18 h (Szarek et al., Tetrahedron Lett. 1986, 27, 3827) or Dowex 50 WX8 (H+), MeOH, RT (about 20-25°C), 3 days) in 72% yield. The anomeric mixture thus obtained was benzoylated with benzoyl chloride/pyridine to afford the 2,3-di-O-benzoyl derivative, which was subjected to mild acetolysis conditions (Walczak et al., Synthesis, 1993, 790-792) (Ac2O, AcOH, H2SO4, EtOAc, 0°C The desired 1 -O-acetyl-2,3-di-O- benzoyl-D-ribofu ranose difluoromethylphosphonate (5) was obtained in quantitative yield as an anomeric mixture. These derivatives were used for selective glycosylation of silylated uracil and N4-acetylcytosine under Vorbrüggen conditions (Vorbrüggen, Nucleoside Analogs. Chemistry, Biology and Medical Applications, NATO ASI Series A, 26, Plenum Press, New York, London, 1980; pp. 35-69. The use of F3CSO2OSi(CH3)3 as a glycosylation catalyst is precluded because it is expected to lead to the undesired 1-ethyluracil or 9-ethyladenine byproducts: Podyukova, et al., Tetrahedron Lett. 1987, 28, 3623-3626 and references cited therein) (SnCl4 as a catalyst, boiling acetonitrile) to yield β-nucleosides (62% 6a, 75% 6b). Glycosylation of silylated N6-benzoyladenine under the same conditions yielded a mixture of N-9 isomer 6c and N-7 isomer 7 in 34% and 15% yield, respectively. The above nucleotides were successfully deprotected using trimethylsilylbromide for the cleavage of the ethyl groups, followed by treatment with ammonia-methanol to remove the acyl protecting groups. Nucleoside 5'-deoxy-5'-difluoromethylphosphonates 8 were finally purified on a DEAE Sephadex A-25 (HCO3-) column using a 0.01-0.25 M TEAB gradient for elution and obtained as their sodium salts (82% 8a; 87% 8b; 82% 8c).
Selected analytical data: 31 P-NMR (31 P) and 1 H-NMR (1 H) were recorded on a Varian Gemini 400. Chemical shifts in ppm refer to H3PO4 and TMS, respectively. Solvent was CDCI3 unless otherwise noted. 5: 1 H δ 8.07-7.28 (m, Bz), 6.66 (d, J 1 ,2 4.5, αH1), 6.42 (s, βH1), 5.74 (d, J2 ,3 4.9, βH2), 5.67 (dd, J3,2 4.9, J3,4 6.6, βH3), 5.63 (dd, J3,2 6.7, J3,4 3.6, αH3), 5.57 (dd, J2,1 4.5, J2,3 6.7, αH2), 4.91 (m, H4), 4.30 (m, CH2CH3), 2.64 (m, CH2CF2), 2.18 (s, βAc), 2.12 (s, αAc), 1.39 (m, CH2CH3). 31 P δ 7.82 (t, JP ,F 105.2), 7.67 (t, JP,F 106.5). 6a: 1 H δ 9.11 (s, 1 H, NH), 8.01 (m, 11H, Bz, H6), 5.94 (d, J 1 ',2' 4.1 , 1 H, H1'), 5.83 (dd, J5,6 8.1 , 1 H, H5), 5.79 (dd,
J2', 1 ' 4.1 , J2',3' 6.5, 1 H, H2'), 5.71 (dd, J3',2' 6.5, J3',4' 6.4, 1 H, H3'), 4.79 (dd, J4',3. 6.4, J4',F 11.6, 1 H, H4'), 4.31 (m, 4H, CH2CH3), 2.75 (tq, JH ,F 19.6, 2H, CH2CF2), 1.40 (m, 6H, CH2CH3). 31 P δ 7.77 (t, JP,F 104.0). 8c: 31 P (vs DSS) (D2O) δ 5.71 (t, JP,F 87.9). Compound 7 was deacylated with methanolic ammonia yielding the product that showed λmax (H2O) 271 nm and λmjn 233 nm, confirming that the site of glycosylation was N-7.
Example 91 :Synthesis of Nucleic Acids Containing Modified Nucleotide Containing Cores
The method of synthesis used follows the procedure for normal RNA synthesis as described in Usman et al., J. Am. Chem. Soc. 1987, 109, 7845-7854 and in Scaringe et al., Nucleic Acids Res. 1990, 18, 5433-5441 and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end (Figure 88 and Janda et al., Science 1989, 244:437-440.). These nucleoside 5'-deoxy-5'-difluoromethylphosphonates may be incorporated not only into hammerhead ribozymes, but also into hairpin, hepatitis delta virus, Group 1 or Group 2 introns, or into antisense oligonucleotides. They are, therefore, of general use in any nucleic acid structure. Example 92: Synthesis of Modified Triphosphate
The triphosphate derivatives of the above nucleotides can be formed as shown in Fig. 89. according to known procedures. Nucleic Acid Chem.,
Leroy B. Townsend, John Wiley & Sons, New York 1991 , pp. 337-340;
Nucleotide Analogs, Karl Heinz Scheit; John Wiley & Sons New York 1980, pp. 211-218.
Equivalent synthetic schemes for 3' dihalophosphonates are shown in Figures 90 and 91 using art recognized nomenclature. The conditions can be optimized by standard procedures.
The nucleoside dihalophosphonates described herein are advantageous as modified nucleotides in any nucleic acid structure, e.g., catalytic or antisense, since they are resistant to exo- and endonucleases that normally degrade unmodified nucleic acids in vivo. They also do not perturb the normal structure of the nucleic acid in which they are incorporated thereby maintaining any activity associated with that structure. These compounds may also be of use as monomers as antiviral and/or antitumor drugs.
Oligonucleotides with Amido or Peptido Modification
This invention replaces 2'-hydroxyl group of a ribonucleotide moiety with a 2'-amido or 2'-peptido moiety. In other embodiments, the 3' and 5' portions of the sugar of a nucleotide may be substituted, or the phosphate group may be substituted with amido or peptido moieties. Generally, such a nucleotide has the general structure shown in Formula I below:
Figure imgf000148_0001
FORMULA I
The base (B) is any one of the standard bases or is a modified nucleotide base known to those in the art, or can be a hydrogen group. In addition, either R1 or R2 is H or an alkyl, alkene or alkyne group containing between 2 and 10 carbon atoms, or hydrogen, an amine (primary, secondary or tertiary, e.g., R3NR4 where each R3 and R4 independently is hydrogen or an alkyl, alkene or alkyne having between 2 and 10 carbon atoms, or is a residue of an amino acid, Le,., an amide), an alkyl group, or an amino acid (D or L forms) or peptide containing between 2 and 5 amino acids. The zigzag lines represent hydrogen, or a bond to another base or other chemical moiety known in the art. Preferably, one of R 1 , R2 and R3 is an H, and the other is an amino acid or peptide.
Applicant has recognized that RNA can assume a much more complex structural form than DNA because of the presence of the 2'-hydroxyl group in RNA. This group is able to provide additional hydrogen bonding with other hydrogen donors, acceptors and metal ions within the
RNA molecule. Applicant now provides molecules which have a modified amine group at the 2' position, such that significantly more complex structures can be formed by the modified oligonucleotide. Such modification with a 2'-amido or peptido group leads to expansion and enrichment of the side-chain hydrogen bonding network. The amide and peptide moieties are responsible for complex structural formation of the oligonucleotide and can form strong complexes with other bases, and interfere with standard base pairing interactions. Such interference will allow the formation of a complex nucleic acid and protein conglomerate. Oligonucleotides of this invention are significantly more stable than existing oligonucleotides and can potentially form biologically active bioconjugates not previously possible for oligonucleotides. They may also be used for in vitro selection of unique aptamers, that is, randomly generated oligonucleotides which can be folded into an effective ligand for a target protein, nucleic acid or polysaccharide.
Thus, in one aspect, the invention features an oligonucleotide containing the modified base shown in Formula I, above.
In other aspects, the oligonucleotide may include a 3' or 5' nucleotide having a 3' or 5' located amino acid or aminoacyl group. In all these aspects, as well as the 2'-modified nucleotide, it will be evident that various standard modifications can be made. For example, an "O" may be replaced with an S, the sugar may lack a base (i.e., abasic) and the phosphate moiety may be modified to include other substitutions (see Sproat, supra).
Example 93: General procedure for the preparation of 2'-aminoacyl-2'-deoxy-2'-aminonucleoside conjugates.
Referring to Fig. 92. to the solution of 2'-deoxy-2'-amino nucleoside (1 mmol) and N-Fmoc L- (or D-) amino acid (1 mmol) in methanol [dimethylformamide (DMF) and tetrahydrofuran (THF) can also be used], 1-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ) [or 1 -isobutyloxycarbonyl-2-isobutyloxy-1 ,2-dihydroquinoline (IIDQ)] (2 mmol) is added and the reaction mixture is stirred at room temperature or up to 50°C from 3-48 hours. Solvents are removed under reduced pressure and the residual syrup is chromatographed on the column of silica-gel using 1-10 % methanol in dichloromethane. Fractions containing the product are concentrated yielding a white foam with yields ranging from 85 to 95 %. Structures are confirmed by 1 H NMR spectra of conjugates which show correct chemical shifts for nucleoside and aminoacyl part of the molecule. Further proofs of the structures are obtained by cleaving the aminoacyl protecting groups under appropriate conditions and assigning 1H NMR resonances for the fully deprotected conjugate.
Partially protected conjugates described above are converted into their 5'-O-dimethoxytrityl derivatives and into 3'-phosphoramidites using standard procedures (Oligonucleotide Synthesis: A Practical Approach, M.J. Gait ed.; IRL Press, Oxford, 1984). Incorporation of these phosphoramidites into RNA was performed using standard protocols (Usman et al., 1987 supra).
A general deprotection protocol for oligonucleotides of the present invention is described in Fig. 93.
The scheme shows synthesis of conjugate of 2'-d-2'-aminouridine.
This is meant to be a non-limiting example, and those skilled in the art will recognize that, variations to the synthesis protocol can be readily generated to synthesize other nucelotides (e.g., adenosine, cytidine, guanosine) and/or abasic moieties.
Example 94: RNA cleavage bv hammerhead ribozymes containing 2'-aminoacyl modifications.
Hammerhead ribozymes targeted to site N (see Fig. 94) are synthesized using solid-phase synthesis, as described above. U4 and U7 positions are modified, individually or in combination, with either 2'-NH-alanine or 2'-NH-lysine.
RNA cleavage assay in vitro: Substrate RNA is 5' end-labeled using [γ-32P] ATP and T4 polynucleotide kinase (US Biochemicals). Cleavage reactions were carried out under ribozyme "excess" conditions. Trace amount (≤ 1 nM) of 5' end-labeled substrate and 40 nM unlabeled ribozyme are denatured and renatured separately by heating to 90°C for 2 min and snap-cooling on ice for 10 -15 min. The ribozyme and substrate are incubated, separately, at 37°C for 10 min in a buffer containing 50 mM Tris-HCl and 10 mM MgCl2. The reaction is initiated by mixing the ribozyme and substrate solutions and incubating at 37°C Aliquots of 5 μl are taken at regular intervals of time and the reaction is quenched by mixing with equal volume of 2X formamide stop mix. The samples are resolved on 20 % denaturing polyacrylamide gels. The results are quantified and percentage of target RNA cleaved is plotted as a function of time.
Referring to Fig. 95. hammerhead ribozymes containing 2'-NH-alanine or 2'-NH-lysine modifications at U4 and U7 positions cleave the target RNA efficiently. Sequences listed in Figure 94 and the modifications described in Figure 95 are meant to be non-limiting examples. Those skilled in the art will recognize that variants (base-substitutions, deletions, insertions, mutations, chemical modifications) of the ribozyme and RNA containing other 2'-hydroxyl group modifications, including but not limited to amino acids, peptides and cholesterol, can be readily generated using techniques known in the art, and are within the scope of the present invention.
Example 95: Aminoacylation of 3'-ends of RNA
I. Referring to Fig. 96. 3'-OH group of the nucleotide is converted to succinate as described by Gait, supra. This can be linked with amino-alkyl solid support (for example: CpG). Zig-zag line indicates linkage of 3'OH group with the solid support.
1L Preparation of aminoacyl-derivatized solid support
A) Synthesis of O-Dimethoxytrityl (O-DMT) amino acids
Referring to Fig. 97, to a solution of L- (or P-) serine, tyrosine or threonine (2 mmol) in dry pyridine (15 ml) 4,4'-dimethoxytrityl chloride (3 mmol) is added and the reaction mixture is stirred at RT (about 20-25°C) for 16 h. Methanol (10 ml) is then added and the solution evaporated under reduced pressure. The residual syrup was partitioned between 5% aq. NaHCO3 and dichloromethane, organic layer was washed with brine, dried (Na2SO4) and concentrated in vacuo. The residue is purified by flash silicagel column chromatography using 2-10% methanol in dichloromethane (containing 0.5 % pyridine). Fractions containing product are combined and concentrated in vacuo to yield white foam (75-85 % yield).
B) Preparation of the solid support and its derivatization with amino acids
Referring to Fig. 97, the modified solid support (has an OH group instead of the standard NH2 end group) was prepared according to Haralambidis et al., Tetrahedron Lett. 1987, 28, 5199, (P denotes aminopropyl CPG or polystyrene type support). O-DMT or NH-monomethoxytrityl (NH-MMT amino acid was attached to the above solid support using standard procedures for derivatization of the solid support (Gait, 1984, supra) creating a base-labile ester bond between amino acids and the support. This support is suitable for the construction of RNA/DNA chain using suitably protected nucleoside phosphoramidites.
Example 96: Aminoacylation of 5'-ends of RNA
I. Referring to Fig. 98. 5'-amino-containing sugar moiety was synthesized as described (Mag and Engels, 1989 Nucleic Acids Res. 17,
5973). Aminoacylation of the 5'-end of the monomer was achieved as described above and RNA phosphoramidite of the 5'-aminoacylated monomer was prepared as described by Usman et al., 1987 supra. The phosphoramidite was then incorporated at the 5'-end of the oligonucleotide using standard solid-phase synthesis protocols described above.
II. Referring to Fig. 99, aminoacyl group(s) is attached to the phosphate group at the 5'-end of the RNA using standard procedures described above.
VII. Reversing Genetic Mutations Modification of existing nucleic acid sequences can be achieved by homologous recombination. In this process a transfected sequence recombines with homologous chromosomal sequences and can replace the endogenous cellular sequence. Boggs, 8 International J. Cell Cloning 80, 1990, describes targeted gene modification. It reviews the use of homologous DNA recombination to correct genetic defects. Banga and Boyd, 89 Proc. Natl. Acad. Sci. U.S.A. 1735, 1992, describe a specific example of in vivo site-directed mutagenesis using a 50 base oligonucleotide. In this methodology a gene or gene segment is essentially replaced by the oligonucleotide used. This invention uses a complementary oligonucleotide to position a nucleotide base changing activity at a particular site on a gene (RNA or genomic DNA), such that the nucleotide modifying activity will change (or revert) a mutation to wild-type, or its equivalent. By reversion or change of a mutation, we refer to reversion in a broad sense, such as when a mutation at a second site which leads to functional reversion to a wild type phenotype. Also, due to the degeneracy of the genetic code, a revertant may be achieved by changing any one of the three codon positions. Additionally, creation of a stop codon in a deleterious gene (or transcript) is defined here as reverting a mutant phenotype to wild-type. An example of this type of reversion is creating a stop codon in a critical HIV proviral gene in a human.
Referring to Figures 100 and 101 , broadly there are two approaches to causing a site directed change in order to revert a mutation to wild-type. In one (Fig. 100) the oligonucleotide is used to target RNA specifically. RNA is provided with a complementary (Watson-crick) oligonucleotide sequence to that in the target molecule. In this case the sequence modifying oligonucleotide would (analogously to an antisense oligonucleotide or ribozyme) have to be continuously present to revert the RNA as it is made by the cell. Such a reversion would be transient and would potentially require continuous addition of more sequence modifying oligonucleotide. The transient nature of this approach is an advantage, in that treatment could be stopped by simply removing the sequence modifying oligonucleotide (as with a traditional drug). A second approach targets DNA (Fig. 101) and has the advantage that changes may be permanently encoded in the target cell's genetic code. Thus, a single course (or several courses) of treatment may lead to permanent reversion of the genetic disease. If inadvertent chromosomal mutations are introduced this may cause cancer, mutate other genes, or cause genetic changes in the germ-line (in patients of reproductive age). However, if the base changing activity is a specific methylation that may modulate gene expression it would not necessarily lead to germ-line transmission. See Lewin, Genes, 1983 John Wilely & Sons, Inc. NY pp 493-496. Complementary base pairing to single-stranded PNA or RNA is one method of directing an oligonucleotide to a particular site of PNA. This could occur by a strand displacement mechanism or by targeting PNA when it is single-stranded (such as during replication, or transcription). Another method is using triple-strand binding (triplex formation) to double-stranded PNA, which is an established technique for binding polypyrimidine tracts, and can be extended to recognize all 4 nucleotides. See Povsic, T., Strobel, S., & Dervan, P. (1992). Sequence-specific double-strand alkylation and cleavage of DNA mediated by triple-helix formation. J. Am. Chem. Soc. 114. 5934-5944 (1992). Knorre, D.G., Valentin, V.V., Valentina, F.Z., Lebedev, AN. & Federova, O.S. Design and targeted reactions of oligonucleotide derivatives 1-366 (CRC Press, Novosibirsk, 1993) describe conjugation of reactive groups or enzyme to oligonucleotides and can be used in the methods described herein.
Recently, antisense oligonucleotides have been used to redirect an incorrect splice into order to obtain correct splicing of a splice mutant globin gene in vitro. Dominski Z; Kole R (1993) Restoration of correct splicing in thalassemia pre-mRNA by antisense oligonucleotides. Proc Natl Acad Sci U S A 90:8673-7. Analogously, in one preferred embodiment of this invention a complementary oligomer is used to correct an existiing mutant RNA, instead of the traditional approach of inhibiting that RNA by antisense.
In either the RNA or DNA mode, after binding to a particular site on the RNA or DNA the oligonucleotide will modify the nucleic acid sequence. This can be accomplished by activating an endogenous enzyme (see Figure 102). by appropriate positioning of an enzyme (or ribozyme) conjugated (or activated by the duplex) to the oligonucleotide, or by appropriate positioning of a chemical mutagen. Specific mutagens, such as nitrous acid which deaminates C to U, are most useful, but others can also be used if inactivation of a harmful RNA is desired.
RNA editing is an naturally occurring event in mammalian cells in which a sequence modifying activity edits a RNA to its proper sequence post-transcriptionally. Higuchi, M.,, Single, F., Kohler, M., Sommer, B., and Seeburg, P. (1993) RNA Editing of AMPA Receptor Subunit GluR-B: A base-paired intron-exon structure determines position and efficiency Cell 75:1361-1370. The machinery involved in RNA editing can be co-opted by a suitable oligonucleotide in order to promote chemical modification.
The changes in the base created by the methods of this invention cause a change in the nucleotide sequence, either directly, or after DNA repair by normal cellular mechanisms. These changes functionally correct a genetic defect or introduce a stop codon. Thus, the invention is distinct from techniques in which an active chemical group (e.g., an alkylator) is attached to an antisense or triple strand oligonucleotide in order to chemically inactivate the target RNA or DNA.
Thus, this invention creates an alteration to an existing base in a nucleic acid molecule so that the base is read in vivo as a different base. This includes correcting a sequence instead of inactivating a gene but can also include inactivating a deleterious gene.
Thus, in one aspect, the invention features a method for altering in vivo the nucleotide base sequence of a naturally occurring mutant nucleic acid molecule. The method includes contacting the nucleic acid molecule in vivo with an oligonucleotide or peptide nucleic acid or other sequence specific binding molecules able to form a duplex or triplex molecule with the nucleic acid molecule. After formation of the duplex or triplex molecule a base modifying activity chemically or enzymatically alters the targeted base directly, or after nucleic acid repair in vivo. This results in the functional alteration of the nucleic acid sequence.
By "alter", as it is used in this context, is meant that one or more chemical moieties in a targeted base, or bases, is altered so that the mutant nucleic acid will be functionally different. Thus, this is distinct from prior methods of correcting defects in DNA, such as homologous recombination, in which an entire segment of the targeted sequence is replaced with a segment of DNA from the transfected nucleic acid. This is also distinct from other methods that use reactive groups to inactivate a RNA or DNA target, in that this method functionally corrects the sequence of the target, instead of merely damaging it, by causing it to be read by a polymerase as a different base from the original base. As noted above, the naturally occurring enzymes in a cell can be utilized to cause the chemical alteration, examples of which are provided below.
By "functionally alter" is meant that the ability of the target nucleic acid to perform its normal function (i.e.., transcription or translation control) is changed. For example, an RNA molecule may be altered so that it can cause production of a desired protein, or a DNA molecule can be altered so that upon DNA repair, the DNA sequence is changed.
By "mutant" it is meant a nucleic acid molecule which is altered in some way compared to equivalent molecules present in a normal individual. Such mutants may be well known in the art, and include, molecules present in individuals with known genetic deficiencies, such as muscular dystrophy, or diabetes and the like. It also includes individuals with diseases or conditions characterized by abnormal expression of a gene, such as cancer, thalassemia's and sickle cell anemia, and cystic fibrosis. It allows modulation of lipid metabolism to reduce artery disease, treatment of integrated AIDS genomes, and AIDs RNA, and Alzeimer's disease. Thus, this invention concerns alteration of a base in a mutant to provide a "wild type" phenotype and/or genotype. For deleterious conditions this involves altering a base to allow expression or prevent expression as is necassary. When treating an infection, such as HIV, it concerns inactivation of a gene in the HIV RNA by mutation of the mutant (i.e., non-human gene) to a wild type (i.e., no production of a non-human protein). Such modification is performed in trans rather than in cis as in prior methods.
In preferred embodiments, the oligonucleotide is of a length (at least 12 bases, preferably 17 - 22) sufficient to activate dsRNA deaminase in vivo to cause conversion of an adenine base to inosine; the oligonucleotide is an enzymatic nucleic acid molecule that is active to chemically modify a base (see below); the nucleic acid molecule is DNA or RNA; the oligonucleotide includes a chemical mutagen, e.g., the mutagen is nitrous acid; and the oligonucleotide causes deamination of 5-methylcytosine to thymidine, cytosine to uracil, or adenine to inosine, or methtylation of cytosine to 5-methylcytosine. In a most preferred embodiment, the invention features correction of a mutation, rather than inactivation of a target by causing a mutation.
Using in vitro directed evolution, it is possible to screen for ribozymes with catalytic activities different than RNA cleavage. Bartel, D. and Szostak, J. (1993) Isolation of new ribozymes from a large pool of random sequences. Science 261 :1411-1418. Using these methods of in vitro directed evolution, an enzymatic nucleic acid molecule, or ribozyme that mutates bases, instead of cleaving the phosphodiester backbone can be selected. This is a convenient method of obtaining an enzyme with the appropriate base sequence modifying activities for use in the present invention.
Sequence modifying activities can change one nucleotide to another
(or modify a nucleotide so that it will be repaired by the cellular machinery to another nucleotide). Sequence modifying activities could also delete or add one or more nucleotides to a sequence. A specific embodiment of adding sequences is described by Sullenger and Cech, PCT/US94/12976 hereby incorporated by reference herein), in which entire exons with wild-type sequence are spliced into a mutant transcript. The present invention features only the addition of a few bases (1 - 3).
Thus, in another aspect, the invention features ribozymes or enzymatic nucleic acid molecules active to change the chemical structure of an existing base in a separate nucleic acid molecule. Applicant is the first to determine that such molecules would be useful, and to provide a description of how such molecules might be isolated.
Molecules used to achieve in situ reversion can be delivered using the existing means employed for delivering antisense molecules and ribozymes, including liposomes and cationic lipid complexes. If the in situ reverting molecule is composed only of RNA, then expression vectors can be used in a gene therapy protocol to produce the reverting molecules endogenously, analogously to antisense or ribozymes expression vectors. There are several advantages of using such an expression vector, rather than simply replacing the gene through standard gene therapy. Firstly, this approach would limit the production of the corrected gene to cells that already express that gene. Furthermore, the corrected gene would be properly regulated by its natural transcriptional promoter. Lastly, reversion can be used when the mutant RNA creates a dominant gain of function protein (e.g., in sickle cell anemia), where correction of the mutant RNA is necessary to stop the production of the deleterious mutant protein, and allow production of the corrected protein.
Endogenous Mammalian RNA Editing System It was observed in the mid-1980s that the sequence of certain cellular
RNAs were different from the DNA sequence that encodes them. By a process called RNA editing, cellular RNA are post-transcriptionally modified to a) create a translation initiation and termination codons, b) enable tRNA and rRNA to fold into a functional conformation (for a review see Bass, B. L. (1993) In The RNA World. R. Gesteland, R. and Atkins, J. eds. (Cold Spring Harbor, New York; CSH Lab. Press) pp. 383-418). The process of RNA editing includes base modification, deletion and insertion of nucleotides.
Although, the RNA editing process is widespread among lower eukaryotes, very few RNAs (four) have been reported to undergo editing in mammals (Bass, supra). The predominant mode of RNA editing in mammalian system is base modification (C → U and A→ G). The mechanism of RNA editing in the mammalian system is postulated to be that C→U conversion is catalyzed by cytidine deaminase. The mechanism of conversion of A→G has recently been reported for glutamate receptor B subunit (gluR-B) in rat PC12 cells (Higuchi, M. et al. (1993) Cell 75, 1361-1370). According to Higuchi gluR-B mRNA precursor attains a structure such that intron 11 and exon 11 can form a stable stem-loop structure. This stem-loop structure is a substrate for a nuclear double strand-specific adenosine deaminase enzyme. The deamination will result in the conversion of A→l. Reverse transcription followed by double strand synthesis will result in the incorporation of G in place of A.
In the present invention, the endogenous deaminase activity or other such activities can be utilized to achieve targeted base modification. The following are examples of the invention to illustrate different methods by which in vivo conversion of a base can be achieved. These are provided only to clarify specific embodiments of the invention and are not limiting to the invention. Those in the art will recognize that equivalent methods can be readily devised within the scope of the claims. Example 97: Exploiting cellular dsRNA dependent Adenine to Inosine converter:
An endogenous activity in most mammalian cells and Xenopus oocytes converts about 50% of adenines to inosines in double stranded RNA. (Bass, B. L., & Weintraub, H. (1988). An unwinding activity that covalently modifies it double-stranded RNA substrate. Cell, 55, 1089-1098.). This activity can be used to cause an in situ reversion of a mutation at the RNA level. Referring to Figures 102 and 104, for demonstration purposes a stop codon is incorporated into the coding region of dystrophin, which is fused to the reporter gene luciferase. This stop codon can be reverted by targeting an antisense RNA which is long enough to activate the dsRNA deaminase, which converts Adenines to Inosines. The A to I transition will be read by the ribosome as an A to G transition in some cases and will thereby functionally revert the stop codon. While other A's in this region may be converted to I's and read as G, converting an A to I (G) cannot create a stop codon. The A to I transitions in the region surrounding the target mutation will create some point mutations, however, the function of the dystrophin protein is rarely inactivated by point mutations.
The reverted mRNA was then translated in a cell lysate and assayed for luciferase activity. As evidenced by the dramatic increase in luciferase counts in the graph in figure 103, the A to I transition was read by the ribosome as an A to G transition and the stop codon has successfully been reverted with the lysate treated complex. As a control, an irrelevant non-complementary RNA oligonucleotide was added to the dystrophin/luciferase mRNA. As expected, in this case no translation (luciferase activity) is observed because of the stop codon. As an additional control, the hybrid was not treated with extract, and again no translation (Iuciferase activity) is observed (Figure 103).
While other A's in the targeted region may have been converted to I's and read as G, converting an A to I (G) cannot create a stop codon, so the ribosome will still read through the region. Dystrophin is not generally sensitive to point mutations if the open reading frame is maintained, so a dystrophin protein made from an mRNA reverted by this method should retain full activity. The following detail specifics of the methodology: RNA oligonucleotides were synthesized on a 394 (ABI) synthesizer using phosphoramidite chemistry. The sequence of the synthetic complementary RNA that binds to the mutant dystrophin sequence is as follows (5' to 3'):
CCCGCGGTAGATCTTTCTGGAGGCTTACAGTTTTCTACAAACCTCC CTTCAAA (Seq. ID No. 1)
Referring to Figure 104, fifty-nine base pairs of a human dystrophin mutant sequence containing a stop codon was fused in frame to the luciferase coding region using standard cloning technology, into the Hind III and Not I sites of pRC-CMV (Invitrogen, San Diego, CA). The AUG of luciferase was deleted. The sequences of the insert from the Hind III site to the start of the Iuciferase coding region is (5' to 3'):
GCCCCTGAGGAGCGATGGAGGCCTTGAAGGGAGGTTTGTGGAAAA CTGTAAGCCTCCAGAAAGATCTACCGCGG (Seq ID No. 2) This corresponds to base pairs 3649-3708 of normal dystrophin (Entrez IP # 311627) with a Sac II site at the 3' end. This plasmid was used as a template for in vitro transcription of mRNA using T7 polymerase with the manufacturers protocol (Promega, Madison, WI). Xenopus nuclear extracts were prepared in 0.5X TGKED buffer (0.5X=
25mM Tris (pH 7.9), 12.5% glycerol, 25 mM KCl, 0.25mM PTT and 0.05mM EDTA), by vortexing nuclei and resuspended in a volume of 0.5X TGKEP equal to total cytoplasm volume of the oocytes. Bass, B.L. & Weintraub, H. Cell 55, 1089-1098 (1988). The target mRNA at 500ng/ul was pre-annealed to 1 micromolar complementary or irrelevant RNA oligonucleotide by heating to 70°C, and allowing it to slowly cool to 37°C over 30 minutes. Fifty nanograms of mRNA pre-annealed to the RNA oligonucleotides was added to 7ul of nuclear extracts containing 1 mM ATP, 15mM EDTA, 1600un/ml RNasin and 12.5mM Tris pH 8 to a total volume of 12ul. Bass, B.L. & Weintraub, H. supra. This mixture, which contains the dsRNA deaminase activity, was incubated for 30 minutes at 25°C Next, 1.5ul of this mixture was added to a rabbit reticulocyte lysate in vitro translation mixture and translated for two hours according to the manufacturers protocol (Life Technologies, Gaithersberg, MP), except that an additional 1.3 mM magnesium acetate was added to compensate for the EPTA carried through from the nuclear extract mixture. Luciferase assays were performed on 15ul of extract with the Promega luciferase assay system (Promega, Madison, WI), and luminescence was detected with a 96 well luminometer, and the results are displayed in the graph in figure 102.
Example 98: Base changing activities
The chemical synthesis of antisense and triple-strand forming oligomers conjugated to reactive groups is well studied and characterized (Knorre, D.G., Valentin, V.V., Valentina, F.Z., Lebedev, AN. & Federova, O.S. Design and targeted reactions of oligonucleotide derivatives 1-366 (CRC Press, Novosibirsk, 1993) and Povsic, T., Strobel, S. & Dervan, P. Sequence-specific double-strand alkylation and cleavage of DNA mediated by triple-helix formation J. Am. Chem. Soc. 114, 5934-5944 (1992). Reactive groups such as alkylators that can modify nucleotide bases in targeted RNA or DNA have been conjugated to oligonucleotides. Additionally enzymes that modify nucleic acids have been conjugated to oligonucleotides. (Knorre, P.G., Valentin, V.V., Valentina, F.Z., Lebedev, AN. & Federova, O.S. Design and targeted reactions of oligonucleotide derivatives 1 -366 (CRC Press, Novosibirsk, 1993). In the past these conjugated chemical groups or enzymes have been used to inactivate DNA or RNA that is specifically targeted by antisense or triple-strand interactions. Below is a list of useful base changing activities that could be used to change the sequence of DNA or RNA targeted by antisense or triple strand interactions, in order to achieve in situ reversion of mutations, as described herein (see figure 100-104).
1. Deamination of 5-methylcytosine to create thymidine
(performed by the enzyme cytidine deaminase (Bass, B.L. in The RNA World (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1993). Also, nitrous acid or related compounds promote oxidative deamination of C to be read at T(Microbial Genetics, David Freifelder, Jones and Bartlett Publishers, Inc., Boston, 1987, PP.226-230.). Additionally hydroxylamine or related compounds can transform C to be read at T (Microbial Genetics, David Freifelder, Jones and Bartlett Publishers, Inc., Boston, 1987, PP.226-230.) 2. Deamination of cytosine to create uracil (performed by the enzyme cytidine deaminase (Bass, B.L. in The RNA World (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1993) or by chemical groups similar to nitrous acid that promote oxidative deamination (Microbial Genetics, David Freifelder, Jones and Bartlett Publishers, Inc., Boston, 1987, PP.226-230.)
3. Deamination of Adenine to be read like G (Inosine) (as done by the adenosine deaminase, AMP deaminase or the dsRNA deaminating activity ( Bass, B.L. in The RNA World (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1993). 4. Methylation of cytosine to 5-methylcytosine
5. Transforming thymidine (or uracil) to O2-methyl thymidine (or
O2-methyl uracil), to be read as cytosine by alkynitrosoureas (Xu, and Swann, Tetrahedron Letters 35:303-306 (1994)). 6. Transforming guanine to 6-O-methyl (or other alkyls) to be read as adenine (Mehta and Ludlum, Biochimica et Biophysica Acta, 521 :770-778 (1978) which can be done with the mutagen ethyl methane sulfonate (EMS) Microbial Genetics, David Freifelder, Jones and Bartlett Publishers, Inc., Boston, 1987, PP.226-230.
7. Amination of uracil to cytosine (as performed by the cellular enzyme CTP synthetase (Bass, B.L. in The RNA World (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1993).
The following are examples of useful chemical modifications that can be utilized in the present invention. There are a few preferred straightforward chemical modifications that can change one base to another base. Appropriate mutagenic chemicals are placed on the targetting oligonucleotide, e.g., nitrous acid, or a suitable protein with such activity. Such chemicals and proteins can be attatched by standard procedures. These include molecules which introduce fundamental chemical changes, that would be useful independent of the particular technical approach. See Lewin, Genes.1983 John Wilely & Sons, Inc. NY pp 42-48.
The following matrix shows that the chemical modifications noted can cause transversion reversions (pyrimidine to pyrimidine, or purine to purine) in RNA or DNA. The transversions (pyrimidine to purine, or purine to pyrimidine) are not preferred because these are more difficult chemical transformations. The footnotes refer to the specific desired chemical transformations. The bold footnotes refer to the reaction on the opposite DNA strand. For example, if one desires to change an A to a G, this can be accomplished at the DNA level by using reaction #5 to change a T to a C in the opposing strand. In this example an A/T base pair goes to A/C , then when the DNA is replicated, or mismatch repair occurs this can become G/C, thus the original A has been converted to a G. ISR matrix
Reverted Base
Mutant base A T(U) C G
Figure imgf000163_0001
1 Deamination of 5-methylcytosine to create thymidine.
2 Deamination of cytosine to create uracil.
3 Deamination of Adenine to be read like G (Inosine).
4 Methylation of cytosine to 5-methylcytosine.
5 Transforming thymidine (or uracil) to O2-methyl thymidine (or O2-methyl uracil), to be read as cytosine (Xu, and Swann, Tetrahedron Letters 35:303-306 (1994)).
6 Transforming guanine to 6-O-methyl (or other alkyls) to be read as adenine (Mehta and Ludlum, Biochimica et Biophysica Acta, 521 :770-778 (1978)).
7. Amination of uracil to cytosine. Bass supra, fig. 6c.
In Vitro Selection Strategy
Referring to Figure 105. there is provided a schematic describing an approach to selecting for a ribozyme with such base changing activity. An
RNA is designed that folds back on itself (this is similar to approaches already used to select for RNA ligases, Bartel, D. and Szostak, J. (1993)
Isolation of new ribozymes from a large pool of random sequences.
Science 261 :1411-1418). A degenerate loop opposing the base to be modified provides for diversity. After incubating this library of molecules in a buffer, the RNA is reverse transcribed into DNA (that is, using standard in vitro evolution protocol. Tuerk and Gold, 249 Science 505, 1990) , and then the DNA is selected for having a base change. A restriction enzyme cleavage and size selection or its equivalent is used to isolate the fraction of DNAs with the appropriate base change. The cycle could then be repeated many times. The in vitro selection (evolution) strategy is similar to approaches developed by Joyce (Beaudry, A. A. and Joyce, G.F. (1992) Science 257, 635-641 ; Joyce, G. F. (1992) Scientific American 267. 90-97) and Szostak (Bartel, D. and Szostak, J. (1993) Science 261 :1411-1418; Szostak, J. W. (1993) TIBS 17, 89-93). Briefly, a random pool of nucleic acids is synthesized wherein, each member contains two domains: a) one domain consists of a region with defined (known) nucleotide sequence; b) the second domain consists of a region with degenerate (random) sequence. The known nucleotide sequence domain enables: 1) the nucleic acid to bind to its target (the region flanking the mutant nucleotide), 2) complimentary DNA (cDNA) synthesis and PCR amplification of molecules selected for their base modifying activity, 3) introduction of restriction endonuclease site for the purpose of cloning. The degenerate domain can be created to be completely random (each of the four nucleotides represented at every position within the random region) or the degeneracy can be partial (Beaudry, A. A. and Joyce, G.F. (1992) Science 257, 635-641). In this invention, the degenerate domain is flanked by regions containing known sequences (see Figure 105), such that the degenerate domain is placed across from the mutant base (the base that is targeted for modification). This random library of nucleic acids is incubated under conditions that ensure folding of the nucleic acids into conformations that facilitate the catalysis of base modification (the reaction protocol may also include certain cofactors like ATP or GTP or an S-adenosyl-methionine (if methylation is desired) in order to make the selection more stringent). Following incubation, nucleic acids are converted into complimentary DNA (if the starting pool of nucleic acids is RNA). Nucleic acids with base modification (at the mutant base position) can be separated from rest of the population of nucleic acids by using a variety of methods. For example, a restriction endonuclease cleavage site can either be created or abolished as a result of base modification. If a restriction endonuclease site is created as a result of base modification, then the library can be digested with the restriction endonuclease (RE). The fraction of the population that is cleaved by the RE is the population that has been able to catalyze the base modification reaction (active pool). A new piece of DNA (containing oligonucleotide primer binding sites for PCR and RE sites for cloning) is ligated to the termini of the active pool to facilitate PCR amplification and subsequent cycles (if necessary) of selection. The final pool of nucleic acids with the best base modifying activity is cloned in to a plasmid vector and transformed into bacterial hosts. Recombinant plasmids can then be isolated from transformed bacteria and the identity of clones can be determined using DNA sequencing techniques.
Base modifying enzymatic nucleic acids (identified via in vitro selection) can be used to cause the chemical modification in vivo.
In addition, the ribozyme could be evolved to specifically bind a protein having an enzymatic base changing acitivity.
Such ribozymes can be used to cause the above chemical modifications in vivo. The ribozymes or above noted antisense-type molecules can be administered by methods discussed in the above referenced art.
VIII. Administration of Nucleic Acids
Applicant has determined that double-stranded nucleic acid lacking a transcription termination signal can be used for continuous expression of the encoded RNA. This is achieved by use of an R-loop, i.e., an RNA molecule non-covalently associated with the double-stranded nucleic acid and which causes localized denaturation ("bubble" formation) within the double stranded nucleic acid (Thomas et al., 1976 Proc. Natl. Acad. Sci. USA 73, 2294). In addition, applicant has determined that that the RNA portion of the R-loop can be used to target the whole R-loop complex to a desirable intracellular or cellular site, and aid in cellular uptake of the complex. Further, applicant indicates that expression of enzymatically active RNA or ribozymes can be significantly enhanced by use of such R-loop complexes. Thus, in one aspect, the invention features a method for introduction of enzymatic nucleic acid into a cell or tissue. A complex of a first nucleic acid encoding the enzymatic nucleic acid and a second nucleic acid molecule is provided. The second nucleic acid molecule has sufficient complementarity with the first nucleic acid to be able to form an R-loop base pair structure under physiological conditions. The R-loop is formed in a region of the first nucleic acid molecule which promotes expression of RNA from the first nucleic acid under physiological conditions. The method further includes contacting the complex with a cell or tissue under conditions in which the enzymatic nucleic acid is produced within the cell or tissue.
By "complex" is simply meant that the two nucleic acid molecules interact by intermolecular bond formation (such as by hydrogen bonding) between two complementary base-paired sequences. The complex will generally be stable under physiological condition such that it is able to cause initiation of transcription from the first nucleic acid molecule.
The first and second nucleic acid molecules may be formed from any desired nucleotide bases, either those naturally occurring (such as adenine, guanine, thymine and cytosine), or other bases well known in the art, or may have modifications at the sugar or phosphate moieties to allow greater stability or greater complex formation to be achieved. In addition, such molecules may contain non-nucleotides in place of nucleotides. Such modifications are well known in the art, see e.g., Eckstein et al., International Publication No. WO 92/07065; Perrault et al., 1990 Nature 344, 565; Pieken et al., 1991 Science. 253, 314; Usman and Cedergren, 1992 Trends in Biochem. Sci. 17, 334; Usman et al., International Publication No. WO 93/15187; and Rossi et al., International Publication No. WO 91/03162, as well as Sproat.B. European Patent Application 92110298.4 which describe various chemical modifications that can be made to the sugar moieties of enzymatic RNA molecules. All these publications are hereby incorporated by reference herein.
By "sufficient complementarity" is meant that sufficient base pairing occurs so that the R-loop base pair structure can be formed under the appropriate conditions to cause transcription of the enzymatic nucleic acid. Those in the art will recognize routine tests by which such sufficient base pairs can be determined. In general, between about 15 - 80 bases is sufficient in this invention.
By "physiological condition" is meant the condition in the cell or tissue to be targeted by the first nucleic acid molecule, although the R-loop complex may be formed under many other conditions. One example is use of a standard physiological saline at 37°C, but it is simply desirable in this invention that the R-loop structure exists to some extent at the site of action so that the expression of the desired nucleic acid will be achieved at that site of action. While it is preferred that the R-loop structure be stable under those conditions, even a minimal amount of formation of the R-loop structure to cause expression will be sufficient. Those in the art will recognize that measurement of such expression is readily achieved, especially in the absence of any promoter or leader sequence on the first nucleic acid molecule (Daube and von Hippel, 1992 Science 258, 1320). Such expression can thus only be achieved if an R-loop structure is truly formed with the second nucleic acid. If a promoter of leader sequence is provided, then it is preferred that the R-loop be formed at a site distant from those regions so that transcription is enhanced. In a related aspect, the invention features a method for introduction of ribonucleic acid within a cell or tissue by forming an R-loop base-paired structure (as described above) with the first nucleic acid molecule lacking any promoter region or transcription termination signal such that once expression is initiated it will continue until the first nucleic acid is degraded. In another related aspect, the invention features a method in which the second nucleic acid is provided with a localization factor, such as a protein, e.g., an antibody, transferin, a nuclear localization peptide, or folate, or other such compounds well known in the art, which will aid in targeting the R-loop complex to a desired cell or tissue. In preferred embodiments, the first nucleic acid is a plasmid, e.g., one without a promoter or a transcription termination signal ; the second nucleic acid is of length between about 40-200 bases and is formed of ribonucleotides at a majority of positions; and the second nucleic is covalently bonded with a ligand such as a nucleic acid, protein, peptide, lipid, carbohydrate, cellular receptor, nuclear localization factor, or is attached to maleimide or a thiol group: the first nucleic acid is an expression plasmid lacking a promoter able to express a desired gene, e.g., it is a double-stranded molecule formed with a majority of deoxyribonucleic acids; the R-loop complex is a RNA/DNA heteroduplex; no promoter or leader region is provided in the first nucleic acid; and the R-loop is adapted to prevent nucleosome assembly and is designed to aid recruitment of cellular transcription machinery.
In other preferred embodiments, the first nucleic acid encodes one or more enzymatic nucleic acids, e.g., it is formed with a plurality of intramolecular and intermolecular cleaving enzymatic nucleic acids to allow release of therapeutic enzymatic nucleic acid in vivo.
In a further related aspect, the invention features a complex of the above first nucleic acid molecules and second nucleic acid molecules. R-loop complex
An R-loop complex is designed to provide a non-integrating plasmid so that, when an RNA polymerase binds to the plasmid, transcription is continuous until the plasmid is degraded. This is achieved by hybridizing an RNA molecule, 40 to 200 nucleotides in length, to a DNA expression plasmid resulting in an R-loop structure (see figure 106). This RNA, when conjugated with a ligand that binds to a cell surface receptor, triggers internalization of the plasmid/RNA-ligand complex. Formation of R-loops in general is described by DeWet, 1987 Methods in Enzymol. 145, 235; Neuwald et al., 1977 J. Virol. 21 ,1019; and Meyer et al., 1986 J. Ult. Mol. Str. Res. 96, 187. Thus, those in the art can readily design complexes of this invention following the teachings of the art.
Promoters placed in retroviral genomes have not always behaved as planned in that the additional promoter will serve as a stop signal or reverses the direction of the polymerase. Applicant was told that creation of an R-loop between the promoter and the reporter gene increased the transfection efficiency. Incubation of an RNA molecule with a double-stranded DNA molecule, containing a region of complementarity with the RNA will result in the formation of a stable RNA-DNA hetroduplex and the DNA strand that has a sequence identical to the RNA will be displaced into a loop-like structure called the R-loop. This displacement of DNA strand occurs because an RNA-DNA duplex is more stable compared to a DNA-DNA duplex. Applicant was also told that an 80 nt long RNA was used to generate a R-loop structure in a plasmid encoding the β-galactosidase gene. The R-loop was initiated either in the promoter region or in the leader sequence. Plasmids containing an R-loop structure were microinjected into the cytoplasm of COS cells and the gene expression was assayed. R-loop formation in the promoter region of the plasmid inhibited expression of the gene. RNA that hybridized to the leader sequence between the promoter and the gene, or directly to the first 80 nucleotides of the mRNA increased the expression levels 8-10 fold. The proposed mechanism is that R-loop formation prevents nucleosome assembly, thus making the DNA more accessible for transcription. Alternatively, the R-loop may resemble a RNA primer promoting either DNA replication or transcription (Daube and von Hippel, 1992, supra). One of the salient features of this invention is to generate R-loops in expression vectors of choice and introduce them into cells to achieve enhanced expression from the expression vector. The presence of an R-loop may aid in the recruitment of cellular transcription machinery. Once an RNA polymerase binds to the plasmid and initiates transcription, the process will continue until a termination signal is reached, or the plasmid is degraded.
This invention will increase the expression of ribozymes inside a cell. The idea is to construct a plasmid with no transcription termination signal, such that a transcript-containing multiple ribozyme units can be generated. In order to liberate unit length ribozymes, self-processing ribozymes can be cloned downstream of each therapeutic ribozyme (see figure 107) as described by Draper supra.
Ligand Targeting
Another salient feature of this invention is that the RNA used to generate R-loop structures can be covalently linked to a ligand (nucleic acid, proteins, peptides, lipids, carbohydrates, etc). Specific ligands can be chosen such that the ligand can bind selectively to a desired cell surface receptor. This ligand-receptor interaction will help internalize a plasmid containing an R-loop. Thus, RNA is used to attach the ligand to the DNA such that localization of the gene to certain regions of the cell is achieved. One of several methods can be used to attach a ligand to RNA. This includes the incorporation of deoxythymidine containing a 6 carbon spacer having a terminal primary amine into the RNA (see figure 108). This amino group can be directly derivatized with the ligand, such as folate (Lee and Low, 1994 J. Biol. Chem. 269, 3198-3204). The RNA containing a 6 carbon spacer with a terminal amine group is mixed with folate and the mixture is reacted with activators like 1 -(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC). This reaction should be carried out in the presence of 1-Hydroxybenzotriazole hydrate (HOBT) to prevent any undesirable side reactions. The RNA can also be derivatized with a heterobifuctional crosslinking agent (or linker) like succinimidyl 4-(p-maleimidophenyl)butyrate (SMPB). The SMPB introduces a maleimide into the RNA. This maleimide can then react with a thiol moiety either in a peptide or in a protein. Thiols can also be introduced into proteins or peptides that lack naturally occurring thiols using succinylacetylthioacetate. The amino linker can be attached at the 5' end or 3' end of the RNA. The RNA can also contain a series of nucleotides that do not hybridize to the DNA and extend the linker away from the RNA/DNA complex, thus increasing the accessibility of the ligand for its receptor and not interfering with the hybridization. These techniques can be used to link peptides such as nuclear localization signal (NLS) peptides (Lanford et al., 1984 Cell 37, 801-813; Kalderon et al., 1984 Cell 39, 499-509; Goldfarb et al., 1986 Nature 322, 641-644)and/or proteins like the transferrin (Curiel et al., 1991 Proc. Natl. Acad. Sci. USA 88, 8850-8854; Wagner et al., 1992 Proc. Natl. Acad. Sci. USA 89, 6099-6103; Giulio et al., 1994 Cell. Signal. 6, 83-90) to the ends of R-loop forming RNA in order to facilitate the uptake and localization of the R-loop-DNA complex. To link a protein to the ends of R-loop forming RNA, an intrinsic thiol can be used to react with the maleimide or the thiols can be introduced into the protein itself using either iminothiolate or succinimidyl acetyl thioacetate (SATA; Duncan et al., 1983 Anal. Biochem 132, 68). The SATA requires an additional deprotection step using 0.5 M hydroxylamine.
In addition liposomes can be used to cause an R-loop complex to be delivered to an appropriate intracellular cite by techniques well known in the art. For example, pH-sensitive liposomes (Connor and Huang, 1986 Cancer Res. 46, 3431-3435) can be used to facilitate DNA transfection.
Calcium phosphate mediated or electroporation-mediated delivery of the R-loop complex in to desired cells can also be readily acomplished. In vitro Selection
In vitro selection strategies can be used to select nucleic acids that a) can form stable R-loops b) selectively bind to specific cell surface receptors. These nucleic acids can then be covalently linked to each other.
This will help internalize the R-loop-containing plasmid efficiently using receptor-mediated endocytosis. The in vitro selection (evolution) strategy is similar to approaches developed by Joyce (Beaudry and Joyce, 1992 Science 257, 635-641 ; Joyce, 1992 Scientific American 267, 90-97) and Szostak (Barrel and Szostak, 1993 Science 261 :1411 -1418; Szostak, 1993 TI BS 17, 89-93). Briefly, a random pool of nucleic acids is synthesized wherein each member contains two domains: a) one domain consists of a region with defined (known) nucleotide sequence; b) the second domain consists of a region with degenerate (random) sequence. The known nucleotide sequence domain enables: 1) the nucleic acid to bind to its target (a specific region of the double strand DNA), 2) complimentary DNA (cDNA) synthesis and PCR amplification of molecules selected for their affinity to form R-loop and/or their ability to bind to a specific receptor, 3) introduction of a restriction endonuclease site for the purpose of cloning. The degenerate domain can be created to be completely random (each of the four nucleotides represented at every position within the random region) or the degeneracy can be partial (Beaudry and Joyce, 1992 Science 257, 635-641). In this invention, the degenerate domain is flanked by regions containing known sequences. This random library of nucleic acids is incubated under conditions that ensure equilibrium binding to either double-stranded DNA or cell surface receptor. Following incubation, nucleic acids are converted into complementary DNA (if the starting pool of nucleic acids is RNA). Nucleic acids with desired characteristics can be separated from the rest of the population of nucleic acids by using a variety of methods (Joyce, 1992 supra). The desired pool of nucleic acids can then be carried through subsequent rounds of selection to enrich the population with the most desired traits. These molecules are then cloned in to appropriate vectors. Recombinant plasmids can then be isolated from transformed bacteria and the identity of clones can be determined using DNA sequencing techniques. Other embodiments are within the following claims. TABLE 1
Characteristics of Ribozymes
Group I Introns
Size: ~200 to >1000 nucleotides.
Requires a U in the target sequence immediately 5' of the cleavage site.
Binds 4-6 nucleotides at 5' side of cleavage site.
Over 75 known members of this class. Found in Tetrahymena thermophila rRNA, fungal mitochondria, chloroplasts, phage T4, blue-green algae, and others.
RNAseP RNA (M1 RNA)
Size: ~290 to 400 nucleotides.
RNA portion of a ribonucleoprotein enzyme. Cleaves tRNA precursors to form mature tRNA.
Roughly 10 known members of this group all are bacterial in origin.
Hammerhead Ribozyme
Size: -13 to 40 nucleotides.
Requires the target sequence UH immediately 5' of the cleavage site.
Binds a variable number nucleotides on both sides of the cleavage site.
14 known members of this class. Found in a number of plant pathogens (virusoids) that use RNA as the infectious agent (Figures 1 and 2)
Hairpin Ribozyme
Size: ~50 nucleotides.
Requires the target sequence GUC immediately 3' of the cleavage site.
Binds 4-6 nucleotides at 5' side of the cleavage site and a variable number to the 3' side of the cleavage site.
Only 3 known member of this class. Found in three plant pathogen
(satellite RNAs of the tobacco ringspot virus, ara bis mosaic virus and chicory yellow mottle virus) which uses RNA as the
infectious agent (Figure 3).
Hepatitis Delta Virus (HDV) Ribozyme
Size: 50 - 60 nucleotides (at present).
Cleavage of target RNAs recently demonstrated.
Sequence requirements not fully determined.
Binding sites and structural requirements not fully determined, although no sequences 5' of cleavage site are required.
Only 1 known member of this class. Found in human HDV (Figure 4).
Neurospora VS RNA Ribozyme
Size: ~144 nucleotides (at present) Cleavage of target RNAs recently demonstrated.
Sequence requirements not fully determined.
Binding sites and structural requirements not fully determined. Only 1 known member of this class. Found in Neurospora VS RNA (Figure 5).
Table 2
Human ICAM HH Target sequence
nt. Position Target Sequences nt. Position Target S Sequence:
11 CCCCAGU C GACGCUG 386 ACCGUGU A CUGGACU
23 CUGAGCU C CUCUGCU 394 CUGGACU C CAGAACG
26 AGCUCCU C UGCUACU 420 CACCCCU C CCCUCUU
31 CUCUGCU A CUCAGAG 425 CUCCCCU C UUGGCAG
34 UGCUACU C AGAGUUG 427 CCCCUCU U GGCAGCC
40 UCAGAGU U GCAACCU 450 AGAACCU U ACCCUAC
48 GCAACCU C AGCCUCG 451 GAACCUU A CCCUACG
54 UCAGCCU C GCUAUGG 456 UUACCCU A CGCUGCC
58 CCUCGCU A UGGCUCC 495 CCAACCU C ACCGUGG
64 UAUGGCU C CCAGCAG 510 UGCUGCU C CGUGGGG
96 CCGCACU C CUGGUCC 564 CUGAGGU C ACGACCA
102 UCCUGGU C CUGCUCG 592 GAGAGAU C ACCAUGG
108 UCCUGCU C GGGGCCC 607 AGCCAAU U UCUCGUG
115 CGGGGCU C UGUUCCC 608 GCCAAUU U CUCGUGC
119 GCUCUGU U CCCAGGA 609 CCAAUUU C UCGUGCC
120 CUCUGUU C CCAGGAC 611 AAUUUCU C GUGCCGC
146 CAGACAU C UGUGUCC 656 GAGCUGU U UGAGAAC
152 UCUGUGU C CCCCUCA 657 AGCUGUU U GAGAACA
158 UCCCCCU C AAAAGUC 668 AACACCU C GGCCCCC
165 CAAAAGU C AUCCUGC 677 GCCCCCU A CCAGCUC
168 AAGUCAU C CUGCCCC 684 ACCAGCU C CAGACCU
185 GGAGGCU C CGUGCUG 692 CAGACCU U UGUCCUG
209 AGCACCU C CUGUGAC 693 AGACCUU U GUCCUGC
227 CCCAAGU U GUUGGGC 696 CCUUUGU C CUGCCAG
230 AAGUUGU U GGGCAUA 709 AGCGACU C CCCCACA
237 UGGGCAU A GAGACCC 720 CACAACU U GUCAGCC
248 ACCCCGU U GCCUAAA 723 AACUUGU C AGCCCCC
253 GUUGCCU A AAAAGGA 735 CCCGGGU C CUAGAGG
263 AAGGAGU U GCUCCUG 738 GGGUCCU A GAGGUGG
267 AGUUGCU C CUGCCUG 765 CCGUGGU C UGUUCCC
293 AAGGUGU A UGAACUG 769 GGUCUGU U CCCUGGA
319 AGAAGAU A GCCAACC 770 GUCUGUU C CCUGGAC
335 AUGUGCU A UUCAAAC 785 GGGCUGU U CCCAGUC
337 GUGCUAU U CAAACUG 786 GGCUGUU C CCAGUCU
338 UGCUAUU C AAACUGC 792 UCCCAGU C UCGGAGG
359 GGGCAGU C AACAGCU 794 CCAGUCU C GGAGGCC
367 AACAGCU A AAACCUU 807 CCCAGGU C CACCUGG
374 AAAACCU U CCUCACC 833 CAGAGGU U GAACCCC
375 AAACCUU C CUCACCG 846 CCACAGU C ACCUAUG
378 CCUUCCU C ACCGUGU 851 GUCACCU A UGGCAAC 863 AACGACU C CUUCUCG 1408 UCGAGAU C UUGAGGG
866 GACUCCU U CUCGGCC 1410 GAGAUCU U GAGGGCA
867 ACUCCUU C UCGGCCA 1421 GGCACCU A CCUCUGU
869 UCCUUCU C GGCCAAG 1425 CCUACCU C UGUCGGG
881 AAGGCCU C AGUCAGU 1429 CCUCUGU C GGGCCAG
885 CCUCAGU C AGUGUGA 1444 GAGCACU C AAGGGGA
933 GUGCAGU A AUACUGG 1455 GGGAGGU C ACCCGCG
936 CAGUAAU A CUGGGGA 1482 AUGUGCU C UCCCCCC
978 UGACCAU C UACAGCU 1484 GUGCUCU C CCCCCGG
980 ACCAUCU A CAGCUUU 1493 CCCCGGU A UGAGAUU
986 UACAGCU U UCCGGCG 1500 AUGAGAU U GUCAUCA
987 ACAGCUU U CCGGCGC 1503 AGAUUGU C AUCAUCA
988 CAGCUUU C CGGCGCC 1506 UUGUCAU C AUCACUG
1005 ACGUGAU U CUGACGA 1509 UCAUCAU C ACUGUGG
1006 CGUGAUU C UGACGAA 1518 CUGUGGU A GCAGCCG
1023 CAGAGGU C UCAGAAG 1530 CCGCAGU C AUAAUGG
1025 GAGGUCU C AGAAGGG 1533 CAGUCAU A AUGGGCA
1066 CCACCCU A GAGCCAA 1551 CAGGCCU C AGCACGU
1092 AUGGGGU U CCAGCCC 1559 AGCACGU A CCUCUAU
1093 UGGGGUU C CAGCCCA 1563 CGUACCU C UAUAACC
1125 CCCAGCU C CUGCUGA 1565 UACCUCU A UAACCGC
1163 CGCAGCU U CUCCUGC 1567 CCUCUAU A ACCGCCA
1164 GCAGCUU C UCCUGCU 1584 GGAAGAU C AAGAAAU
1166 AGCUUCU C CUGCUCU 1592 AAGAAAU A CAGACUA
1172 UCCUGGU C UGCAACC 1599 ACAGACU A CAACAGG
1200 GCCAGCU U AUACACA 1651 CACGCCU C CCUGAAC
1201 CCAGCUU A UACACAA 1661 UGAACCU A UCCCGGG
1203 AGCUUAU A CACAAGA 1663 AACCUAU C CCGGGAC
1227 GGGAGCU U CGUGUCC 1678 AGGGCCU C UUCCUCG
1228 GGAGCUU C GUGUCCU 1680 GGCCUCU U CCUCGGC
1233 UUCGUGU C CUGUAUG 1681 GCCUCUU C CUCGGCC
1238 GUCCUGU A UGGCCCC 1684 UCUUCCU C GGCCUUC
1264 GAGGGAU U GUCCGGG 1690 UCGGCCU U CCCAUAU
1267 GGAUUGU C CGGGAAA 1691 CGGCCUU C CCAUAUU
1294 AGAAAAU U CCCAGCA 1696 UUCCCAU A UUGGUGG
1295 GAAAAUU C CCAGCAG 1698 CCCAUAU U GGUGGCA
1306 GCAGACU C CAAUGUG 1737 AAGACAU A UGCCAUG
1321 CCAGGCU U GGGGGAA 1750 UGCAGCU A CACCUAC
1334 AACCCAU U GCCCGAG 1756 UACACCU A CCGGCCC
1344 CCGAGCU C AAGUGUC 1787 AGGGCAU U GUCCUCA
1351 CAAGUGU C UAAAGGA 1790 GCAUUGU C CUCAGUC
1353 AGUGUCU A AAGGAUG 1793 UUGUCCU C AGUCAGA
1366 UGGCACU U UCCCACU 1797 CCUCAGU C AGAUACA
1367 GGCACUU U CCCACUG 1802 GUCAGAU A CAACAGC
1368 GCACUUU C CCACUGC 1812 ACAGCAU U UGGGGCC
1380 UGCCCAU C GGGGAAU 1813 CAGCAUU U GGGGCCA
1388 GGGGAAU C AGUGACU 1825 CCAUGGU A CCUGCAC
1398 UGACUGU C ACUCGAG 1837 CACACCU A AAACACU
1402 UGUCACU C GAGAUCU 1845 AAACACU A GGCCACG 1856 CACGCAU C UGAUCUG 2189 UAUUUAU U GAGUGUC
1861 AUCUGAU C UGUAGUC 2196 UGAGUGU C UUUUAUG
1865 GAUCUGU A GUCACAU 2198 AGUGUCU U UUAUGDA
1868 CUGUAGU C ACAUGAC 2199 GUGUCUU U UAUGUAG
1877 CAUGACU A AGCCAAG 2200 UGUCUUU U AUGUAGG
1901 CAAGACU C AAGACAU 2201 GUCUUUU A UGUAGGC
1912 ACAUGAU U GAUGGAU 2205 UUUAUGU A GGCUAAA
1922 UGGAUGU U AAAGUCU 2210 GUAGGCU A AAUGAAC
1923 GGAUGUU A AAGUCUA 2220 UGAACAU A GGCCUCU
1928 UUAAAGU C UAGCCUG 2224 CAUAGGU C UCUGGCC
1930 AAAGUCU A GCCUGAU 2226 UAGGUCU C UGGCCUC
1964 GAGACAU A GCCCCAC 2233 CUGGCCU C ACGGAGC
1983 AGGACAU A CAACUGG 2242 CGGAGCU C CCAGUCC
1996 GGGAAAU A CUGAAAC 2248 UCCCAGU C CAUGUCA
2005 UGAAACU U GCUGCCU 2254 UCCAUGU C ACAUUCA
2013 GCUGCCU A UUGGGUA 2259 GUCACAU U CAAGGUC
2015 UGCCUAU U GGGUAUG 2260 UCACAUU C AAGGUCA
2020 AUUGGGU A UGCUGAG 2266 UCAAGGU C ACCAGGU
2039 ACAGACU U ACAGAAG 2274 ACCAGGU A CAGUUGU
2040 CAGACUU A CAGAAGA 2279 GUACAGU U GUACAGG
2057 UGGCCCU C CAUAGAC 2282 CAGUUGU A CAGGUUG
2061 CCUCCAU A GACAUGU 2288 UACAGGU U GUACACU
2071 CAUGUGU A GCAUCAA 2291 AGGUUGU A CACUGCA
2076 GUAGCAU C AAAACAC 2321 AAAAGAU C AAAUGGG
2097 CCACACU U CCUGACG 2338 UGGGACU U CUCAUUG
2098 CACACUU C CUGACGG 2339 GGGACUU C UCAUUGG
2115 GCCAGCU U GGGCACU 2341 GACUUCU C AUUGGCC
2128 CUGCUGU C UACUGAC 2344 UUCUCAU U GGCCAAC
2130 GCUGUCU A CUGACCC 2358 CCUGCCU U UCCCCAG
2145 CAACCCU U GAUGAUA 2359 CUGCCUU U CCCCAGA
2152 UGAUGAU A UGUAUUU 2360 UGCCUUU C CCCAGAA
2156 GAUAUGU A UUUAUUC 2376 GAGUGAU U UUUCUAU
2158 UAUGUAU U UAUUCAU 2377 AGUGAUU U UUCUAUC
2159 AUGUAUU U AUUCAUU 2378 GUGAUUU U UCUAUCG
2160 UGUAUUU A UUCAUUU 2379 UGAUUUU U CUAUCGG
2162 UAUUUAU U CAUUUGU 2380 GAUUUUU C UAUCGGC
2163 AUUUAUU C AUUUGUU 2382 UUUUUCU A UCGGCAC
2166 UAUUCAU U UGUUAUU 2384 UUUCUAU C GGCACAA
2167 AUUCAUU U GUUAUUU 2399 AAGCACU A UAUGGAC
2170 CAUUUGU U AUUUUAC 2401 GCACUAU A UGGACUG
2171 AUUUGUU A UUUUACC 2411 GACUGGU A AUGGUUC
2173 UUGUUAU U UUACCAG 2417 UAAUGGU U CACAGGU
2174 UGUUAUU U UACCAGC 2418 AAUGGUU C ACAGGUU
2175 GUUAUUU U ACCAGCU 2425 CACAGGU U CAGAGAU
2176 UUAUUUU A CCAGCUA 2426 ACAGGUU C AGAGAUU
2183 ACCAGCU A UUUAUUG 2433 CAGAGAU U ACCCAGU
2185 CAGCUAU U UAUUGAG 2434 AGAGAUU A CCCAGUG
2186 AGCUAUU U AUUGAGU 2448 GAGGCCU U AUUCCUC
2187 GCUAUUU A UUGAGUG 2449 AGGCCUU A UUCCUCC 2451 GCCUUAU U CCUCCCU 2750 UAUGUGU A GACAAGC
2452 CCUUAUU C CUCCCUU 2759 ACAAGCU C UCGCUCU
2455 UAUUCCU C CCUUCCC 2761 AAGCUCU C GCUCUGU
2459 CCUCCCU U CCCCCCA 2765 UCUCGCU C UGUCACC
2460 CUCCCUU C CCCCCAA 2769 GCUCUGU C ACCCAGG
2479 GACACCU U UGUUAGC 2797 GUGCAAU C AUGGUUC
2480 ACACCUU U GUUAGCC 2803 UCAUGGU U CACUGCA
2483 CCUUUGU U AGCCACC 2804 CAUGGUU C ACUGCAG
2484 CUUUGUU A GCCACCU 2813 CUGCAGU C UUGACCU
2492 GCCACCU C CCCACCC 2815 GCAGUCU U GACCUUU
2504 CCCACAU A CAUUUCU 2821 UUGACCU U UUGGGCU
2508 CAUACAU U UCUGCCA 2822 UGACCUU U UGGGCUC
2509 AUACAUU U CUGCCAG 2823 GACCUUU U GGGCUCA
2510 UACAUUU C UGCCAGU 2829 UUGGGCU C AAGUGAU
2520 CCAGUGU U CACAAUG 2837 AAGUGAU C CUCCCAC
2521 CAGUGUU C ACAAUGA 2840 UGAUCCU C CCACCUC
2533 UGACACU C AGCGGUC 2847 CCCACCU C AGCCUCC
2540 CAGCGGU C AUGUCUG 2853 UCAGCCU C CUGAGUA
2545 GUCAUGU C UGGACAU 2860 CCUGAGU A GCUGGGA
2568 AGGGAAU A UGCCCAA 2872 GGACCAU A GGCUCAC
2579 CCAAGCU A UGCCUUG 2877 AUAGGCU C ACAACAC
2585 UAUGCCU U GUCCUCU 2899 GGCAAAU U CGAUUUU
2588 GCCUUGU C CUCUUGU 2900 GCAAAUU U GAUUUUU
2591 UUGUCCU C UUGUCCU 2904 AUUUGAU U UUUUUUU
2593 GUCCUCU U GUCCUGU 2905 UUUGAUU U UUUUUUU
2596 CUCUUGU C CUGUUUG 2906 UUGAUUU U UUUUUUU
2601 GUCCUGU U UGCAUUU 2907 UGAUUUU U UUUUUUU
2602 UCCUGUU U GCAUUUC 2908 GAUUUUU U UUUUUUU
2607 UUUGCAU U UCACUGG 2909 AUUUUUU U UUUUUUU
2608 UUGCAUU U CACUGGG 2910 UUUUUUU U UUUUUUU
2609 UGCAUUU C ACUGGGA 2911 UUUUUUU U UUUUUUU
2620 GGGAGCU U GCACUAU 2912 UUUUUUU U UUUUUUC
2626 UUGCACU A UUGCAGC 2913 UUUUUUU U UUUUUCA
2628 GCACUAU U GCAGCUC 2914 UUUUUUU U UUUUCAG
2635 UGCAGCU C CAGUUUC 2915 UUUUUUU U UUUCAGA
2640 CUCCAGU U UCCUGCA 2916 UUUUUUU U UUCAGAG
2641 UCCAGUU U CCUGCAG 2917 UUUUUUU U UCAGAGA
2642 CCAGUUU C CUGCAGU 2918 UUUUUUU U CAGAGAC
2653 CAGUGAU C AGGGUCC 2919 UUUUUUU C AGAGACG
2659 UCAGGGU C CUGCAAG 2931 ACGGGGU C UCGCAAC
2689 CCAAGGU A UUGGAGG 2933 GGGGUCU C GCAACAU
2691 AAGGUAU U GGAGGAC 2941 GCAACAU U GCCCAGA
2700 GAGGACU C CCUCCCA 2951 CCAGACU U CCUUUGU
2704 ACUCCCU C CCAGCUU 2952 CAGACUU C CUUUGUG
2711 CCCAGCU U UGGAAGG 2955 ACUUCCU U UGUGUUA
2712 CCAGCUU U GGAAGGG 2956 CUUCCUU U GUGUUAG
2721 GAAGGGU C AUCCGCG 2961 UUUGUGU U AGUUAAU
2724 GGGUCAU C CGCGUGU 2962 UUGUGUU A GUUAAUA
2744 UGUGUGU A UGUGUAG 2965 UGUUAGU U AAUAAAG 2966 GUUAGUU A AUAAAGC
2969 AGUUAAU A AAGCUUU
2975 UAAAGCU U UCUCAAC
2976 AAAGCUU U CUCAACU
2977 AAGCUUU C UCAACUG
2979 GCUUUCU C AACUGCC
Table 3
Mouse ICAM HH Target Sequence
nt. Position Target Sequence nt. Position Target Sequence 11 CCCugGU C acCGuUG 367 AAugGCU u cAACCcg 23 CaGuGgU u CUCUGCU 374 gAAgCCU U CCUgcCC 26 uGgUuCU C UGCUcCU 375 AAgCCUU C CUgcCCc 31 CUCUGCU c CUCcaca 378 CuaaCaU C ACCGUGU 34 UuCUcaU a AGgGUcG 386 ACCGUGU A uUcGuuU 40 gCAcAcU U GuAgCCU 394 CcGGACU u ucGAuCu 48 aggACCU C AGCCUgG 420 CACaCuU C CCCcCcg 54 UggGCCU C GugAUGG 425 CaCCCCU C ccaGCAG 58 CaUgcCU u UaGCUCC 427 CagCUCU c aGCAGug 64 cAcccCU C CCAGCAG 450 AGgACCU c ACCCUgC 96 CucugCU C CUGGcCC 451 GAAaCcU u uCCUuuG 102 UgCcaGU a CUGCUgG 456 UUACCCU c aGCcaCu 108 cuCUGCU C cuGGCcC 495 CuAcCaU C ACCGUGu 115 uGGuuCU C UGcUCCu 510 UGCUGCU C CGUGGGG 119 GgaaUGU c aCCAGGA 564 CUcAGGU a uCcAuCc 120 CUCUGcU C CugGccC 592 GAaAGAU C ACaugGG 146 CAGuCgU C cGcuUCC 607 AGCCAAU U UCUCaUG 152 UCUGUGU C agCCaCu 608 GCCAAUU U CUCaUGC 158 UCCuguU u AAAAacC 609 CCAAUUU C UCaUGCC 165 CAgAAGU u gUuuUGC 611 AAUUUCU C aUGCCGC 168 AAGcCuU C CUGCCCC 656 aAGCUGU U UGAGcug 185 GGuGGgU C CGUGCaG 657 AGCUGUU U GAGcugA 209 gcCACuU C CUcUGgC 668 cgagCCU a GGCCaCC 227 CagAAGU U GUUuuGC 677 GaCCuCU A CCAGCcu 230 AAGUUGU U uuGCucc 684 uuCAGCU C CgGuCCU 237 UGuGCuU u GAGAaCu 692 CgGACuU U cGauCUu 248 AaCCCaU c uCCUAAA 693 AGgaCcU c acCCUGC 253 ccUGCCU A AggAaGA 696 CCUgUuU C CUGCCuc 263 AgGGuuU c uCUaCUG 709 gGCGgCU C CaCCuCA 267 AGggGCU C CUGCCUa 720 uACAACU U uUCAGCu 293 AAGcUGU u UGAgCUG 723 AACUUuU C AGCuCCg 319 AGgAGAU A cugAgCC 735 aCCaGaU C CUgGAGa 335 cUGUGCU u UgagAAC 738 uGGgCCU c GuGaUGG 337 GUcCaAU U CAcACUG 765 CaGUcGU C cGcUuCC 338 aGCUgUU u gAgCUGa 769 GGcCUGU U uCCUGcc 359 GuGCAGU C guCcGCU 770 uUuUGcU C CCUGGAa 785 GGcCUGU U uCCuGcC 1353 AGUGggU c gAaGgUG 786 GcCUGUU u CCuGcCU 1366 UaaCAgU c UaCaACU 792 UggagGU C UCGGAaG 1367 aGCACcU c CCCACcu 794 CugGgCU u GGAGaCu 1368 GuACUgU a CCACUcu 807 CuCgGaU a uACCUGG 1380 UGCCCAU C GGGGugg 833 CAaAGcU c GAca.CCC 1388 GGaGAcU C AGUGgCU 846 CCcugGU C ACCguUG 1398 UGgCUGU C ACagaAc 851 GagACCU c UacCAgC 1402 UGUgcuU u GAGAaCU 863 AgCcACU u CcUCUgG 1408 gCGAGAU C ggGgaGG
866 GAagCCU U CcuGcCC 1410 GAGgUCU c GgaaGgg
867 AuUCgUU u cCGGagA 1421 ccCACCU A CuUuUGU
869 UCuUcCU C augCAAG 1425 aCUgCCU u gGUaGaG
881 AuGGCuU C AacCcGU 1429 uCUCUaU u GccCCuG
885 CCUugGU a gagGUGA 1444 GAaggCU C AgGaGGA
933 cUauAaU c AUuCUGG 1455 GGaAuGU C ACCaGga
936 uAaUcAU u CUGGuGc 1482 AguUGuU u UgCuCCC
978 UaACagU C UACAaCU 1484 cUGuUCU u CCuCauG
980 ACagUCU A CAaCUUU 1493 CuguGcU u UGAGAac
986 UACAaCU U UuCaGCu 1500 AUGAaAU c aUggUCc
987 ACAaCUU U uCaGCuC 1503 gGAcUaU a AUCAUuc
988 CAaCUUU u CaGCuCC 1506 UUaUguU u AUaACcG
1005 ACcaGAU c CUGgaGA 1509 cuAcCAU C ACcGUGu
1006 uGaGAgU C UGggGAA 1518 ucaUGGU c cCAGgCG
1023 ugGAGGU C UCgGAAG 1530 CuauAaU C AUucUGG
1025 GAGGUCU C gGAAGGG 1533 ugGUCAU u gUGGGCc
1066 CCACuCU c aAaauAA 1551 CAuGCCU u AGCAgcU
1092 AcuGGaU c uCAGgCC 1559 AGCACcU c CCcaccU
1093 UGGaccU u CAGCCaA 1563 CuUAugU u UAUAACC
1125 CCCAaCU C uUcuUGA 1555 UAugUuU A UAACCGC
1163 CGaAGCU U CUuuUGC 1567 ugUuUAU A ACCGCCA
1164 GaAGCUU C UuuUGCU 1584 GaAAGAU C AgGAuAU
1156 AGCUUCU u uUGCUCU 1592 AgGAuAU A CAaguUA
1172 UCCUGuU u aaaAACC 1599 ACAaguU A CAgaAGG
1200 cuCuGCU c cUcCACA 1651 CcCaCCU C CCUGAgC
1201 gCuGCUU u UgaACAg 1661 gaAACCU u UCCuuuG
1203 AcuUUuU u CACcAGu 1663 AACCUuU C CuuuGAa
1227 GGuAcaU a CGUGUgC 1678 AGGaCCU C agCCUgG
1228 GaAGCUU C uUuUgCU 1680 aGCCaCU U CCUCuGg
1233 UUCGUuU C CgGagaG 1681 GCCaCUU C CUCuGgC
1238 GUgCUGU A UGGuCCu 1684 aCUUCCU C uGgCUgu
1264 GAaGGgU c GUgCaaG 1690 cCGGaCU U uCgAUcU
1267 uGAgaGU C uGGGgAA 1691 CGGaCUU u CgAUcUU
1294 AGgAgAU a CugAGCc 1696 UgCCCAU c ggGGUGG
1295 GAggggU C uCAGCAG 1698 CggAUAU a ccUGGag
1306 GCAGACU C ugAaaUG 1737 gAGACcU c UaCCAgc
1321 gaAGGCU c aGGaGgA 1750 gGCgGCU c CACCUca
1334 AACCCAU c uCCuaAa 1756 gAagCCU u CCuGCCC
1344 auGAGCU C gAGaGUg 1787 gaGaCAU U GUCCcCA
1351 ugAaUGU a UAAguuA 1790 GCAUUGU u CUCuaau
1793 UgGUCCU C gGcugGA 2173 UUagagU U UUACCAG
1797 CacCAGU C AcAUAaA 2174 UagagUU U UACCAGC
1802 acCAGAU c CuggAGa 2175 agagUUU U ACCAGCU
1812 ACuGgAU c UcaGGCC 2176 gagUUUU A CCAGCUA
1813 CAGCAUU U acccuCA 2183 ACCAGCU A UUUAUUG
1825 CCAcGcU A CCUcugC 2185 CAGCUAU U UAUUGAG 1837 CAugCCU u uAgCuCc 2186 AGCUAUU U AUUGAGU
1845 cgAgcCU A GGCCACc 2187 GCUAUUU A UUGAGUa 1856 CggaCuU u cGAUCUu 2189 UAUUUAU U GAGUacC
1861 AcaUGAU a UccAGUa 2196 caAcUcU u cUUgAUG
1865 cAcuUGU A GcCuCAg 2198 gcaGcCU c UUAUGUu
1863 CaccAGU C ACAUaAa 2199 GccUCUU a UgUuUAu
1877 CAUGcCU u AGCagcu 2200 UcUuccU c AUGcAaG
1901 uAAaACU C AAGggAc 2201 aagUUUU A UGUcGGC
1912 AuAUagU a GAUcagU 2205 UUUAUGU c GGCcugA
1922 UGaAUGU a uAAGUua 2210 GgAGaCU c AgUGgcu
1923 uGAUGcU c AgGUaUc 2220 cuggCAU u GuUCUCU
1928 UUAgAGU u UuaCCaG 2224 CucAGGU a UCcauCC
1930 AgAGUuU u aCCaGcU 2226 UgGaUCU C aGGCCgC
1964 GAGACAU u GuCCCca 2233 CUGaCCU C cuGGAGg
1983 AGGAuAU A CAAgUua 2242 uGGAGCU a gCgGaCC
1996 aGGAgAU A CUGAgcC 2248 UauCcaU CAUccCA
2005 UGgAgCU a GCgGaCc 2254 UCCAauU C ACAcUgA
2013 GCUauuU A UUGaGUA 2259 aUCACAU u CAcGGUg
2015 UGCCcAU c GGGgugG 2260 UCACAUU C AcGGUgc
2020 ggUGGuU c UuCUGAG 2266 ggAAuGU C ACCAGGa
2039 gCuGgCU a gCAGAgG 2274 ACCAGaU c CuGgaGa
2040 CuGACcU c CuGgAGg 2279 GaAggGU c GUgCAaG
2057 UGcuCCU C CAcAucC 2282 aAGcUGU u ugaGcUG
2061 CuaCCAU c acCgUGU 2288 UAuAaGU U aUggcCU
2071 CAcuUGU A GCcUCAg 2291 caGUgGU u CuCUGCu
2076 GUAGCcU C AgAgCua 2321 gAAAGAU C AcAUGGG
2097 CaACuCU u CuUGAuG 2338 UGaGACU c CUgccUG
2098 CACACUU c CcccCcG 2339 GaaACcU u UCcUUuG
2115 GCCAGCU c GGaggaU 2341 GACcUCU a ccaGcCu
2128 CaGCUaU u UAuUGAg 2344 UUucgAU c uuCCAgC
2130 cCUGUuU c CUGcCuC 2358 CCcagCU c UCagCAG
2145 CAACuCU U cuUGAUg 2359 CUGCuUU U gaaCAGA
2152 UauUaAU u UagAgUU 2360 aaCCUUU C CuuuGAA
2156 uugAUGU A UUUAUUa 2376 agGUGgU U cUUCUga
2158 gAUGUAU U UAUUaAU 2377 gGUGgUU c UUCUgag
2159 AUGUAUU U AUUaAUU 2378 agGgUUU c UCUAcuG
2160 UGUAUUU A UUaAUUU 2379 UGcUUUU c ucAUaaG
2162 UAUUUAU U aAUUUag 2380 aAgUUUU a UgUCGGC
2163 AUgUAUU u AUUaaUU 2382 aUUcUCU A UuGcCcC
2166 acUUCAU U cucUAUU 2384 aUcCagU a GaCACAA
2167 AUguAUU U aUUAaUU 2399 AAaCACU A UgUGGAC
2170 uAUUUaU u AaUUUAg 2401 aagCUgU u UGagCUG
2171 AgUUGUU u UgcUcCC 2411 uACUGGU c AgGaUgC
2417 gAAUGGU a CAuAcGU 2691 AAuGUcU c cGAGGcC
2418 AcUGGaU C uCAGGcc 2700 GAaGcCU u CCUgCCc
2425 CAugGGU c gAGgGuU 2704 gacCuCU a CCAGCcU
2426 AuuaaUU u AGAGuUU 2711 CCCAGCU c UcagcaG
2433 uAGAGuU U uaCCAGc 2712 gagGucU c GGAAGGG
2434 AGAGuUU u aCCAGcu 2721 GAAGGGU C gUgCaaG
2448 GAaGCCU U ccUgCcC 2724 GGuaCAU a CGuGUGc
2449 AaGCCUU c cUgCcCC 2744 gGUGgGU c cGUGcAG 2451 GCCUguU U CCUgCCU 2750 UAUuUaU u GAguAcC
2452 CCUguUU C CUgCCUc 2759 cCggaCU u UCGaUCU
2455 gAagCCU u CCUgCCC 2761 AgGacCU C aCcCUGc
2459 CCaCaCU U CCCCCCc 2765 UuUuGCU C UGcCgCu
2460 CaCaCUU C CCCCCcg 2769 agUCUGU C AaaCAGG
2479 GAgACCU c UaccAGC 2797 aUGaAAU c AUGGUcC
2480 uCACCgU U GUgAuCC 2803 UCAUGGU c CcagGCg
2483 CCaaUGU c AGCCACC 2804 ggUGGgU c cgUGCAG
2484 CUUUuUU c aCCAguc 2813 CUcCgGU c cUGACCc
2492 agCACCU C CCCACCu 2815 aCAGUCU a cAaCUUU
2504 CCCACcU A CuUUUgU 2821 cUGACCU c cUGGagg
2508 uAUcCAU c caUcCCA 2822 gGAgCcU c cGGaCUu
2509 uUAgAgU U uUaCCAG 2823 ugCCUUU a GcuCcCA
2510 UAgAgUU u UaCCAGc 2829 cUGGaCU a uAaUcAU
2520 CuuuUGU U CcCAAUG 2837 AgGUGgU u CUuCuga
2521 CAGcaUU u ACccUcA 2840 UGAgaCU C CugCCUg
2533 UGAugCU C AGguaUC 2847 CCaAugU C AGCCaCC
2540 CAGCaGU C cgcUgUG 2853 gCAGCCU C uUauGUu
2545 GUgcUGU a UGGuCcU 2860 gCcaAGU A aCUGuGA
2568 guGaAgU c UGuCaAA 2872 GGACCuU c aGCcaAg
2579 auAAGuU A UGgCcUG 2877 uUccGCU a cCAuCAC
2585 cugGCaU U GUuCUCU 2899 cGgAcuU U cGAUcUU
2588 GCaUUGU u CUCUaaU 2900 uuAAuUU a GAgUUUU
2591 UgGUuCU C UgcUCCU 2904 AcUUcAU U cUcUaUU
2593 cUuCUuU U GcuCUGc 2905 cUUcAUU c UcUaUUg
2596 CUuUUGU u CccaaUG 2906 UUGAUgU a UUUaUUa
2601 acCgUGU a UuCgUUU 2907 UGuaUUU a UUaaUUU
2602 UCCaGcU a cCAUccC 2908 GAagcUU c UUUUgcU
2607 cUcGgAU a UacCUGG 2909 AgcUUcU U UUgcUcU
2608 caGCAgU c CgCUGuG 2910 UgUaUUU a UUaaUUU
2609 gGaAUgU C ACcaGGA 2911 UgUaUUU a UUaaUUU
2620 aGGAcCU c aCcCUgc 2912 UUgUUcU c UaaUgUC
2626 UUuCgaU c UUcCAGC 2913 UUUcUcU a cUggUCA
2628 GCACacU U GuAGCcu 2914 UgcUUUU c UcaUaAG
2635 UuCAGCU C CgGUccu 2915 aUUUaUU a aUUuAGA
2640 ggCCuGU U UCCUGCc 2916 UaUUcgU U UcCgGAG
2641 cCCAGcU c uCaGCAG 2917 aUUcgUU U cCgGAGA
2642 CCuGUUU C CUGCcuc 2918 UUcgUUU c CgGAGAg
2653 uAcUGgU C AGGaUgC 2919 UUcUcaU a AGgGuCG
2659 gaAGGGU C gUGCAAG 2931 ugGaGGU C UCGgAAg
2689 CuAAuGU c UccGAGG 2933 GaGGUCU C GgAAggg
2941 GagACAU U GuCCccA
2951 CCAcgCU a CCUcUGc
2952 CAGcagU C CgcUGUG
2955 AgUgaCU c UGUGUcA
2956 uUUCCUU U GaaUcAa
2961 UcUGUGU c AGccAcU
2962 aUGUaUU u aUUAAUu
2965 UuUgAaU c AAUAAAG 2966 GcUgGcU A gcAgAGg
2969 AaUcAAU A AAGuUUU
2975 UAgAGuU U UacCAgC
2976 gAgGgUU U CUCuACU
2977 AAGCUgU u UgAgCUG
2979 uCaUUCU C uAuUGCC
Table 4
Human ICAM HH Ribozyme Sequences
nt. Position Ribozyme Sequence
11 CAGCGUC CUGAUGAGGCCGAAAGGCCGAA ACUGGGG
23 AGCAGAG CUGAUGAGGCCGAAAGGCCGAA AGCUCAG
26 AGUAGCA CUGAUGAGGCCGAAAGGCCGAA AGGAGCU
31 CUCUGAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
34 CAACUCU CUGAUGAGGCCGAAAGGCCGAA AGUAGCA
40 AGGUUGC CUGAUGAGGCCGAAAGGCCGAA ACUCUGA
48 CGAGGCU CUGAUGAGGCCGAAAGGCCGAA AGGUUGC
54 CCAUAGC CUGAUGAGGCCGAAAGGCCGAA AGGCUGA
58 GGAGCCA CUGAUGAGGCCGAAAGGCCGAA AGCGAGG
54 CUGCUGG CUGAUGAGGCCGAAAGGCCGAA AGCCAUA
96 GGACCAG CUGAUGAGGCCGAAAGGCCGAA AGUGCGG
102 CGAGCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGA
108 GAGCCCC CUGAUGAGGCCGAAAGGCCGAA AGCAGGA
115 GGGAACA CUGAUGAGGCCGAAAGGCCGAA AGCCCCG
119 UCCUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGAGC
120 GUCCUGG CUGAUGAGGCCGAAAGGCCGAA AACAGAG 146 GGACACA CUGAUGAGGCCGAAAGGCCGAA AUGUCUG 152 UGAGGGG CUGAUGAGGCCGAAAGGCCGAA ACACAGA 158 GACUUUU CUGAUGAGGCCGAAAGGCCGAA AGGGGGA 165 GCAGGAU CUGAUGAGGCCGAAAGGCCGAA ACUUUUG 158 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AUGACUU 185 CAGCACG CUGAUGAGGCCGAAAGGCCGAA AGCCUCC 209 GUCACAG CUGAUGAGGCCGAAAGGCCGAA AGGUGCU 227 GCCCAAC CUGAUGAGGCCGAAAGGCCGAA ACUUGGG 230 UAUGCCC CUGAUGAGGCCGAAAGGCCGAA ACAACUU 237 GGGUCUC CUGAUGAGGCCGAAAGGCCGAA AUGCCCA 248 UUUAGGC CUGAUGAGGCCGAAAGGCCGAA ACGGGGU 253 UCCUUUU CUGAUGAGGCCGAAAGGCCGAA AGGCAAC 263 CAGGAGC CUGAUGAGGCCGAAAGGCCGAA ACUCCUU 267 CAGGCAG CUGAUGAGGCCGAAAGGCCGAA AGCAACU 293 CAGUUCA CUGAUGAGGCCGAAAGGCCGAA ACACCUU 319 GGUUGGC CUGAUGAGGCCGAAAGGCCGAA AUCUUCU 335 GUUUGAA CUGAUGAGGCCGAAAGGCCGAA AGCACAU
337 CAGUUUG CUGAUGAGGCCGAAAGGCCGAA AUAGCAC
338 GCAGUUU CUGAUGAGGCCGAAAGGCCGAA AAUAGCA 359 AGCUGUU CUGAUGAGGCCGAAAGGCCGAA ACUGCCC 367 AAGGUUU CUGAUGAGGCCGAAAGGCCGAA AGCUGUU
374 GGUGAGG CUGAUGAGGCCGAAAGGCCGAA AGGUUUU
375 CGGUGAG CUGAUGAGGCCGAAAGGCCGAA AAGGUUU 378 ACACGGU CUGAUGAGGCCGAAAGGCCGAA AGGAAGG 386 AGUCCAG CUGAUGAGGCCGAAAGGCCGAA ACACGGU 394 CGUUCUG CUGAUGAGGCCGAAAGGCCGAA AGUCCAG 420 AAGAGGG CUGAUGAGGCCGAAAGGCCCAA AGGGGUG 425 CUGCCAA CUGAUGAGGCCGAAAGGCCGAA AGGGGAG 427 GGCUGCC CUGAUGAGGCCGAAAGGCCGAA AGAGGGG
450 GUAGGGU CUGAUGAGGCCGAAAGGCCGAA AGGUUCU
451 CGUAGGG CUGAUGAGGCCGAAAGGCCGAA AAGGUUC 456 GGCAGCG CUGAUGAGGCCGAAAGGCCGAA AGGGUAA 495 CCACGGU CUGAUGAGGCCGAAAGGCCGAA AGGUUGG 510 CCCCACG CUGAUGAGGCCGAAAGGCCGAA AGCAGCA 564 UGGUCGU CUGAUGAGGCCGAAAGGCCGAA ACCUCAG 592 CCAUGGU CUGAUGAGGCCGAAAGGCCGAA AUCUCUC
607 CACGAGA CUGAUGAGGCCGAAAGGCCGAA AUUGGCU
608 GCACGAG CUGAUGAGGCCGAAAGGCCGAA AAUUGGC
609 GGCACGA CUGAUGAGGCCGAAAGGCCGAA AAAUUGG 611 GCGGCAC CUGAUGAGGCCGAAAGGCCGAA AGAAAUU
656 GUUCUCA CUGAUGAGGCCGAAAGGCCGAA ACAGCUC
657 UGUUCUC CUGAUGAGGCCGAAAGGCCGAA AACAGCU 668 GGGGGCC CUGAUGAGGCCGAAAGGCCGAA AGGUGUU 677 GAGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGGC 684 AGGUCUG CUGAUGAGGCCGAAAGGCCGAA AGCUGGU
692 CAGGACA CUGAUGAGGCCGAAAGGCCGAA AGGUCUG
693 GCAGGAC CUGAUGAGGCCGAAAGGCCGAA AAGGUCU 696 CUGGCAG CUGAUGAGGCCGAAAGGCCGAA ACAAAGG 709 UGUGGGG CUGAUGAGGCCGAAAGGCCGAA AGUCGCU 720 GGCUGAC CUGAUGAGGCCGAAAGGCCGAA AGUUGUG 723 GGGGGCU CUGAUGAGGCCGAAAGGCCGAA ACAAGUU 735 CCUCUAG CUGAUGAGGCCGAAAGGCCGAA ACCCGGG 738 CCACCUC CUGAUGAGGCCGAAAGGCCGAA AGGACCC 765 GGGAACA CUGAUGAGGCCGAAAGGCCGAA ACCACGG
769 UCCAGGG CUGAUGAGGCCGAAAGGCCGAA ACAGACC
770 GUCCAGG CUGAUGAGGCCGAAAGGCCGAA AACAGAC
785 GACUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGCCC
786 AGACUGG CUGAUGAGGCCGAAAGGCCGAA AACAGCC 792 CCUCCGA CUGAUGAGGCCGAAAGGCCGAA ACUGGGA 794 GGCCUCC CUGAUGAGGCCGAAAGGCCGAA AGACUGG 807 CCAGGUG CUGAUGAGGCCGAAAGGCCGAA ACCUGGG 833 GGGGUUC CUGAUGAGGCCGAAAGGCCGAA ACCUCUG 846 CAUAGGU CUGAUGAGGCCGAAAGGCCGAA ACUGUGG 851 GUUGCCA CUGAUGAGGCCGAAAGGCCGAA AGGUGAC 863 CGAGAAG CUGAUGAGGCCGAAAGGCCGAA AGUCGUU
866 GGCCGAG CUGAUGAGGCCGAAAGGCCGAA AGGAGUC
867 UGGCCGA CUGAUGAGGCCGAAAGGCCGAA AAGGAGU 869 CUUGGCC CUGAUGAGGCCGAAAGGCCGAA AGAAGGA 881 ACUGACU CUGAUGAGGCCGAAAGGCCGAA AGGCCUU 885 UCACACU CUGAUGAGGCCGAAAGGCCGAA ACUGAGG 933 CCAGUAU CUGAUGAGGCCGAAAGGCCGAA ACUGCAC 936 UCCCCAG CUGAUGAGGCCGAAAGGCCGAA AUUACUG 978 AGCUGUA CUGAUGAGGCCGAAAGGCCGAA AUGGUCA 980 AAAGCUG CUGAUGAGGCCGAAAGGCCGAA AGAUGGU
986 CGCCGGA CUGAUGAGGCCGAAAGGCCGAA AGCUGUA
987 GCGCCGG CUGAUGAGGCCGAAAGGCCGAA AAGCUGU
988 GGCGCCG CUGAUGAGGCCGAAAGGCCGAA AAAGCUG 1005 UCGUCAG CUGAUGAGGCCGAAAGGCCGAA AUCACGU
1006 UUCGUCA CUGAUGAGGCCGAAAGGCCGAA AAUCACG
1023 CUUCUGA CUGAUGAGGCCGAAAGGCCGAA ACCUCUG
1025 CCCUUCU CUGAUGAGGCCGAAAGGCCGAA AGACCUC
1066 UUGGCUC CUGAUGAGGCCGAAAGGCCGAA AGGGUGG
1092 GGGCUGG CUGAUGAGGCCGAAAGGCCGAA ACCCCAU
1093 UGGGCUG CUGAUGAGGCCGAAAGGCCGAA AACCCCA
1125 UCAGCAG CUGAUGAGGCCGAAAGGCCGAA AGCUGGG
1153 GCAGGAG CUGAUGAGGCCGAAAGGCCGAA AGCUGCG
1154 AGCAGGA CUGAUGAGGCCGAAAGGCCGAA AAGCUGC
1156 AGAGCAG CUGAUGAGGCCGAAAGGCCGAA AGAAGCU
1172 GGUUGCA CUGAUGAGGCCGAAAGGCCGAA AGCAGGA
1200 UGUGUAU CUGAUGAGGCCGAAAGGCCGAA AGCUGGC
1201 UUGUGUA CUGAUGAGGCCGAAAGGCCGAA AAGCUGG
1203 UCUUGUG CUGAUGAGGCCGAAAGGCCGAA AUAAGCU
1227 GGACACG CUGAUGAGGCCGAAAGGCCGAA AGCUCCC
1228 AGGACAC CUGAUGAGGCCGAAAGGCCGAA AAGCUCC
1233 CAUACAG CUGAUGAGGCCGAAAGGCCGAA ACACGAA
1238 GGGGCCA CUGAUGAGGCCGAAAGGCCGAA ACAGGAC
1264 CCCGGAC CUGAUGAGGCCGAAAGGCCGAA AUCCCUC
1267 UUUCCCG CUGAUGAGGCCGAAAGGCCGAA ACAAUCC
1294 UGCUGGG CUGAUGAGGCCGAAAGGCCGAA AUUUUCU
1295 CUGCUGG CUGAUGAGGCCGAAAGGCCGAA AAUUUUC
1306 CACAUUG CUGAUGAGGCCGAAAGGCCGAA AGUCUGC
1321 UUCCCCC CUGAUGAGGCCGAAAGGCCGAA AGCCUGG
1334 CUCGGGC CUGAUGAGGCCGAAAGGCCGAA AUGGGUU
1344 GACACUU CUGAUGAGGCCGAAAGGCCGAA AGCUCGG
1351 UCCUUUA CUGAUGAGGCCGAAAGGCCGAA ACACUUG
1353 CAUCCUU CUGAUGAGGCCGAAAGGCCGAA AGACACU
1366 AGUGGGA CUGAUGAGGCCGAAAGGCCGAA AGUGCCA
1367 CAGUGGG CUGAUGAGGCCGAAAGGCCGAA AAGUGCC
1368 GCAGUGG CUGAUGAGGCCGAAAGGCCGAA AAAGUGC
1380 AUUCCCC CUGAUGAGGCCGAAAGGCCGAA AUGGGCA
1388 AGUCACU CUGAUGAGGCCGAAAGGCCGAA AUUCCCC
1398 CUCGAGU CUGAUGAGGCCGAAAGGCCGAA ACAGUCA
1402 AGAUCUC CUGAUGAGGCCGAAAGGCCGAA AGUGACA
1408 CCCUCAA CUGAUGAGGCCGAAAGGCCGAA AUCUCGA
1410 UGCCCUC CUGAUGAGGCCGAAAGGCCGAA AGAUCUC
1421 ACAGAGG CUGAUGAGGCCGAAAGGCCGAA AGGUGCC
1425 CCCGACA CUGAUGAGGCCGAAAGGCCGAA AGGUAGG
1429 CUGGCCC CUGAUGAGGCCGAAAGGCCGAA ACAGAGG
1444 UCCCCUU CUGAUGAGGCCGAAAGGCCGAA AGUGCUC
1455 CGCGGGU CUGAUGAGGCCGAAAGGCCGAA ACCUCCC
1482 GGGGGGA CUGAUGAGGCCGAAAGGCCGAA AGCACAU
1484 CCGGGGG CUGAUGAGGCCGAAAGGCCGAA AGAGCAC
1493 AAUCUCA CUGAUGAGGCCGAAAGGCCGAA ACCGGGG
1500 UGAUGAC CUGAUGAGGCCGAAAGGCCGAA AUCUCAU
1503 UGAUGAU CUGAUGAGGCCGAAAGGCCGAA ACAAUCU
1506 CAGUGAU CUGAUGAGGCCGAAAGGCCGAA AUGACAA 1509 CCACAGU CUGAUGAGGCCGAAAGGCCGAA AUGAUGA
1518 CGGCUGC CUGAUGAGGCCGAAAGGCCGAA ACCACAG
1530 CCAUUAU CUGAUGAGGCCGAAAGGCCGAA ACUGCGG
1533 UGCCCAU CUGAUGAGGCCGAAAGGCCGAA AUGACUG
1551 ACGUGCU CUGAUGAGGCCGAAAGGCCGAA AGGCCUG
1559 AUAGAGG CUGAUGAGGCCGAAAGGCCGAA ACGUGCU
1563 GGUUAUA CUGAUGAGGCCGAAAGGCCGAA AGGUACG
1565 GCGGUUA CUGAUGAGGCCGAAAGGCCGAA AGAGGUA
1567 UGGCGGU CUGAUGAGGCCGAAAGGCCGAA AUAGAGG
1584 AUUUCUU CUGAUGAGGCCGAAAGGCCGAA AUCUCCC
1592 UAGUCUG CUGAUGAGGCCGAAAGGCCGAA AUUUCUU
1599 CCUGUUG CUGAUGAGGCCGAAAGGCCGAA AGUCUGU
1551 GUUCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCGUG
1661 CCCGGGA CUGAUGAGGCCGAAAGGCCGAA AGGUUCA
1663 GUCCCGG CUGAUGAGGCCGAAAGGCCGAA AUAGGUU
1678 CGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGGCCCU
1680 GCCGAGG CUGAUGAGGCCGAAAGGCCGAA AGAGGCC
1681 GGCCGAG CUGAUGAGGCCGAAAGGCCGAA AAGAGGC
1684 GAAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGAAGA
1690 AUAUGGG CUGAUGAGGCCGAAAGGCCGAA AGGCCGA
1691 AAUAUGG CUGAUGAGGCCGAAAGGCCGAA AAGGCCG
1696 CCACCAA CUGAUGAGGCCGAAAGGCCGAA AUGGGAA
1698 UGCCACC CUGAUGAGGCCGAAAGGCCGAA AUAUGGG
1737 CAUGGCA CUGAUGAGGCCGAAAGGCCGAA AUGUCUU
1750 GUAGGUG CUGAUGAGGCCGAAAGGCCGAA AGCUGCA
1756 GGGCCGG CUGAUGAGGCCGAAAGGCCGAA AGGUGUA
1787 UGAGGAC CUGAUGAGGCCGAAAGGCCGAA AUGCCCU
1790 GACUGAG CUGAUGAGGCCGAAAGGCCGAA ACAAUGC
1793 UCUGACU CUGAUGAGGCCGAAAGGCCGAA AGGACAA
1797 UGUAUCU CUGAUGAGGCCGAAAGGCCGAA ACUGAGG
1802 GCUGUUG CUGAUGAGGCCGAAAGGCCGAA AUCUGAC
1812 GGCCCCA CUGAUGAGGCCGAAAGGCCGAA AUGCUGU
1813 UGGCCCC CUGAUGAGGCCGAAAGGCCGAA AAUGCUG
1825 GUGCAGG CUGAUGAGGCCGAAAGGCCGAA ACCAUGG
1837 AGUGUUU CUGAUGAGGCCGAAAGGCCGAA AGGUGUG
1845 CGUGGCC CUGAUGAGGCCGAAAGGCCGAA AGUGUUU
1856 CAGAUCA CUGAUGAGGCCGAAAGGCCGAA AUGCGUG
1861 GACUACA CUGAUGAGGCCGAAAGGCCGAA AUCAGAU
1865 AUGUGAC CUGAUGAGGCCGAAAGGCCGAA ACAGAUC
1868 GUCAUGU CUGAUGAGGCCGAAAGGCCGAA ACUACAG
1877 CUUGGCU CUGAUGAGGCCGAAAGGCCGAA AGUCAUG
1901 AUGUCUU CUGAUGAGGCCGAAAGGCCGAA AGUCUUG
1912 AUCCAUC CUGAUGAGGCCGAAAGGCCGAA AUCAUGU
1922 AGACUUU CUGAUGAGGCCGAAAGGCCGAA ACAUCCA
1923 UAGACUU CUGAUGAGGCCGAAAGGCCGAA AACAUCC
1928 CAGGCUA CUGAUGAGGCCGAAAGGCCGAA ACUUUAA
1930 AUCAGGC CUGAUGAGGCCGAAAGGCCGAA AGACUUU
1964 GUGGGGC CUGAUGAGGCCGAAAGGCCGAA AUGUCUC
1983 CCAGUUG CUGAUGAGGCCGAAAGGCCGAA AUGUCCU 1996 GUUUCAG CUGAUGAGGCCGAAAGGCCGAA AUUUCCC
2005 AGGCAGC CUGAUGAGGCCGAAAGGCCGAA AGUUUCA
2013 UACCCAA CUGAUGAGGCCGAAAGGCCGAA AGGCAGC
2015 CAUACCC CUGAUGAGGCCGAAAGGCCGAA AUAGGCA
2020 CUCAGCA CUGAUGAGGCCGAAAGGCCGAA ACCCAAU
2039 CUUCUGU CUGAUGAGGCCGAAAGGCCGAA AGUCUGU
2040 UCUUCUG CUGAUGAGGCCGAAAGGCCGAA AAGUCUG
2057 GUCUAUG CUGAUGAGGCCGAAAGGCCGAA AGGGCCA
2061 ACAUGUC CUGAUGAGGCCGAAAGGCCGAA AUGGAGG
2071 UUGAUGC CUGAUGAGGCCGAAAGGCCGAA ACACAUG
2076 GUGUUUU CUGAUGAGGCCGAAAGGCCGAA AUGCUAC
2097 CGUCAGG CUGAUGAGGCCGAAAGGCCGAA AGUGUGG
2098 CCGUCAG CUGAUGAGGCCGAAAGGCCGAA AAGUGUG
2115 AGUGCCC CUGAUGAGGCCGAAAGGCCCAA AGCUGGC
2128 GUCAGUA CUGAUGAGGCCGAAAGGCCGAA ACAGCAG
2130 GGGUCAG CUGAUGAGGCCGAAAGGCCGAA AGACAGC
2145 UAUCAUC CUGAUGAGGCCGAAAGGCCGAA AGGGUUG
2152 AAAUACA CUGAUGAGGCCGAAAGGCCGAA AUCAUCA
2156 GAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACAUAUC
2158 AUGAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUA
2159 AAUGAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
2160 AAAUGAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
2162 ACAAAUG CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
2163 AACAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
2166 AAUAACA CUGAUGAGGCCGAAAGGCCGAA AUGAAUA
2167 AAAUAAC CUGAUGAGGCCGAAAGGCCGAA AAUGAAU
2170 GUAAAAU CUGAUGAGGCCGAAAGGCCGAA ACAAAUG
2171 GGUAAAA CUGAUGAGGCCGAAAGGCCGAA AACAAAU
2173 CUGGUAA CUGAUGAGGCCGAAAGGCCGAA AUAACAA
2174 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AAUAACA
2175 AGCUGGU CUGAUGAGGCCGAAAGGCCGAA AAAUAAC
2176 UAGCUGG CUGAUGAGGCCGAAAGGCCGAA AAAAUAA
2183 CAAUAAA CUGAUGAGGCCGAAAGGCCGAA AGCUGGU
2185 CUCAAUA CUGAUGAGGCCGAAAGGCCGAA AUAGCUG
2186 ACUCAAU CUGAUGAGGCCGAAAGGCCGAA AAUAGCU
2187 CACUCAA CUGAUGAGGCCGAAAGGCCGAA AAAUAGC
2189 GACACUC CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
2196 CAUAAAA CUGAUGAGGCCGAAAGGCCGAA ACACUCA
2198 UACAUAA CUGAUGAGGCCGAAAGGCCGAA AGACACU
2199 CUACAUA CUGAUGAGGCCGAAAGGCCGAA AAGACAC
2200 CCUACAU CUGAUGAGGCCGAAAGGCCGAA AAAGACA
2201 GCCUACA CUGAUGAGGCCGAAAGGCCGAA AAAAGAC
2205 UUUAGCC CUGAUGAGGCCGAAAGGCCGAA ACAUAAA
2210 GUUCAUU CUGAUGAGGCCGAAAGGCCGAA AGCCUAC
2220 AGAGACC CUGAUGAGGCCGAAAGGCCGAA AUGUUCA
2224 GGCCAGA CUGAUGAGGCCGAAAGGCCGAA ACCUAUG
2226 GAGGCCA CUGAUGAGGCCGAAAGGCCGAA AGACCUA
2233 GCUCCGU CUGAUGAGGCCGAAAGGCCGAA AGGCCAG
2242 GGACUGG CUGAUGAGGCCGAAAGGCCGAA AGCUCCG 2248 UGACAUG CUGAUGAGGCCGAAAGGCCGAA ACUGGGA
2254 UGAAUGU CUGAUGAGGCCGAAAGGCCGAA ACAUGGA
2259 GACCUUG CUGAUGAGGCCGAAAGGCCGAA AUGUGAC
2260 UGACCUU CUGAUGAGGCCGAAAGGCCGAA AAUGUGA
2266 ACCUGGU CUGAUGAGGCCGAAAGGCCGAA ACCUUGA
2274 ACAACUG CUGAUGAGGCCGAAAGGCCGAA ACCUGGU
2279 CCUGUAC CUGAUGAGGCCGAAAGGCCGAA ACUGUAC
2282 CAACCUG CUGAUGAGGCCGAAAGGCCGAA ACAACUG
2288 AGUGUAC CUGAUGAGGCCGAAAGGCCGAA ACCUGUA
2291 UGCAGUG CUGAUGAGGCCGAAAGGCCGAA ACAACCU
2321 CCCAUUU CUGAUGAGGCCGAAAGGCCGAA AUCUUUU
2338 CAAUGAG CUGAUGAGGCCGAAAGGCCGAA AGUCCCA
2339 CCAAUGA CUGAUGAGGCCGAAAGGCCGAA AAGUCCC
2341 GGCCAAU CUGAUGAGGCCGAAAGGCCGAA AGAAGUC
2344 GUUGGCC CUGAUGAGGCCGAAAGGCCGAA AUGAGAA
2358 CUGGGGA CUGAUGAGGCCGAAAGGCCGAA AGGCAGG
2359 UCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGGCAG
2360 UUCUGGG CUGAUGAGGCCGAAAGGCCGAA AAAGGCA
2376 AUAGAAA CUGAUGAGGCCGAAAGGCCGAA AUCACUC
2377 GAUAGAA CUGAUGAGGCCGAAAGGCCGAA AAUCACU
2378 CGAUAGA CUGAUGAGGCCGAAAGGCCGAA AAAUCAC
2379 CCGAUAG CUGAUGAGGCCGAAAGGCCGAA AAAAUCA
2380 GCCGAUA CUGAUGAGGCCGAAAGGCCGAA AAAAAUC
2382 GUGCCGA CUGAUGAGGCCGAAAGGCCGAA AGAAAAA
2384 UUGUGCC CUGAUGAGGCCGAAAGGCCGAA AUAGAAA
2399 GUCCAUA CUGAUGAGGCCGAAAGGCCGAA AGUGCUU
2401 CAGUCCA CUGAUGAGGCCGAAAGGCCGAA AUAGUGC
2411 GAACCAU CUGAUGAGGCCGAAAGGCCGAA ACCAGUC
2417 ACCUGUG CUGAUGAGGCCGAAAGGCCGAA ACCAUUA
2418 AAGCUGU CUGAUGAGGCCGAAAGGCCGAA AACCAUU
2425 AUCUCUG CUGAUGAGGCCGAAAGGCCGAA ACCUGUG
2426 AAUCUCU CUGAUGAGGCCGAAAGGCCGAA AACCUGU
2433 ACUGGGU CUGAUGAGGCCGAAAGGCCGAA AUCUCUG
2434 CACUGGG CUGAUGAGGCCGAAAGGCCGAA AAUCUCU
2448 GAGGAAU CUGAUGAGGCCGAAAGGCCGAA AGGCCUC
2449 GGAGGAA CUGAUGAGGCCGAAAGGCCGAA AAGGCCU
2451 AGGGAGG CUGAUGAGGCCGAAAGGCCGAA AUAAGGC
2452 AAGGGAG CUGAUGAGGCCGAAAGGCCGAA AAUAAGG
2455 GGGAAGG CUGAUGAGGCCGAAAGGCCGAA AGGAAUA
2459 UGGGGGG CUGAUGAGGCCGAAAGGCCGAA AGGGAGG
2460 UUGGGGG CUGAUGAGGCCGAAAGGCCGAA AAGGGAG
2479 GCUAACA CUGAUGAGGCCGAAAGGCCGAA AGGUGUC
2480 GGCUAAC CUGAUGAGGCCGAAAGGCCGAA AAGGUGU
2483 GGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACAAAGG
2484 AGGUGGC CUGAUGAGGCCGAAAGGCCGAA AACAAAG
2492 GGGUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUGGC
2504 AGAAAUG CUGAUGAGGCCGAAAGGCCGAA AUGUGGG
2508 UGGCAGA CUGAUGAGGCCGAAAGGCCGAA AUGUAUG
2509 CUGGCAG CUGAUGAGGCCGAAAGGCCGAA AAUGUAU 2510 ACUGGCA CUGAUGAGGCCGAAAGGCCGAA AAAUGUA
2520 CAUUGUG CUGAUGAGGCCGAAAGGCCGAA ACACUGG
2521 UCAUUGU CUGAUGAGGCCGAAAGGCCGAA AACACUG
2533 GACCGCU CUGAUGAGGCCGAAAGGCCGAA AGUGUCA
2540 CAGACAU CUGAUGAGGCCGAAAGGCCGAA ACCGCUG
2545 AUGUCCA CUGAUGAGGCCGAAAGGCCGAA ACAUGAC
2558 UUGGGCA CUGAUGAGGCCGAAAGGCCGAA AUUCCCU
2579 CAAGGCA CUGAUGAGGCCGAAAGGCCGAA AGCUUGG
2585 AGAGGAC CUGAUGAGGCCGAAAGGCCGAA AGGCAUA
2588 ACAAGAG CUGAUGAGGCCGAAAGGCCGAA ACAAGGC
2591 AGGACAA CUGAUGAGGCCGAAAGGCCGAA AGGACAA
2593 ACAGGAC CUGAUGAGGCCGAAAGGCCGAA AGAGGAC
2596 CAAACAG CUGAUGAGGCCGAAAGGCCGAA ACAAGAG
2601 AAAUGCA CUGAUGAGGCCGAAAGGCCGAA ACAGGAC
2602 GAAAUGC CUGAUGAGGCCGAAAGGCCGAA AACAGGA
2607 CCAGUGA CUGAUGAGGCCGAAAGGCCGAA AUGCAAA
2608 CCCAGUG CUGAUGAGGCCGAAAGGCCGAA AAUGCAA
2609 UCCCAGU CUGAUGAGGCCGAAAGGCCGAA AAAUGCA
2620 AUAGUGC CUGAUGAGGCCGAAAGGCCGAA AGCUCCC
2626 GCUGCAA CUGAUGAGGCCGAAAGGCCGAA AGUGCAA
2628 GAGCUGC CUGAUGAGGCCGAAAGGCCGAA AUAGUGC
2635 GAAACUG CUGAUGAGGCCGAAAGGCCGAA AGCUGCA
2640 UGCAGGA CUGAUGAGGCCGAAAGGCCGAA ACUGGAG
2641 CUGCAGG CUGAUGAGGCCGAAAGGCCGAA AACUGGA
2642 ACUGCAG CUGAUGAGGCCGAAAGGCCGAA AAACUGG
2653 GGACCCU CUGAUGAGGCCGAAAGGCCGAA AUCACUG
2659 CUUGCAG CUGAUGAGGCCGAAAGGCCGAA ACCCUGA
2689 CCUCCAA CUGAUGAGGCCGAAAGGCCGAA ACCUUGG
2691 GUCCUCC CUGAUGAGGCCGAAAGGCCGAA AUACCUU
2700 UGGGAGG CUGAUGAGGCCGAAAGGCCGAA AGUCCUC
2704 AAGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGAGU
2711 CCUUCCA CUGAUGAGGCCGAAAGGCCGAA AGCUGGG
2712 CCCUUCC CUGAUGAGGCCGAAAGGCCGAA AAGCUGG
2721 CGCGGAU CUGAUGAGGCCGAAAGGCCGAA ACCCUUC
2724 ACACGCG CUGAUGAGGCCGAAAGGCCGAA AUGACCC
2744 CUACACA CUGAUGAGGCCGAAAGGCCGAA ACACACA
2750 GCUUGUC CUGAUGAGGCCGAAAGGCCGAA ACACAUA
2759 AGAGCGA CUGAUGAGGCCGAAAGGCCGAA AGCUUGU
2761 ACAGAGC CUGAUGAGGCCGAAAGGCCGAA AGAGCUU
2765 GGUGACA CUGAUGAGGCCGAAAGGCCGAA AGCGAGA
2769 CCUGGGU CUCAUGAGGCCGAAAGGCCGAA ACAGAGC
2797 GAACCAU CUGAUGAGGCCGAAAGGCCGAA AUUGCAC
2803 UGCAGUG CUGAUGAGGCCGAAAGGCCGAA ACCAUGA
2804 CUGCAGU CUGAUGAGGCCGAAAGGCCGAA AACCAUG
2813 AGGUCAA CUGAUGAGGCCGAAAGGCCGAA ACUGCAG
2815 AAAGGUC CUGAUGAGGCCGAAAGGCCGAA AGACUGC
2821 AGCCCAA CUGAUGAGGCCGAAAGGCCGAA AGGUCAA
2822 GAGCCCA CUGAUGAGGCCGAAAGGCCGAA AAGGUCA
2823 UGAGCCC CUGAUGAGGCCGAAAGGCCGAA AAAGGUC 2829 AUCACUU CUGAUGAGGCCGAAAGGCCGAA AGCCCAA
2837 GUGGGAG CUGAUGAGGCCGAAAGGCCGAA AUCACUU
2840 GAGGUGG CUGAUGAGGCCGAAAGGCCGAA AGGAUCA
2847 GGAGGCU CUGAUGAGGCCGAAAGGCCGAA AGGUGGG
2853 UACUCAG CUGAUGAGGCCGAAAGGCCGAA AGGCUGA
2860 UCCCAGC CUGAUGAGGCCGAAAGGCCGAA ACUCAGG
2872 GUGAGCC CUGAUGAGGCCGAAAGGCCGAA AUGGUCC
2877 GUGUUGU CUGAUGAGGCCGAAAGGCCGAA AGCCUAU
2899 AAAAUCA CUGAUGAGGCCGAAAGGCCGAA AUUUGCC
2900 AAAAAUC CUGAUGAGGCCGAAAGGCCGAA AAUUUGC
2904 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AUCAAAU
2905 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAUCAAA
2906 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAUCAA
2907 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAUCA
2908 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAUC
2909 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAU
2910 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2911 AAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2912 GAAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2913 UGAAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2914 CUGAAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2915 UCUGAAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2916 CUCUGAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2917 UCUCUGA CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2918 GUCUCUG CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2919 CGUCUCU CUGAUGAGGCCGAAAGGCCGAA AAAAAAA
2931 GUUGCGA CUGAUGAGGCCGAAAGGCCGAA ACCCCGU
2933 AUGUUGC CUGAUGAGGCCGAAAGGCCGAA AGACCCC
2941 UCUGGGC CUGAUGAGGCCGAAAGGCCGAA AUGUUGC
2951 ACAAAGG CUGAUGAGGCCGAAAGGCCGAA AGUCUGG
2952 CACAAAG CUGAUGAGGCCGAAAGGCCGAA AAGUCUG
2955 UAACACA CUGAUGAGGCCGAAAGGCCGAA AGGAAGU
2956 CUAACAC CUGAUGAGGCCGAAAGGCCGAA AAGGAAG
2961 AUUAACU CUGAUGAGGCCGAAAGGCCGAA ACACAAA
2962 UAUUAAC CUGAUGAGGCCGAAAGGCCGAA AACACAA
2965 CUUUAUU CUGAUGAGGCCGAAAGGCCGAA ACUAACA
2966 GCUUUAU CUGAUGAGGCCGAAAGGCCGAA AACUAAC
2969 AAAGCUU CUGAUGAGGCCGAAAGGCCGAA AUUAACU
2975 GUUGAGA CUGAUGAGGCCGAAAGGCCGAA AGCUUUA
2976 AGUUGAG CUGAUGAGGCCGAAAGGCCGAA AAGCUUU
2977 CAGUUGA CUGAUGAGGCCGAAAGGCCGAA AAAGCUU
2979 GGCAGUU CUGAUGAGGCCGAAAGGCCGAA AGAAAGC Table 5
Mouse ICAM HH Ribozyme Sequence
nt. Position Ribozyme Sequence
11 CAACGGU CUGAUGAGGCCGAAAGGCCGAA ACCAGGG
23 AGCAGAG CUGAUGAGGCCGAAAGGCCGAA ACCACUG
26 AGGAGCA CUGAUGAGGCCGAAAGGCCGAA AGAACCA
31 UGUGGAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
34 CGACCCU CUGAUGAGGCCGAAAGGCCGAA AUGAGAA
40 AGGCUAC CUGAUGAGGCCGAAAGGCCGAA AGUGUGC
48 CCAGGCU CUGAUGAGGCCGAAAGGCCGAA AGGUCCU
54 CCAUCAC CUGAUGAGGCCGAAAGGCCGAA AGGCCCA
58 GGAGCUA CUGAUGAGGCCGAAAGGCCGAA AGGCAUG
64 CUGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUG
96 GGGCCAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
102 CCAGCAG CUGAUGAGGCCGAAAGGCCGAA ACUGGCA
108 GGGCCAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
115 AGGAGCA CUGAUGAGGCCGAAAGGCCGAA AGAACCA
119 UCCUGGU CUGAUGAGGCCGAAAGGCCGAA ACAUUCC
120 GGGCCAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG 146 GGAAGCG CUGAUGAGGCCGAAAGGCCGAA ACGACUG 152 AGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACACAGA 158 GGUUUUU CUGAUGAGGCCGAAAGGCCGAA AACAGGA 165 GCAAAAC CUGAUGAGGCCGAAAGGCCGAA ACUUCUG 168 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUU 185 CUGCACG CUGAUGAGGCCGAAAGGCCGAA ACCCACC 209 GCCAGAG CUGAUGAGGCCGAAAGGCCGAA AAGUGGC 227 GCAAAAC CUGAUGAGGCCGAAAGGCCGAA ACUUCUG 230 GGAGCAA CUGAUGAGGCCGAAAGGCCGAA ACAACUU 237 AGUUCUC CUGAUGAGGCCGAAAGGCCGAA AAGCACA 248 UUUAGGA CUGAUGAGGCCGAAAGGCCGAA AUGGGUU 253 UCUUCCU CUGAUGAGGCCGAAAGGCCGAA AGGCAGG 263 CAGUAGA CUGAUGAGGCCGAAAGGCCGAA AAACCCU 267 UAGGCAG CUGAUGAGGCCGAAAGGCCGAA AGCCCCU 293 CAGCUCA CUGAUGAGGCCGAAAGGCCGAA ACAGCUU 319 GGCUCAG CUGAUGAGGCCGAAAGGCCGAA AUCUCCU 335 GUUCUCA CUGAUGAGGCCGAAAGGCCGAA AGCACAG
337 CAGUGUG CUGAUGAGGCCGAAAGGCCGAA AUUGGAC
338 UCAGCUC CUGAUGAGGCCGAAAGGCCGAA AACAGCU 359 AGCGGAC CUGAUGAGGCCGAAAGGCCGAA ACUGCAC 367 CGGGUUG CUGAUGAGGCCGAAAGGCCGAA AGCCAUU
374 GGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUC
375 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUU 378 ACACGGU CUGAUGAGGCCGAAAGGCCGAA AUGGUAG 386 AAACGAA CUGAUGAGGCCGAAAGGCCGAA ACACGGU 394 AGAUCGA CUGAUGAGGCCGAAAGGCCGAA AGUCCGG 420 CGGGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUGUG 425 CUGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUG 427 CACUGCU CUGAUGAGGCCGAAAGGCCGAA AGAGCUG
450 GCAGGGU CUGAUGAGGCCGAAAGGCCGAA AGGUCCU
451 CAAAGGA CUGAUGAGGCCGAAAGGCCGAA AGGUUUC 456 AGUGGCU CUGAUGAGGCCGAAAGGCCGAA AGGGUAA 495 ACACGGU CUGAUGAGGCCGAAAGGCCGAA AUGGUAG 510 CCCCACG CUGAUGAGGCCGAAAGGCCGAA AGGAGCA 554 GGAUGGA CUGAUGAGGCCGAAAGGCCGAA ACCUGAG 592 CCCAUGU CUGAUGAGGCCGAAAGGCCGAA AUCUUUC
607 CAUGAGA CUGAUGAGGCCGAAAGGCCGAA AUUGGCU
608 GCAUGAG CUGAUGAGGCCGAAAGGCCGAA AAUUGGC
609 GGCAUGA CUGAUGAGGCCGAAAGGCCGAA AAAUUGG 611 GCGGCAU CUGAUGAGGCCGAAAGGCCGAA AGAAAUU
656 CAGCUCA CUGAUGAGGCCGAAAGGCCGAA ACAGCUU
657 UCAGCUC CUGAUGAGGCCGAAAGGCCGAA AACAGCU 668 GGUGGCC CUGAUGAGGCCGAAAGGCCGAA AGGCUCG 677 AGGCUGG CUGAUGAGGCCGAAAGGCCGAA AGAGGUC 684 AGGACCG CUGAUGAGGCCGAAAGGCCGAA AGCUGAA
692 AAGAUCG CUGAUGAGGCCGAAAGGCCGAA AAGUCCG
693 GCAGGGU CUGAUGAGGCCGAAAGGCCGAA AGGUCCU 696 GAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGG 709 UGAGGUG CUGAUGAGGCCGAAAGGCCGAA AGCCGCC 720 AGCUGAA CUGAUGAGGCCGAAAGGCCGAA AGUUGUA 723 CGGAGCU CUGAUGAGGCCGAAAGGCCGAA AAAAGUU
735 UCUCCAG CUGAUGAGGCCGAAAGGCCGAA AUCUGGU 738 CCAUCAC CUGAUGAGGCCGAAAGGCCGAA AGGCCCA 765 GGAAGCG CUGAUGAGGCCGAAAGGCCGAA ACGACUG
769 GGCAGGA CUGAUGAGGCCGAAAGGCCGAA ACAGGCC
770 UUCCAGG CUGAUGAGGCCGAAAGGCCGAA AGCAAAA 785 GGCAGGA CUGAUGAGGCCGAAAGGCCGAA ACAGGCC
736 AGGCAGG CUGAUGAGGCCGAAAGGCCGAA AACAGGC 792 CUUCCGA CUGAUGAGGCCGAAAGGCCGAA ACCUCCA 794 AGUCUCC CUGAUGAGGCCGAAAGGCCGAA AGCCCAG 807 CCAGGUA CUGAUGAGGCCGAAAGGCCGAA AUCCGAG 833 GGGUGUC CUGAUGAGGCCGAAAGGCCGAA AGCUUUG 846 CAACGGU CUGAUGAGGCCGAAAGGCCGAA ACCAGGG 851 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AGGUCUC 863 CCAGAGG CUGAUGAGGCCGAAAGGCCGAA AGUGGCU
866 GGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUC
867 UCUCCGG CUGAUGAGGCCGAAAGGCCGAA AACGAAU 869 CUUGCAU CUGAUGAGGCCGAAAGGCCGAA AGGAAGA 881 ACGGGUU CUGAUGAGGCCGAAAGGCCGAA AAGCCAU 885 UCACCUC CUGAUGAGGCCGAAAGGCCGAA ACCAAGG 933 CCAGAAU CUGAUGAGGCCGAAAGGCCGAA AUUAUAG 936 GCACCAG CUGAUGAGGCCGAAAGGCCGAA AUGAUUA 978 AGUUGUA CUGAUGAGGCCGAAAGGCCGAA ACUGUUA 980 AAAGUUG CUGAUGAGGCCGAAAGGCCGAA AGACUGU
986 AGCUGAA CUGAUCAGGCCGAAAGGCCGAA AGUUGUA
987 CAGCUCA CUGAUGAGGCCGAAAGGCCGAA AAGUUGU 388 GGAGCUG CUGAUGAGGCCGAAAGGCCGAA AAAGUUG 1005 UCUCCAG CUGAUGAGGCCGAAAGGCCGAA AUCUGGU
1006 UUCCCCA CUGAUGAGGCCGAAAGGCCGAA ACUCUCA
1023 CUUCCGA CUGAUGAGGCCGAAAGGCCGAA ACCUCCA
1025 CCCUUCC CUGAUGAGGCCGAAAGGCCGAA AGACCUC
1066 UUAUUUU CUGAUGAGGCCGAAAGGCCGAA AGAGUGG
1092 GGCCUGA CUGAUGAGGCCGAAAGGCCGAA AUCCAGU
1093 UUGGCUG CUGAUGAGGCCGAAAGGCCGAA AGGUCCA
1125 UCAAGAA CUGAUGAGGCCGAAAGGCCGAA AGUUGGG
1163 GCAAAAG CUGAUGAGGCCGAAAGGCCGAA AGCUUCG
1164 AGCAAAA CUGAUGAGGCCGAAAGGCCGAA AAGCUUC
1166 AGAGCAA CUGAUGAGGCCGAAAGGCCGAA AGAAGCU
1172 GGUUUUU CUGAUGAGGCCGAAAGGCCGAA AACAGGA
1200 UGUGGAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
1201 CUGUUCA CUGAUGAGGCCGAAAGGCCGAA AAGCAGC
1203 ACUGGUG CUGAUGAGGCCGAAAGGCCGAA AAAAAGU
1227 GCACACG CUGAUGAGGCCGAAAGGCCGAA AUGUACC
1228 AGCAAAA CUGAUGAGGCCGAAAGGCCGAA AAGCUUC
1233 CUCUCCG CUGAUGAGGCCGAAAGGCCGAA AAACGAA
1238 AGGACCA CUGAUGAGGCCGAAAGGCCGAA ACAGCAC
1264 CUUGCAC CUGAUGAGGCCGAAAGGCCGAA ACCCUUC
1267 UUCCCCA CUGAUGAGGCCGAAAGGCCGAA ACUCUCA
1294 GGCUCAG CUGAUGAGGCCGAAAGGCCGAA AUCUCCU
1295 CUGCUGA CUGAUGAGGCCGAAAGGCCGAA ACCCCUC
1306 CAUUUCA CUGAUGAGGCCGAAAGGCCGAA AGUCUGC
1321 UCCUCCU CUGAUGAGGCCGAAAGGCCGAA AGCCUUC
1334 UUUAGGA CUGAUGAGGCCGAAAGGCCGAA AUGGGUU
1344 CACUCUC CUGAUGAGGCCGAAAGGCCGAA AGCUCAU
1351 UAACUUA CUGAUGAGGCCGAAAGGCCGAA ACAUUCA
1353 CACCUUC CUGAUGAGGCCGAAAGGCCGAA ACCCACU
1366 AGUUGUA CUGAUGAGGCCGAAAGGCCGAA ACUGUUA
1367 AGGUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUGCU
1368 AGAGUGG CUGAUGAGGCCGAAAGGCCGAA ACAGUAC
1380 CCACCCC CUGAUGAGGCCGAAAGGCCGAA AUGGGCA
1388 AGCCACU CUGAUGAGGCCGAAAGGCCGAA AGUCUCC
1398 GUUCUGU CUGAUGAGGCCGAAAGGCCGAA ACAGCCA
1402 AGUUCUC CUGAUGAGGCCGAAAGGCCGAA AAGCACA
1408 CCUCCCC CUGAUGAGGCCGAAAGGCCGAA AUCUCGC
1410 CCCUUCC CUGAUGAGGCCGAAAGGCCGAA AGACCUC
1421 ACAAAAG CUGAUGAGGCCGAAAGGCCGAA AGGUGGG
1425 CUCUACC CUGAUGAGGCCGAAAGGCCGAA AGGCAGU
1429 CAGGGGC CUGAUGAGGCCGAAAGGCCGAA AUAGAGA
1444 UCCUCCU CUGAUGAGGCCGAAAGGCCGAA AGCCUUC
1455 UCCUGGU CUGAUGAGGCCGAAAGGCCGAA ACAUUCC
1482 GGGAGCA CUGAUGAGGCCGAAAGGCCGAA AACAACU
1484 CAUGAGG CUGAUGAGGCCGAAAGGCCGAA AGAACAG
1493 GUUCUCA CUGAUGAGGCCGAAAGGCCGAA AGCACAG
1500 GGACCAU CUGAUGAGGCCGAAAGGCCGAA AUUUCAU
1503 GAAUGAU CUGAUGAGGCCGAAAGGCCGAA AUAGUCC
1506 CGGUUAU CUGAUGAGGCCGAAAGGCCGAA AACAUAA 1509 ACACGGU CUGAUGAGGCCGAAAGGCCGAA AUGGUAG
1518 CGCCUGG CUGAUGAGGCCGAAAGGCCGAA ACCAUGA
1530 CCAGAAU CUGAUGAGGCCGAAAGGCCGAA AUUAUAG
1533 GGCCCAC CUGAUGAGGCCGAAAGGCCGAA AUGACCA
1551 AGCUGCU CUGAUGAGGCCGAAAGGCCGAA AGGCAUG
1559 AGGUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUGCU
1563 GGUUAUA CUGAUGAGGCCGAAAGGCCGAA ACAUAAG
1565 GCGGUUA CUGAUGAGGCCGAAAGGCCGAA AAACAUA
1557 UGGCGGU CUGAUGAGGCCGAAAGGCCGAA AUAAACA
1584 AUAUCCU CUGAUGAGGCCGAAAGGCCGAA AUCUUUC
1592 UAACUUG CUGAUGAGGCCGAAAGGCCGAA AUAUCCU
1599 CCUUCUG CUGAUGAGGCCGAAAGGCCGAA AACUUGU
1651 GCUCAGG CUGAUGAGGCCGAAAGGCCGAA AGGUGGG
1661 CAAAGGA CUGAUGAGGCCGAAAGGCCGAA AGGUUUC
1663 UUCAAAG CUGAUGAGGCCGAAAGGCCGAA AAAGGUU
1678 CCAGGCU CUGAUGAGGCCGAAAGGCCGAA AGGUCCU
1680 CCAGAGG CUGAUGAGGCCGAAAGGCCGAA AGUGGCU
1681 GCCAGAG CUGAUGAGGCCGAAAGGCCGAA AAGUGGC
1684 ACAGCCA CUGAUGAGGCCGAAAGGCCGAA AGGAAGU
1690 AGAUCGA CUCAUGAGGCCGAAAGGCCGAA AGUCCGG
1691 AAGAUCG CUGAUGAGGCCGAAAGGCCGAA AAGUCCG
1696 CCACCCC CUGAUGAGGCCGAAAGGCCGAA AUGGGCA
1698 CUCCAGG CUGAUGAGGCCGAAAGGCCGAA AUAUCCG
1737 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AGGUCUC
1750 UGAGGUG CUGAUGAGGCCGAAAGGCCGAA AGCCGCC
1756 GGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUC
1787 UGGGGAC CUGAUGAGGCCGAAAGGCCGAA AUGUCUC
1790 AUUAGAG CUGAUGAGGCCGAAAGGCCGAA ACAAUGC
1793 UCCAGCC CUGAUGAGGCCGAAAGGCCGAA AGGACCA
1797 UUUAUGU CUGAUGAGGCCGAAAGGCCGAA ACUGGUG
1802 UCUCCAG CUGAUGAGGCCGAAAGGCCGAA AUCUGGU
1812 GGCCUGA CUGAUGAGGCCGAAAGGCCGAA AUCCAGU
1813 UGAGGGU CUGAUGAGGCCGAAAGGCCGAA AAUGCUG
1825 GCAGAGG CUGAUGAGGCCGAAAGGCCGAA AGCGUGG
1837 GGAGCUA CUGAUGAGGCCGAAAGGCCGAA AGGCAUG
1845 GGUGGCC CUGAUGAGGCCGAAAGGCCGAA AGGCUCG
1856 AAGAUCG CUGAUGAGGCCGAAAGGCCGAA AAGUCCG
1861 UACUGGA CUGAUGAGGCCGAAAGGCCGAA AUCAUGU
1865 CUGAGGC CUGAUGAGGCCGAAAGGCCGAA ACAAGUG
1868 UUUAUGU CUGAUGAGGCCGAAAGGCCGAA ACUGGUG
1877 AGCUGCU CUGAUGAGGCCGAAAGGCCGAA AGGCAUG
1901 GUCCCUU CUGAUGAGGCCGAAAGGCCGAA AGUUUUA
1912 ACUGAUC CUGAUGAGGCCGAAAGGCCGAA ACUAUAU
1922 UAACUUA CUGAUGAGGCCGAAAGGCCGAA ACAUUCA
1923 GAUACCU CUGAUGAGGCCGAAAGGCCGAA AGCAUCA
1928 CUGGUAA CUGAUGAGGCCGAAAGGCCGAA ACUCUAA
1930 AGCUGGU CUGAUGAGGCCGAAAGGCCGAA AAACUCU
1964 UGGGGAC CUCAUGAGGCCGAAAGGCCCAA AUGUCUC
1383 UAACUUG CUGAUGAGGCCGAAAGGCCGAA AUAUCCU 1996 GGCUCAG CUGAUGAGGCCGAAAGGCCGAA AUCUCCU
2005 GGUCCGC CUGAUGAGGCCGAAAGGCCGAA AGCUCCA
2013 UACUCAA CUGAUGAGGCCGAAAGGCCGAA AAAUAGC
2015 CCACCCC CUGAUGAGGCCGAAAGGCCGAA AUGGGCA
2020 CUCAGAA CUGAUGAGGCCGAAAGGCCGAA AACCACC
2039 CCUCUGC CUGAUGAGGCCGAAAGGCCGAA AGCCAGC
2040 CCUCCAG CUGAUGAGGCCGAAAGGCCGAA AGGUCAG
2057 GGAUGUG CUGAUGAGGCCGAAAGGCCGAA AGGAGCA
2061 ACACGGU CUGAUGAGGCCGAAAGGCCGAA AUGGUAG
2071 CUGAGGC CUGAUGAGGCCGAAAGGCCGAA ACAAGUG
2076 UAGCUCU CUGAUGAGGCCGAAAGGCCGAA AGGCUAC
2097 CAUCAAG CUGAUGAGGCCGAAAGGCCGAA AGAGUUG
2098 CGGGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUGUG
2115 AUCCUCC CUGAUGAGGCCGAAAGGCCGAA AGGUGGC
2128 CUCAAUA CUGAUGAGGCCGAAAGGCCGAA AUAGCUG
2130 GAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGG
2145 CAUCAAG CUGAUGAGGCCGAAAGGCCGAA AGAGUUG
2152 AACUCUA CUGAUGAGGCCGAAAGGCCGAA AUUAAUA
2156 UAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACAUCAA
2158 AUUAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUC
2159 AAUUAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
2160 AAAUUAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
2162 CUAAAUU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
2163 AAUUAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
2166 AAUAGAG CUGAUGAGGCCGAAAGGCCGAA AUGAAGU
2167 AAUUAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
2170 CUAAAUU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
2171 GGGAGCA CUGAUGAGGCCGAAAGGCCGAA AACAACU
2173 CUGGUAA CUGAUGAGGCCGAAAGGCCGAA ACUCUAA
2174 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AACUCUA
2175 AGCUGGU CUGAUGAGGCCGAAAGGCCGAA AAACUCU
2176 UAGCUGG CUGAUGAGGCCGAAAGGCCGAA AAAACUC
2183 CAAUAAA CUGAUGAGGCCGAAAGGCCGAA AGCUGGU
2185 CUCAAUA CUGAUGAGGCCGAAAGGCCGAA AUAGCUG
2186 ACUCAAU CUGAUGAGGCCGAAAGGCCGAA AAUAGCU
2187 UACUCAA CUGAUGAGGCCGAAAGGCCGAA AAAUAGC
2189 GGUACUC CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
2196 CAUCAAG CUGAUGAGGCCGAAAGGCCGAA AGAGUUG
2198 AACAUAA CUGAUGAGGCCGAAAGGCCGAA AGGCUGC
2199 AUAAACA CUGAUGAGGCCGAAAGGCCGAA AAGAGGC
2200 CUUGCAU CUGAUGAGGCCGAAAGGCCGAA AGGAAGA
2201 GCCGACA CUGAUGAGGCCGAAAGGCCGAA AAAACUU
2205 UCAGGCC CUGAUGAGGCCGAAAGGCCGAA ACAUAAA
2210 AGCCACU CUGAUGAGGCCGAAAGGCCGAA AGUCUCC
2220 AGAGAAC CUGAUGAGGCCGAAAGGCCGAA AUGCCAG
2224 GGAUGGA CUGAUGAGGCCGAAAGGCCGAA ACCUGAG
2226 GCGGCCU CUGAUGAGGCCCAAAGGCCGAA AGAUCCA
2233 CCUCCAG CUCAUGAGGCCCAAAGGCCCAA AGGUCAG
2242 GGUCCGC CUGAUGAGGCCGAAAGGCCGAA AGCUCCA 2248 UGGGAUG CUGAUGAGGCCGAAAGGCCGAA AUGGAUA
2254 UCAGUGU CUGAUGAGGCCGAAAGGCCGAA AAUUGGA
2259 CACCGUG CUGAUGAGGCCGAAAGGCCGAA AUGUGAU
2260 GCACCGU CUGAUGAGGCCGAAAGGCCGAA AAUGUGA
2266 UCCUGGU CUGAUGAGGCCGAAAGGCCGAA ACAUUCC
2274 UCUCCAG CUGAUGAGGCCGAAAGGCCGAA AUCUGGU
2279 CUUGCAC CUGAUGAGGCCGAAAGGCCGAA ACCCUUC
2282 CAGCUCA CUGAUGAGGCCGAAAGGCCGAA ACAGCUU
2288 AGGCCAU CUGAUGAGGCCGAAAGGCCGAA ACUUAUA
2291 AGCAGAG CUGAUGAGGCCGAAAGGCCGAA ACCACUG
2321 CCCAUGU CUGAUGAGGCCGAAAGGCCGAA AUCUUUC
2338 CAGGCAG CUGAUGAGGCCGAAAGGCCGAA AGUCUCA
2339 CAAAGGA CUGAUGAGGCCGAAAGGCCGAA AGGUUUC
2341 AGGCUGG CUGAUGAGGCCGAAAGGCCGAA AGAGGUC
2344 GCUGGAA CUGAUGAGGCCGAAAGGCCGAA AUCGAAA
2358 CUGCUGA CUGAUGAGGCCGAAAGGCCGAA AGCUGGG
2359 UCUGUUC CUGAUGAGGCCGAAAGGCCGAA AAAGCAG
2360 UUCAAAG CUGAUGAGGCCGAAAGGCCGAA AAAGGUU
2376 UCAGAAG CUGAUGAGGCCGAAAGGCCGAA ACCACCU
2377 CUCAGAA CUGAUGAGGCCGAAAGGCCGAA AACCACC
2378 CAGUAGA CUGAUGAGGCCGAAAGGCCGAA AAACCCU
2379 CUUAUGA CUGAUGAGGCCGAAAGGCCGAA AAAAGCA
2380 GCCGACA CUGAUGAGGCCGAAAGGCCGAA AAAACUU
2382 GGGGCAA CUGAUGAGGCCGAAAGGCCGAA AGAGAAU
2384 UUGUGUC CUGAUGAGGCCGAAAGGCCGAA ACUGGAU
2399 GUCCACA CUGAUGAGGCCGAAAGGCCGAA AGUGUUU
2401 CAGCUCA CUGAUGAGGCCGAAAGGCCGAA ACAGCUU
2411 GCAUCCU CUGAUGAGGCCGAAAGGCCGAA ACCAGUA
2417 ACGUAUG CUGAUGAGGCCGAAAGGCCGAA ACCAUUC
2418 GGCCUGA CUGAUGAGGCCGAAAGGCCGAA AUCCAGU
2425 AACCCUC CUGAUGAGGCCGAAAGGCCGAA ACCCAUG
2426 AAACUCU CUGAUGAGGCCGAAAGGCCGAA AAUUAAU
2433 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AACUCUA
2434 AGCUGGU CUGAUGAGGCCGAAAGGCCGAA AAACUCU
2448 GGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUC
2449 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUU
2451 AGGCAGG CUGAUGAGGCCGAAAGGCCGAA AACAGGC
2452 GAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGG
2455 GGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUC
2459 GGGGGGG CUGAUGAGGCCGAAAGGCCGAA AGUGUGG
2460 CGGGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUGUG
2479 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AGGUCUC
2480 GGAUCAC CUGAUGAGGCCGAAAGGCCGAA ACGGUGA
2483 GGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACAUUGG
2484 GACUGGU CUGAUGAGGCCGAAAGGCCGAA AAAAAAG
2492 AGGUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUGCU
2504 ACAAAAG CUGAUGAGGCCGAAAGGCCGAA AGGUGGG
2508 UGGGAUG CUGAUGAGGCCCAAAGGCCGAA AUGGAUA
2509 CUGGUAA CUGAUGAGGCCGAAAC-GCCGAA ACUCUAA 2510 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AACUCUA
2520 CAUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAAAAG
2521 UGAGGGU CUGAUGAGGCCGAAAGGCCGAA AAUGCUG
2533 GAUACCU CUGAUGAGGCCGAAAGGCCGAA AGCAUCA
2540 CACAGCG CUGAUGAGGCCGAAAGGCCGAA ACUGCUG
2545 AGGACCA CUGAUGAGGCCGAAAGGCCGAA ACAGCAC
2568 UUUGACA CUGAUGAGGCCGAAAGGCCGAA ACUUCAC
2579 CAGGCCA CUGAUGAGGCCGAAAGGCCGAA AACUUAU
2585 AGAGAAC CUGAUGAGGCCGAAAGGCCGAA AUGCCAG
2588 AUUAGAG CUGAUGAGGCCGAAAGGCCGAA ACAAUGC
2591 AGGAGCA CUGAUGAGGCCGAAAGGCCGAA AGAACCA
2593 GCAGAGC CUGAUGAGGCCGAAAGGCCGAA AAAGAAG
2596 CAUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAAAAG
2601 AAACGAA CUGAUGAGGCCGAAAGGCCGAA ACACGGU
2602 GGGAUGG CUGAUGAGGCCGAAAGGCCGAA AGCUGGA
2607 CCAGGUA CUGAUGAGGCCGAAAGGCCGAA AUCCGAG
2608 CACAGCG CUGAUGAGGCCGAAAGGCCGAA ACUGCUG
2609 UCCUGGU CUGAUGAGGCCGAAAGGCCGAA ACAUUCC
2620 GCAGGGU CUGAUGAGGCCGAAAGGCCGAA AGGUCCU
2626 GCUGGAA CUGAUGAGGCCGAAAGGCCGAA AUCGAAA
2628 AGGCUAC CUGAUGAGGCCGAAAGGCCGAA AGUGUGC
2635 AGGACCG CUGAUGAGGCCGAAAGGCCGAA AGCUGAA
2640 GGCAGGA CUGAUGAGGCCGAAAGGCCGAA ACAGGCC
2641 CUGCUGA CUGAUGAGGCCGAAAGGCCGAA AGCUGGG
2642 GAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGG
2653 GCAUCCU CUGAUGAGGCCGAAAGGCCGAA ACCAGUA
2659 CUUGCAC CUGAUGAGGCCGAAAGGCCGAA ACCCUUC
2689 CCUCGGA CUGAUGAGGCCGAAAGGCCGAA ACAUUAG
2691 GGCCUCG CUGAUGAGGCCGAAAGGCCGAA AGACAUU
2700 GGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUC
2704 AGGCUGG CUGAUGAGGCCGAAAGGCCGAA AGAGGUC
2711 CUGCUGA CUGAUGAGGCCGAAAGGCCGAA AGCUGGG
2712 CCCUUCC CUGAUGAGGCCGAAAGGCCGAA AGACCUC
2721 CUUGCAC CUGAUGAGGCCGAAAGGCCGAA ACCCUUC
2724 GCACACG CUGAUGAGGCCGAAAGGCCGAA AUGUACC
2744 CUGCACG CUGAUGAGGCCGAAAGGCCGAA ACCCACC
2750 GGUACUC CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
2759 AGAUCGA CUGAUGAGGCCGAAAGGCCGAA AGUCCGG
2761 GCAGGGU CUGAUGAGGCCGAAAGGCCGAA AGGUCCU
2765 AGCGGCA CUGAUGAGGCCGAAAGGCCGAA AGCAAAA
2769 CCUGUUU CUGAUGAGGCCGAAAGGCCGAA ACAGACU
2797 GGACCAU CUGAUGAGGCCGAAAGGCCGAA AUUUCAU
2803 CGCCUGG CUGAUGAGGCCGAAAGGCCGAA ACCAUGA
2804 CUGCACG CUGAUGAGGCCGAAAGGCCGAA ACCCACC
2813 GGGUCAG CUGAUGAGGCCGAAAGGCCGAA ACCGGAG
2815 AAAGUUG CUGAUGAGGCCGAAAGGCCGAA AGACUGU
2821 CCUCCAG CUGAUGAGGCCGAAAGGCCGAA AGGUCAG
2322 AAGUCCG CUCAUGAGGCCCAAAGGCCGAA AGGCUCC
2323 UGGGAGC CUGAUCAGGCCGAAAGGCCGAA AAAGGCA 2829 AUGAUUA CUGAUGAGGCCGAAAGGCCGAA AGUCCAG
2837 UCAGAAG CUGAUGAGGCCGAAAGGCCGAA ACCACCU
2840 CAGGCAG CUGAUGAGGCCGAAAGGCCGAA AGUCUCA
2847 GGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACAUUGG
2853 AACAUAA CUGAUGAGGCCGAAAGGCCGAA AGGCUGC
2860 UCACAGU CUGAUGAGGCCGAAAGGCCGAA ACUUGGC
2872 CUUGGCU CUGAUGAGGCCGAAAGGCCGAA AAGGUCC
2877 GUGAUGG CUGAUGAGGCCGAAAGGCCGAA AGCGGAA
2899 AAGAUCG CUGAUGAGGCCGAAAGGCCGAA AAGUCCG
2900 AAAACUC CUGAUGAGGCCGAAAGGCCGAA AAAUUAA
2904 AAUAGAG CUGAUGAGGCCGAAAGGCCGAA AUGAAGU
2905 CAAUAGA CUGAUGAGGCCGAAAGGCCGAA AAUGAAG
2906 UAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACAUCAA
2907 AAAUUAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
2908 AGCAAAA CUGAUGAGGCCGAAAGGCCGAA AAGCUUC
2909 AGAGCAA CUGAUGAGGCCGAAAGGCCGAA AGAAGCU
2910 AAAUUAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
2911 AAAUUAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
2912 GACAUUA CUGAUGAGGCCGAAAGGCCGAA AGAACAA
2913 UGACCAG CUGAUGAGGCCGAAAGGCCGAA AGAGAAA
2914 CUUAUGA CUGAUGAGGCCGAAAGGCCGAA AAAAGCA
2915 UCUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
2916 CUCCGGA CUGAUGAGGCCGAAAGGCCGAA ACGAAUA
2917 UCUCCGG CUGAUGAGGCCGAAAGGCCGAA AACGAAU
2918 CUCUCCG CUGAUGAGGCCGAAAGGCCGAA AAACGAA
2919 CGACCCU CUGAUGAGGCCGAAAGGCCGAA AUGAGAA
2931 CUUCCGA CUGAUGAGGCCGAAAGGCCGAA ACCUCCA
2933 CCCUUCC CUGAUGAGGCCGAAAGGCCGAA AGACCUC
2941 UGGGGAC CUGAUGAGGCCGAAAGGCCGAA AUGUCUC
2951 GCAGAGG CUGAUGAGGCCGAAAGGCCGAA AGCGUGG
2952 CACAGCG CUGAUGAGGCCGAAAGGCCGAA ACUGCUG
2955 UGACACA CUGAUGAGGCCGAAAGGCCGAA AGUCACU
2956 UUGAUUC CUGAUGAGGCCGAAAGGCCGAA AAGGAAA
2961 AGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACACAGA
2962 AAUUAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
2965 CUUUAUU CUGAUGAGGCCGAAAGGCCGAA AUUCAAA
2966 CCUCUGC CUGAUGAGGCCGAAAGGCCGAA AGCCAGC
2969 AAAACUU CUGAUGAGGCCGAAAGGCCGAA AUUGAUU
2975 GCUGGUA CUGAUGAGGCCGAAAGGCCGAA AACUCUA
2976 AGUAGAG CUGAUGAGGCCGAAAGGCCGAA AACCCUC
2977 CAGCUCA CUGAUGAGGCCGAAAGGCCGAA ACAGCUU
2979 GGCAAUA CUGAUGAGGCCGAAAGGCCGAA AGAAUGA Table 6
Human ICAM Hairpin Ribozyme/Substrate Sequences
nt. Hairpin Ribozyme Sequence Substrate Position
70 GGGCCGGG AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCA GCC CCCGGCCC
86 GGAGUGCG AGAA GCGC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GCGCU GCC CGCACUCC 343 CCCAUCAG AGAA GUUU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AAACU GCC CUGAUGGG 635 GCCCUUGG AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCG GCC CCAAGGGC 653 UGUUCUCA AGAA GCUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAGCU GUU UGAGAACA 782 AGACUGGG AGAA GCCC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GGGCU GUU CCCAGUCU 920 CUGCACAC AGAA GCCG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GGGCU GAC GUGUGCAG 1301 ACAUUGGA AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCA GAC UCCAAUGU 1373 CCCCGAUG AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACU GCC CAUCGGGG 1521 AUGACUGC AGAA GCUA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UAGCA GCC GCAGUCAU 1594 CUGUUGUA AGAA GUAU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AUACA GAC UACAACAG 2008 ACCCAAUA AGAA GCAA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UUGCU GCC UAUUGGGU 2034 UUCUGUAA AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACA GAC UUACAGAA 2125 GGUCAGUA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCU GUC UACUGACC 2132 GGGUUGGG AGAA GUAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUACU GAC CCCAACCC 2276 ACCUGUAC AGAA GUAC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GUACA GUU GUACAGGU 2810 AAGGUCAA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCA GUC UUGACCUU
Table 7
Mouse ICAM Hairpin Ribozyme/Substrate Sequences
nt. Hairpin Ribozyme Sequence Substrate Position
76 GGGAUCAC AGAA GUGA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UCACC GUU GUGAUCCC 164 UGAGGAAG AGAA GUUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAACU GUU CUUCCUCA 252 UCAGCUCA AGAA GCUU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AAGCU GUU UGAGCUGA 284 GCACAGCG AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCA GUC CGCUGUGC 318 AAGCGGAC AGAA GCAC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GUGCA GUC GUCCGCUU 447 AGAGCUGG AGAA GCGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCGCG GAC CCAGCUCU 804 UCUCCUGG AGAA GCAU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AUGCC GAC CCAGGAGA 847 UCUACCAA AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACU GCC UUGGUAGA 913 AGGAUCUG AGAA GCUA ACCAGAGLAAACACACGUUGUGGUACAUUACCUGGUA UAGCG GAC CAGAUCCU 946 AAGUUGUA AGAA GUUA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UAACA GUC UACAACUU 1234 CCCAAGCA AGAA GUCU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AGACG GAC UGCUUGGG 1275 AUUUCAGA AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCA GAC UCUGAAAU 1325 UGCCUUCC AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCA GAC GGAAGGCA 1350 CCCCGAUG AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCU GCC CAUCGGGG 1534 ACAUAAGA AGAA GCCA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UGGCA GCC UCUUAUGU 1851 GUCCACCG AGAA GUAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUACA GCC CGGUGGAC 1880 AGAAUGAA AGAA GCGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACGCU GAC UUCAUUCU
Table 8
Rat ICAM Hairpin Ribozyme/Substrate Sequences
nt. Hairpin Ribozyme Sequence Substrate
Position
5 AAAGUGCA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCU GCC UGCACUUU
59 GGAGCAGA AGAA GCAU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AUGCU GCC UCUGCUCC
84 GGGAUCAC AGAA GCGA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UCGCC GUU GUGAUCCC
295 GCACAGUG AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCA GAC CACUGUGC
329 AAGCCGAG AGAA GCGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACGCA GUC CUCGGCUU
433 UUCCACCA AGAA GCGC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GCGCU GCC UGGUGGAA
626 CAUUCUUG AGAA GUGA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UCACU GUU CAAGAAUG
806 UCUCCAGG AGAA GCAU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AUGCU GAC CCUGGAGA
849 UCCACUGA AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACU GCC UCAGUGGA
915 AGGGUCUG AGAA GCCA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UGGCG GAC CAGACCCU
1182 ACCUCCAA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCG GCC UUGGAGGU
1307 AUGUAAGA AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCA GAC UCUUACAU
1357 UGCUUUCC AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCA GCC GGAAAGCA
1382 UCCCGAUA AGAA GCGG ACCΛGAGΛAACACACGUUGUGGUACAUUACCUGGUA CCGCU GCC UAUCGGGA
1858 GCCCACCA AGAA GUAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUACA GCC UGGUGGGC
1887 AGAAGGAA AGAA GCCU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AGGCU GAC UUCCUUCU
2012 GAGUUGGG AGAA GUGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACACU GUC CCCAACUC
2303 AGACUCCA AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACA GCC UGGAGUCU
2539 CCUCCCAC AGAA GCUU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AAGCU GUU GUGGGAGG
Table 9: Rat ICAM HH Ribozyme Target Sequence
nt. HH Target sequence nt. HH Target Sequence Position Position
11 GAUCCAAU U CACACUGA 394 GUGGUGCU U CUGAACAG 23 GCUGACUU C CUUCUCUA 420 GCACCCCU C CCAGCGCA 26 GAACUGCU C UUCCUCUU 425 CCUCGGCU U CUGCCACC 31 CCUCUGCU C CUGGUCCU 427 UCCCUGUU U AAAAACCA 34 CUGAAGCU C AGAUAUAC 450 AAGAACCU C AUCCUGCG 40 CUCAAGGU A CAAGCCCC 451 GGGUACUU C CCCCAGGC 48 GAGAACCU C GGCCUGGG 456 CUCGGCUU C UGCCACCA 54 CCCCGCCU C CCUGAGCC 495 GCCACCAU C ACUUUGUA 58 CCGUGCCU U UAGCUCCC 510 GUGCUGCU C CGUGGGAA 64 CAAUGGCU U CAACCCGU 564 GAAAAUGU U CCAACCAC 96 CCUCUGCU C CUGGUCCU 592 GGGAGUAU C ACCAGGGA 102 CUCCUGGU C CUGGUCGC 607 GAGCCAAU U UCUCAUGC
108 GGACUGCU U GGGGAACU 608 AGCCAAUU U CUCAUGCU
115 UCCUACCU U UGUUCCCA 609 GCCAAUUU C UCAUGCUU
119 GACACUGU C CCCAACUC 611 CAAUUUCU C AUGCUUCA
120 GUUGUGAU C CCCGGGCC 656 GUCACUGU U CAAGAAUG 146 CCAGACCU U GGAACUCC 657 UCACUGUU C AAGAAUGC 152 ACCCGGCU C CACCUCAA 668 GAACUGCU C UUCCUCUU 158 AUUUCUUU C ACGAGUCA 677 GCACCCCU C CCAGCGCA 165 UGAACAGU A CUUCCCCC 684 AGGCAGCU C CGGACUUU 168 GAAGCCUU C CUGCCUCG 692 CCAGACCU U GGAACUCC 185 GGGUGGAU C CGUGCAGG 693 CGGACUUU C GAUCUUCC 209 CAGCCCCU A AUCUGACC 696 GCCUGUUU C CUGCCUCU 227 GACCAAGU A ACUGUGAA 709 CAGCAUUU A CCCCUCAC 230 CAAGCUGU U GUGGGAGG 720 CUACAACU U UUCAGCUC 237 CUGAAGCU C GACACCCC 723 CAACUUUU C AGCUCCCA 248 GGCCCCCU A CCUUAGGA 735 CUCCUGGU C CUGGUCGC 253 CACUGCCU C AGUGGAGG 738 UCCUGCCU C GGGGUGGA 263 GAGCCAAU U UCUCAUGC 765 ACUGUGCU U UGAGAACU 267 GAAGCCUU C CUGCCUCG 769 UCUUGUGU U CCCUGGAA 293 GAAGCUCU U CAAGCUGA 770 CUUGUGUU C CCUGGAAG 319 CGGAGGAU C ACAAACGA 785 AGGCCUGU U UCCUGCCU 335 ACUGUGCU U UGAGAACU 786 GGCCUGUU U CCUGCCUC
337 UGUGCUAU A UGGUCCUC 792 CUCCUGGU C CUGGUCGC
338 AAGCUCUU C AAGCUGAG 794 UCCUGCCU C UGAAGCUA 359 CACGCAGU C CUCGGCUU 807 GCUCAGAU A UACCUGGA 367 CAAUGGCU U CAACCCGU 833 CCUGGGGU U GGAGACUA
374 UUACCCCU C ACCCACCU 846 CUGACAGU U AUUUAUUG
375 AGAAGCCU U CCUGCCUC 851 GCUCACCU U UAGCAGCU 378 ACCCACCU C ACAGGGUA 863 CAAUGGCU U CAACCCGU 386 CGCUGUGU U UUGGAGCU 866 CCAUGCUU C CUCUGACA 867 GACCACCU C CCCACCUA 1421 GGGUACUU C CCCCAGGC 869 CUCUUCCU C UUGCGAAG 1425 ACCCACCU C CUCUGGCU 881 AAUGGCUU C AACCCGUG 1429 AUACUUGU A GCCUCAGG 885 GACCAAGU A ACUGUGAA 1444 AGAAGGCU C AGGAGGAG 933 UGUGUAUU C GUUCCCAG 1455 GGGAGUAU C ACCAGGGA 936 GCAGAGAU U UUGUGUCA 1482 AGGGUACU U CCCCCAGG 978 UUGAGAAU C UACAACUU 1484 ACUGCUCU U CCUCUUGC 980 GAGAAUCU A CAACUUUU 1493 CCUGGGGU U GGAGACUA 986 CUACAACU U UUCAGCUC 1500 CG-UGAAAU U AUGGUCAA 987 UACAACUU U UCAGCUCC 1503 GAAAAUGU U CCAACCAC 988 ACAACUUU U CAGCUCCC 1506 UGGGUCAU A AUUGUUGG 1005 UUCGUGAU c GUGGCGUC 1509 GCCACCAU C ACUFUGUA 1006 GUGGGAGU A UCACCAGG 1518 GUCCUGGU C GCCGUUGU 1023 CCGGAGGU C UCAGAAGG 1530 ACCUGGGU C AUAAUUGU 1025 GGAGGUCU C AGAAGGGG 1533 CUGAUCAU U GCGGGCUU 1066 CCUACCUU U GUUCCCAA 1551 GUGGCCCU C UGCUCGUA 1092 AGAGGGGU C UCAGCAGA 1559 UGGGAAGU C CCUGUUUA 1093 AGGGGAAU C CAGCCCCU 1563 UCCUACCU U UGUUCCCA 1125 CCCCAACU C UUGUUGAU 1565 UUACACCU A UUACCGCC 1163 ACGACGCU U CUUUUGCU 1567 ACACCUAU U ACCGCCAG 1164 CGACGCUU C UUUUGCUC 1584 AGGAAGAU C AGGAUAUA 1166 ACGCUUCU U UUGCUCUG 1592 CAGGAUAU A CAAGUUAC 1172 CUUUUGCU C UGCGGCCU 1599 UACAAGUU A CAGAAGGC 1200 AUCCAAUU C ACACUGAA 1651 CCCCGCCU C CCUGAGCC 1201 UUGGGCUU C UCCACAGG 1661 CUGCACUU U GCCCUGGU 1203 GGGCUUCU C CACAGGUC 1663 GAACAGAU C AAUGGACA 1227 UUGGAACU C CAUGUGCU 1678 GAGAACCU C GGCCUGGG 1228 GCGGGCUU C GUGAUCGU 1680 GGGCUUCU C CACAGGUC 1222 CUCCUGGU C CUGGUCGC 1681 GGCCUGUU U CCUGCCUC 1238 UGUGCUAU A UGGUCCUC 1684 CUGCUCGU A GACCUCUC 1264 GGAAAGAU C AUACGGGU 1690 CCCCACCU A CAUACAUU 1267 GUCACUGU U CAAGAAUG 1691 CCGGACUU U CGAUCUUC 1294 CAGAGAUU UGUGUCAG 1696 CUCCUGGU C CUGGUCGC 1295 AGAGGGGU C UCAGCAGA 1698 UCAGAUAU A CCUGGAGA 1306 AGCAGACU C UUACAUGC 1737 GAUCACAU U CACGGUGC 1321 AACAGAGU C UGGGGAAA 1750 GUCCAUUU A CACCUAUU 1334 GUAUUCGU U CCCAGAGC 1756 CCUCUGCU C CUGGUCCU 1344 UCGGUGCU C AGGUAUCC 1787 GAGAACCU C GGCCUGGG 1351 UCAGGCCU A AGAGGACU 1790 GACACUGU C CCCAACUC 1353 UAGCAGCU C AACAAUGG 1793 AUGGUCCU C ACCUGGAC 1366 AGGGUACU CCCCCAGG 1797 UCCCUGUU U AAAAACCA 1367 GGGUACUU C CCCCAGGC 1802 GCUCAGAU A UACCUGGA 1368 GAUGGUGU C CCGCUGCC 1812 AACAGAGU C UGGGGAAA 1380 CUGCCUAU c GGGAUGGU 1813 GCGGGCUU C GUGAUCGU 1388 UGGAGACU A ACUGGAUG 1825 GCCACCAU C ACUGUGUA 1398 CUGGCUGU C ACAGGACA 1837 ACCCACCU C ACAGGGUA 1402 CUGUGCUU U GAGAACUG 1845 AGAGGACU C GGAGGGGC 1408 UUCGUGAU C GUGGCGUC 1856 C CCCCUAAU C UGACCUGC 1410 CGAACUAU C GAGUGGAC 1861 CCAUGUGCU A UAUGGUCC 1865 UAUCCGGU A GACACAAG 2198 GAAUGUCU C CGAGGUCA
1868 UCACGAGU C AUAUAAAU 2199 AGACUCUU A CAUGCCAG
1877 ACAGUACU U CCCCCAGG 2200 GGGUACUU C CCCCAGGC
1901 CUAAAACU C AAGGUACA 2201 GGGCUUCU C CACAGGUC
1912 GAACAGAU C AAUGGACA 2205 UUUUGUGU C AGCCACUG
1922 AUGUAAGU U AUUGCCUA 2210 UGGAGACU A ACUGGAUG
1923 UGGACGCU C ACCUUUAG 2220 GAGAACCU C GGCCUGGG
1928 GCUCAGAU A UACCUGGA 2224 ACAUACAU U CCUACCUU
1930 UGGAGACU A ACUGGAUG 2226 CUGGACCU C AGGCCACA
1964 AGAGAUUU U GUGUCAGC 2233 UCAUGCUU C ACAGAACU
1983 GAGAACCU C GGCCUGGG 2242 ACACAGCU C UCAGUAGU
1996 UGGAAGCU C UUCAAGCU 2248 CUCCUGGU C CUGGUCGC
2005 AUGUAAGU U AUUGCCUA 2254 AUCCAAUU C ACACUGAA
2013 CGCUGCCU A UCGGGAUG 2259 GAUCACAU U CACGGUGC
2015 CUGCCUAU C GGGADΞGU 2260 AUCACAUU C ACGGUGCU
2020 UAUUGAGU A CCCUGUAC 2266 AUCAGGAU A UACAAGUU 2039 CGGAGGAU C ACAAACGA 2274 GAGCAGGU U AACAUGUA 2040 CCUGACCU C CUGGAGGU 2279 GGAAAGAU C AUACGGGU 2057 CUGGUCCU C CAAUGGCU 2282 ACAGUUAU U UAUUGAGU 2061 GCGUCCAU U UACACCUA 2288 GCCCUGGU C CUCCAAUG 2071 AUACUUGU A GCCUCAGG 2291 CAGGAUAU A CAAGUUAC 2076 CGUAGCCU C AGGCCUAA 2321 GGAAAGAU C AUACGGGU 2097 CCAACUCU U GUUGAUGU 2338 UUGGGCUU C UCCACAGG 2098 CCUGACCU C CUGGAGGU 2339 GGGUACUU C CCCCAGGC 2115 UUCCGACU A GGGUCCUG 2341 GGGCCUGU C GGUGCUCA 2128 AGUGCUGU A CCAUGAUC 2344 CUGCUCGU A GACCUCUC 2130 GCCUGUUU C CUGCCUCU 2358 CCCUGCCU C CUCCCACA 2145 CCAACUCU U GUUGAUGU 2359 CCAUCCAU C CCACAGAA 2152 UUGAGAAU C UACAACUU 2360 CUUGUGUU C CCUGGAAG 2156 UGACAGUU A UUUAUUGA 2376 GAACUGCU C UUCCUCUU 2158 UGAUGUAU U UAUUAAUU 2377 GACUUCCU U CUCUAUUA 2159 GAUGUAUU U AUUAAUUC 2378 GCUGAUUU C UUUCACGA 2160 AUGUAUUU A UUAAUUCA 2379 CUGCUCUU C CUCUUGCG 2162 ACAUUCCU A CCUUUGUU 2380 UGAUUUCU U UCACGAGU 2163 UAUUUAUU A AUUCAGAG 2382 AUUUCUUU C ACGAGUCA 2166 UGAUGUAU U UAUUAAUU 2384 UAUCCGGU A GACACAAG 2167 GAUGUAUU U AUUAAUUC 2399 UAAAUACU A UGUGGACG 2170 GUAUUUAU U AAUUCAGA 2401 UGUGCUAU A UGGUCCUC 2171 CAGUUAUU U AUUGAGUA 2411 CAAUUUCU C AUGCUUCA 2173 UGUGCUAU A UGGUCCUC 2417 AUCAGGAU A UACAAGUU 2174 UCUCUAUU A CCCCUGCU 2418 UCAUGCUU C ACAGAACU 2175 AUUUCUUU C ACGAGUCA 2425 UUAUUAAU U CAGAGUUC 2176 GAAAAUGU U CCAACCAC 2426 CCUGGGGU U GGAGACUA 2183 UGACAGUU A UUUAUUGA 2433 UCAGAGUU C UGACAGUU 2185 ACAGUUAU U UAUUGAGU 2434 CGGAGGAU C ACAAACGA2186 CAGUUAUU U AUUGAGUA 2448 UGAACAGU A CUUCCCCC 2187 AGUUAUUU A UUGAGUAC 2449 GAAGCCUU C CUGCCUCG 2189 UUAUUUAU U GAGUACCC 2451 GGCCUGUU U CCUGCCUC2196 CUGACAGU U AUUUAUUG 2452 GCCUGUUU C CUGCCUCU 2455 ACAUUCCU A CCUUUGUU 2761 CGGACUUU C GAUCUUCC
2459 CCCUGCCU C CUCCCACA 2765 CUUUUGCU C UGCGGCCU
2460 CCUACCUU U GUUCCCAA 2769 UUCUCUAU U ACCCCUGC
2479 UUACACCU A UUACCGCC 2797 CGUGAAAU U AUGGUCAA
2480 GUCGCCGU U GUGAUCCC 2803 CUCAUGCU U CACAGAAC
2483 ACCUUUGU U CCCAAUGU 2804 UCAUGCUU C ACAGAACU
2484 CCUUUGUU C CCAAUGUC 2813 GCUCCCAU C CUGACCCU
2492 GACCACCU C CCCACCUA 2815 CGGACUUU C GAUCUUCC
2504 ACCUACAU A CAUUCCUA 2821 CCUGACCU C CUGGAGGU
2508 ACAUACAU U CCUACCUU 2822 UACAACUU U UCAGCUCC
2509 CAUACAUU C CUACCUUU 2823 CAACUUUU C AGCUCCCA
2510 GUCCAUUU A CACCUAUU 2829 UCGGUGCU C AGGUAUCC
2520 ACCUUUGU U CCCAAUGU 2837 CACAGGGU A CUUCCCCC
2521 CCUUUGUU C CCAAUGUC 2840 GCACCCCU C CCAGCGCA
2533 ACAGCAUU U ACCCCUCA 2847 UUACCCCU C ACCCACCU
2540 UCGGUGCU C AGGUAUCC 2853 UUCGAUCU U CCGACUAG
2545 AGGCAGCU C CGGACUUU 2860 UCUUGUGU U CCCUGGAA
2563 CAGAGAUU U UGUGUCAG 2872 GGGCCUGU C GGUGCUCA
2579 CCUGCACU U UGCCCUGG 2877 UGGAGUCU C CCAGCACC
2585 CUGCUCGU A GACCUCUC 2899 AGGCAGCU C CGGACUUU
2588 UGCCUCCU C CCACAGCC 2900 GGCUGACU U CCUUCUCU
2591 CUCUUCCU C UUGCGAAG 2904 GAACUGCU C UUCCUCUU
2593 UCUCUAUU A CCCCUGCU 2905 GGCUGACU U CCUUCUCU
2596 CUCCUGGU C CUGGUCGC 2906 GUUGAUGU A UUUAUUAA
2601 UGUGCUAU A UGGUCCUC 2907 CUGCUCUU C CUCUUGCG
2602 GUCCUGGU C GCCGUUGU 2908 UGAUGUAU U UAUUAAUU
2607 GUGGGAGU A UCACCAGG 2909 GAACUGCU C UUCCUCUU
2608 CUUUAGCU C CCGUGGGA 2910 ACUUCCUU C UCUAUUAC
2609 UGGAGACU A ACUGGAUG 2911 UUCCUUCU C UAUUACCC
2620 UCAGAGUU C UGACAGUU 2912 AUGUAUUU A UUAAUUCA
2626 CUCUCAGU A GUGCUGCU 2913 UGUGUAUU C GUUCCCAG
2628 UACAACUU U UCAGCUCC 2914 GUAUUUAU U AAUUCAGA
2635 UCACAGAU C CAAUUCAC 2915 UAUUUAUU A AUUCAGAG
2640 GCUCAGGU A UCCAUCCA 2916 CUCUUCCU C UUGCGAAG
2641 CCCCACCU A CAUACAUU 2917 CUUCCUCU U GCGAAGAC
2642 GCCUGUUU C CUGCCUCU 2918 AUUUCUUU C ACGAGUCA
2653 CCACAGGU c AGGGUGCU 2919 UUUUGUGU C AGCCACUG
2659 AGAAGGGU c CUGCAAGC 2931 GAUGGUGU C CCGCUGCC
2689 ACUAGGGU c CUGAAGCU 2933 UGGAGUCU C CCAGCACC
2691 UCAGGCCU A AGAGGACU 2941 CAGUACUU C CCCCAGGC
2700 AGGGUACU U CCCCCAGG 2951 ACCAUGCU U CCUCUGAC
2704 GACCACCU C CCCACCUA 2952 CCGGACUU U CGAUCUUC
2711 CCCUACCU U AGGAAGGU 2955 UGCUUCCU C UGACAUGG
2712 CCUACCUU A GGAAGGUG 2956 CUUUCCUU U GAAUCAAU
2721 GGAAAGAU C AUACGGGU 2961 UUUUGUGU C AGCCACUG
2724 AAGAUCAU A CGGGUUUG 2962 UGUGUAUU C GUUCCCAG
2744 GGGUGGAU C CGUGCAGG 2965 CUUUGAAU C AAUAAAGU
2750 GUCCCUGU U UAAAAACC 2966 UGGAAGCU C UUCAAGCU
2759 GACGAACU A UCGAGUGG 2969 GAAUCAAU A AAGUUUUA 2975 UGGAAGCU C UUCAAGCU
2976 UAUAUGGU C CUCACCUG
2977 GAAGCUCU U CAAGCUGA
Table 10: Rat ICAM HH Ribozyme Sequences
nt. Rat HH Ribozyme Sequence
Position
11 UCAGUGUG CUGAUGAGGCCGAAAGGCCGAA AϋUGGAUC
23 UAGAGAAG CUGAUGAGGCCGAAAGGCCGAA AAGUCAGC
26 AAGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGCAGUUC
31 AGGACCAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAGG
34 GUAUAUCU CUGAUGAJGGCCGAAAGGCCGAA AGCϋUCAG
40 GGGGCUUG CUGAUGAGGCCGAAAGGCCGAA ACCUUGAG
48 CCCAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGUUCUC
54 GGCUCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCGGGG
58 GGGAGCUA CUGAUGAGGCCGAAAGGCCGAA AGGCACGG
64 ACGGGUUG CUGAUGAGGCCGAAAGGCCGAA AGCCAϋUG
96 AGGACCAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAGG
102 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG
108 AGUUCCCC CUGAUGAGGCCGAAAGGCCGAA AGCAGUCC
115 UGGGAACA CUGAϋGAC^GCCGAAAGGCCGAA AGGUAGGA
119 GAGUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGUGUC
120 GGCCCGGG CUGAUGAGGCCGAAAGGCCGAA AUCACAAC 146 GGAGUUCC CUGAUGAGGCCGAAAGGCCGAA AGGUCUGG 152 UUGAGGUG CUGAUGAGGCCGAAAGGCCGAA AGCCGGGU 158 UGACUCGU CUGAUGAGGCCGAAAGGCCGAA AAAGAAAU 165 GGGGGAAG CUGAUGAGGCCGAAAGGCCGAA ACUGUUCA 163 CGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUUC 185 CCUGCACG CUGAUGAGGCCGAAAGGCCGAA AUCCACCC 209 GGUCAGAU CUGAUGAGGCCGAAAGGCCGAA AGGGGCUG 227 UUCACAGU CUGAUGAGGCCGAAAGGCCGAA ACUUGGUC 230 CCUCCCAC CUGAUGAGGCCGAAAGGCCGAA ACAGCUUG 237 GGGGUGUC CUGAUGAGGCCGAAAGGCCGAA AGCUUCAG 248 UCCUAAGG CUGAUGAGGCCGAAAGGCCGAA AGGGGGCC 253 CCUCCACU CUGAUGAGGCCGAAAGGCCGAA AGGCAGUG 263 GCAUGAGA CUGAUGAGGCCGAAAGGCCGAA AUUGGCUC 267 CGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUUC 293 UCAGCUUG CUGAUGAGGCCGAAAGGCCGAA AGAGCUUC 319 UCGUUUGU CUGAUGAGGCCGAAAGGCCGAA AUCCUCCG 335 AGUUCUCA CUGAUGAGGCCGAAAGGCCGAA AGCACAGU
337 GAGGACCA CUGAUGAGGCCGAAAGGCCGAA AUAGCACA
338 CUCAGCUU CUGAUGAGGCCGAAAGGCCGAA AAGAGCUU 359 AAGCCGAG CUGAUGAGGCCGAAAGGCCGAA ACUGCGUG 367 ACGGGUUG CUGAUGAGGCCGAAAGGCCGAA AGCCAUUG
374 AGGUGGGU CUGAUGAGGCCGAAAGGCCGAA AGGGGUAA
375 GAGGCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCUUCU 378 UACCCUGU CUGAUGAGGCCGAAAGGCCGAA AGGUGGGU 386 AGCUCCAA CUGAUGAGGCCGAAAGGCCGAA ACACAGCG 394 CUGUUCAG CUGAUGAGGCCGAAAGGCCGAA AGCACCAC
420 UGCGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUGC
425 GGUGGCAG CUGAUGAGGCCGAAAGGCCGAA AGCCGAGG
427 UGGUUUUU CUGAUGAGGCCGAAAGGCCGAA AACAGGGA
450 CGCAGGAU CUGAUGAGGCCGAAAGGCCGAA AGGUUCUU
451 GCCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUACCC 456 UGGUGGCA CUGAUGAGGCCGAAAGGCCGAA AAGCCGAG 495 UACACAGU CUGAUGAGGCCGAAAGGCCGAA AUGGUGGC 510 UUCCCACG CUGAUGAGGCCGAAAGGCCGAA AGCAGCAC 564 GUGGUUGG CUGAUGAGGCCGAAAGGCCGAA ACAUUUUC 592 UCCCUGGU CUGAUGAGGCCGAAAGGCCGAA AUACUCCC
607 GCAUGAGA CUGAUGAGGCCGAAAGGCCGAA AUUGGCUC
608 AGCAUGAG CUGAUGAGGCCGAAAGGCCGAA AAUUGGCU
609 AAGCAUGA CUGAUGAGGCCGAAAGGCCGAA AAAUUGGC 611 UGAAGCAU CUGAUGAGGCCGAAAGGCCGAA AGAAAUUG
656 CAUUCUUG CUGAUGAGGCCGAAAGGCCGAA ACAGUGAC
657 ACAUUCUU CUGAUGAGGCCGAAAGGCCGAA AACAGUGA 668 AAGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGCAGUUC 677 UGCGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUGC 684 AAAGUCCG CUGAUGAGGCCGAAAGGCCGAA AGCUGCCU
692 GGAGUUCC CUGAUGAGGCCGAAAGGCCGAA AGGUCUGG
693 GGAAGAUC CUGAUGAGGCCGAAAGGCCGAA AAAGUCCG 696 AGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGGC 709 GUGAGGGG CUGAUGAGGCCGAAAGGCCGAA AAAUGCUG 720 GAGCUGAA CUGAUGAGGCCGAAAGGCCGAA AGUUGUAG 723 UGGGAGCU CUGAUGAGGCCGAAAGGCCGAA AAAAGUUG 735 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG 738 UCCACCCC CUGAUGAGGCCGAAAGGCCGAA AGGCAGGA 765 AGUUCUCA CUGAUGAGGCCGAAAGGCCGAA AGCACAGU
769 UUCCAGGG CUGAUGAGGCCGAAAGGCCGAA ACACAAGA
770 CUUCCAGG CUGAUGAGGCCGAAAGGCCGAA AACACAAG
785 AGGCAGGA CUGAUGAGGCCGAAAGGCCGAA ACAGGCCU
786 GAGGCAGG CUGAUGAGGCCGAAAGGCCGAA AACAGGCC 792 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG 794 GAGCUUCA CUGAUGAGGCCGAAAGGCCGAA AGGCAGGA 807 UCCAGGUA CUGAUGAGGCCGAAAGGCCGAA AUCUGAGC 833 UAGUCUCC CUGAUGAGGCCGAAAGGCCGAA ACCCCAGG 846 CAAUAAAU CUGAUGAGGCCGAAAGGCCGAA ACUGUCAG 851 AGCUGCUA CUGAUGAGGCCGAAAGGCCGAA AGGUGAGC 863 ACGGGUUG CUGAUGAGGCCGAAAGGCCGAA AGCCAUUG
866 UGUCAGAG CUGAUGAGGCCGAAAGGCCGAA AAGCAUGG
867 UAGGUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUGGUC 869 CUUCGCAA CUGAUGAGGCCGAAAGGCCGAA AGGAAGAG 881 CACGGGUU CUGAUGAGGCCGAAAGGCCGAA AAGCCAUU 885 UUCACAGU CUGAUGAGGCCGAAAGGCCGAA ACUUGGUC 933 CUGGGAAC CUGAUGAGGCCGAAAGGCCGAA AAUACACA 936 UGACACAA CUGAUGAGGCCGAAAGGCCGAA AUCUCUGC 978 AAGUUGUA CUGAUGAGGCCGAAAGGCCGAA AUUCUCAA 980 AAAAGUUG CUGAUGAGGCCGAAAGGCCGAA AGAUUCUC 986 GAGCUGAA CUGAUGAGGCCGAAAGGCCGAA AGUUGUAG
987 GGAGCUGA CUGAUGAGGCCGAAAGGCCGAA AAGUUGUA
988 GGGAGCUG CUGAUGAGGCCGAAAGGCCGAA AAAGUUGU
1005 GACGCCAC CUGAUGAGGCCGAAAGGCCGAA AUCACGAA
1006 CCUGGUGA CUGAUGAGGCCGAAAGGCCGAA ACUCCCAC 1023 CCUUCUGA CUGAUGAGGCCGAAAGGCCGAA ACCUCCGG 1025 CCCCUUCU CUGAUGAGGCCGAAAGGCCGAA AGACCUCC 1066 UUGGGAAC CUGAUGAGGCCGAAAGGCCGAA AAGGUAGG
1092 UCUGCUGA CUGAUGAGGCCGAAAGGCCGAA ACCCCUCU
1093 AGGGGCUG CUGAUGAGGCCGAAAGGCCGAA AUUCCCCU 1125 AUCAACAA CUGAUGAGGCCGAAAGGCCGAA AGUUGGGG 1153 AGCAAAAG CUGAUGAGGCCGAAAGGCCGAA AGCGUCGU 1164 GAGCAAAA CUGAUGAGGCCGAAAGGCCGAA AAGCGUCG 1166 CAGAGCAA CUGAUGAGGCCGAAAGGCCGAA AGAAGCGU 1172 AGGCCGCA CUGAUGAGGCCGAAAGGCCGAA AGCAAAAG
1200 UUCAGUGU CUGAUGAGGCCGAAAGGCCGAA AAUUGGAU
1201 CCUGUGGA CUGAUGAGGCCGAAAGGCCGAA AAGCCCAA 1203 GACCUGUG CUGAUGAGGCCGAAAGGCCGAA AGAAGCCC
1227 AGCACAUG CUGAUGAGGCCGAAAGGCCGAA AGUUCCAA
1228 ACGAUCAC CUGAUGAGGCCGAAAGGCCGAA AAGCCCGC 1233 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG 1238 GAGGACCA CUGAUGAGGCCGAAAGGCCGAA AUAGCACA 1264 ACCCGUAU CUGAUGAGGCCGAAAGGCCGAA AUCUUUCC 1267 CAUUCUUG CUGAUGAGGCCGAAAGGCCGAA ACAGUGAC
1294 CUGACACA CUGAUGAGGCCGAAAGGCCGAA AAUCUCUG
1295 UCUGCUGA CUGAUGAGGCCGAAAGGCCGAA ACCCCUCU 1306 GCAUGUAA CUGAUGAGGCCGAAAGGCCGAA AGUCUGCU 1321 UUUCCCCA CUGAUGAGGCCGAAAGGCCGAA ACUCUGUU 1334 GCUCUGGG CUGAUGAGGCCGAAAGGCCGAA ACGAAUAC 1344 GGAUACCU CUGAUGAGGCCGAAAGGCCGAA AGCACCGA 1351 AGUCCUCU CUGAUGAGGCCGAAAGGCCGAA AGGCCUGA 1353 CCAUUGUU CUGAUGAGGCCGAAAGGCCGAA AGCUGCUA
1366 CCUGGGGG CUGAUGAGGCCGAAAGGCCGAA AGUACCCU
1367 GCCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUACCC 1363 GGCAGCGG CUGAUGAGGCCGAAAGGCCGAA ACACCAUC 1380 ACCAUCCC CUGAUGAGGCCGAAAGGCCGAA AUAGGCAG 1388 CAUCCAGU CUGAUGAGGCCGAAAGGCCGAA AGUCUCCA 1398 UGUCCUGU CUGAUGAGGCCGAAAGGCCGAA ACAGCCAG 1402 CAGUUCUC CUGAUGAGGCCGAAAGGCCGAA AAGCACAG 1408 GACGCCAC CUGAUGAGGCCGAAAGGCCGAA AUCACGAA 1410 GUCCACUC CUGAUGAGGCCGAAAGGCCGAA AUAGUUCG 1421 GCCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUACCC 1425 AGCCAGAG CUGAUGAGGCCGAAAGGCCGAA .AGGUGGGU 1429 CCUGAGGC CUGAUGAGGCCGAAAGGCCGAA ACAAGUAU 1444 CUCCUCCU CUGAUGAGGCCGAAAGGCCGAA AGCCUUCU 1455 UCCCUGGU CUGAUGAGGCCGAAAGGCCGAA AUACUCCC 1482 CCUGGGGG CUGAUGAGGCCGAAAGGCCGAA AGUACCCU 1484 GCAAGAGG CUGAUGAGGCCGAAAGGCCGAA AGAGCAGU 1493 UAGUCUCC CUGAUGAGGCCGAAAGGCCGAA ACCCCAGG 1500 UUGACCAU CUGAUGAGGCCGAAAGGCCGAA AUUUCACG
1503 GUGGUUGG CUGAUGAGGCCGAAAGGCCGAA ACAUUUUC
1506 CCAACAAU CUGAUGAGGCCGAAAGGCCGAA AUGACCCA
1509 UACACAGU CUGAUGAGGCCGAAAGGCCGAA AUGGUGGC
1518 ACAACGGC CUGAUGAGGCCGAAAGGCCGAA ACCAGGAC
1530 ACAAUUAU CUGAUGAGGCCGAAAGGCCGAA ACCCAGGU
1533 AAGCCCGC CUGAUGAGGCCGAAAGGCCGAA AUGAUCAG
1551 UACGAGCA CUGAUGAGGCCGAAAGGCCGAA AGGGCCAC
1559 UAAACAGG CUGAUGAGGCCGAAAGGCCGAA ACUUCCCA
1563 UGGGAACA CUGAUGAGGCCGAAAGGCCGAA AGGUAGGA
1565 GGCGGUAA CUGAUGAGGCCGAAAGGCCGAA AGGUGUAA
1567 CUGGCGGU CUGAUGAGGCCGAAAGGCCGAA AUAGGUGU
1584 UAUAUCCU CUGAUGAGGCCGAAAGGCCGAA AUCUUCCU
1592 GUAACUUG CUGAUGAGGCCGAAAGGCCGAA AUAUCCUG
1599 GCCUUCUG CUGAUGAGGCCGAAAGGCCGAA AACUUGUA
1651 GGCUCAGG CUGAUGAGGCCGAAAGGCCGAA AGGCGGGG
1661 ACCAGGGC CUGAUGAGGCCGAAAGGCCGAA AAGUGCAG
1663 UGUCCAUU CUGAUGAGGCCGAAAGGCCGAA AUCUGUUC
1678 CCCAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGUUCUC
1680 GACCUGUG CUGAUGAGGCCGAAAGGCCGAA AGAAGCCC
1681 GAGGCAGG CUGAUGAGGCCGAAAGGCCGAA AACAGGCC
1684 GAGAGGUC CUGAUGAGGCCGAAAGGCCGAA ACGAGCAG
1690 AAUGUAUG CUGAUGAGGCCGAAAGGCCGAA AGGUGGGG
1691 GAAGAUCG CUGAUGAGGCCGAAAGGCCGAA AAGUCCGG
1696 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG
1698 UCUCCAGG CUGAUGAGGCCGAAAGGCCGAA AUAUCUGA
1737 GCACCGUG CUGAUGAGGCCGAAAGGCCGAA AUGUGAUC
1750 AAUAGGUG CUGAUGAGGCCGAAAGGCCGAA AAAUGGAC
1756 AGGACCAG CUGAUGAGGCCGAAAGGCCGAA AGCAGAGG
1787 CCCAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGUUCUC
1790 GAGUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGUGUC
1793 GUCCAGGU CUGAUGAGGCCGAAAGGCCGAA AGGACCAU
1797 UGGUUUUU CUGAUGAGGCCGAAAGGCCGAA AACAGGGA
1802 UCCAGGUA CUGAUGAGGCCGAAAGGCCGAA AUCUGAGC
1812 UUUCCCCA CUGAUGAGGCCGAAAGGCCGAA ACUCUGUU
1813 ACGAUCAC CUGAUGAGGCCGAAAGGCCGAA AAGCCCGC
1825 UACACAGU CUGAUGAGGCCGAAAGGCCGAA AUGGUGGC
1837 UACCCUGU CUGAUGAGGCCGAAAGGCCGAA AGGUGGGU
1845 GCCCCUCC CUGAUGAGGCCGAAAGGCCGAA AGUCCUCU
1856 GCAGGUCA CUGAUGAGGCCGAAAGGCCGAA AUUAGGGG
1861 GGACCAUA CUGAUGAGGCCGAAAGGCCGAA AGCACAUG
1865 CUUGUGUC CUGAUGAGGCCGAAAGGCCGAA ACCGGAUA
1868 AUUUAUAU CUGAUGAGGCCGAAAGGCCGAA ACUCGUGA
1877 CCUGGGGG CUGAUGAGGCCGAAAGGCCGAA AGUACUGU
1901 UGUACCUU CUGAUGAGGCCGAAAGGCCGAA AGUUUUAG
1912 UGUCCAUU CUGAUGAGGCCGAAAGGCCGAA AUCUGUUC
1922 UAGGCAAU CUGAUGAGGCCGAAAGGCCGAA ACUUACAU
1923 CUAAAGGU CUGAUGAGGCCGAAAGGCCGAA AGCGUCCA
1928 UCCAGGUA CUGAUGAGGCCGAAAGGCCGAA AUCUGAGC 1930 CAUCCAGU CUGAUGAGGCCGAAAGGCCGAA AGUCUCCA
1964 GCUGACAC CUGAUGAGGCCGAAAGGCCGAA AAAUCUCU
1983 CCCAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGUUCUC
1996 AGCUUGAA CUGAUGAGGCCGAAAGGCCGAA AGCUUCCA
2005 UAGGCAAU CUGAUGAGGCCGAAAGGCCGAA ACUUACAU
2013 CAUCCCGA CUGAUGAGGCCGAAAGGCCGAA AGGCAGCG
2015 ACCAUCCC CUGAUGAGGCCGAAAGGCCGAA AUAGGCAG
2020 GUACAGGG CUGAUGAGGCCGAAAGGCCGAA ACUCAAUA
2039 UCGUUUGU CUGAUGAGGCCGAAAGGCCGAA AUCCUCCG
2040 ACCUCCAG CUGAUGAGGCCGAAAGGCCGAA AGGUCAGG
2057 AGCCAUUG CUGAUGAGGCCGAAAGGCCGAA AGGACCAG
2061 UAGGUGUA CUGAUGAGGCCGAAAGGCCGAA AUGGACGC
2071 CCUGAGGC CUGAUGAGGCCGAAAGGCCGAA ACAAGUAU
2076 UUAGGCCU CUGAUGAGGCCGAAAGGCCGAA AGGCUACA
2097 ACAUCAAC CUGAUGAGGCCGAAAGGCCGAA AGAGUUGG
2098 ACCUCCAG CUGAUGAGGCCGAAAGGCCGAA AGGUCAGG
2115 CAGGACCC CUGAUGAGGCCGAAAGGCCGAA AGUCGGAA
2128 GAUCAUGG CUGAUGAGGCCGAAAGGCCGAA ACAGCACU
2130 AGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGGC
2145 ACAUCAAC CUGAUGAGGCCGAAAGGCCGAA AGAGUUGG
2152 AAGUUGUA CUGAUGAGGCCGAAAGGCCGAA AUUCUCAA
2156 UCAAUAAA CUGAUGAGGCCGAAAGGCCGAA AACUGUCA
2158 AAUUAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUCA
2159 GAAUUAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAUC
2150 UGAAUUAA CUGAUGAGGCCGAAAGGCCGAA AAAUACAU
2162 AACAAAGG CUGAUGAGGCCGAAAGGCCGAA AGGAAUGU
2153 CUCUGAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAUA
2166 AAUUAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUCA
2167 GAAUUAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAUC
2170 UCUGAAUU CUGAUGAGGCCGAAAGGCCGAA AUAAAUAC
2171 UACUCAAU CUGAUGAGGCCGAAAGGCCGAA AAUAACUG
2173 GAGGACCA CUGAUGAGGCCGAAAGGCCGAA AUAGCACA
2174 AGCAGGGG CUGAUGAGGCCGAAAGGCCGAA AAUAGAGA
2175 UGACUCGU CUGAUGAGGCCGAAAGGCCGAA AAAGAAAU
2176 GUGGUUGG CUGAUGAGGCCGAAAGGCCGAA ACAUUUUC
2183 UCAAUAAA CUGAUGAGGCCGAAAGGCCGAA AACUGUCA
2185 ACUCAAUA CUGAUGAGGCCGAAAGGCCGAA AUAACUGU
2186 UACUCAAU CUGAUGAGGCCGAAAGGCCGAA AAUAACUG
2187 GUACUCAA CUGAUGAGGCCGAAAGGCCGAA AAAUAACu
2189 GGGUACUC CUGAUGAGGCCGAAAGGCCGAA AUAAAUAA
2196 CAAUAAAU CUGAUGAGGCCGAAAGGCCGAA ACUGUCAG
2198 UGACCUCG CUGAUGAGGCCGAAAGGCCGAA AGACAUUC
2199 CUGGCAUG CUGAUGAGGCCGAAAGGCCGAA AAGAGUCU
2200 GCCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUACCC
2201 GACCUGUG CUGAUGAGGCCGAAAGGCCGAA AGAAGCCC
2205 CAGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACACAAAA
2210 CAUCCAGU CUGAUGAGGCCGAAAGGCCGAA AGUCUCCA
2220 CCCAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGUUCUC
2224 AAGGUAGG CUGAUGAGGCCGAAAGGCCGAA AUGUAUGU 2226 UGUGGCCU CUGAUGAGGCCGAAAGGCCGAA AGGUCCAG
2233 AGUUCUGU CUGAUGAGGCCGAAAGGCCGAA AAGCAUGA
2242 ACUACUGA CUGAUGAGGCCGAAAGGCCGAA AGCUGUGU
2248 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG
2254 UUCAGUGU CUGAUGAGGCCGAAAGGCCGAA AAUUGGAU
2259 GCACCGUG CUGAUGAGGCCGAAAGGCCGAA AUGUGAUC
2260 AGCACCGU CUGAUGAGGCCGAAAGGCCGAA AAUGUGAU
2266 AACUUGUA CUGAUGAGGCCGAAAGGCCGAA AUCCUGAU
2274 UACAUGUU CUGAUGAC4GCCGAAAGGCCGAA ACCUGCUC
2279 ACCCGUAU CUGAUGAGGCCGAAAGGCCGAA AUCUUUCC
2282 ACUCAAUA CUGAUGAGGCCGAAAGGCCGAA AUAACUGU
2288 CAUUGGAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGGC
2291 GUAACUUG CUGAUGAGGCCGAAAGGCCGAA AUAUCCUG
2321 ACCCGUAU CUGAUGAGGCCGAAAGGCCGAA AUCUUUCC
2338 CCUGUGGA CUGAUGAGGCCGAAAGGCCGAA AAGCCCAA
2339 GCCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUACCC
2341 UGAGCACC CUGAUGAGGCCGAAAGGCCGAA ACAGGCCC
2344 GAGAGGUC CUGAUGAGGCCGAAAGGCCGAA ACGAGCAG
2358 UGUGGGAG CUGAUGAGGCCGAAAGGCCGAA AGGCAGGG
2359 UUCUGUGG CUGAUGAGGCCGAAAGGCCGAA AUGGAUGG
2360 CUUCCAGG CUGAUGAGGCCGAAAGGCCGAA AACACAAG
2376 AAGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGCAGUUC
2377 UAAUAGAG CUGAUGAGGCCGAAAGGCCGAA AGGAAGUC
2378 UCGUGAAA CUGAUGAGGCCGAAAGGCCGAA AAAUCAGC
2379 CGCAAGAG CUGAUGAGGCCGAAAGGCCGAA AAGAGCAG
2380 ACUCGUGA CUGAUGAGGCCGAAAGGCCGAA ACAAAUCA
2382 UGACUCGU CUGAUGAGGCCGAAAGGCCGAA AAAGAAAU
2384 CUUGUGUC CUGAUGAGGCCGAAAGGCCGAA ACCGGAUA
2399 CGUCCACA CUGAUGAGGCCGAAAGGCCGAA AGUAUUUA
2401 GAGGACCA CUGAUGAGGCCGAAAGGCCGAA AUAGCACA
2411 UGAAGCAU CUGAUGAGGCCGAAAGGCCGAA AGAAAUUG
2417 AACUUGUA CUGAUGAGGCCGAAAGGCCGAA AUCCUGAU
2418 AGUUCUGU CUGAUGAGGCCGAAAGGCCGAA AAGCAUGA
2425 GAACUCUG CUGAUGAGGCCGAAAGGCCGAA AUUAAUAA
2426 UAGUCUCC CUGAUGAGGCCGAAAGGCCGAA ACCCCAGG
2433 AACUGUCA CUGAUGAGGCCGAAAGGCCGAA AACUCUGA
2434 UCGUUUGU CUGAUGAGGCCGAAAGGCCGAA AUCCUCCG
2448 GGGGGAAG CUGAUGAGGCCGAAAGGCCGAA ACUGUUCA
2449 CGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUUC
2451 GAGGCAGG CUGAUGAGGCCGAAAGGCCGAA AACAGGCC
2452 AGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGGC
2455 AACAAAGG CUGAUGAGGCCGAAAGGCCGAA AGGAAUGU
2459 UGUGGGAG CUGAUGAGGCCGAAAGGCCGAA AGGCAGGG
2460 UUGGGAAC CUGAUGAGGCCGAAAGGCCGAA AAGGUAGG
2479 GGCGGUAA CUGAUGAGGCCGAAAGGCCGAA AGGUGUAA
2480 GGGAUCAC CUGAUGAGGCCGAAAGGCCGAA ACGGCGAC
2483 ACAUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAAAGGU
2484 GACAUUGG CUGAUGAGGCCGAAAGGCCGAA AACAAAGG
2492 UAGGUGGG CUGAUGAGGCCGAAAGGCCGAA A.GGUGGUC 2504 UAGGAAUG CUGAUGAGGCCGAAAGGCCGAA AUGUAGGU
2508 AAGGUAGG CUGAUGAGGCCGAAAGGCCGAA AUGUAUGU
2509 AAAGGUAG CUGAUGAGGCCGAAAGGCCGAA AAUGUAUG
2510 AAUAGGUG CUGAUGAGGCCGAAAGGCCGAA AAAUGGAC
2520 ACAUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAAAGGU
2521 GACAUUGG CUGAUGAGGCCGAAAGGCCGAA AACAAAGG
2533 UGAGGGGU CUGAUGAGGCCGAAAGGCCGAA AAUGCUGU
2540 GGAUACCU CUGAUGAGGCCGAAAGGCCGAA AGCACCGA
2545 AAAGUCCG CUGAUGAGGCCUAAAGGCCGAA AGCUGCCU
2568 CUGACACA CUGAUGAGGCCGAAAGGCCGAA AAUCUCUG
2579 CCAGGGCA CUGAUGAGGCCGAAAGGCCGAA AGUGCAGG
2585 GAGAGGUC CUGAUGAGGCCGAAAGGCCGAA ACGAGCAG
2588 GGCUGUGG CUGAUGAGGCCGAAAGGCCGAA AGGAGGCA
2591 CUUCGCAA CUGAUGAGGCCGAAAGGCCGAA AGGAAGAG
2593 AGCAGGGG CUGAUGAGGCCGAAAGGCCGAA AAUAGAGA
2596 GCGACCAG CUGAUGAGGCCGAAAGGCCGAA ACCAGGAG
2601 GAGGACCA CUGAUGAGGCCGAAAGGCCGAA AUAGCACA
2602 ACAACGGC CUGAUGAGGCCGAAAGGCCGAA ACCAGGAC
2607 CCUGGUGA CUGAUGAGGCCGAAAGGCCGAA ACUCCCAC
2608 UCCCACGG CUGAUGAGGCCGAAAGGCCGAA AGCUAAAG
2609 CAUCCAGU CUGAUGAGGCCGAAAGGCCGAA AGUCUCCA
2620 AACUGUCA CUGAUGAGGCCGAAAGGCCGAA AACUCUGA
2626 AGCAGCAC CUGAUGAGGCCGAAAGGCCGAA ACUGAGAG
2628 GGAGCUGA CUGAUGAGGCCGAAAGGCCGAA AAGUUGUA
2635 GUGAAUUG CUGAUGAGGCCGAAAGGCCGAA AUCUGUGA
2640 UGGAUGGA CUGAUGAGGCCGAAAGGCCGAA ACCUGAGC
2641 AAUGUAUG CUGAUGAGGCCGAAAGGCCGAA AGGUGGGG
2642 AGAGGCAG CUGAUGAGGCCGAAAGGCCGAA AAACAGGC
2653 AGCACCCU CUGAUGAGGCCGAAAGGCCGAA ACCUGUGG
2659 GCUUGCAG CUGAUGAGGCCGAAAGGCCGAA ACCCUUCU
2689 AGCUUCAG CUGAUGAGGCCGAAAGGCCGAA ACCCUAGU
2691 AGUCCUCU CUGAUGAGGCCGAAAGGCCGAA AGGCCUGA
2700 CCUGGGGG CUGAUGAGGCCGAAAGGCCGAA AGUACCCU
2704 UAGGUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUGGUC
2711 ACCUUCCU CUGAUGAGGCCGAAAGGCCGAA AGGUAGGG
2712 CACCUUCC CUGAUGAGGCCGAAAGGCCGAA AAGGUAGG
2721 ACCCGUAU CUGAUGAGGCCGAAAGGCCGAA AUCUUUCC
2724 CAAACCCG CUGAUGAGGCCGAAAGGCCGAA AUGAUCUU
2744 CCUGCACG CUGAUGAGGCCGAAAGGCCGAA AUCCACCC
2750 GGUUUUUA CUGAUGAGGCCGAAAGGCCGAA ACAGGGAC
2759 CCACUCGA CUGAUGAGGCCGAAAGGCCGAA AGUUCGUC
2761 GGAAGAUC CUGAUGAGGCCGAAAGGCCGAA AAAGUCCG
2765 AGGCCGCA CUGAUGAGGCCGAAAGGCCGAA AGCAAAAG
2769 GCAGGGGU CUGAUGAGGCCGAAAGGCCGAA AUAGAGAA
2797 UUGACCAU CUGAUGAGGCCGAAAGGCCGAA AUUUCACG
2803 GUUCUGUG CUGAUGAGGCCGAAAGGCCGAA AGCAUGAG
2804 AGUUCUGU CUGAUGAGGCCGAAAGGCCGAA AAGCAUGA
2813 AGGGUCAG CUGAUGAGGCCGAAAGGCCGAA AUGGGAGC
2815 GGAAGAUC CUGAUGAGGCCGAAAGGCCGAA AAAGUCCG 2821 ACCUCCAG CUGAUGAGGCCGAAAGGCCGAA AGGUCAGG 2822 GGAGCUGA CUGAUGAGGCCGAAAGGCCGAA AAGUUGUA 2823 UGGGAGCU CUGAUGAGGCCGAAAGGCCGAA AAAAGUUG 2829 GGAUACCU CUGAUGAGGCCGAAAGGCCGAA AGCACCGA 2837 GGGGGAAG CUGAUGAGGCCGAAAGGCCGAA ACCCUGUG 2840 UGCGCUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUGC 2847 AGGUGGGU CUGAUGAGGCCGAAAGGCCGAA AGGGGUAA 2853 CUAGUCGG CUGAUGAGGCCGAAAGGCCGAA AGAUCGAA 2860 UUCCAGGG CUGAUGAGGCCGAAAGGCCGAA ACACAAGA 2372 UGAGCACC CUGAUGAGGCCGAAAGGCCGAA ACAGGCCC 2877 GGUGCUGG CUGAUGAGGCCGAAAGGCCGAA AGACUCCA 2899 AAAGUCCG CUGAUGAGGCCGAAAGGCCGAA AGCUGCCU 2900 AGAGAAGG CUGAUGAGGCCGAAAGGCCGAA AGUCAGCC 2904 AAGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGCAGUUC 2905 AGAGAAGG CUGAUGAGGCCGAAAGGCCGAA AGUCAGCC 2906 UUAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACAUCAAC 2907 CGCAAGAG CUGAUGAGGCCGAAAGGCCGAA AAGAGCAG 2908 AAUUAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUCA 2909 AAGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGCAGUUC 2910 GUAAUAGA CUGAUGAGGCCGAAAGGCCGAA AAGGAAGU 2911 GGGUAAUA CUGAUGAGGCCGAAAGGCCGAA AGAAGGAA 2912 UGAAUUAA CUGAUGAGGCCGAAAGGCCGAA AAAUACAU 2913 CUGGGAAC CUGAUGAGGCCGAAAGGCCGAA AAUACACA 2914 UCUGAAUU CUGAUGAGGCCGAAAGGCCGAA AUAAAUAC
2915 CUCUGAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAUA 2916 CUUCGCAA CUGAUGAGGCCGAAAGGCCGAA AGGAAGAG 2917 GUCUUCGC CUGAUGAGGCCGAAAGGCCGAA AGAGGAAG 2913 UGACUCGU CUGAUGAGGCCGAAAGGCCGAA AAAGAAAU 2919 CAGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACACAAAA 2931 GGCAGCGG CUGAUGAGGCCGAAAGGCCGAA ACACCAUC 2933 GGUGCUGG CUGAUGAGGCCGAAAGGCCGAA AGACUCCA 2941 GCCUGGGG CUGAUGAGGCCGAAAGGCCGAA AAGUACUG 2951 GUCAGAGG CUGAUGAGGCCGAAAGGCCGAA AGCAUGGU 2952 GAAGAUCG CUGAUGAGGCCGAAAGGCCGAA AAGUCCGG 2955 CCAUGUCA CUGAUGAGGCCGAAAGGCCGAA AGGAAGCA 2956 AUUGAUUC CUGAUGAGGCCGAAAGGCCGAA AAGGAAAG 2961 CAGUGGCU CUGAUGAGGCCGAAAGGCCGAA ACACAAAA 2962 CUGGGAAC CUGAUGAGGCCGAAAGGCCGAA AAUACACA 2965 ACUUUAUU CUGAUGAGGCCGAAAGGCCGAA AUUCAAAG 2966 AGCUUGAA CUGAUGAGGCCGAAAGGCCGAA AGCUUCCA 2969 UAAAACUU CUGAUGAGGCCGAAAGGCCGAA AUUGAUUC 2975 AGCUUGAA CUGAUGAGGCCGAAAGGCCGAA AGCUUCCA 2975 CAGGUGAG CUGAUGAGGCCGAAAGGCCGAA ACCAUAUA 2977 UCAGCUUG CUGAUGAGGCCGAAAGGCCGAA AGAGCUUC Table 11: Human IL-5 HH Target Sequence
nt. HH Target Sequence nt. HH Target Sequence Position Position
8 AUGCACU U UCUUUGC 245 AAGAAAU C UUUCAGG
9 UGCACUU U CUUUGCC 247 GAAAUCU U UCAGGGA
10 GCACUUU C UUUGCCA 248 AAAUCUU U CAGGGAA
12 ACUUUCU U UGCCAAA 249 AAUCUUU C AGGGAAU
13 CUUUCUU U GCCAAAG 257 AGGGAAU A GGCACAC
36 AGAACGU U UCAGAGC 273 GGAGAGU C AAACUGU
37 GAACGUU U CAGAGCC 291 AGGGGGU A CUGUGGA
38 AACGUUU C AGAGCCA 305 AAAGACU A UUCAAAA
56 GGAUGCU U CUGCAUU 307 AGACUAU U CAAAAAC
57 GAUGCUU C UGCAUUU 308 GACUAUU C AAAAACU
63 UCUGCAU U UGAGUUU 316 AAAAACU U GUCCUUA
64 CUGCAUU U GAGUUUG 319 AACUUGU C CUUAAUA
69 UUUGAGU U UGCUAGC 322 UUGUCCU U AAUAAAG
70 UUGAGUU U GCUAGCU 323 UGUCCUU A AUAAAGA
74 GUUUGCU A GCUCUUG 326 CCUUAAU A AAGAAAU
78 GCUAGCU C UUGGAGC 334 AAGAAAU A CAUUGAC
80 UAGCUCU U GGAGCUG 338 AAUACAU U GACGGCC
91 GCUGCCU A CGUGUAU 380 GGAGAGU A AACCAAU
97 UACGUGU A UGCCAUC 388 AACCAAU U CCUAGAC
104 AUGCCAU C CCCACAG 389 ACCAAUU C CUAGACU
116 CAGAAAU U CCCACAA 392 AAUUCCU A GACUACC
117 AGAAAUU C CCACAAG 397 CUAGACU A CCUGCAA
130 AGUGCAU U GGUGAAA 409 CAAGAGU U UCUUGGU
145 GAGACCU U GGCACUG 410 AAGAGUU U CUUGGUG
155 CACUGCU U UCUACUC 411 AGAGUUU C UUGGUGU
156 ACUGCUU U CUACUCA 413 AGUUUCU U GGUGUAA
157 CUGCUUU C UACUCAU 419 UUGGUGU A AUGAACA
159 GCUUUCU A CUCAUCG 437 AGUGGAU A AUAGAAA
162 UUCUACU C AUCGAAC 440 GGAUAAU A GAAAGUU
165 UACUCAU C GAACU CU 447 AGAAAGU U GAGACUA
171 UCGAACU C UGCUGAU 454 UGAGACU A AACUGGU
179 UGCUGAU A GCCAAUG 462 AACUGGU U UGUUGCA
192 UGAGACU C UGAGGAU 463 ACUGGUU U GUUGCAG
200 UGAGGAU U CCUGUUC 466 GGUUUGU U GCAGCCA
201 GAGGAUU C CUGUUCC 479 CAAAGAU U UUGGAGG
206 UUCCUGU U CCUGUAC 480 AAAGAUU U UGGAGGA
207 UCCUGUU C CUGUACA 481 AAGAUUU U GGAGGAG
212 UUCCUGU A CAUAAAA 497 AGGACAU U UUACUGC
216 UGUACAU A AAAAUCA 498 GGACAUU U UACUGCA
222 UAAAAAU C ACCAACU 499 GACAUUU U ACUGCAG 500 ACAUUUU A CUGCAGU 684 UACUUUU U UCUUAUU 531 AAAGAGU C AGGCCUU 685 ACUUUUU U CUUAUUU 538 CAGGCCU U AAUUUUC 686 CUUUUUU C UUAUUUA 539 AGGCCUU A AUUUUCA 688 UUUUUCU U AUUUAAC 542 CCUUAAU U UUCAAUA 689 UUUUCUU A UUUAACU 543 CUUAAUU U UCAAUAU 691 UUCUUAU U UAACUUA 544 UUAAUUU U CAAUAUA 692 UCUUAUU U AACUUAA 545 UAAUUUU C AAUAUAA 693 CUUAUUU A ACUUAAC 549 UUUCAAU A UAAUUUA 697 UUUAACU U AACAUUC 551 UCAAUAU A AUUUAAC 698 UUAACUU A ACAUUCU 554 AUAUAAU U UAACUUC 703 UUAACAU U CUGUAAA 555 UAUAAUU U AACUUCA 704 UAACAUU C UGUAAAA 556 AUAAUUU A ACUUCAG 708 AUUCUGU A AAAUGUC 560 UUUAACU U CAGAGGG 715 AAAAUGU C UGUUAAC 561 UUAACUU C AGAGGGA 719 UGUCUGU U AACUUAA 573 GGAAAGU A AAUAUUU 720 GUCUGUU A ACUUAAU 577 AGUAAAU A UUUCAGG 724 GUUAACU U AAUAGUA 579 UAAAUAU U UCAGGCA 725 UUAACUU A AUAGUAU 580 AAAUAUU U CAGGCAU 728 ACUUAAU A GUAUUUA 581 AAUAUUU C AGGCAUA 731 UAAUAGU A UUUAUGA 588 CAGGCAU A CUGACAC 733 AUAGUAU U UAUGAAA 597 UGACACU U UGCCAGA 734 UAGUAUU U AUGAAAU 598 GACACUU U GCCAGAA 735 AGUAUUU A UGAAAUG611 AAAGCAU A AAAUUCU 745 AAAUGGU U AAGAAUU616 AUAAAAU U CUUAAAA 746 AAUGGUU A AGAAUUU 617 UAAAAUU C UUAAAAU 752 UAAGAAU U UGGUAAA619 AAAUUCU U AAAAUAU 753 AAGAAUU U GGUAAAU 620 AAUUCUU A AAAUAUA 757 AUUUGGU A AAUUAGU 625 UUAAAAU A UAUUUCA 761 GGUAAAU U AGUAUUU 627 AAAAUAU A UUUCAGA 762 GUAAAUU A GUAUUUA 629 AAUAUAU U UCAGAUA 765 AAUUAGU A UUUAUUU 630 AUAUAUU U CAGAUAU 767 UUAGUAU U UAUUUAA 631 UAUAUUU C AGAUAUC 768 UAGUAUU U AUUUAAU636 UUCAGAU A UCAGAAU 769 AGUAUUU A UUUAAUG638 CAGAUAU C AGAAUCA 771 UAUUUAU U UAAUGUU644 UCAGAAU C AUUGAAG 772 AUUUAUU U AAUGUUA647 GAAUCAU U GAAGUAU 773 UUUAUUU A AUGUUAU653 UUGAAGU A UUUUCCU 778 UUAAUGU U AUGUUGU655 GAAGUAU U UUCCUCC 779 UAAUGUU A UGUUGUG656 AAGUAUU U UCCUCCA 783 GUUAUGU U GUGUUCU657 AGUAUUU U CCUCCAG 788 GUUGUGU U CUAAUAA658 GUAUUUU C CUCCAGG 789 UUGUGUU C UAAUAAA661 UUUUCCU C CAGGCAA 791 GUGUUCU A AUAAAAC672 GCAAAAU U GAUAUAC 794 UUCUAAU A AAACAAA676 AAUUGAU A UACUUUU 805 CAAAAAU A GACAACU678 UUGAUAU A CUUUUUU
681 AUAUACU U UUUUCUU
682 UAUACUU U UUUCUUA Table 12: Human IL-5 HH Ribozyme Sequences
nt . HH Ribozyme Sequence
Position
8 GCAAAGA CUGAUGAGGCCGAAAGGCCGAA AGUGCAU
9 GGCAAAG CUGAUGAGGCCGAAAGGCCGAA AAGUGCA 10 UGGCAAA CUGAUGAGGCCGAAAGGCCGAA AAAGUGC
12 UUUGGCA CUGAUGAGGCCGAAAGGCCGAA AGAAAGU
13 CUUUGGC CUGAUGAGGCCGAAAGGCCGAA AAGAAAG
36 GCUCUGA CUGAUGAGGCCGAAAGGCCGAA ACGUUCU
37 GGCUCUG CUGAUGAGGCCGAAAGGCCGAA AACGUUC
38 UGGCUCU CUGAUGAGGCCGAAAGGCCGAA AAACGUU
56 AAUGCAG CUGAUGAGGCCGAAAGGCCGAA AGCAUCC
57 AAAUGCA CUGAUGAGGCCGAAAGGCCGAA AAGCAUC
63 AAACUCA CUGAUGAGGCCGAAAGGCCGAA AUGCAGA
64 CAAACUC CUGAUGAGGCCGAAAGGCCGAA AAUGCAG
69 GCUAGCA CUGAUGAGGCCGAAAGGCCGAA ACUCAAA
70 AGCUAGC CUGAUGAGGCCGAAAGGCCGAA AACUCAA 74 CAAGAGC CUGAUGAGGCCGAAAGGCCGAA AGCAAAC 78 GCUCCAA CUGAUGAGGCCGAAAGGCCGAA AGCUAGC 80 CAGCUCC CUGAUGAGGCCGAAAGGCCGAA AGAGCUA 91 AUACACG CUGAUGAGGCCGAAAGGCCGAA AGGCAGC 97 GAUGGCA CUGAUGAGGCCGAAAGGCCGAA ACACGUA
104 CUGUGGG CUGAUGAGGCCGAAAGGCCGAA AUGGCAU
116 UUGUGGG CUGAUGAGGCCGAAAGGCCGAA AUUUCUG
117 CUUGUGG CUGADGAGGCCGAAAGGCCGAA AAUUUCU 130 UUUCACC CUGAUGAGGCCGAAAGGCCGAA AUGCACU 145 CAGUGCC CUGAUGAGGCCGAAAGGCCGAA AGGUCUC
155 GAGUAGA CUGAUGAGGCCGAAAGGCCGAA AGCAGUG
156 UGAGUAG CUGAUGAGGCCGAAAGGCCGAA AAGCAGU
157 AUGAGUA CUGAUGAGGCCGAAAGGCCGAA AAAGCAG 159 CGAUGAG CUGAUGAGGCCGAAAGGCCGAA AGAAAGC 162 GUUCGAU CUGAUGAGGCCGAAAGGCCGAA AGUAGAA 165 AGAGUUC CUGAUGAGGCCGAAAGGCCGAA AUGAGUA 171 AUCAGCA CUGAUGAGGCCGAAAGGCCGAA AGUUCGA 179 CAUUGGC CUGAUGAGGCCGAAAGGCCGAA AUCAGCA 192 AUCCUCA CUGAUGAGGCCGAAAGGCCGAA AGUCUCA
200 GAACAGG CUGAUGAGGCUGAAAGGCCGAA AUCCUCA
201 GGAACAG CUGAUGAGGCCGAAAGGCCGAA AAUCCUC
206 GUACAGG CUGAUGAGGCCGAAAGGCCGAA ACAGGAA
207 UGUACAG CUGAUGAGGCCGAAAGGCCGAA AACAGGA 212 UUUUAUG CUGAUGAGGCCGAAAGGCCGAA ACAGGAA 216 UGAUUUU CUGAUGAGGCCGAAAGGCCGAA AUGUACA 222 AGUUGGU CUGAUGAGGCCGAAAGGCCGAA AUUUUUA 245 CCUGAAA CUGAUGAGGCCGAAAGGCCCAA AUUUCUU 247 UCCCUGA CUGAUGAGGCCGAAAGGCCGAA AGAUUUC
248 UUCCCUG CUGAUGAGGCCGAAAGGCCGAA AAGAUUU
249 AUUCCCU CUGAUGAGGCCGAAAGGCCGAA AAAGAUU 257 GUGUGCC CUGAUGAGGCCGAAAGGCCGAA AUUCCCU 273 ACAGUUU CUGAUGAGGCCGAAAGGCCGAA ACUCUCC 291 UCCACAG CUGAUGAGGCCGAAAGGCCGAA ACCCCCU 305 UUUUGAA CUGAUGAGGCCGAAAGGCCGAA AGUCUUU
307 GUUUUUG CUGAUGAGGCCGAAAGGCCGAA AUAGUCU
308 AGUUUUU CUGAUGAGGCCGAAAGGCCGAA AAUAGUC 315 UAAGGAC CUGAUGAGGCCGAAAGGCCGAA AGUUUUU 319 UAUUAAG CUGAUGAGGCCGAAAGGCCGAA ACAAGUU
322 CUUUAUU CUGAUGAGGCCGAAAGGCCGAA AGGACAA
323 UCUUUAU CUGAUGAGGCCGAAAGGCCGAA AAGGACA 326 AUUUCUU CUGAUGAGGCCGAAAGGCCGAA AUUAAGG 334 GUCAAUG CUGAUGAGGCCGAAAGGCCGAA AUUUCUU 338 GGCCGUC CUGAUGAGGCCGAAAGGCCGAA AUGUAUU 380 AUUGGUU CUGAUGAGGCCGAAAGGCCGAA ACUCUCC
388 GUCUAGG CUGAUGAGGCCGAAAGGCCGAA AUUGGUU
389 AGUCUAGCUGAUGAGGCCGAAAGGCCGAA AAUUGGU 392 GGUAGUC CUGAUGAGGCCGAAAGGCCGAA AGGAAUU 397 UUGCAGG CUGAUGAGGCCGAAAGGCCGAA AGUCUAG
409 ACCAAGA CUGAUGAGGCCGAAAGGCCGAA ACUCUUG
410 CACCAAG CUGAUGAGGCCGAAAGGCCGAA AACUCUU
411 ACACCAA CUGAUGAGGCCGAAAGGCCGAA AAACUCU 413 UUACACCCUGAUGAGGCCGAAAGGCCGAA AGAAACU 419 UGUUCAU CUGAUGAGGCCGAAAGGCCGAA ACACCAA 437 UUUCUAU CUGAUGAGGCCGAAAGGCCGAA AUCCACU 440 AACUUUC CUGAUGAGGCCGAAAGGCCGAA AUUAUCC 447 UAGUCUC CUGAUGAGGCCGAAAGGCCGAA ACUUUCU 454 ACCAGUU CUGAUGAGGCCGAAAGGCCGAA AGUCUCA
462 UGCAACA CUGAUGAGGCCGAAAGGCCGAA ACCAGUU
463 CUGCAAC CUGAUGAGGCCGAAAGGCCGAA AACCAGU 466 UGGCUGC CUGAUGAGGCCGAAAGGCCGAA ACAAACC
479 CCUCCAA CUGAUGAGGCCGAAAGGCCGAA AUCUUUG
480 UCCUCCA CUGAUGAGGCCGAAAGGCCGAA AAUCUUU
481 CUCCUCC CUGAUGAGGCCGAAAGGCCGAA AAAUCUU
497 GCAGUAA CUGAUGAGGCCGAAAGGCCGAA AUGUCCU
498 UGCAGUA CUGAUGAGGCCGAAAGGCCGAA AAUGUCC
499 CUGCAGU CUGAUGAGGCCGAAAGGCCGAA AAAUGUC
500 ACUGCAG CUGAUGAGGCCGAAAGGCCGAA AAAAUGU 531 AAGGCCU CUGAUGAGGCCGAAAGGCCGAA ACUCUUU
538 GAAAAUU CUGAUGAGGCCGAAAGGCCGAA AGGCCUG
539 UGAAAAU CUGAUGAGGCCGAAAGGCCGAA AAGGCCU
542 UAUUGAA CUGAUGAGGCCGAAAGGCCGAA AUUAAGG
543 AUAUUGA CUGAUGAGGCCGAAAGGCCGAA AAUUAAG
544 UAUAUUG CUGAUGAGGCCGAAAGGCCGAA AAAUUAA
545 UUAUAUU CUGAUGAGGCCGAAAGGCCGAA AAAAUUA 549 UAAAUUA CUGAUGAGGCCGAAAGGCCGAA AUUGAAA 551 GUUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAUUGA 554 GAAGUUA CUGAUGAGGCCGAAAGGCCGAA AUUAUAU
555 UGAAGUU CUGAUGAGGCCGAAAGGCCGAA AAUUAUA
556 CUGAAGU CUGAUGAGGCCGAAAGGCCGAA AAAUUAU
560 CCCUCUG CUGAUGAGGCCGAAAGGCCGAA AGUUAAA
561 UCCCUCU CUGAUGAGGCCGAAAGGCCGAA AAGUUAA 573 AAAUAUU CUGAUGAGGCCGAAAGGCCGAA ACUUUCC 577 CCUGAAA CUGAUGAGGCCGAAAGGCCGAA AUUUACU
579 UGCCUGA CUGAUGAGGCCGAAAGGCCGAA AUAUUUA
580 AUGCCUG CUGAUGAGGCCGAAAGGCCGAA AAUAUUU
581 UAUGCCU CUGAUGAGGCCGAAAGGCCGAA AAAUAUU 588 GUGUCAG CUGAUGAGGCCGAAAGGCCGAA AUGCCUG
597 UCUGGCA CUGAUGAGGCCGAAAGGCCGAA AGUGUCA
598 UUCUGGC CUGAUGAGGCCGAAAGGCCGAA AAGUGUC 611 AGAAUUU CUGAUGAGGCCGAAAGGCCGAA AUGCUUU
616 UUUUAAG CUGAUGAGGCCGAAAGGCCGAA AUUUUAU
617 AUUUUAA CUGAUGAGGCCGAAAGGCCGAA AAUUUUA
619 AUAUUUU CUGAUGAGGCCGAAAGGCCGAA AGAAUUU
620 UAUAUUU CUGAUGAGGCCGAAAGGCCGAA AAGAAUU 625 UGAAAUA CUGAUGAGGCCGAAAGGCCGAA AUUUUAA 627 UCUGAAA CUGAUGAGGCCGAAAGGCCGAA AUAUUUU
629 UAUCUGA CUGAUGAGGCCGAAAGGCCGAA AUAUAUU
630 AUAUCUG CUGAUGAGGCCGAAAGGCCGAA AAUAUAU
631 GAUAUCU CUGAUGAGGCCGAAAGGCCGAA AAAUAUA 636 AUUCUGA CUGAUGAGGCCGAAAGGCCGAA AUCUGAA 638 UGAUUCU CUGAUGAGGCCGAAAGGCCGAA AUAUCUG 644 CUUCAAU CUGAUGAGGCCGAAAGGCCGAA AUUCUGA 647 AUACUUC CUGAUGAGGCCGAAAGGCCGAA AUGAUUC 653 AGGAAAA CUGAUGAGGCCGAAAGGCCGAA ACUUCAA
655 GGAGGAA CUGAUGAGGCCGAAAGGCCGAA AUACUUC
656 UGGAGGA CUGAUGAGGCCGAAAGGCCGAA AAUACUU
657 CUGGAGG CUGAUGAGGCCGAAAGGCCGAA AAAUACU
658 CCUGGAG CUGAUGAGGCCGAAAGGCCGAA AAAAUAC 661 UUGCCUG CUGAUGAGGCCGAAAGGCCGAA AGGAAAA 672 GUAUAUC CUGAUGAGGCCGAAAGGCCGAA AUUUUGC 676 AAAAGUA CUGAUGAGGCCGAAAGGCCGAA AUCAAUU 678 AAAAAAG CUGAUGAGGCCGAAAGGCCGAA AUAUCAA
681 AAGAAAA CUGAUGAGGCCGAAAGGCCGAA AGUAUAU
682 UAAGAAA CUGAUGAGGCCGAAAGGCCGAA AAGUAUA
683 AUAAGAA CUGAUGAGGCCGAAAGGCCGAA AAAGUAU
684 AAUAAGA CUGAUGAGGCCGAAAGGCCGAA AAAAGUA
685 AAAUAAG CUGAUGAGGCCGAAAGGCCGAA AAAAAGU
686 UAAAUAA CUGAUGAGGCCGAAAGGCCGAA AAAAAAG
688 GUUAAAU CUGAUGAGGCCGAAAGGCCGAA AGAAAAA
689 AGUUAAA CUGAUGAGGCCGAAAGGCCGAA AAGAAAA
691 UAAGUUA CUGAUGAGGCCGAAAGGCCGAA AUAAGAA
692 UUAAGUU CUGAUGAGGCCGAAAGGCCGAA AAUAAGA
693 GUUAAGU CUGAUGAGGCCGAAAGGCCGAA AAAUAAG
697 GAAUGUU CUGAUGAGGCCGAAAGGCCGAA AGUUAAA
698 AGAAUGU CUGAUGAGGCCGAAAGGCCGAA AAGUUAA 703 UUUACAG CUGAUGAGGCCGAAAGGCCGAA AUGUUAA
704 UUUUACA CUGAUGAGGCCGAAAGGCCGAA AAUGUUA
708 GACAUUU CUGAUGAGGCCGAAAGGCCGAA ACAGAAU
715 GUUAACA CUGAUGAGGCCGAAAGGCCGAA ACAUUUU
719 UUAAGUU CUGAUGAGGCCGAAAGGCCGAA ACAGACA
720 AUUAAGU CUGAUGAGGCCGAAAGGCCGAA AACAGAC
724 UACUAUU CUGAUGAGGCCGAAAGGCCGAA AGUUAAC
725 AUACUAU CUGAUGAGGCCGAAAGGCCGAA AAGUUAA
728 UAAAUAC CUGAUGAGGCCGAAAGGCCGAA AUUAAGU
731 UCAUAAA CUGAUGAGGCCGAAAGGCCGAA ACUAUUA
733 UUUCAUA CUGAUGAGGCCGAAAGGCCGAA AUACUAU
734 AUUUCAU CUGAUGAGGCCGAAAGGCCGAA AAUACUA
735 CAUUUCA CUGAUGAGGCCGAAAGGCCGAA AAAUACU
745 AAUUCUU CUGAUGAGGCCGAAAGGCCGAA ACCAUUU
746 AAAUUCU CUGAUGAGGCCGAAAGGCCGAA AACCAUU
752 UUUACCA CUGAUGAGGCCGAAAGGCCGAA AUUCUUA
753 AUUUACC CUGAUGAGGCCGAAAGGCCGAA AAUUCUU
757 ACUAAUU CUGAUGAGGCCGAAAGGCCGAA ACCAAAU
761 AAAUACU CUGAUGAGGCCGAAAGGCCGAA AUUUACC
762 UAAAUAC CUGAUGAGGCCGAAAGGCCGAA AAUUUAC
765 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACUAAUU
767 UUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUACUAA
768 AUUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUACUA
769 CAUUAAA CUGAUGAGGCCGAAAGGCCGAA AAAUACU
771 AACAUUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
772 UAACAUU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
773 AUAACAU CUGAUGAGGCCGAAAGGCCGAA AAAUAAA
778 ACAACAU CUGAUGAGGCCGAAAGGCCGAA ACAUUAA
779 CACAACA CUGAUGAGGCCGAAAGGCCGAA AACAUUA
783 AGAACAC CUGAUGAGGCCGAAAGGCCGAA ACAUAAC
788 UUAUUAG CUGAUGAGGCCGAAAGGCCGAA ACACAAC
789 UUUAUUA CUGAUGAGGCCGAAAGGCCGAA AACACAA
791 GUUUUAU CUGAUGAGGCCGAAAGGCCGAA AGAACAC
794 UUUGUUU CUGAUGAGGCCGAAAGGCCGAA AUUAGAA
805 AGUUGUC CUGAUGAGGCCGAAAGGCCGAA AUUUUUG
Table 13: Mouse IL-5 HH Ribozyme Target Sequence
nt. HH Target Sequence nt. HH Target Sequence Position Position
8 cGCuCUU c CUUUGCu 253 AGGGgcU A GaCAuAC
11 uCUUcCU U UGCugAA 259 UagACAU a CUGaAgA 12 CUUcCUU U GCugAAG 269 GaAGAaU C AAACUGU 36 GAAgacU U CAGAGuC 269 GaAGAaU c AAaCugU 36 GaAgAcU u cAgAGUc 269 GAAgaAU c aAAcUgU 37 AAgacUU C AGAGuCA 287 UGGGGGU A CUGUGGA 43 UcaGaGU c AUGAgaA 301 AAAugCU A UUCcAAA 58 GGAUGCU U CUGCAcU 301 AAAugCU a uUCCaaA 59 GAUGCUU C UGCAcUU 303 AUGCuAU u CCaAaAc 59 gAUGcUU c uGcAcUU 303 AugCUAU U CcAAAAC 66 CUGCAcU U GAGUgUu 304 ugCUAUU C cAAAACc 82 UgAcucU c aGcUGUG 315 AACcUGU C aUUAAUA 91 GcUgUGU c uggGCCA 318 cUGUCaU U AAUAAAG 112 ugGAgAU U CCCAugA 319 UGUCaUU A AUAAAGA 113 gGAgAUU C CCAugAG 322 CaUUAAU A AAGAAAU 141 GAGACCU U GaCACaG 330 AAGAAAU A CAUUGAC 141 GAgACcU U GaCAcAg 334 AAUACAU U GACcGCC 158 gUCcgCU C AcCGAgC 334 AAUaCaU u GACcgCC 167 cCGAgCU C UGuUGAc 384 AggCAgU U CCUgGAu 196 UGAGGcU U CCUGUcC 385 ggCAgUU c CUgGAuU 197 GAGGcUU C CUGUcCC 393 CUgGAuU A CCUGCAA 197 gAGGCuU c CUGuCcC 405 CAAGAGU U cCUUGGU 202 UUCCUGU c CCUacuC 406 AAGAGUU c CUUGGUG 202 UUCCUGU c CcUAcuc 409 AGUUcCU U GGUGUgA 206 UGUCccU a cuCaUAA 481 UcaCAAU u UAAgUUA 212 UACUCAU a aAAaUCa 482 cAcAAUU U AAgUUaA 212 UacuCAU A AAAAUCA 483 AcAAUUU A AgUUaAa 218 UaaAaaU c aCcAGCU 483 AcAAUuU a aGUUAAa 218 UAAAAAU C ACCAgCU 495 AAAUUgU c AAcAgAU 218 uAAAAAU c acCAgCU 553 GCUGuuU c CaUuUAU 232 uaUGCAU U GGaGAAA 557 UuUcCAU U UauaUUU 241 gAGAAAU C UUUCAGG 564 UUauAuU u aUgUCCU 241 gAgAaAU c UUucAGG 564 UUAuaUU u AugUcCU 241 gagAAAU c UUUCAGG 565 uaUAUUU a ugUCCuG 241 gAgAaAU c UUUCAGg 565 UAUAuUU a UgUCcUg 243 gaAAucU U UCAGgGg 569 UUuAUGU c cUGUaGU 243 GAAAUCU U UCAGGGg 569 UUUAUGU c cUGUagU 244 AAAUCUU U CAGGGgc 613 AAAGuGU u uaaCCUU 245 AAUCUUU C AGGGgcU 614 AAgUGuU u aACcUUU 620 UUAACcU u uUuGUAU 1407 cCAgUUU A CUcCAGg 793 caAGgCU u UGuGcAU 1407 ccAgUUU a CUCCAGG 816 CUGagUU a UACUCcc 1410 gUUUaCU C CAGGaAA 818 GAguUAU a cUCCcuC 1434 AUgCUUU U aUuUaAU 825 ACUcCcU c CccCUCA 1434 aUgcUuU U AUUUAAu 825 aCUccCU c CcCcUCa 1434 aUgcuUU u AuUUAAU
839 AuCcucU U cGUUGCA 1435 UgCUUUU a UuUaAUU
840 uCcucUU c GUUGCAu 1435 ugcUUUU a uUUAaUU
863 cAAgUAU U cCAGGCu 1438 UuUUAUU U AAuUcug
864 AAgUAUU c CAGGCug 1438 uUUUAUU U AAUucUg 864 AAGUAUU c caggCug 1439 UUUAUUU A AUucUgU 913 gAaCUCU U GGucCaG 1443 UUUaAuU c UGuaAGa 917 UcUuggU c CAGAuGG 1447 AUUCUGU A AgAUGUu 957 UUagcAU c CUUUcUc 1458 ugUUcaU a UUAUUUA 960 GCAuccU u UcUcCuA 1458 ugUUcAU A uUAUUUA 960 GcaUcCU u uCUCcUa 1460 UucAUAU u AUUUAug 962 AUcCuuU c UCcUaGC 1461 UcAUAuU A UUUAUGA 975 gcccCUU u AgAUAgA 1463 AUAuUAU U UAUGAug 987 aGaOGAU A cuuAAUG 1475 AuGgAUU c aGUAAgU 990 UGAuACU u AAugacU 1479 AUUcaGU A AgUUAaU
1000 UGACuCU c UugCuGA 1483 aGuAAGU u AAUAUUU
1027 CgggGCU U cCUgCUC 1483 aGUAAgU U AaUAUUU
1034 UCCUGcU C CUaUcuA 1484 GUAAgUU A aUAUUUA
1037 UgcUCcU A UcUAACU 1487 agUUAAU a UUuAuUA
1039 cUccuAU c UAACUUC 1487 AgUUAaU A UUUAUUa
1039 cUCcUAU c UAACUUc 1489 UUAAUaU U uAuUAcA
1041 CcUAUcU A ACUUcAa 1489 UUAAuAU u UAUUaCA
1051 UUcAAuU U AAuAccC 1489 UUAaUAU U UAUUacA 1148 uGAcUUU u cUuaUGU 1490 UAAUaUU u AuUAcAc
1213 GCUgGaU u UUGGAaa 1490 UAaUAUU U AUuAcAc
1213 gcUGGAU u uUgGAAA 1490 UAaUAUU U AUUacAc
1214 cugGAUU U UGGAaaA 1491 AAUAUUU a uuaCAcg
1215 ugGAUUU U GGAaaAG 1491 AAUAUuU a UuAcAcg 1234 gGGACAU c UccuUGC 1491 AaUAUUU A UuAcAcG 1236 GACAUcU c cuUGCAG 1491 AaUAUUU A UUacAcG
1275 ugGGCCU U AcUUcUC 1494 AUuUAUU a CAcgUAU
1276 gGGCCUU A cUUcUCc 1502 cACGUaU A UaauAUu 1280 CUUAcUU c UCcgUgU 1502 cAcgUAU a UAAUaUU 1298 UgAACUU a AGAaGcA 1507 AUAUAaU a UUcUaaU 1310 gcAAAGU a aAuACcA 1509 AUAAuAU U CUaAuAA 1310 GCAAAgU a aAUAcca 1509 aUaaUaU U CUAAUAA 1310 GcaAAgU a AAUAccA 1510 UAAuAUU C UaAuAAa 1350 AAAGCAU A AAAUggU 1510 UAAuAUU C UaauAAA 1358 AAAUGGU U ggGAugU 1510 UAAuAuU c UaaUAAA 1370 UgUuaUU C AGgUAUC 1510 UaaUaUU C UAAUAAA 1375 UUCAGgU A UCAGggU 1512 aUaUUCU A AUAAAgC 1377 CAGgUAU C AGggUCA 1515 UUCUAAU A AAgCAgA 1383 UCAGggU C AcUGgAG
1405 cccCAgU U UACUcCA
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Table 17
Mouse rel A HH Target sequence
nt. Position HH Target Sequence nt. Position HH Target Sequence
19 AAUGGCU a caCaGgA 467 cCAGGCU c cuguUCg 22 aGCUCcU a cGUgGUG 469 AaGCcAU u AGcCAGC 26 CcUCcaU u GcGgACa 473 UuUgAGU C AGauCAg 93 GAuCUGU U uCCCCUC 481 AGCGaAU C CAGACCA 94 AuCUGUU u CCCCUCA 501 AACCCCU U uCAcGUU 100 UuCCCCU C AUCUUuC 502 ACCCCUU u CAcGUUC 103 CCCUCAU C UUuCCcu 508 UuCAcGU U CCUAUAG 105 CUCAUCU U uCCcuCA 509 uCAcGUU C CUAUAGA 106 UCAUCUU u CCcuCAG 512 cGUUCCU A UAGAgGA 129 CAGGCuU C UGGgCCu 514 UUCCUAU A GAgGAGC 138 GGgCCuU A UGUGGAG 534 GGGGACU A uGACuUG 148 UGGAGAU C AUcGAaC 556 UGCGcCU C UGCUUCC 151 AGAUCAU c GAaCAGC 561 CUCUGCU U CCAGGUG 180 AUGCGaU U CCGCUAu 562 UCUGCUU C CAGGUGA 181 UGCGaUU C CGCUAuA 585 aAgCCAU u AGcCAGc 186 UUCCGCU A uAAaUGC 598 GGCCCCU C CuCCUGa 204 GGGCGCU C aGCGGGC 613 CcCCUGU C CUcuCaC 217 GCAGuAU u CCuGGCG 616 CUGUCCU c uCaCAUC 239 CACAGAU A CCACCAA 617 gucCCUU C CUCAgCC 262 CCACCAU C AAGAUCA 620 CCUUCCU C AgCCaug 268 UCAAGAU C AAUGGCU 623 UCCUgcU u CCAUCUc 276 AAUGGCU A CACAGGA 628 AUCCgAU u UUUGAuA 301 UuCGaAU C UCCCUGG 630 CCgAUuU U UGAuAAc 303 CGaAUCU C CCUGGUC 631 CgAUuUU U GAuAAcC 310 CCCUGGU C ACCAAGG 638 UGgCcAU u GUGuuCC 323 GGcCCCU C CUCcuga 661 CCGAGCU C AAGAUCU 326 uCCaCCU C ACCGGCC 667 UCAAGAU C UGCCGAG 335 CCGGCCU C AuCCaCA 687 CGgAACU C UGGgAGC 349 AuGAaCU U GUgGGgA 700 GCUGCCU C GGUGGGG 352 AGaUcaU c GaAcAGc 715 AUGAGAU C UUCuUgC 375 GAUGGCU a CUAUGAG 717 GAGAUCU U CuUgCUG 376 AUGGucU C UccGgaG 713 AGAUCUU C uUgCUGU 378 GGCUaCU A UGAGGCU 721 UucUCCU c CauUGcG 391 CUGAcCU C UGCCCaG 751 AaGACAU U GAGGUGU 409 GCaGuAU C CAuAGcU 759 GAGGUGU A UUUCACG 416 CCgCAGU a UCCAuAg 761 GGUGUAU U UCACGGG 417 CAuAGcU U CCAGAAC 762 GUGUAUU U CACGGGA 418 AuAGcUU C CAGAACC 763 UGUAUUU C ACGGGAC 433 UGGGgAU C CAGUGUG 792 CGAGGCU C CUUUUCu 795 GGCUCCU U UUCuCAA 1167 GAUGAGU U UuCCcCC 796 GCUCCUU U UCuCAAG 1163 AUGAGUU U uCCcCCA 797 CUCCUUU U CuCAAGC 1169 UGAGUUU u CCcCCAU 798 UCCUUUU C uCAAGCU 1182 AUGcUGU U aCCaUCa 829 UGGCCAU U GUGUUCC 1183 UGcUGUU a CCaUCaG 834 AUUGUGU U CCGGACu 1184 GGccccU C CUcCUGa
835 UUGUGUU C CGGACuC 1187 GUccCuU c CUcaGCc
845 GACuCCU C CgUACGC 1188 UUaCCaU C aGGGCAG
849 CCUCCgU A CGCcGAC 1198 GGgAGuU u AGuCuGa
872 CCAGGCU C CUGUuCG 1209 CAGcCCU a caCCUUc
883 UuCGaGU C UCCAUGC 1215 cuGGCCU U aGCaCCG
885 CGaGUCU C CAUGCAG 1229 GGuCCCU u CCucAGc
905 GCGGCCU U CuGAuCG 1237 CCCAgcU C CUGCCCC
906 CGGCCUU C uGAuCGc 1250 CCAGcCU C CAGgCuC
919 GcGAGCU C AGUGAGC 1268 CCCaGCU C CuGCCcc
936 AUGGAgU U CCAGUAC 1279 CCAUGGU c cCuuCcu
937 UGGAgUU C CAGUACu 1281 gUGGgcU C AGCUgcG
942 UUCCAGU A CuUGCCA 1286 AUgAGuU u UccCCCA
953 GCCucAU c CAcAuGA 1309 CuCCUGU u CgAGUCu
962 AGAuGAU C GcCACCG 1315 cCCCAGU u CUAaCCC
965 CagUacU u gCCaGAc 1318 CAGUuCU A aCCCCgG
973 ACCGGAU U GAaGAGA 1331 gGGuCCU C CcCAGuC
986 GAgACcU u cAAGagu 1334 CuuUuCU C AaGCUGa
996 AGGACcU A UGAGACC 1389 ACGCUGU C gGAaGCC
1005 GAGACCU U CAAGAGu 1413 CUGCAGU U UGAUGcU
1006 AGACCUU C AAGAGuA 1414 UGCAGUU U GAUGcUG
1015 AGAGuAU C AUGAAGA 1437 GGGGCCU U GCUUGGC
1028 GAAGAGU C CUUUCAa 1441 CCUUGCU U GGCAACA
1031 GAGUCCU U UCAauGG 1467 GgaGUGU U CACAGAC
1032 AGUCCUU U CAauGGA 1468 gaGUGUU C ACAGACC
1033 GUCCUUU C AauGGAC 1482 CUGGCAU C uGUgGAC
1058 CCGGCCU C CAaCcCG 1486 CuUCgGU a GggAACU
1064 UaCACCU u GAucCAa 1494 GACAACU C aGAGUUU
1072 GgCGuAU U GCUGUGC 1500 UCaGAGU U UCAGCAG
1082 UGUGCCU a CCCGaAa 1501 CaGAGUU U CAGCAGC
1083 aaGCCUU C CCGaAGu 1502 aGAGUUU C AGCAGCU
1092 CGaAaCU C AaCUUCU 1525 gGuGCAU c CCUGUGu
1097 CUCAaCU U CUGUCCC 1566 AUGGAGU A CCCUGAa
1098 UCAaCUU C UGOCCCC 1577 UGAaGCU A UAACUCG
1102 CUUCUGU C CCCAAGC 1579 AaGCUAU A ACUCGCC
1125 CAGCCCU A caCCUUc 1583 UAUAACU C GCCUgGU
1127 GCCaUAU a gCcUUAC 1588 CUCuCCU A GaGAggG
1131 cAUCCCU c agCacCA 1622 CCCAGCU C CUGCcCC
1132 AcaCCUU c cCagCAU 1628 UCCUGCU u CggUaGG
1133 UCCaUcU c CagCuUC 1648 CGGGGCU U CCCAAUG
1137 UUUACuU u AgCgCgc 1660 cUGaCCU C ugccCAG
1140 cCagCAU C CCUcAGC 1663 cuCUgCU U cCAGGuG
1153 GCACCAU C AACUuUG 1664 uCUgCUU c CAGGuGA
1158 AUCAACU U UGAUGAG 1665 CUCgcUU u cGGAGgU
1680 GAAGACU U CUCCUCC
1681 AAGACUU C UCCUCCA
1683 GACUUCU C CUCCAUU
1686 UUCUCCU C CAUUGCG
1690 CCUCCAU U GCGGACA 1704 AUGGACU U CUCuGCu
1705 UGGACUU C UCuGCuC
1707 GACUUCU C uGCuCUu
1721 UUUGAGU C AGAUCAG
1726 GUCAGAU C AGCUCCU
1731 AUCAGCU C CUAAGGu
1734 AGCUCCU A AGGuGcU
1754 CaGugCU C CCaAGAG
Table 18
Human rel A HH Target Sequences
nt. Position HH Target Sequence nt. Position HH Target Sequence
19 AAUGGCU C GUCUGUA 467 GCAGGCU A UCAGUCA
22 GGCUCGU C UGUAGUG 469 AGGCUAU C AGUCAGC
26 CGUCUGU A GUGCACG 473 UAUCAGU C AGCGCAU
93 GAACUGU U CCCCCUC 481 AGCGCAU C CAGACCA
94 AACUGUU C CCCCUCA 501 AACCCCU U CCAAGUU
100 UCCCCCU C AUCUUCC 502 ACCCCUU C CAAGUUC
103 CCCUCAU C UUCCCGG 508 UCCAAGU U CCUAUAG
105 CUCAUCU U CCCGGCA 509 CCAAGUU C CUAUAGA
106 UCAUCUU C CCGGCAG 512 AGUUCCU A UAGAAGA
129 CAGGCCU C UGGCCCC 514 UUCCUAU A GAAGAGC
138 GGCCCCU A UGUGGAG 534 GGGGACU A CGACCUG
148 UGGAGAU C AUUGAGC 556 UGCGGCU C UGCUUCC
151 AGAUCAU U GAGCAGC 561 CUCUGCU U CCAGGUG
180 AUGCGCU U CCGCUAC 562 UCUGCUU C CAGGUGA
181 UGCGCUU C CGCUACA 585 GACCCAU C AGGCAGG
186 UUCCGCU A CAAGUGC 598 GGCCCCU C CGCCUGC
204 GGGCGCU C CGCGGGC 613 CGCCUGU C CUUCCUC
217 GCAGCAU C CCAGGCG 616 CUGUCCU U CCUCAUC
239 CACAGAU A CCACCAA 617 UGUCCUU C CUCAUCC
262 CCACCAU C AAGAUCA 620 CCUUCCU C AUCCCAU
268 UCAAGAU C AAUGGCU 623 UCCUCAU C CCAUCUU
276 AAUGGCU A CACAGGA 628 AUCCCAU C UUUGACA
301 UGCGCAU C UCCCUGG 630 CCCAUCU U UGACAAU
303 CGCAUCU C CCUGGUC 631 CCAUCUU U GACAAUC
310 CCCUGGU C ACCAAGG 638 UGACAAU C GUGCCCC
323 GGACCCU C CUCACCG 661 CCGAGCU C AAGAUCU
326 CCCUCCU C ACCGGCC 667 UCAAGAU C UGCCGAG
335 CCGGCCU C ACCCCCA 687 CGAAACU C UGGCAGC
349 AGGAGCU U GUAGGAA 700 GCUGCCU C GGUGGGG
352 AGCUUGU A GGAAAGG 715 AUGAGAU C UUCCUAC
375 GAUGGCU U CUAUGAG 717 GAGAUCU U CCUACUG
376 AUGGCUU C UAUGAGG 718 AGAUCUU C CUACUGU
378 GGCUUCU A UGAGGCU 721 UCUUCCU A CUGUGUG
391 CUGAGCU C UGCCCGG 751 AGGACAU U GAGGUGU
409 GCUGCAU C CACAGUU 759 GAGGUGU A UUUCACG
416 CCACAGU U UCCAGAA 761 GGUGUAU U UCACGGG
417 CACAGUU U CCAGAAC 762 GUGUAUU U CACGGGA
418 ACAGUUU C CAGAACC 763 UGUAUUU C ACGGGAC
433 UGGGAAU C CAGUGUG 792 CGAGGCU C CUUUUCG
795 GGCUCCU U UUCGCAA 1167 GAUGAGU U UCCCACC
796 GCUCCUU U UCGCAAG 1168 AUGAGUU U CCCACCA
797 CUCCUUU U CGCAAGC 1169 UGAGUUU C CCACCAU
798 UCCUUUU C GCAAGCU 1182 AUGGUGU U UCCUUCU
829 UGGCCAU U GUGUUCC 1183 UGGUGUU U CCUUCUG
834 AUUGUGU U CCGGACC 1184 GGUGUUU C CUUCUGG 835 UUGUGUU C CGGACCC 1187 GUUUCCU U CUGGGCA
845 GACCCCU C CCUACGC 1188 UUUCCUU C UGGGCAG
849 CCUCCCU A CGCAGAC 1198 GGCAGAU C AGCCAGG
872 GCAGGCU C CUGUGCG 1209 CAGGCCU C GGCCUUG
883 UGCGUGU C UCCAUGC 1215 UCGGCCU U GGCCCCG
885 CGUGUCU C CAUGCAG 1229 GGCCCCU C CCCAAGU
905 GCGGCCU U CCGACCG 1237 CCCAAGU C CUGCCCC
906 CGGCCUU C CGACCGG 1250 CCAGGCU C CAGCCCC
919 GGGAGCU C AGUGAGC 1268 CCCUGCU C CAGCCAU
936 AUGGAAU U CCAGUAC 1279 CCAUGGU A UCAGCUC
937 UGGAAUU C CAGUACC 1281 AUGGUAU C AGCUCUG
942 UUCCAGU A CCUGCCA 1286 AUCAGCU C UGGCCCA
953 GCCAGAU A CAGACGA 1309 CCCCUGU C CCAGUCC
962 AGACGAU C GUCACCG 1315 UCCCAGU C CUAGCCC
965 CGAUCGU C ACCGGAU 1318 CAGUCCU A GCCCCΑG
973 ACCGGAU U GAGGAGA 1331 AGGCCCU C CUCAGGC
986 GAAACGU A AAAGGAC 1334 CCCUCCU C AGGCUGU
996 AGGACAU A UGAGACC 1389 ACGCUGU C AGAGGCC
1005 GAGACCU U CAAGAGC 1413 COGCAGU U UGAUGAU
1006 AGACCUU C AAGAGCA 1414 UGCAGUU U GAUGAUG
1015 AGAGCAU C AUGAAGA 1437 GGGGCCU U GCUUGGC
1028 GAAGAGU C CUUUCAG 1441 CCUUGCU U GGCAACA
1031 GAGUCCU U UCAGCGG 1467 GCUGUGU U CACAGAC
1032 AGUCCUU U CAGCGGA 1468 CUGUGUU C ACAGACC
1033 GUCCUUU C AGCGGAC 1482 CUGGCAU C CGUCGAC
1058 CCGGCCU C CACCUCG 1486 CAUCCGU C GACAACU
1064 UCCACCU C GACGCAU 1494 GACAACU C CGAGUUU
1072 GACGCAU U GCUGUGC 1500 UCCGAGU U UCAGCAG
1082 UGUGCCU U CCCGCAG 1501 CCGAGUU U CAGCAGC
1083 GUGCCUU C CCGCAGC 1502 CGAGUUU C AGCAGCU
1092 CGCAGCU C AGCUUCU 1525 AGGGCAU A CCUGUGG
1097 CUCAGCU U COGUCCC 1566 AUGGAGU A CCCUGAG
1098 UCAGCUU C UGUCCCC 1577 UGAGGCU A UAACUCG
1102 CUUCUGU C CCCAAGC 1579 AGGCUAU A ACUCGCC
1125 CAGCCCU A UCCCUUU 1583 UAUAACU C GCCUAGU
1127 GCCCUAU C CCUUUAC 1588 CUCGCCU A GUGACAG
1131 UAUCCCU U UACGUCA 1622 CCCAGCU C CUGCUCC
1132 AUCCCUU U ACGUCAU 1628 UCCUGCU C CACUGGG
1133 UCCCUUU A CGUCAUC 1648 CGGGGCU C CCCAAUG
1137 UUUACGU C AUCCCUG 1660 AUGGCCU C CUUUCAG
1140 ACGUCAU C CCUGAGC 1663 GCCUCCU U UCAGGAG
1153 GCACCAU C AACUAUG 1664 CCUCCUU U CAGGAGA
1158 AUCAACU A UGAUGAG 1665 CUCCUUU C AGGAGAU
1680 GAAGACU U CUCCUCC
1681 AAGACUU C UCCUCCA
1683 GACUUCU C CUCCAUU
1686 UUCUCCU C CAUUGCG
1690 CCUCCAU U GCGGACA
1704 AUGGACU U CUCAGCC 1705 UGGACUU C UCAGCCC
1707 GACUUCU C AGCCCUG
1721 GCUGAGU C AGAUCAG
1726 GUCAGAU C AGCUCCU
1731 AUCAGCU C CUAAGGG
1734 AGCUCCU A AGGGGGU
1754 CUGCCCU C CCCAGAG
Table 19
Mouse rel A HH -Ribozyme Sequences
nt HH Ribozyme Sequence
Sequence
19 UCCUGUG CUGAUGAGGCCGAAAGGCCGAA AGCCAUU
22 CACCACG CUGAUGAGGCCGAAAGGCCGAA AGGAGCU
26 UGUCCGC CUGAUGAGGCCGAAAGGCCGAA AUGGAGG
93 GAGGGGA CUGAUGAGGCCGAAAGGCCGAA ACAGAUC
94 UGAGGGG CUGAUGAGGCCGAAAGGCCGAA AACAGAU 100 GAAAGAU CUGAUGAGGCCGAAAGGCCGAA AGGGGAA 103 AGGGAAA CUGAUGAGGCCGAAAGGCCGAA AUGAGGG
105 UGAGGGA CUGAUGAGGCCGAAAGGCCGAA AGAUGAG
106 CUGAGGG CUGAUGAGGCCGAAAGGCCGAA AAGAUGA 129 AGGCCCA CUGAUGAGGCCGAAAGGCCGAA AAGCCUG 138 CUCCACA CUGAUGAGGCCGAAAGGCCGAA AAGGCCC 148 GUUCGAU CUGAUGAGGCCGAAAGGCCGAA AUCUCCA 151 GCUGUUC CUGAUGAGGCCGAAAGGCCGAA AUGAUCU
180 AUAGCGG CUGAUGAGGCCGAAAGGCCGAA AUCGCAU
181 UAUAGCG CUGAUGAGGCCGAAAGGCCGAA AAUCGCA 186 GCAUUUA CUGAUGAGGCCGAAAGGCCGAA AGCGGAA 204 GCCCGCU CUGAUGAGGCCGAAAGGCCGAA AGCGCCC 217 CGCCAGG CUGAUGAGGCCGAAAGGCCGAA AUACUGC 239 UUGGUGG CUGAUGAGGCCGAAAGGCCGAA AUCUGUG 262 UGAUCUU CUGAUGAGGCCGAAAGGCCGAA AUGGUGG 268 AGCCAUU CUGAUGAGGCCGAAAGGCCGAA AUCUUGA 276 UCCUGUG CUGAUGAGGCCGAAAGGCCGAA AGCCAUU 301 CCAGGGA CUGAUGAGGCCGAAAGGCCGAA AUUCGAA 303 GACCAGG CUGAUGAGGCCGAAAGGCCGAA AGAUUCG 310 CCUUGGU CUGAUGAGGCCGAAAGGCCGAA ACCAGGG 323 UCAGGAG CUGAUGAGGCCGAAAGGCCGAA AGGGGCC 326 GGCCGGU CUGAUGAGGCCGAAAGGCCGAA AGGUGGA 335 UGUGGAU CUGAUGAGGCCGAAAGGCCGAA AGGCCGG 349 UCCCCAC CUGAUGAGGCCGAAAGGCCGAA AGUUCAU 352 GCUGUUC CUGAUGAGGCCGAAAGGCCGAA AUGAUCU
375 CUCAUAG CUGAUGAGGCCGAAAGGCCGAA AGCCAUC
376 CUCCGGA CUGAUGAGGCCGAAAGGCCGAA AGACCAU 378 AGCCUCA CUGAUGAGGCCGAAAGGCCGAA AGUAGCC 391 CUGGGCA CUGAUGAGGCCGAAAGGCCGAA AGGUCAG 409 AGCUAUG CUGAUGAGGCCGAAAGGCCGAA AUACUGC
416 CUAUGGA CUGAUGAGGCCGAAAGGCCGAA ACUGCGG
417 GUUCUGG CUGAUGAGGCCGAAAGGCCGAA AGCUAUG
418 GGUUCUG CUGAUGAGGCCGAAAGGCCGAA AAGCUAU 433 CACACUG CUGAUGAGGCCGAAAGGCCGAA AUCCCCA 467 CGAACAG CUGAUGAGGCCGAAAGGCCGAA AGCCUGG 469 GCUGGCU CUGAUGAGGCCGAAAGGCCGAA AUGGCUU 473 CUGAUCU CUGAUGAGGCCGAAAGGCCGAA ACUCAAA 481 UGGUCUG CUGAUGAGGCCGAAAGGCCGAA AUUCGCU 501 AACGUGA CUGAUGAGGCCGAAAGGCCGAA AGGGGUU
502 GAACGUG CUGAUGAGGCCGAAAGGCCGAA AAGGGGU
508 CUAUAGG CUGAUGAGGCCGAAAGGCCGAA ACGUGAA
509 UCUAUAG CUGAUGAGGCCGAAAGGCCGAA AACGUGA 512 UCCUCUA CUGAUGAGGCCGAAAGGCCCAA AGGAACG 514 GCUCCUC CUGAUGAGGCCGAAAGGCCGAA AUAGGAA 534 CAAGUCA CUGAUGAGGCCGAAAGGCCGAA AGUCCCC 556 GGAAGCA CUGAUGAGGCCGAAAGGCCGAA AGGCGCA
561 CACCUGG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
562 UCACCUG CUGAUGAGGCCGAAAGGCCGAA AAGCAGA 585 GCUGGCU CUGAUGAGGCCGAAAGGCCGAA AUGGCUU 598 UCAGGAG CUGAUGAGGCCGAAAGGCCGAA AGGGGCC 613 GUGAGAG CUGAUGAGGCCGAAAGGCCGAA ACAGGGG
616 GAUGUGA CUGAUGAGGCCGAAAGGCCGAA AGGACAG
617 GGCUGAG CUGAUGAGGCCGAAAGGCCGAA AAGGGAC 620 CAUGGCU CUGAUGAGGCCGAAAGGCCGAA AGGAAGG 623 GAGAUGG CUGAUGAGGCCGAAAGGCCGAA AGCAGGA 628 UAUCAAA CUGAUGAGGCCGAAAGGCCGAA AUCGGAU
630 GUUAUCA CUGAUGAGGCCGAAAGGCCGAA AAAUCGG
631 GGUUAUC CUGAUGAGGCCGAAAGGCCGAA AAAAUCG 638 GGAACAC CUGAUGAGGCCGAAAGGCCGAA AUGGCCA 661 AGAUCUU CUGAUGAGGCCGAAAGGCCGAA AGCUCGG 667 CUCGGCA CUGAUGAGGCCGAAAGGCCGAA AUCUUGA 687 GCUCCCA CUGAUGAGGCCGAAAGGCCGAA AGUUCCG 700 CCCCACC CUGAUGAGGCCGAAAGGCCGAA AGGCAGC 715 GCAAGAA CUGAUGAGGCCGAAAGGCCGAA AUCUCAU
717 CAGCAAG CUGAUGAGGCCGAAAGGCCGAA AGAUCUC
718 ACAGCAA CUGAUGAGGCCGAAAGGCCGAA AAGAUCU 721 CGCAAUG CUGAUGAGGCCGAAAGGCCGAA AGGAGAA 751 ACACCUC CUGAUGAGGCCGAAAGGCCGAA AUGUCUU 759 CGUGAAA CUGAUGAGGCCGAAAGGCCGAA ACACCUC
761 CCCGUGA CUGAUGAGGCCGAAAGGCCGAA AUACACC
762 UCCCGUG CUGAUGAGGCCGAAAGGCCGAA AAUACAC
763 GUCCCGU CUGAUGAGGCCGAAAGGCCGAA AAAUACA 792 AGAAAAG CUGAUGAGGCCGAAAGGCCGAA AGCCUCG
795 UUGAGAA CUGAUGAGGCCGAAAGGCCGAA AGGAGCC
796 CUUGAGA CUGAUGAGGCCGAAAGGCCGAA AAGGAGC
797 GCUUGAG CUGAUGAGGCCGAAAGGCCGAA AAAGGAG
798 AGCUUGA CUGAUGAGGCCGAAAGGCCGAA AAAAGGA 829 GGAACAC CUGAUGAGGCCGAAAGGCCGAA AUGGCCA
834 AGUCCGG CUGAUGAGGCCGAAAGGCCGAA ACACAAU
835 GAGUCCG CUGAUGAGGCCGAAAGGCCGAA AACACAA 845 GCGUACG CUGAUGAGGCCGAAAGGCCGAA AGGAGUC 849 GUCGGCG CUGAUGAGGCCGAAAGGCCGAA ACGGAGG 872 CGAACAG CUGAUGAGGCCGAAAGGCCGAA AGCCUGG 883 GCAUGGA CUGAUGAGGCCGAAAGGCCGAA ACUCGAA 885 CUGCAUG CUGAUGAGGCCGAAAGGCCGAA AGACUCG
905 CGAUCAG CUGAUGAGGCCGAAAGGCCGAA AGGCCGC
906 GCGAUCA CUGAUGAGGCCGAAAGGCCGAA AAGGCCG 919 GCUCACU CUGAUGAGGCCGAAAGGCCGAA AGCUCGC
936 GUACUGG CUGAUGAGGCCGAAAGGCCGAA ACUCCAU
937 AGUACUG CUGAUGAGGCCGAAAGGCCGAA AACUCCA 942 UGGCAAG CUGAUGAGGCCGAAAGGCCGAA ACUGGAA 953 UCAUGUG CUGAUGAGGCCGAAAGGCCGAA AUGAGGC 962 CGGUGGC CUGAUGAGGCCGAAAGGCCGAA AUCAUCU 965 GUCUGGC CUGAUGAGGCCGAAAGGCCGAA AGUACUG 973 UCUCUUC CUGAUGAGGCCGAAAGGCCGAA AUCCGGU 986 ACUCUUG CUGAUGAGGCCGAAAGGCCGAA AGGUCUC 996 GGUCUCA CUGAUGAGGCCGAAAGGCCGAA AGGUCCU
1005 ACUCUUG CUGAUGAGGCCGAAAGGCCGAA AGGUCUC
1006 UACUCUU CUGAUGAGGCCGAAAGGCCGAA AAGGUCU 1015 UCUUCAU CUGAUGAGGCCGAAAGGCCGAA AUACUCU 1028 UUGAAAG CUGAUGAGGCCGAAAGGCCGAA ACUCUUC
1031 CCAUUGA CUGAUGAGGCCGAAAGGCCGAA AGGACUC
1032 UCCAUUG CUGAUGAGGCCGAAAGGCCGAA AAGGACU
1033 GUCCAUU CUGAUGAGGCCGAAAGGCCGAA AAAGGAC 1058 CGGGUUG CUGAUGAGGCCGAAAGGCCGAA AGGCCGG 1064 UUGGAUC CUGAUGAGGCCGAAAGGCCGAA AGCUGUA 1072 GCACAGC CUGAUGAGGCCGAAAGGCCGAA AUACGCC
1082 UUUCGGG CUGAUGAGGCCGAAAGGCCGAA AGGCACA
1083 ACUUCGG CUGAUGAGGCCGAAAGGCCGAA AAGGCUU 1092 AGAAGUU CUGAUGAGGCCGAAAGGCCGAA AGUUUCG
1097 GGGACAG CUGAUGAGGCCGAAAGGCCGAA AGUUGAG
1098 GGGGACA CUGAUGAGGCCGAAAGGCCGAA AAGUUGA 1102 GCUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGAAG 1125 GAAGGUG CUGAUGAGGCCGAAAGGCCGAA AGGGCUG 1127 GUAAGGC CUGAUGAGGCCGAAAGGCCGAA AUAUGGC
1131 UGGUGCU CJGAUGAGGCCGAAAGGCCGAA AGGGAUG
1132 AUGCUGG CUGAUGAGGCCGAAAGGCCGAA AAGGUGU
1133 GAAGCUG CUGAUGAGGCCGAAAGGCCGAA AGAUGGA 1137 GCGCGCU CUGAUGAGGCCGAAAGGCCGAA AAGUAAA 1140 GCUGAGG CUGAUGAGGCCGAAAGGCCGAA AUGCUGG 1153 CAAAGUU CUGAUGAGGCCGAAAGGCCGAA AUGGUGC 1158 CUCAUCA CUGAUGAGGCCGAAAGGCCGAA AGUUGAU
1167 GGGGGAA CUGAUGAGGCCGAAAGGCCGAA ACUCAUC
1168 UGGGGGA CUGAUGAGGCCGAAAGGCCGAA AACUCAU
1169 AUGGGGG CUGAUGAGGCCGAAAGGCCGAA AAACUCA
1182 UGAUGGU CUGAUGAGGCCGAAAGGCCGAA ACAGCAU
1183 CUGAUGG CUGAUGAGGCCGAAAGGCCGAA AACAGCA
1184 UCAGGAG CUGAUGAGGCCGAAAGGCCGAA AGGGGCC
1187 GGCUGAG CUGAUGAGGCCGAAAGGCCGAA AAGGGAC
1188 CUGCCCU CUGAUGAGGCOSAAAGGCCGAA AUGGUAA 1198 UCAGACU CUGAUGAGGCCGAAAGGCCGAA AACUCCC 1209 GAAGGUG CUGAUGAGGCCGAAAGGCCGAA AGGGCUG 1215 CGGUGCU CUGAUGAGGCCGAAAGGCCGAA AGGCCAG 1229 GCUGAGG CUGAUGAGGCCGAAAGGCCGAA AGGGACC 1237 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AGCUGGG 1250 GAGCCUG CUGAUGAGGCCGAAAGGCCGAA AGGCUGG 1268 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AGCUGGG
1279 AGGAAGG CUGAUGAGGCCGAAAGGCCGAA ACCAUGG
1281 CGCAGCU CUGAUGAGGCCGAAAGGCCGAA AGCCCAC
1286 UGGGGGA CUGAUGAGGCCGAAAGGCCGAA AACUCAU
1309 AGACUCG CUGAUGAGGCCGAAAGGCCGAA ACAGGAG
1315 GGGUUAG CUGAUGAGGCCGAAAGGCCGAA ACUGGGG
1318 CCGGGGU CUGAUGAGGCCGAAAGGCCGAA AGAACUG
1331 GACUGGG CUGAUGAGGCCGAAAGGCCGAA AGGACCC
1334 UCAGCUU CUGAUGAGGCCGAAAGGCCGAA AGAAAAG
1389 GGCUUCC CUGAUGAGGCCGAAAGGCCGAA ACAGCGU
1413 AGCAUCA CUGAUGAGGCCGAAAGGCCGAA ACUGCAG
1414 CAGCAUC CUGAUGAGGCCGAAAGGCCGAA AACUGCA 1437 GCCAAGC CUGAUGAGGCCGAAAGGCCGAA ΑGGCCCC 1441 UGUUGCC CUGAUGAGGCCGAAAGGCCGAA AGCAAGG
1467 GUCUGUG CUGAUGAGGCCGAAAGGCCGAA ACACUCC
1468 GGUCUGU CUGAUGAGGCCGAAAGGCCGAA AACACUC 1482 GUCCACA CUGAUGAGGCCGAAAGGCCGAA AUGCCAG 1486 AGUUCCC CUGAUGAGGCCGAAAGGCCGAA ACCGAAG 1494 AAACUCU CUGAUGAGGCCGAAAGGCCGAA AGUUGUC
1500 CUGCUGA CUGAUGAGGCCGAAAGGCCGAA ACUCUGA
1501 GCUGCUG CUGAUGAGGCCGAAAGGCCGAA AACUCUG
1502 AGCUGCU CUGAUGAGGCCGAAAGGCCGAA AAACUCU 1525 ACACAGG CUGAUGAGGCCGAAAGGCCGAA AUGCACC 1566 UUCAGGG CUGAUGAGGCCGAAAGGCCGAA ACUCCAU 1577 CGAGUUA CUGAUGAGGCCGAAAGGCCGAA AGCUUCA 1579 GGCGAGU CUGAUGAGGCCGAAAGGCCGAA AUAGCUU 1583 ACCAGGC CUGAUGAGGCCGAAAGGCCGAA AGUUAUA 1588 CCCUCUC CUGAUGAGGCCGAAAGGCCGAA AGGAGAG 1622 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA AGCUGGG 1628 CCUACCG CUGAUGAGGCCGAAAGGCCGAA AGCAGGA 1648 CAUUGGG CUGAUGAGGCCGAAAGGCCGAA AGCCCCG 1660 CUGGGCA CUGAUGAGGCCGAAAGGCCGAA AGGUCAG
1663 CACCUGG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
1664 UCACCUG CUGAUGAGGCCGAAAGGCCGAA AAGCAGA
1665 ACCUCCG CUGAUGAGGCCGAAAGGCCGAA AAGCGAG
1680 GGAGGAG CUGAUGAGGCCGAAAGGCCGAA AGUCUUC
1681 UGGAGGA CUGAUGAGGCCGAAAGGCCGAA AAGUCUU 1683 AAUGGAG CUGAUGAGGCCGAAAGGCCGAA AGAAGUC 1686 CGCAAUG CUGAUGAGGCCGAAAGGCCGAA AGGAGAA 1690 UGUCCGC CUGAUGAGGCCGAAAGGCCGAA AUGGAGG
1704 AGCAGAG CUGAUGAGGCCGAAAGGCCGAA AGUCCAU
1705 GAGCAGA CUGAUGAGGCCGAAAGGCCGAA AAGUCCA 1707 AAGAGCA CUGAUGAGGCCGAAAGGCCGAA AGAAGUC 1721 CUGAUCU CUGAUGAGGCCGAAAGGCCGAA ACUCAAA 1726 AGGAGCU CUGAUGAGGCCGAAAGGCCGAA AUCUGAC 1731 ACCUUAG CUGAUGAGGCCGAAAGGCCGAA AGCUGAU 1734 AGCACCU CUGAUGAGGCCGAAAGGCCGAA AGGAGCU 1754 CUCUUGG CUGAUGAGGCCGAAAGGCCCAA AGCACUG Table 20
Human rel A HH Ribozyme Sequences
nt. Position HH Ribozyme Sequences
19 UACAGAC CUGAUGAGGCCGAAAGGCCGAA AGCCAUU
22 CACUACA CUGAUGAGGCCGAAAGGCCGAA ACGAGCC
26 CGUGCAC CUGAUGAGGCCGAAAGGCCGAA ACAGACG
93 GAGGGGG CUGAUGAGGCCGAAAGGCCGAA ACAGUUC
94 UGAGGGG CUGAUGAGGCCGAAAGGCCGAA AACAGUU 100 GGAAGAU CUGAUGAGGCCGAAAGGCCGAA AGGGGGA 103 CCGGGAA CUGAUGAGGCCGAAAGGCCGAA AUGAGGG
105 UGCCGGG CUGAUGAGGCCGAAAGGCCGAA AGAUGAG
106 CUGCCGG CUGAUGAGGCCGAAAGGCCGAA AAGAUGA 129 GGGGCCA CUGAUGAGGCCGAAAGGCCGAA AGGCCUG 138 CUCCACA CUGAUGAGGCCGAAAGGCCGAA AGGGGCC 148 GCTCAAU CUGAUGAGGCCGAAAGGCCGAA AUCUCCA 151 GCUGCUC CUGAUGAGGCCGAAAGGCCGAA AUGAUCU
180 GUAGCGG CUGAUGAGGCCGAAAGGCCGAA AGCGCAU
181 UGUAGCG CUGAUGAGGCCGAAAGGCCGAA AAGCGCA 186 GCACUUG CUGAUGAGGCCGAAAGGCCGAA AGCGGAA 204 GCCCGCG CUGAUGAGGCCGAAAGGCCGAA AGCGCCC 217 CGCCUGG CUGAUGAGGCCGAAAGGCCGAA AUGCUGC 239 UUGGUGG CUGAUGAGGCCGAAAGGCCGAA AUCUGUG 262 UGAUCUU CUGAUGAGGCCGAAAGGCCGAA AUGGUGG 268 AGCCAUU CUGAUGAGGCCGAAAGGCCGAA AUCUUGA 276 UCCUGUG CUGAUGAGGCCGAAAGGCCGAA AGCCAUU 301 CCAGGGA CUGAUGAGGCCGAAAGGCCGAA AUGCGCA 303 GACCAGG CUGAUGAGGCCGAAAGGCCGAA AGAUGCG 310 CCUUGGU CUGAUGAGGCCGAAAGGCCGAA ACCAGGG 323 CGGUGAG CUGAUGAGGCCGAAAGGCCGAA AGGGUCC 326 GGCCGGU CUGAUGAGGCCGAAAGGCCGAA AGGAGGG 335 UGGGGGU CUGAUGAGGCCGAAAGGCCGAA AGGCCGG 349 UUCCUAC CUGAUGAGGCCGAAAGGCCGAA AGCUCGU 352 CCUUUCC CUGAUGAGGCCGAAAGGCCGAA ACAAGCU
375 CUCAUAG CUGAUGAGGCCGAAAGGCCGAA AGCCAUC
376 CCUCAUA CUGAUGAGGCCGAAAGGCCGAA AAGCCAU 378 AGCCUCA CUGAUGAGGCCGAAAGGCCGAA AGAAGCC 391 CCGGGCA CUGAUGAGGCCGAAAGGCCGAA AGCUCAG 409 AACUGUG CUGAUGAGGCCGAAAGGCCGAA AUGCAGC
416 UUCUGGA CUGAUGAGGCCGAAAGGCCGAA ACUGUGG
417 GUUCUGG CUGAUGAGGCCGAAAGGCCGAA AACUGUG
418 GGUUCUG CUGAUGAGGCCGAAAGGCCGAA AAACUGU 433 CACACUG CUGAUGAGGCCGAAAGGCCGAA AUUCCCA 467 UGACUGA CUGAUGAGGCCGAAAGGCCGAA AGCCUGC 469 GCUGACU CUGAUGAGGCCGAAAGGCCGAA AUAGCCU 473 AUGCGCU CUGAUGAGGCCGAAAGGCCGAA ACUGAUA 481 UGGUCUG CUGAUGAGGCCGAAAGGCCGAA AUGCGCU 501 AACUUGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUU 502 GAACUUG CUGAUGAGGCCGAAAGGCCGAA AAGGGGU
508 CUAUAGG CUGAUGAGGCCGAAAGGCCGAA ACUUGGA
509 UCUAUAG CUGAUGAGGCCCAAAGGCCGAA AACUUGG 512 UCUUCUA CUGAUGAGGCCGAAAGGCCGAA AGGAACU 514 GCUCUUC CUGAUGAGGCCGAAAGGCCGAA AUAGGAA 534 CAGGUCG CUGAUGAGGCCGAAAGGCCGAA AGUCCCC 556 GGAAGCA CUGAUGAGGCCGAAAGGCCGAA AGCCGCA
561 CACCUGG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG
562 UCACCUG CUGAUGAGGCCGAAAGGCCGAA AAGCAGA 585 CCUGCCU CUGAUGAGGCCGAAAGGCCGAA AUGGGUC 598 GCAGGCG CUGAUGAGGCCGAAAGGCCGAA AGGGGCC 613 GAGGAAG CUGAUGAGGCCGAAAGGCCGAA ACAGGCG
616 GAUGAGG CUGAUGAGGCCGAAAGGCCGAA AGGACAG
617 GGAUGAG CUGAUGAGGCCGAAAGGCCGAA AAGGACA 620 AUGGGAU CUGAUGAGGCCGAAAGGCCGAA AGGAAGG 623 AAGAUGG CUGAUGAGGCCGAAAGGCCGAA AUGAGGA 628 UGUCAAA CUGAUGAGGCCGAAAGGCCGAA AUGGGAU
630 AUUGUCA CUGAUGAGGCCGAAAGGCCGAA AGAUGGG
631 GAUUGUC CUGAUGAGGCCGAAAGGCCGAA AAGAUGG 638 GGGGCAC CUGAUGAGGCCGAAAGGCCGAA AUUGUCA 661 AGAUCUU CUGAUGAGGCCGAAAGGCCGAA AGCUCGG 667 CUCGGCA CUGAUGAGGCCGAAAGGCCGAA AUCUUGA 687 GCUGCCA CUGAUGAGGCCGAAAGGCCGAA AGUUUCG 700 CCCCACC CUGAUGAGGCCGAAAGGCCGAA AGGCAGC 715 GUAGGAA CUGAUGAGGCCGAAAGGCCGAA AUCUCAU
717 CAGUAGG CUGAUGAGGCCGAAAGGCCGAA AGAUCUC
718 ACAGUAG CUGAUGAGGCCGAAAGGCCGAA AAGAUCU 721 CACACAG CUGAUGAGGCCGAAAGGCCGAA AGGAAGA 751 ACACCUC CUGAUGAGGCCGAAAGGCCGAA AUGUCCU 759 CGUGAAA CUGAUGAGGCCGAAAGGCCGAA ACACCUC
761 CCCGUGA CUGAUGAGGCCGAAAGGCCGAA AUACACC
762 UCCCGUG CUGAUGAGGCCGAAAGGCCGAA AAUACAC
763 GUCCCGU CUGAUGAGGCCGAAAGGCCGAA AAAUACA 792 CGAAAAG CUGAUGAGGCCGAAAGGCCGAA AGCCUCG
795 UUGCGAA CUGAUGAGGCCGAAAGGCCGAA AGGAGCC
796 CUUGCGA CUGAUGAGGCCGAAAGGCCGAA AAGGAGC
797 GCUUGCG CUGAUGAGGCCGAAAGGCCGAA AAAGGAG
798 AGCUUGC CUGAUGAGGCCGAAAGGCCGAA AAAAGGA 829 GGAACAC CUGAUGAGGCCGAAAGGCCGAA AUGGCCA
834 GGUCCGG CUGAUGAGGCCGAAAGGCCGAA ACACAAU
835 GGGUCCG CUGAUGAGGCCGAAAGGCCGAA AACACAA 845 GCGUAGG CUGAUGAGGCCGAAAGGCCGAA AGGGGUC 849 GUCUGCG CUGAUGAGGCCGAAAGGCCGAA AGGGAGG 372 CGCACAG CUGAUGAGGCCGAAAGGCCGAA AGCCUGC 883 GCAUGGA CUGAUGAGGCCGAAAGGCCGAA ACACGCA 885 CUGCAUG CUGAUGAGGCCGAAAGGCCGAA AGACACG
905 CGGUCGG CUGAUGAGGCCGAAAGGCCGAA AGGCCGC
906 CCGGUCG CUGAUGAGGCCGAAAGGCCGAA AAGGCCG 919 GCUCACU CUGAUGAGGCCGAAAGGCCGAA AGCUCCC 936 GUACUGG CUGAUGAGGCCGAAAGGCCCAA AUUCCAU
937 GGUACUG CUGAUGAGGCCGAAAGGCCGAA AAUUCCA 942 UGGCAGG CUGAUGAGGCCGAAAGGCCGAA ACUGGAA 953 UCGUCUG CUGAUGAGGCCGAAAGGCCGAA AUCUGGC 962 CGGUGAC CUGAUGAGGCCGAAAGGCCGAA AUCGUCU 965 AUCCGGU CUGAUGAGGCCGAAAGGCCGAA ACGAUCG 973 UCUCCUC CUGAUGAGGCCGAAAGGCCGAA AUCCGGU 986 GUCCUUU CUGAUGAGGCCGAAAGGCCGAA ACGUUUC 996 GGUCUCA CUGAUGAGGCCGAAAGGCCGAA AUGUCCU
1005 GCUCUUG CUGAUGAGGCCGAAAGGCCGAA AGGUCUC
1006 UGCUCUU CUGAUGAGGCCGAAAGGCCGAA AAGGUCU 1015 UCUUCAU CUGAUGAGGCCGAAAGGCCGAA AUGCUCU 1028 CUGAAAG CUGAUGAGGCCGAAAGGCCGAA ACUCUUC
1031 CCGCUGA CUGAUGAGGCCGAAAGGCCGAA AGGACUC
1032 UCCGCUG CUGAUGAGGCCGAAAGGCCGAA AAGGACU
1033 GUCCGCU CUGAUGAGGCCGAAAGGCCGAA AAAGGAC 1058 CGAGGUG CUGAUGAGGCCGAAAGGCCGAA AGGCCGG 1064 AUGCGUC CUGAUGAGGCCGAAAGGCCGAA AGGUGGA 1072 GCACAGC CUGAUGAGGCCGAAAGGCCGAA AUGCGUC
1082 CUGCGGG CUGAUGAGGCCGAAAGGCCGAA AGGCACA
1083 GCUGCGG CUGAUGAGGCCGAAAGGCCGAA AAGGCAC 1092 AGAAGCU CUGAUGAGGCCGAAAGGCCGAA AGCUGCG
1097 GGGACAG CUGAUGAGGCCGAAAGGCCGAA AGCUGAG
1098 GGGGACA CUGAUGAGGCCGAAAGGCCGAA AAGCUGA 1102 GCUUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGAAG 1125 AAAGGGA CUGAUGAGGCCGAAAGGCCGAA AGGGCUG 1127 GUAAAGG CUGAUGAGGCCGAAAGGCCGAA AUAGGGC
1131 UGACGUA CUGAUGAGGCCGAAAGGCCGAA AGGGAUA
1132 AUGACGU CUGAUGAGGCCGAAAGGCCGAA AAGGGAU
1133 GAUGACG CUGAUGAGGCCGAAAGGCCGAA AAAGGGA 1137 CAGGGAU CUGAUGAGGCCGAAAGGCCGAA ACGUAAA 1140 GCUCAGG CUGAUGAGGCCGAAAGGCCGAA AUGACGU 1153 CAUAGUU CUGAUGAGGCCGAAAGGCCGAA AUGGUGC 1158 CUCAUCA CUGAUGAGGCCGAAAGGCCGAA AGUUGAU
1167 GGUGGGA CUGAUGAGGCCGAAAGGCCGAA ACUCAUC
1168 UGGUGGG CUGAUGAGGCCGAAAGGCCGAA AACUCAU
1169 AUGGUGG CUGAUGAGGCCGAAAGGCCGAA AAACUCA
1182 AGAAGGA CUGAUGAGGCCGAAAGGCCGAA ACACCAU
1183 CAGAAGG CUGAUGAGGCCGAAAGGCCGAA AACACCA
1184 CCAGAAG CUGAUGAGGCCGAAAGGCCGAA AAACACC
1187 UGCCCAG CUGAUGAGGCCGAAAGGCCGAA AGGAAAC
1188 CUGCCCA CUGAUGAGGCCGAAAGGCCGAA AAGGAAA 1198 CCUGGCU CUGAUGAGGCCGAAAGGCCGAA AUCUGCC 1209 CAAGGCC CUGAUGAGGCCGAAAGGCCGAA AGGCCUG 1215 CGGGGCC CUGAUGAGGCCGAAAGGCCGAA AGGCCGA 1229 ACUUGGG CUGAUGAGGCCGAAAGGCCGAA AGGGGCC 1237 GGGGCAG CUGAUGAGGCCGAAAGGCCGAA ACUUGGG 1250 GGGGCUG CUGAUGAGGCCGAAAGGCCGAA AGCCUGG 1268 AUGGCUG CUGAUGAGGCCGAAAGGCCGAA AGCAGGG 1279 GAGCUGA CUGAUGAGGCCGAAAGGCCGAA ACCAUGG
1281 CAGAGCU CUGAUGAGGCCGAAAGGCCGAA AUACCAU
1286 UGGGCCA CUGAUGAGGCCGAAAGGCCGAA AGCUGAU
1309 GGACUGG CUGAUGAGGCCGAAAGGCCGAA ACAGGGG
1315 GGGCUAG CUGAUGAGGCCGAAAGGCCGAA ACUGGGA
1318 CUGGGGC CUGAUGAGGCCGAAAGGCCGAA AGGACUG
1331 GCCUGAG CUGAUGAGGCCGAAAGGCCGAA AGGGCCU
1334 ACAGCCU CUGAUGAGGCCGAAAGGCCGAA AGGAGGG
1389 GGCCUCU CUGAUGAGGCCGAAAGGCCGAA ACAGCGU
1413 AUCAUCA CUGAUGAGGCCGAAAGGCCGAA ACUGCAG
1414 CAUCAUC CUGAUGAGGCCGAAAGGCCGAA AACUGCA 1437 GCCAAGC CUGAUGAGGCCGAAAGGCCGAA AGGCCCC 1441 UGUUGCC CUGAUGAGGCCGAAAGGCCGAA AGCAAGG
1467 GUCUGUG CUGAUGAGGCCGAAAGGCCGAA ACACAGC
1468 GGUCUGU CUGAUGAGGCCGAAAGGCCGAA AACACAG 1482 GUCGACG CUGAUGAGGCCGAAAGGCCGAA AUGCCAG I486 AGUUGUC CUGAUGAGGCCGAAAGGCCGAA ACGGAUG 1494 AAACUCG CUGAUGAGGCCGAAAGGCCGAA AGUUGUC
1500 CUGCUGA CUGAUGAGGCCGAAAGGCCGAA ACUCGGA
1501 GCUGCUG CUGAUGAGGCCGAAAGGCCGAA AACUCGG
1502 AGCUGCU CUGAUGAGGCCGAAAGGCCGAA AAACUCG 1525 CCACAGG CUGAUGAGGCCGAAAGGCCGAA AUGCCCU 1566 CUCAGGG CUGAUGAGGCCGAAAGGCCGAA ACUCCAU 1577 CGAGUUA CUGAUGAGGCCGAAAGGCCGAA AGCCUCA 1579 GGCGAGU CUGAUGAGGCCGAAAGGCCGAA AUAGCCU 1583 ACUAGGC CUGAUGAGGCCGAAAGGCCGAA AGUUAUA 1588 CUGUCAC CUGAUGAGGCCGAAAGGCCGAA AGGCGAG 1622 GGAGCAG CUGAUGAGGCCGAAAGGCCGAA AGCUGGG 1628 CCCAGUG CUGAUGAGGCCGAAAGGCCGAA AGCAGGA 1648 CAUUGGG CUGAUGAGGCCGAAAGGCCGAA AGCCCCG 1660 CUGAAAG CUGAUGAGGCCGAAAGGCCGAA AGGCCAU
1663 CUCCUGA CUGAUGAGGCCGAAAGGCCGAA AGGAGGC
1664 UCUCCUG CUGAUGAGGCCGAAAGGCCGAA AAGGAGG
1665 AUCUCCU CUGAUGAGGCCGAAAGGCCGAA AAAGGAG
1680 GGAGGAG CUGAUGAGGCCGAAAGGCCGAA AGUCUUC
1681 UGGAGGA CUGAUGAGGCCGAAAGGCCGAA AAGUCUU 1683 AAUGGAG CUGAUGAGGCCGAAAGGCCGAA AGAAGUC 1686 CGCAAUG CUGAUGAGGCCGAAAGGCCGAA AGGAGAA 1690 UGUCCGC CUGAUGAGGCCGAAAGGCCGAA AUGGAGG
1704 GGCUGAG CUGAUGAGGCCGAAAGGCCGAA AGUCCAU
1705 GGGCUGA CUGAUGAGGCCGAAAGGCCGAA AAGUCCA 1707 CAGGGCU CUGAUGAGGCCGAAAGGCCGAA AGAAGUC 1721 CUGAUCU CUGAUGAGGCCGAAAGGCCGAA ACUCAGC 1726 AGGAGCU CUGAUGAGGCCGAAAGGCCGAA AUCUGAC 1731 CCCUUAG CUGAUGAGGCCGAAAGGCCGAA AGCUGAU 1734 ACCCCCU CUGAUGAGGCCGAAAGGCCGAA AGGAGCU 1754 CUCUGGG CUGAUGAGGCCGAAAGGCCGAA AGGGCAG Table 21
Human rel A Hairpin Ribozyme/Tar)
nt. Position HHaaiirrppin Ribozyme sequence Substrate
90 UGAGGGGG AGAA GUUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAACU GUU CCCCCUCA
156 GCUGCUUG AGAA GCUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAGCA GCC CAAGCAGC
362 GCCAUCCC AGAA GUCC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GGACU GCC GGGAUGGC
413 GUUCUGGA AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACA GUU UCCAGAAC
606 GAAGGACA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCC GCC UGUCCUUC
652 UUGAGCUC AGAA GUGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACACU GCC GAGCUCAA
695 CCCACCGA AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCU GCC UCGGUGGG
853 AGGCUGGG AGAA GCGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACGCA GAC CCCAGCCU
900 GGUCGGAA AGAA GCCG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CGGCG GCC UUCCGACC
955 UGACGAUC AGAA GUAU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AUACA GAC GAUCGUCA
1037 GUCGGUGG AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCG GAC CCACCGAC
1045 GGCCGGGG AGAA GUGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCACC GAC CCCCGGCC
1410 CAUCAUCA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCA GUU UGAUGAUG
1453 ACAGCUGG AGAA GUGC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GCACA GAC CCAGCUGU
1471 GAUGCCAG AGAA GUGA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UCACA GAC CUGGCAUC
Table 22
Mouse rel A Hairpin Ribozyme/Target Sequences
nt. Position Hairpin Ribozyme sequence Substrate
137 GUUGCUUC AGAA GUUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAACA GCC GAAGCAAC
273 GAGAUUCG AGAA GUUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAACA GUU CGAAUCUC
343 GCCAUCCC AGAA GUCC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GGACU GCC GGGAUGGC
366 GGGCAGAG AGAA GCCU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AGGCU GAC CUCUGCCC
633 UUGAGCUC AGAA GUGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACACU GCC GAGCUCAA
676 CCCACCGA AGAA GCUC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GAGCU GCC UCGGUGGG
834 AGGCUGGG AGAA GCGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACGCC GAC CCCAGCCU
881 GAUCAGAA AGAA GCCG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CGGCG GCC UUCUGAUC
1100 AGGUGUAG AGAA GCGG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CCGCA GCC CUACACCU
1205 GGGCAGAG AGAA GUGC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GCACC GUC CUCUGCCC
1361 GGGCUUCC AGAA GCGU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA ACGCU GUC GGAAGCCC
1385 CAGCAUCA AGAA GCAG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CUGCA GUU UGAUGCUG
1431 ACUCCUGG AGAA GUGC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GCACA GAC CCAGGAGU
1449 GAUGCCAG AGAA GUGA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UCACA GAC CUGGCAUC
1802 AAGUCGGG AGAA GCUG ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA CAGCU GCC CCCGACUU
2009 UGGCUCCA AGAA GUCC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GGACA GAC UGGAGCCA
2124 UGGUGUCG AGAA GCAC ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA GUGCU GCC CGACACCA
2233 AUUCUGAA AGAA GCCA ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA UGGCC GCC UUCAGAAU
2354 UCAGUAAA AGAA GUCU ACCAGAGAAACACACGUUGUGGUACAUUACCUGGUA AGACA GCC UUUACUGA
Table 23: Human TNF-α HH Ribozyme Target Sequence
nt. HH Target Sequence nt. HH Target Sequence
Position Position
28 GGCAGGU U CUCUUCC
29 GCAGGUU C UCUUCCU 321 GUCAGAU C AUCUUCU
31 AGGUUCU C UUCCUCU 324 AGAUCAU C UUCUCGA
33 GUUCUCU U CCUCUCA 326 AUCAUCU U CUCGAAC
34 UUCUCUU C CUCUCAC 327 UCAUCUU C UCGAACC
37 UCUUCCU C UCACAUA 329 AUCUUCU C GAACCCC
39 UUCCUCU C ACAUACU 352 AGCCUGU A GCCCAUG
44 CUCACAU A CUGACCC 361 CCCAUGU U GUAGCAA
58 CACGGCU C CACCCUC 364 AUGUUGU A GCAAACC
65 CCACCCU C UCUCCCC 374 AAACCCU C AAGCUGA
67 ACCCUCU C UCCCCUG 391 GGCAGCU C CAGUGGC
69 CCUCUCU C CCCUGGA 421 AUGCCCU C CUGGCCA
106 GCAUGAU C CGGGACG 449 GAGAGAU A ACCAGCU
136 AGGCGCU C CCCAAGA 468 GUGCCAU C AGAGGGC
165 CAGGGCU C CAGGCGG 480 GGCCUGU A CCUCAUC
177 CGGUGCU U GUUCCUC 484 UGUACCU C AUCUACU
180 UGCUUGU U CCUCAGC 487 ACCUCAU C UACUCCC
181 GCUUGUU C CUCAGCC 489 CUCAUCU A CUCCCAG 184 UGUUCCU C AGCCUCU 492 AUCUACU C CCAGGUC
190 UCAGCCU C UUCUCCU 499 CCCAGGU C CUCUUCA
192 AGCCUCU U CUCCUUC 502 AGGUCCU C UUCAAGG
193 GCCUCUU C UCCUUCC 504 GUCCUCU U CAAGGGC
195 CUCUUCU C CUUCCUG 505 UCCUCUU C AAGGGCC
198 UUCUCCU U CCUGAUC 525 UGCCCCU C CACCCAU
199 UCUCCUU C CUGAUCG 538 AUGUGCU C CUCACCC
205 UCCUGAU C GUGGCAG 541 UGCUCCU C ACCCACA
226 CCACGCU C UUCUGCC 553 ACACCAU C AGCCGCA
228 ACGCUCU U CUGCCUG 562 GCCGCAU C GCCGUCU
229 CGCUCUU C UGCCUGC 568 UCGCCGU C UCCUACC
243 CUGCACU U UGGAGUG 570 GCCGUCU C CUACCAG
244 UGCACUU U GGAGUGA 573 GUCUCCU A CCAGACC
253 GAGUGAU C GGCCCCC 586 CCAAGGU C AACCUCC
273 GAAGAGU C CCCCAGG 592 UCAACCU C CUCUCUG
286 GGGACCU C UCUCUAA 595 ACCUCCU C UCUGCCA
288 GACCUCU C UCUAAUC 597 CUCCUCU C UGCCAUC
290 CCUCUCU C UAAUCAG 604 CUGCCAU C AAGAGCC
292 UCUCUCU A AUCAGCC 657 CCCUGGU A UGAGCCC
295 CUCUAAU C AGCCCUC 667 AGCCCAU C UAUCUGG
302 CAGCCCU C UGGCCCA 669 CCCAUCU A UCUGGGA 671 CAUCUAU C UGGGAGG 960 UGGGAUU C AGGAAUG
682 GAGGGGU C UUCCAGC 1001 AACCACU A AGAAUUC
684 GGGGUCU U CCAGCUG 1007 UAAGAAU U CAAACUG
685 GGGUCUU C CAGCUGG 1008 AAGAAUU C AAACUGG
709 ACCGACU C AGCGCUG 1021 GGGGCCU C CAGAACU
721 CUGAGAU C AAUCGGC 1029 CAGAACU C ACUGGGG
725 GAUCAAU C GGCCCGA 1040 GGGGCCU A CAGCUUU
735 CCCGACU A UCUCGAC 1046 UACAGCU U UGAUCCC
737 CGACUAU C UCGACUU 1047 ACAGCUU U GAUCCCU
739 ACUAUCU C GACUUUG 1051 CUUUGAU C CCUGACA
744 CUCGACU U UGCCGAG 1060 CUGACAU C UGGAAUC
745 UCGACUU U GCCGAGU 1067 CUGGAAU C UGGAGAC
753 GCCGAGU C UGGGCAG 1085 GGAGCCU U UGGUUCU
763 GGCAGGU C UACUUUG 1086 GAGCCUU U GGUUCUG
765 CAGGUCU A CUUUGGG 1090 CUUUGGU U CUGGCCA
768 GUCUACU U UGGGAUC 1091 UUUGGUU C UGGCCAG
769 UCUACUU U GGGAUCA 1113 CAGGACU U GAGAAGA
775 UUGGGAU C AUUGCCC 1124 AAGACCU C ACCUAGA
778 GGAUCAU U GCCCUGU 1129 CUCACCU A GAAAUUG
801 CGAACAU C CAACCUU 1135 UAGAAAU U GACACAA
808 CCAACCU U CCCAAAC 1151 UGGACCU U AGGCCUU
809 CAACCUU C CCAAACG 1152 GGACCUU A GGCCUUC
820 AACGCCU C CCCUGCC 1158 UAGGCCU U CCUCUCU
833 CCCCAAU C CCUUUAU 1159 AGGCCUU C CUCUCUC
837 AAUCCCU U UAUUACC 1162 CCUUCCU C UCUCCAG
838 AUCCCUU U AUUACCC 1164 UUCCUCU C UCCAGAU
839 UCCCUUU A UUACCCC 1166 CCUCUCU C CAGAUGU
841 CCUUUAU U ACCCCCU 1174 CAGAUGU U UCCAGAC
842 CUUUAUU A CCCCCUC 1175 AGAUGUU U CCAGACU
849 ACCCCCU C CUUCAGA 1176 GAUGUUU C CAGACUU
852 CCCUCCU U CAGACAC 1183 CCAGACU U CCUUGAG
853 CCUCCUU C AGACACC 1184 CAGACUU C CUUGAGA
863 ACACCCU C AACCUCU 1187 ACUUCCU U GAGACAC
869 UCAACCU C UUCUGGC 1208 CAGCCCU C CCCAUGG
871 AACCUCU U CUGGCUC 1224 GCCAGCU C CCUCUAU
872 ACCUCUU C UGGCUCA 1228 GCUCCCU C UAUUUAU
878 UCUGGCU C AAAAAGA 1230 UCCCUCU A UUUAUGU
890 AGAGAAU U GGGGGCU 1232 CCUCUAU U UAUGUUU
898 GGGGGCU U AGGGUCG 1233 CUCUAUU U AUGUUUG
899 GGGGCUU A GGGUCGG 1234 UCUAUUU A UGUUUGC
904 UUAGGGU C GGAACCC 1238 UUUAUGU U UGCACUU
917 CCAAGCU U AGAACUU 1239 UUAUGUU U GCACUUG
918 CAAGCUU A GAACUUU 1245 UUGCACU U GUGAUUA
924 UAGAACU U UAAGCAA 1251 UUGUGAU U AUUUAUU
925 AGAACUU U AAGCAAC 1252 UGUGAUU A UUUAUUA 926 GAACUUU A AGCAACA 1254 UGAUUAU U UAUUAUU
945 CACCACU U CGAAACC 1255 GAUUAUU U AUUAUUU
946 ACCACUU C GAAACCU 1256 AUUAUUU A UUAUUUA
959 CUGGGAU U CAGGAAU 1258 UAUUUAU U AUUUAUU 1259 AUUUAUU A UUUAUUU 1440 UGUUUUU U AAAAUAU
1261 UUAUUAU U UAUUUAU 1441 GUUUUUU A AAAUAUU
1262 UAUUAUU U AUUUAUU 1446 UUAAAAU A UUAUCUG
1263 AUUAUUU A UUUAUUA 1448 AAAAUAU U AUCUGAU
1265 UAUUUAU U UAUUAUU 1449 AAAUAUU A UCUGAUU
1266 AUUUAUU U AUUAUUU 1451 AUAUUAU C UGAUUAA
1267 UUUAUUU A UUAUUUA 1456 AUCUGAU U AAGUUGU
1269 UAUUUAU U AUUUAUU 1457 UCUGAUU A AGUUGUC
1270 AUUUAUU A UUUAUUU 1461 AUUAAGU U GUCUAAA
1272 UUAUUAU U UAUUUAU 1464 AAGUUGU C UAAACAA
1273 UAUUAUU U AUUUAUU 1466 GUUGUCU A AACAAUG
1274 AUUAUUU A UUUAUUU 1479 UGCUGAU U UGGUGAC
1276 UAUUUAU U UAUUUAC 1480 GCUGAUU U GGUGACC
1277 AUUUAUU U AUUUACA 1494 CAACUGU C ACUCAUU
1278 UUUAUUU A UUUACAG 1498 UGUCACU C AUCGCUG
1280 UAUUUAU U UACAGAU 1501 CACUCAU U GCUGAGG
1281 AUUUAUU U ACAGAUG 1512 GAGGCCU C UGCUCCC
1282 UUUAUUU A CAGAUGA 1517 CUCUGCU C CCCAGGG
1294 UGAAUGU A UUUAUUU 1528 AGGGAGU U GUGCCUG
1296 AAUGUAU U UAUUUGG 1533 GUUGUGU C UGUAAUC
1297 AUGUAUU U AUUUGGG 1537 UGUCUGU A AUCGGCC
1298 UGUAUUU A UUUGGGA 1540 CUGUAAU C GGCCUAC
1300 UAUUUAU U UGGGAGA 1546 UCGGCCU A CUAUUCA
1301 AUUUAUU U GGGAGAC 1549 GCCUACU A UUCAGUG
1315 CCGGGGU A UCCUGGG 1551 CUACUAU U CAGUGGC
1317 GGGGUAU C CUGGGGG 1552 UACUAUU C AGUGGCG
1334 CCAAUGU A GGAGCUG 1566 GAGAAAU A AAGGUUG
1345 GCUGCCU U GGCUCAG 1572 UAAAGGU U GCUUAGG
1350 CUUGGCU C AGACAUG 1576 GGUUGCU U AGGAAAG
1359 GACAUGU U UUCCGUG 1577 GUUGCUU A GGAAAGA
1360 ACAUGUU U UCCGOGA
1361 CAUGUUU U CCGUGAA
1362 AUGUUUU C CGUGAAA
1386 GAACAAU A GGCUGUU
1393 AGGCUGU U CCCAUGU
1394 GGCUGUU C CCAUGUA
1401 CCCAUGU A GCCCCCU
1414 CUGGCCU C UGUGCCU
1422 UGUGCCU U CUUUUGA
1423 GUGCCUU C UUUUGAU
1425 GCCUUCU U UUGAUUA
1426 CCUUCUU U UGAUUAU
1427 CUUCUUU U GAUUAUG
1431 UUUUGAU U AUGUUUU
1432 UUUGAUU A UGUUUUU
1436 AUUAUGU U UUUUAAA
1437 UUAUGUU U UUUAAAA
1438 UAUGUUU U UUAAAAU Table 24: Human TNF-α Hammerhead Ribozyme Sequences
nt . HH Ribozyme Se quence
Position
28 GGAAGAG CUGAUGAGGCCGAAAGGCCGAA ACCUGCC
29 AGGAAGA CUGAUGAGGCCGAAAGGCCGAA AACCUGC
31 AGAGGAA CUGAUGAGGCCGAAAGGCCGAA AGAACCU
33 UGAGAGG CUGAUGAGGCCGAAAGGCCGAA AGAGAAC
34 GUGAGAG CUGAUGAGGCCGAAAGGCCGAA AAGAGAA
37 UAUGUGA CUGAUGAGGCCGAAAGGCCGAA AGGAAGA
39 AGUAUGU CUGAUGAGGCCGAAAGGCCGAA AGAGGAA
44 GGGUCAG CUGAUGAGGCCGAAAGGCCGAA AUGUGAG
58 GAGGGUG CUGAUGAGGCCGAAAGGCCGAA AGCCGUG
65 GGGGAGA CUGAUGAGGCCGAAAGGCCGAA AGGGUGG
67 CAGGGGA CUGAUGAGGCCGAAAGGCCGAA AGAGGGU
69 UCCAGGG CUGAUGAGGCCGAAAGGCCGAA AGAGAGG
106 CGUCCCG CUGAUGAGGCCGAAAGGCCGAA AUCAUGC
136 UCUUGGG CUGAUGAGGCCGAAAGGCCGAA AGCGCCU
165 CCGCCUG CUGAUGAGGCCGAAAGGCCGAA AGCCCUG
177 GAGGAAC CUGAUGAGGCCGAAAGGCCGAA AGCACCG
130 GCUGAGG CUGAUGAGGCCGAAAGGCCGAA ACAAGCA
181 GGCUGAG CUGAUGAGGCCGAAAGGCCGAA AACAAGC
184 AGAGGCU CUGAUGAGGCCGAAAGGCCGAA AGGAACA
190 AGGAGAA CUGAUGAGGCCGAAAGGCCGAA AGGCUGA
192 GAAGGAG CUGAUGAGGCCGAAAGGCCGAA AGAGGCU
193 GGAAGGA CUGAUGAGGCCGAAAGGCCGAA AAGAGGC
195 CAGGAAG CUGAUGAGGCCGAAAGGCCGAA AGAAGAG
198 GAUCAGG CUGAUGAGGCCGAAAGGCCGAA AGGAGAA
199 CGAUCAG CUGAUGAGGCCGAAAGGCCGAA AAGGAGA
205 CUGCCAC CUGAUGAGGCCGAAAGGCCGAA AUCAGGA
226 GGCAGAA CUGAUGAGGCCGAAAGGCCGAA AGCGUGG
228 CAGGCAG CUGAUGAGGCCGAAAGGCCGAA AGAGCGU
229 GCAGGCA CUGAUGAGGCCGAAAGGCCGAA AAGAGCG
243 CACUCCA CUGAUGAGGCCGAAAGGCCGAA AGUGCAG
244 UCACUCC CUGAUGAGGCCGAAAGGCCGAA AAGUGCA
253 GGGGGCC CUGAUGAGGCCGAAAGGCCGAA AUCACUC
273 CCUGGGG CUGAUGAGGCCGAAAGGCCGAA ACUCUUC
286 UUAGAGA CUGAUGAGGCCGAAAGGCCGAA AGGUCCC
288 GAUUAGA CUGAUGAGGCCGAAAGGCCGAA AGAGGUC
290 CUGAUUA CUGAUGAGGCCGAAAGGCCGAA AGAGAGG
292 GGCUGAU CUGAUGAGGCCGAAAGGCCGAA AGAGAGA
295 GAGGGCU CUGAUGAGGCCGAAAGGCCGAA AUUAGAG
302 UGGGCCA CUGAUGAGGCCGAAAGGCCGAA AGGGCUG 321 AGAAGAU CUGAUGAGGCCGAAAGGCCGAA AUCUGAC
324 UCGAGAA CUGAUGAGGCCGAAAGGCCGAA AUGAUCU
326 GUUCGAG CUGAUGAGGCCGAAAGGCCGAA AGAUGAU
327 GGUUCGA CUGAUGAGGCCGAAAGGCCGAA AAGAUGA 329 GGGGUUC CUGAUGAGGCCGAAAGGCCGAA AGAAGAU 352 CAUGGGC CUGAUGAGGCCGAAAGGCCGAA ACAGGCU 361 UUGCUAC CUGAUGAGGCCGAAAGGCCGAA ACAUGGG 364 GGUUUGC CUGAUGAGGCCGAAAGGCCGAA ACAACAU 374 UCAGCUU CUGAUGAGGCCGAAAGGCCGAA AGGGUUU 391 GCCACUG CUGAUGAGGCCGAAAGGCCGAA AGCUGCC 421 UGGCCAG CUGAUGAGGCCGAAAGGCCGAA AGGGCAU 449 AGCUGGU CUGAUGAGGCCGAAAGGCCGAA AUCUCUC 468 GCCCUCU CUGAUGAGGCCGAAAGGCCGAA AUGGCAC 480 GAUGAGG CUGAUGAGGCCGAAAGGCCGAA ACAGGCC 484 AGUAGAU CUGAUGAGGCCGAAAGGCCGAA AGGUACA 487 GGGAGUA CUGAUGAGGCCGAAAGGCCGAA AUGAGGU 489 CUGGGAG CUGAUGAGGCCGAAAGGCCGAA AGAUGAG 492 GACCUGG CUGAUGAGGCCGAAAGGCCGAA AGUAGAU 499 UGAAGAG CUGAUGAGGCCGAAAGGCCGAA ACCUGGG 502 CCUUGAA CUGAUGAGGCCGAAAGGCCGAA AGGACCU
504 GCCCUUG CUGAUGAGGCCGAAAGGCCGAA AGAGGAC
505 GGCCCUU CUGAUGAGGCCGAAAGGCCGAA AAGAGGA 525 AUGGGUG CUGAUGAGGCCGAAAGGCCGAA AGGGGCA 538 GGGUGAG CUGAUGAGGCCGAAAGGCCGAA AGCACAU 541 UGUGGGU CUGAUGAGGCCGAAAGGCCGAA AGGAGCA 553 UGCGGCU CUGAUGAGGCCGAAAGGCCGAA AUGGUGU
562 AGACGGC CUGAUGAGGCCGAAAGGCCGAA AUGCGGC
563 GGUAGGA CUGAUGAGGCCGAAAGGCCGAA ACGGCGA 570 CUGGUAG CUGAUGAGGCCGAAAGGCCGAA AGACGGC 573 GGUCUGG CUGAUGAGGCCGAAAGGCCGAA AGGAGAC 586 GGAGGUU CUGAUGAGGCCGAAAGGCCGAA ACCUUGG 592 CAGAGAG CUGAUGAGGCCGAAAGGCCGAA AGGUUGA 595 UGGCAGA CUGAUGAGGCCGAAAGGCCGAA AGGAGGU 597 GAUGGCA CUGAUGAGGCCGAAAGGCCGAA AGAGGAG 604 GGCUCUU CUGAUGAGGCCGAAAGGCCGAA AUGGCAG 657 GGGCUCA CUGAUGAGGCCGAAAGGCCGAA ACCAGGG 667 CCAGAUA CUGAUGAGGCCGAAAGGCCGAA AUGGGCU 669 UCCCAGA CUGAUGAGGCCGAAAGGCCGAA AGAUGGG 671 CCUCCCA CUGAUGAGGCCGAAAGGCCGAA AUAGAUG 682 GCUGGAA CUGAUGAGGCCGAAAGGCCGAA ACCCCUC
684 CAGCUGG CUGAUGAGGCCGAAAGGCCGAA AGACCCC
685 CCAGCUG CUGAUGAGGCCGAAAGGCCGAA AAGACCC 709 CAGCGCU CUGAUGAGGCCGAAAGGCCGAA AGUCGGU 721 GCCGAUU CUGAUGAGGCCGAAAGGCCGAA AUCUCAG 725 UCGGGCC CUGAUGAGGCCGAAAGGCCGAA AUUGAUC 735 GUCGAGA CUGAUGAGGCCCAAAGGCCGAA AGUCGGG 737 AAGUCGA CUGAUGAGGCCGAAAGGCCGAA AUAGUCG 739 CAAAGUC CUGAUGAGGCCGAAAGGCCGAA AGAUAGU 744 CUCGGCA CUGAUGAGGCCGAAAGGCCGAA AGUCGAG 745 ACUCGGC CUGAUGAGGCCGAAAGGCCGAA AAGUCGA
753 CUGCCCA CUGAUGAGGCCGAAAGGCCGAA ACUCGGC
763 CAAAGUA CUGAUGAGGCCGAAAGGCCGAA ACCUGCC
765 CCCAAAG CUGAUGAGGCCGAAAGGCCGAA AGACCUG
768 GAUCCCA CUGAUGAGGCCGAAAGGCCGAA AGUAGAC
769 UGAUCCC CUGAUGAGGCCGAAAGGCCGAA AAGUAGA 775 GGGCAAU CUGAUGAGGCCGAAAGGCCGAA AUCCCAA 778 ACAGGCC CUGAUGAGGCCGAAAGGCCGAA AUGAUCC 801 AAGGUUG CUGAUGAGGCCGAAAGGCCGAA AUGUUCG
808 GUUUGGG CUGAUGAGGCCGAAAGGCCGAA AGGUUGG
809 CGUUUGG CUGAUGAGGCCGAAAGGCCGAA AAGGUUG 820 GGCAGCG CUGAUGAGGCCGAAAGGCCGAA AGGCCUU 833 AUAAAGG CUGAUGAGGCCGAAAGGCCGAA AUUGGGG
837 GGUAAUA CUGAUGAGGCCGAAAGGCCGAA AGGGAUU
838 GGGUAAU CUGAUGAGGCCGAAAGGCCGAA AAGGGAU
839 GGGGUAA CUGAUGAGGCCGAAAGGCCGAA AAAGGGA
841 AGGGGGU CUGAUGAGGCCGAAAGGCCGAA AUAAAGG
842 GAGGGGG CUGAUGAGGCCGAAAGGCCGAA AAUAAAG 849 UCUGAAG CUGAUGAGGCCGAAAGGCCGAA AGGGGGU
852 GUGUCUG CUGAUGAGGCCGAAAGGCCGAA AGGAGGG
853 GGUGUCU CUGAUGAGGCCGAAAGGCCGAA AAGGAGG 863 AGAGGUU CUGAUGAGGCCGAAAGGCCGAA AGGGUGU 869 GCCAGAA CUGAUGAGGCCGAAAGGCCGAA AGGUUGA
871 GAGCCAG CUGAUGAGGCCGAAAGGCCGAA AGAGGUU
872 UGAGCCA CUGAUGAGGCCGAAAGGCCGAA AAGAGGU 878 UCUUUUU CUGAUGAGGCCGAAAGGCCGAA AGCCAGA 890 AGCCCCC CUGAUGAGGCCGAAAGGCCGAA AUUCUCU
898 CGACCCU CUGAUGAGGCCGAAAGGCCGAA AGCCCCC
899 CCGACCC CUGAUGAGGCCGAAAGGCCGAA AAGCCCC 904 GGGUUCC CUGAUGAGGCCGAAAGGCCGAA ACCCUAA
917 AAGUUCU CUGAUGAGGCCGAAAGGCCGAA AGCUUGG
918 AAAGUUC CUGAUGAGGCCGAAAGGCCGAA AAGCUUG
924 UUGCUUA CUGAUGAGGCCGAAAGGCCGAA AGUUCUA
925 GUUGCUU CUGAUGAGGCCGAAAGGCCGAA AAGUUCU
926 UGUUGCU CUGAUGAGGCCGAAAGGCCGAA AAAGUUC
945 GGUUUCG CUGAUGAGGCCGAAAGGCCGAA AGUGGUG
946 AGGUUUC CUGAUGAGGCCGAAAGGCCGAA AAGUGGU
959 AUUCCUG CUGAUGAGGCCGAAAGGCCGAA AUCCCAG
960 CAUUCCU CUGAUGAGGCCGAAAGGCCGAA AAUCCCA 1001 GAAUUCU CUGAUGAGGCCGAAAGGCCGAA AGUGGUU
1007 CAGUUUG CUGAUGAGGCCGAAAGGCCGAA AUUCUUA
1008 CCAGUUU CUGAUGAGGCCGAAAGGCCGAA AAUUCUU 1021 AGUUCUG CUGAUGAGGCCGAAAGGCCGAA AGGCCCC 1029 CCCCAGU CUGAUGAGGCCGAAAGGCCGAA AGUUCUG 1040 AAAGCUG CUGAUGAGGCCGAAAGGCCGAA AGGCCCC
1046 GGGAUCA CUGAUGAGGCCGAAAGGCCGAA AGCUGUA
1047 AGGGAUC CUGAUGAGGCCGAAAGGCCGAA AAGCUGU 1051 UGUCAGG CUGAUGAGGCCGAAAGGCCGAA AUCAAAG 1060 GAUUCCA CUGAUGAGGCCGAAAGGCCGAA AUGUCAG 1067 GUCUCCA CUGAUGAGGCCGAAAGGCCGAA AUUCCAG
1085 AGAACCA CUGAUGAGGCCGAAAGGCCGAA AGGCUCC
1086 CAGAACC CUGAUGAGGCCGAAAGGCCGAA AAGGCUC
1090 UGGCCAG CUGAUGAGGCCGAAAGGCCGAA ACCAAAG
1091 CUGGCCA CUGAUGAGGCCGAAAGGCCGAA AACCAAA 1113 UCUUCUC CUGAUGAGGCCGAAAGGCCGAA AGUCCUG 1124 UCUAGGU CUGAUGAGGCCGAAAGGCCGAA AGGUCUU 1129 CAAUUUC CUGAUGAGGCCGAAAGGCCGAA AGGUGAG 1135 UUGUGUC CUGAUGAGGCCGAAAGGCCGAA AUUUCUA
1151 AAGGCCU CUGAUGAGGCCGAAAGGCCGAA AGGUCCA
1152 GAAGGCC CUGAUGAGGCCGAAAGGCCGAA AAGGUCC
1158 AGAGAGG CUGAUGAGGCCGAAAGGCCGAA AGGCCUA
1159 GAGAGAG CUGAUGAGGCCGAAAGGCCGAA AAGGCCU 1162 CUGGAGA CUGAUGAGGCCGAAAGGCCGAA AGGAAGG 1164 AUCUGGA CUGAUGAGGCCGAAAGGCCGAA AGAGGAA 1166 ACAUCUG CUGAUGAGGCCGAAAGGCCGAA AGAGAGG
1174 GUCUGGA CUGAUGAGGCCGAAAGGCCGAA ACAUCUG
1175 AGUCUGG CUGAUGAGGCCGAAAGGCCGAA AACAUCU
1176 AAGUCUG CUGAUGAGGCCGAAAGGCCGAA AAACAUC
1183 CUCAAGG CUGAUGAGGCCGAAAGGCCGAA AGUCUGG
1184 UCUCAAG CUGAUGAGGCCGAAAGGCCGAA AAGUCUG 1187 GUGUCUC CUGAUGAGGCCGAAAGGCCGAA AGGAAGU 1208 CCAUGGG CUGAUGAGGCCGAAAGGCCGAA AGGGCUG 1224 AUAGAGG CUGAUGAGGCCGAAAGGCCGAA AGCUGGC 1228 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AGGGAGC 1230 ACAUAAA CUGAUGAGGCCGAAAGGCCGAA AGAGGGA
1232 AAACAUA CUGAUGAGGCCGAAAGGCCGAA AUAGAGG
1233 CAAACAU CUGAUGAGGCCGAAAGGCCGAA AAUAGAG
1234 GCAAACA CUGAUGAGGCCGAAAGGCCGAA AAAUAGA
1238 AAGUGCA CUGAUGAGGCCGAAAGGCCGAA ACAUAAA
1239 CAAGUGC CUGAUGAGGCCGAAAGGCCGAA AACAUAA 1245 UAAUCAC CUGAUGAGGCCGAAAGGCCGAA AGUGCAA
1251 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUCACAA
1252 UAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUCACA
1254 AAUAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUCA
1255 AAAUAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUC
1256 UAAAUAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAU
1258 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1259 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1261 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
1262 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUA
1263 UAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAU
1265 AAUAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1266 AAAUAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1267 UAAAUAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAA
1269 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1270 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1272 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
1273 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUA 1274 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAU
1276 GUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1277 UGUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1278 CUGUAAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAA
1280 AUCUGUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1281 CAUCUGU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1282 UCAUCUG CUGAUGAGGCCGAAAGGCCGAA AAAUAAA
1294 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACAUUCA
1296 CCAAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUU
1297 CCCAAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
1298 UCCCAAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
1300 UCUCCCA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1301 GUCUCCC CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1315 CCCAGGA CUGAUGAGGCCGAAAGGCCGAA ACCCCGG
1317 CCCCCAG CUGAUGAGGCCGAAAGGCCGAA AUACCCC
1334 CAGCUCC CUGAUGAGGCCGAAAGGCCCAA ACAUUGG
1345 CUGAGCC CUGAUGAGGCCGAAAGGCCGAA AGGCAGC
1350 CAUGUCU CUGAUGAGGCCGAAAGGCCGAA AGCCAAG
1359 CACGGAA CUGAUGAGGCCGAAAGGCCGAA ACAUGUC
1360 UCACGGA CUGAUGAGGCCGAAAGGCCGAA AACAUGU
1361 UUCACGG CUGAUGAGGCCGAAAGGCCGAA. AAACAUG
1362 UUUCACG CUGAUGAGGCCGAAAGGCCGAA AAAACAU
1386 AACAGCC CUGAUGAGGCCGAAAGGCCGAA AUUGUUC
1393 ACAUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGCCU
1394 UACAUGG CUGAUGAGGCCGAAAGGCCGAA AACAGCC
1401 AGGGGGC CUGAUGAGGCCGAAAGGCCGAA ACAUGGG
1414 AGGCACA CUGAUGAGGCCGAAAGGCCGAA AGGCCAG
1422 UCAAAAG CUGAUGAGGCCGAAAGGCCGAA AGGCACA
1423 AUCAAAA CUGAUGAGGCCGAAAGGCCGAA AAGGCAC
1425 UAAUCAA CUGAUGAGGCCGAAAGGCCGAA AGAAGGC
1426 AUAAUCA CUGAUGAGGCCGAAAGGCCGAA AAGAAGG
1427 CAUAAUC CUGAUGAGGCCGAAAGGCCGAA AAAGAAG
1431 AAAACAU CUGAUGAGGCCGAAAGGCCGAA AUCAAAA
1432 AAAAACA CUGAUGAGGCCGAAAGGCCGAA AAUCAAA
1436 UUUAAAA CUGAUGAGGCCGAAAGGCCGAA ACAUAAU
1437 UUUUAAA CUGAUGAGGCCGAAAGGCCGAA AACAUAA
1438 AUUUUAA CUGAUGAGGCCGAAAGGCCGAA AAACAUA
1439 UAUUUUA CUGAUGAGGCCGAAAGGCCGAA AAAACAU
1440 AUAUUUU CUGAUGAGGCCGAAAGGCCGAA AAAAACA
1441 AAUAUUU CUGAUGAGGCCGAAAGGCCGAA AAAAAAC
1446 CAGAUAA CUGAUGAGGCCGAAAGGCCGAA AUUUUAA
1448 AUCAGAU CUGAUGAGGCCGAAAGGCCGAA AUAUUUU
1449 AAUCAGA CUGAUGAGGCCGAAAGGCCGAA AAUAUUU
1451 UUAAUCA CUGAUGAGGCCGAAAGGCCGAA AUAAUAU
1456 ACAACUU CUGAUGAGGCCGAAAGGCCGAA AUCAGAU
1457 GACAACU CUGAUGAGGCCGAAAGGCCGAA AAUCAGA
1461 UUUAGAC CUGAUGAGGCCGAAAGGCCGAA ACUUAAU
1464 UUGUUUA CUGAUGAGGCCGAAAGGCCGAA ACAACUU
1466 CAUUGUU CUGAUGAGGCCCAAAGGCCGAA AGACAAC 1479 GUCACCA CUGAUGAGGCCGAAAGGCCGAA AUCAGCA
1480 GGUCACC CUGAUGAGGCCGAAAGGCCGAA AAUCAGC 1494 AAUGAGU CUGAUGAGGCCGAAAGGCCGAA ACAGUUG 1498 CAGCAAU CUGAUGAGGCCGAAAGGCCGAA AGUGACA 1501 CCUCAGC CUGAUGAGGCCGAAAGGCCGAA AUGAGUG 1512 GGGAGCA CUGAUGAGGCCGAAAGGCCGAA AGGCCUC 1517 CCCUGGG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG 1528 CAGACAC CUGAUGAGGCCGAAAGGCCGAA ACUCCCU 1533 GAUUACA CUGAUGAGGCCGAAAGGCCGAA ACACAAC 1537 GGCCGAU CUGAUGAGGCCGAAAGGCCGAA ACAGACA 1540 GUAGGCC CUGAUGAGGCCGAAAGGCCGAA AUUACAG 1546 UGAAUAG CUGAUGAGGCCGAAAGGCCGAA AGGCCGA 1549 CACUGAA CUGAUGAGGCCGAAAGGCCGAA AGUAGGC
1551 GCCACUG CUGAUGAGGCCGAAAGGCCGAA AUAGUAG
1552 CGCCACU CUGAUGAGGCCGAAAGGCCGAA AAUAGUA 1566 CAACCUU CUGAUGAGGCCGAAAGGCCGAA AUUUCUC 1572 CCUAAGC CUGAUGAGGCCGAAAGGCCGAA ACCUUUA
1576 CUUUCCU CUGAUGAGGCCGAAAGGCCGAA AGCAACC
1577 UCUUUCC CUGAUGAGGCCGAAAGGCCGAA AAGCAAC
Table 25: Mouse TNF-a HH Target Sequences
nt . HH Target Sequence nt . HH Target Sequenc,
Position Position
66 UgGAAAU a GcucCcA 324 GgGUGAU C GGuCCCC 101 GGCAGGU U CUgUcCC 347 GAGAagU u cCCAaaU 101 GGCAGgU u CuGUccC 364 CCUCcCU C UcAUCAG 102 GCAGGUU C UgUcCCU 366 UCcCUCU c AUCAGuu 102 gCAGgUU c ugUCCCU 366 UcCCUCU C auCAGuU 106 GUUCUgU c CCUuUCA 369 CUCUcAU C AGuuCUa 110 UgUcCCU u UCACucA 376 CAGuuCU a UGGCCCA 111 gUCcCUU u CaCUCAC 390 AgACCCU C AcaCUcA 111 guCCCuU u CACuCAc 396 ucaCAcU C AGAUCAU 112 UcCCUuU C ACucACU 401 cUCAGAU C AUCUUCU 116 UuUCACU C AcUGgcc 404 AGAUCAU C UUCUCaA 137 GCCaCAU C uCCcUCc 406 AUCAUCU U CUCaAAa 139 caCAuCU C CCUCcAg 406 AUcAUcU U cUcaAAA 177 GCAUGAU C CGcGACG 407 UCAUCUU C UCaAAau 207 AGGCaCU C CCCcAaA 409 AUCUUCU C aAAauuC 228 GGGGCuU C CAGAACU 409 AuCuuCU c AaAAUUC 228 GGGGCuU c CAGaacU 409 aUcUUcU c AAAauUc 236 CAGaaCU C CAGGCGG 432 AGCCUGU A GCCCAcG 236 CAGaACU c cAGgcGg
249 GGugCCU a UgUCUcA
249 GGuGCCU a UGucUCa 444 AcGUcGU A GCAAACC
501 AcGCCCU C CUGGCCA
261 UCAGCCU C UUCUCaU 560 gGgUUGU a CCUuguC
261 UCAgCCU C UUCUcau 560 GGguUGU A CCUugUC
263 AGCCUCU U CUCaUUC 564 UGUACCU u gUCUACU
263 AgCCUCU U CUcauUC 567 ACCUugU C UACUCCC
264 GCCUCUU C UCaUUCC 569 CUugUCU A CUCCCAG
264 gCCUCUU C UcauUCc 572 gUCUACU C CCAGGUu
266 CUCUUCU C aUUCCUG 572 GUCUaCU c CCAGguu
269 UUCUCaU U CCUGcUu 572 GuCUacU C CCAgGUu
270 UCUCaUU C CUGcUuG 579 CCCAGGU u CUCUUCA
276 UCCUGcU u GUGGGAG 580 CCAGguU c uCUUcAa
297 CCACGCU C UUCUGuC 580 CCaGGuU c UCuUcaa
299 ACGCUCU U CUGuCUa 582 AGGUUCU C UUCaagg
300 CGCUCUU C UGuCUaC 582 AGGUUCU C UUCAAGG
304 CUuCUgU c uAcUGaa 584 GUuCUCU U CAAGGGa
306 UcUGUcU a cUgAAcU 585 UuCUCUU C AAGGGaC
314 CUGaACU U cGGgGUG 608 CcCGaCU a CgugCUC
315 UGaACUU c GGgGUGA 615 aCgUGcU C CUCAcCC
315 uGaaCUU c GGGguGa 615 ACGUGCU C CUCACCC
324 gGGUGaU c GgUCCcC 618 UGCUCCU C ACCCACA 630 ACACCgU C AGCCGau 940 GuCUACU c cUCAGaG
630 ACACCgU C AgCCgaU 943 UACUccU C AGaGcCc
638 agcCgAU u uGCUaUc 972 UCUaaCU u AgAAAGg
643 aUUUGcU a uCUcAuA 972 ucUaaCU u AGAaAgG
645 UuGCuaU C UCaUACC 973 CUaACuU A GAAAggG
647 GCuaUCU C aUACCAG 984 AGgGgAU U auGGcuc
663 agAAaGU C AACCUCC 984 AGGGgaU U aUGgCUc
669 UCAACCU C CUCUCUG 985 GGGGauU a uGGcUCa
669 UcAAccU C cUcUCUG 997 UcAGAgU c CAAcucu
672 ACCUCCU C UCUGCCg 1010 CuguGCU c AGAgCUU
674 CUCCUCU C UGCCgUC 1017 cAGAgCU U UcAaCAA
681 cUGCCgU C AagaGcC 1018 AGAgCUU U cAaCAAC
681 CUGCCgU C AAGAGCC 1019 GAgCUUU c AaCAACu
681 CUGcCgU C aaGAgcC 1073 UgGGCCU c ucAUgCA
734 CCCUGGU A UGAGCCC 1096 AAGgAcU C AAAugGG
734 CccUGGU a ugaGCCc 1106 aUGGGcU U uccGAAU
744 AGCCCAU a UAcCUGG 1107 UGGGcUU u ccGAAUu
746 CCCAUaU A cCUGGGA 1108 GGgCuUU c cGaaUUC
759 GAgGAGU C uuCCAGc 1115 CcGAAuU C ACUGGaG
759 GAGGaGU C UUCCAGC 1133 CGAAugU C CAuuCcU
761 GGaGUCU U CCAGCUG 1164 gagUGgU c AgGUUGc
762 GaGUCUU C CAGCUGG 1180 UcUgUcU c agaAUGA
786 ACCaACU C AGCGCUG 1203 aaGAuCU c AGGCCUU
798 CUGAGgU C AAUCuGC 1210 cAGGCCU U CCUacCU
802 GgUCAAU C uGCCCaA 1211 AGGCCUU C CUacCUu
812 CCCaAgU A cuUaGAC 1214 CCUUCCU a cCUuCAG
816 AgUAcuU a GACUUUG 1218 CcuACcU u CaGACCu
821 uUaGACU U UGCgGAG 1218 COiaCCU U CAGACcu
822 UaGACUU U GCgGAGU 1213 cCuACcU u cAgACCU
830 GCgGAGU C cGGGCAG 1218 CCUacCU u CAGAccU
840 GGCAGGU C UACUUUG 1219 CuaCCUU C AGACcuu
842 CAGGUCU A CUUUGGa 1219 CuAcCUU c agACcUU
842 CAGgucU a CUUugGA 1226 CagACCU U uCCAgAC
842 cagGuCU a CUUUgGA 1226 CAGACCU U UCCAGAC
845 GUCUACU U UGGagUC 1227 agACCUU u CCAgACu
846 UCUACUU U GGagUCA 1227 AGAccUU U CCAGACU
852 UUGGagU C AUUGCuC 1228 GAccUUU C CAGACUc
855 GagUCAU U GCuCUGU 1238 gACUCuU c cCUGAGG
887 AUCCaUU c ucUACCC 1262 CAGCCuU C CuCAcaG
891 AuucuCU a CCCaGCC 1283 CCCCccU C uaUUUAU
905 CCcCaCU C UgaCCCC 1283 cCcCCCU C UAUUUAU
905 cCCCacU c UgACCCC 1285 CCCCUCU A UUUAUaU
905 CcCCACU c uGAccCC 1287 CcuCUAU u UauAuUU
914 GAcCCcU U uacUCUG 1287 CCUCUAU U UAUaUUU
915 ACCCCuU u acUCuGA 1288 CUCUAUU U AUaUUUG
919 CUUUAcU c ugaCCcC 1289 UCUAUUU A UaUUUGC
928 GACCcCU u UaUugUC 1293 UUUAUaU U UGCACUU
928 gAcCCCU U UAUUguC 1293 uUUaUaU u UGcAcUu
932 CCUUUAU U guCuaCU 1294 UUAUaUU U GCACUUa 1300 UUGCACU U aUuAUUu 1462 aCCuUGU u GCCUCCU
1303 CAcuUaU u AuUuAUU 1470 GccuCcU C UUUUGcU
1304 acUuAUU A UUUAUUA 1472 cuCcUCU u UUGcUUA
1306 UuAUUAU U UAUUAUU 1473 uCcUCUU u UGcUUAU
1307 UAUUAUU U AUUAUUU 1474 CcUCUUU u GcUUAUG
1307 UaUUaUU U AuuAUuU 1478 UUUUGcU u AUGUUUa
1308 AUUAUUU A UUAUUUA 1479 UUUGcUU a UGuuuAa
1310 UauUuAU U AUUUAUU 1479 UUUGcUU A UGUUUaa
1310 UAUUUAU U AUUUAUU 1484 UUAUGUU U aaaAcAA
1310 UAUUUAU U AUUUAUU 1498 AAAuauU U AUCUaAc
1311 AUUUAUU A UUUAUUU 1511 AcccAaU U GUCUuAA
1311 AUUUAUU A UUUAUUU 1514 cAaUUGU c UuAAuAA
1311 AuuUAUU A UuUauUU 1516 aUUGUCU u AAuAAcG
1313 UUAUUAU U UAUUUAU 1529 CgcugAU u UGGuGAC
1313 UUAUUAU U UAUUUAU 1529 cGCUGAU U UGGUGAC
1313 uUAUUAU u UauUUAu 1530 gCUGAUU u gGUgacC
1314 UAUUAUU U AUUUAUU 1530 GCUGAUU U GGUGACC
1314 UAUUAUU U AUUUAUU 1563 UgaAcCU c UGcUCCC
1315 AUUAUUU A UUUAUUA 1563 ugaaCCU C UGCUCCC
1317 UAUUUAU U UAUUAUU 1568 CUCUGCU C CCCAcGG
1318 AUUUAUU U AUUAUUU 1589 UGaCUGU A AUuGcCC
1319 UUUAUUU A UUAUUUA 1592 CUGUAAU u GcCCUAC
1326 AUUAUUU A UUUAUUU 1617 GAGAAAU A AAGaUcG
1328 UAUUUAU U UAUUUgC 1623 UAAAGaU c GCUUAaa
1329 AUUUAUU U AUUUgCu 1633 UUAaaaU a aaAAaCC
1330 UUUAUUU A UUUgCuu 25 AgGgaCU a gCCagGA
1332 UAUUUAU U UgCuuAU
1333 AUUUAUU U gCuuAUG
1337 auUUGCU U AuGAAuG
1338 uUUGCUU A uGAAuGu
1346 UGAAUGU A UUUAUUU
1348 AAUGUAU U UAUUUGG
1349 AUGUAUU U AUUUGGa
1350 UGUAUUU A UUUGGaA
1352 uAUuUAU u UGGaAGG
1352 UAUUUAU U UGGaAGg
1353 AUUUAUU U GGaAGgC
1369 GGGGUgU C CUGGaGG
1398 gCUguCU U cAGACAg
1398 GCUGuCU U cagaCAG
1412 GACAUGU U UUCuGUG
1413 ACAUGUU U UCuGUGA
1414 CAUGUUU U CuGUGAA
1415 AUGUUUU C UGUGAAA
1415 AUGUUUU c UgugAaA
1438 gaGCUGU c CCCAccU
1451 CUGGCCU C UcUaCCU
1453 ggCCUCU C UaCCuUG Table 26: Mouse TNF-α Hammerhead Ribozyme Sequences
nt . Mouse HH Ribozyme Sequence
Position
25 UCCUGGC CUGAUGAGGCCGAAAGGCCGAA AGUCCCU
66 UGGGAGC CUGAUGAGGCCGAAAGGCCGAA AUUUCCA
101 GGGACAG CUGAUGAGGCCGAAAGGCCGAA ACCUGCC
101 GGGACAG CUGAUGAGGCCGAAAGGCCGAA ACCUGCC
102 AGGGACA CUGAUGAGGCCGAAAGGCCGAA AACCUGC 102 AGGGACA CUGAUGAGGCCGAAAGGCCGAA AACCUGC 106 UGAAAGG CUGAUGAGGCCGAAAGGCCGAA ACAGAAC
110 UGAGUGA CUGAUGAGGCCGAAAGGCCGAA AGGGACA
111 GϋGAGUG CUGAUGAGGCCGAAAGGCCGAA AAGGGAC
111 GUGAGUG CUGAUGAGGCCGAAAGGCCGAA AAGGGAC
112 AGUGAGU CUGAUGAGGCCGAAAGGCCGAA AAAGGGA 116 GGCCAGU CUGAUGAGGCCGAAAGGCCGAA AGUGAAA 137 GGAGGGA CUGAUGAGGCCGAAAGGCCGAA AUGUGGC 139 CUGGAGG CUGAUGAGGCCGAAAGGCCGAA AGAUGUG 177 CGUCGCG CUGAUGAGGCCGAAAGGCCGAA AUCAUGC 207 UUUGGGG CUGAUGAGGCCGAAAGGCCGAA AGUGCCU 228 AGUUCUG CUGAUGAGGCCGAAAGGCCGAA AAGCCCC 228 AGUUCUG CUGAUGAGGCCGAAAGGCCGAA AAGCCCC 236 CCGCCUG CUGAUGAGGCCGAAAGGCCGAA AGUUCUG 236 CCGCCUG CUGAUGAGGCCGAAAGGCCGAA AGUUCUG 249 UGAGACA CUGAUGAGGCCGAAAGGCCGAA AGGCACC 249 UGAGACA CUGAUGAGGCCGAAAGGCCGAA AGGCACC
261 AUGAGAA CUGAUGAGGCCGAAAGGCCGAA AGGCUGA
261 AUGAGAA CUGAUGAGGCCGAAAGGCCGAA AGGCUGA
263 GAAUGAG CUGAUGAGGCCGAAAGGCCGAA AGAGGCU
263 GAAUGAG CUGAUGAGGCCGAAAGGCCGAA AGAGGCU
264 GGAAUGA CUGAUGAGGCCGAAAGGCCGAA AAGAGGC 264 GGAAUGA CUGAUGAGGCCGAAAGGCCGAA AAGAGGC 266 CAGGAAU CUGAUGAGGCCGAAAGGCCGAA AGAAGAG
269 AAGCAGG CUGAUGAGGCCGAAAGGCCGAA AUGAGAA
270 CAAGCAG CUGAUGAGGCCGAAAGGCCGAA AAUGAGA 276 CUGCCAC CUGAUGAGGCCGAAAGGCCGAA AGCAGGA 297 GACAGAA CUGAUGAGGCCGAAAGGCCGAA AGCGUGG
299 UAGACAG CUGAUGAGGCCGAAAGGCCGAA AGAGCGU
300 GUAGACA CUGAUGAGGCCGAAAGGCCGAA AAGAGCG 304 UUCAGUA CUGAUGAGGCCGAAAGGCCGAA ACAGAAG 306 AGUUCAG CUGAUGAGGCCGAAAGGCCGAA AGACAGA
314 CACCCCG CUGAUGAGGCCGAAAGGCCGAA AGUUCAG
315 UCACCCC CUGAUGAGGCCGAAAGGCCGAA AAGUUCA 315 UCACCCC CUGAUGAGGCCGAAAGGCCGAA AAGUUCA
324 GGGGACC CUGAUGAGGCCGAAAGGCCGAA AUCACCC
324 GGGGACC CUGAUGAGGCCGAAAGGCCGAA AUCACCC
347 AUUUGGG CUGAUGAGGCCGAAAGGCCGAA ACUUCUC
364 CUGAUGA CUGAUGAGGCCGAAAGGCCGAA AGGGAGG
366 AACUGAU CUGAUGAGGCCGAAAGGCCGAA AGAGGGA
366 AACUGAU CUGAUGAGGCCGAAAGGCCGAA AGAGGGA
369 UAGAACU CUGAUGAGGCCGAAAGGCCGAA AUGAGAG
376 UGGGCCA CUGAUGAGGCCGAAAGGCCGAA AGAACUG
390 UGAGUGU CUGAUGAGGCCGAAAGGCCGAA AGGGCCU
396 AUGAUCU CUGAUGAGGCCGAAAGGCCGAA AGUGUGA
401 AGAAGAU CUGAUGAGGCCGAAAGGCCGAA AUCUGAG
404 UUGAGAA CUGAUGAGGCCGAAAGGCCGAA AUGACCU
406 UUUUGAG CUGAUGAGGCCGAAAGGCCGAA AGAUGAU
406 UUUUGAG CUGAUGAGGCCGAAAGGCCGAA AGAUGAU
407 AUUUUGA CUGAUGAGGCCGAAAGGCCGAA AAGAUGA 409 GAAUUUU CUGAUGAGGCCGAAAGGCCGAA AGAAGAU 409 GAAUUUU CUGAUGAGGCCGAAAGGCCGAA AGAAGAU 409 GAAUUUU CUGAUGAGGCCGAAAGGCCGAA AGAAGAU 432 CGUGGGC CUGAUGAGGCCGAAAGGCCGAA ACAGGCU
444 GGUUUGC CUGAUGAGGCCGAAAGGCCGAA ACGACGU
501 UGGCCAG CUGAUGAGGCCGAAAGGCCGAA AGGGCGU
560 GACAAGG CUGAUGAGGCCGAAAGGCCGAA ACAACCC
560 GACAAGG CUGAUGAGGCCGAAAGGCCGAA ACAACCC
564 AGUAGAC CUGAUGAGGCCGAAAGGCCGAA AGGUACA
567 GGGAGUA CUGAUGAGGCCGAAAGGCCGAA ACAAGGU
569 CUGGGAG CUGAUGAGGCCGAAAGGCCGAA AGACAAG 572 AACCUGC CUGAUGAGGCCGAAAGGCCGAA AGUAGAC
572 AACCUGG CUGAUGAGGCCGAAAGGCCGAA AGUAGAC
572 AACCUGG CUGAUGAGGCCGAAAGGCCGAA AGUAGAC
579 UGAAGAG CUGAUGAGGCCGAAAGGCCGAA ACCUGGG 530 UUGAAGA CUGAUGAGGCCGAAAGGCCGAA AACCUGG
580 UUGAAGA CUGAUGAGGCCGAAAGGCCGAA AACCUGG 532 CCUUGAA CUGAUGAGGCCGAAAGGCCGAA AGAACCU 532 CCUUGAA CUGAUGAGGCCGAAAGGCCGAA AGAACCU 534 UCCCUUG CUGAUGAGGCCGAAAGGCCGAA AGAGAAC 585 GUCCCUU CUGAUGAGGCCGAAAGGCCGAA AAGAGAA 608 GAGCACG CUGAUGAGGCCGAAAGGCCGAA AGUCGGG 615 GGGUGAG CUGAUGAGGCCGAAAGGCCGAA AGCACGU 615 GGGUGAG CUGAUGAGGCCGAAAGGCCGAA AGCACGU 618 UGUGGGU CUGAUGAGGCCGAAAGGCCGAA AGGAGCA 630 AUCGGCU CUGAUGAGGCCGAAAGGCCGAA ACGGUGU 630 AUCGGCU CUGAUGAGGCCGAAAGGCCGAA ACGGUGU 638 GAUAGCA CUGAUGAGGCCGAAAGGCCGAA AUCGGCU 643 UAUGAGA CUGAUGAGGCCGAAAGGCCGAA AGCAAAU 645 GGUAUGA CUGAUGAGGCCGAAAGGCCGAA AUAGCAA 647 CUGGUAU CUGAUGAGGCCGAAAGGCCGAA AGAUAGC 663 GGAGGUU CUGAUGAGGCCGAAAGGCCGAA ACUUUCU
669 CAGAGAG CUGAUGAGGCCGAAAGGCCGAA AGGUUGA
669 CAGAGAG CUGAUGAGGCCGAAAGGCCGAA AGGUUGA
672 CGGCAGA CUGAUGAGGCCGAAAGGCCGAA AGGAGGU
674 GACGGCA CUGAUGAGGCCGAAAGGCCGAA AGAGGAG
681 GGCUCUU CUGAUGAGGCCGAAAGGCCGAA ACGGCAG
681 GGCUCUU CUGAUGAGGCCGAAAGGCCGAA ACGGCAG
631 GGCUCUU CUGAUGAGGCCGAAAGGCCGAA ACGGCAG
734 GGGCUCA CUGAUGAGGCCGAAAGGCCGAA ACCAGGG
734 GGGCUCA CUGAUGAGGCCGAAAGGCCGAA ACCAGGG
744 CCAGGUA CUGAUGAGGCCGAAAGGCCGAA AUGGGCU
746 UCCCAGG CUGAUGAGGCCGAAAGGCCGAA ACAUGGG
759 GCUGGAA CUGAUGAGGCCGAAAGGCCGAA ACUCCUC
759 GCUGGAA CUGAUGAGGCCGAAAGGCCGAA ACUCCUC
761 CAGCUGG CUGAUGAGGCCGAAAGGCCGAA AGACCCC
762 CCAGCUG CUGAUGAGGCCGAAAGGCCGAA AAGACUC 786 CAGCGCU CUGAUGAGGCCGAAAGGCCGAA AGUUGGU 798 GCAGAUU CUGAUGAGGCCGAAAGGCCGAA ACCUCAG 802 UUGGGCA CUGAUGAGGCCGAAAGGCCGAA AUUGACC 312 GUCUAAG CUGAUGAGGCCGAAAGGCCGAA ACUUGGG 816 CAAAGUC CUGAUGAGGCCGAAAGGCCGAA AAGUACU
821 CUCCGCA CUGAUGAGGCCGAAAGGCCGAA AGUCUAA
822 ACUCCGC CUGAUGAGGCCGAAAGGCCGAA AAGUCUA 830 CUGCCCG CUGAUGAGGCCGAAAGGCCGAA ACUCCGC 840 CAAAGUA CUGAUGAGGCCGAAAGGCCGAA ACCUGCC 842 UCCAAAG CUGAUGAGGCCGAAAGGCCGAA AGACCUG 842 UCCAAAG CUGAUGAGGCCGAAAGGCCGAA AGACCUG 842 UCCAAAG CUGAUGAGGCCGAAAGGCCGAA AGACCUG
845 GACUCCA CUGAUGAGGCCGAAAGGCCGAA AGUAGAC
846 UGACUCC CUGAUGAGGCCGAAAGGCCGAA AAGUAGA 852 GAGCAAU CUGAUGAGGCCGAAAGGCCGAA ACUCCAA 855 ACAGAGC CUGAUGAGGCCGAAAGGCCGAA AUGACUC 887 GGGUAGA CUGAUGAGGCCGAAAGGCCGAA AAUGGAU 891 GGCUGGG CUGAUGAGGCCGAAAGGCCGAA AGAGAAU 905 GGGGUCA CUGAUGAGGCCGAAAGGCCGAA AGUGGGG 905 GGGCUCA CUGAUGAGGCCGAAAGGCCGAA AGUGGGG 905 GGGGUCA CUGAUGAGGCCGAAAGGCCGAA AGUGGGG
914 CAGAGUA CUGAUGAGGCCGAAAGGCCGAA AGGGGUC
915 UCAGAGU CUGAUGAGGCCGAAAGGCCGAA AAGGGGU 919 GGGGUCA CUGAUGAGGCCGAAAGGCCGAA AGUAAAG 928 GACAAUA CUGAUGAGGCCGAAAGGCCGAA AGGGGUC 928 GACAAUA CUGAUGAGGCCGAAAGGCCGAA AGGGGUC 932 AGUAGAC CUGAUGAGGCCGAAAGGCCGAA AUAAAGG 940 CUCUGAG CUGAUGAGGCCGAAAGGCCGAA AGUAGAC 943 GGGCUCU CUGAUGAGGCCGAAAGGCCGAA AGGAGUA 972 CCUUUCU CUGAUGAGGCCGAAAGGCCGAA AGUUAGA
972 CCUUUCU CUGAUGAGGCCGAAAGGCCGAA AGUUAGA
973 CCCUUUC CUGAUGAGGCCGAAAGGCCGAA AAGUUAG 984 GAGCCAU CUGAUGAGGCCGAAAGGCCGAA AUCCCCU 984 GAGCCAU CUGAUGAGGCCGAAAGGCCGAA AUCCCCU
985 UGAGCCA CUGAUGAGGCCGAAAGGCCGAA AAUCCCC
997 AGAGUUG CUGAUGAGGCCGAAAGGCCGAA ACUCUGA
1010 AAGCUCU CUGAUGAGGCCGAAAGGCCGAA AGCACAG
1017 UUGUUGA CUGAUGAGGCCGAAAGGCCGAA AGCUCUG
1018 GUUGUUG CUGAUGAGGCCGAAAGGCCGAA AAGCUCU
1019 AGUUGUU CUGAUGAGGCCGAAAGGCCGAA AAAGGUC
1073 UGCAUGA CUGAUGAGGCCGAAAGGCCGAA AGGCCCA
1096 CCCAUUU CUGAUGAGGCCGAAAGGCCGAA AGUCCUU
1106 AUUCGGA CUGAUGAGGCCGAAAGGCCGAA AGCCCAU
1107 AAUUCGG CUGAUGAGGCCGAAAGGCCGAA AAGCCCA
1108 GAAUUCG CUGAUGAGGCCGAAAGGCCGAA AAAGCCC
1115 CUCCAGU CUGAUGAGGCCGAAAGGCCGAA AAUUCGG
1133 AGGAAUG CUGAUGAGGCCGAAAGGCCGAA ACAUUCG
1164 GCAACCU CUGAUGAGGCCGAAAGGCCGAA ACCACUC
1180 UCAUUCU CUGAUGAGGCCGAAAGGCCGAA AGACAGA
1203 AAGGCCU CUGAUGAGGCCGAAAGGCCGAA AGAUCUU
1210 AGGUAGG CUGAUGAGGCCGAAAGGCCGAA AGGCCUG
1211 AAGGUAG CUGAUGAGGCCGAAAGGCCGAA AAGGCCU
1214 CUGAAGG CUGAUGAGGCCGAAAGGCCGAA AGGAAGG
1218 AGGUCUG CUGAUGAGGCCGAAAGGCCGAA AGGUAGG
1218 AGGUCUG CUGAUGAGGCCGAAAGGCCGAA AGGUAGG
1218 AGGUCUG CUGAUGAGGCCGAAAGGCCGAA AGGUAGG
1218 AGGUCUG CUGAUGAGGCCGAAAGGCCGAA AGGUAGG
1219 AAGCUCU CUGAUGAGGCCGAAAGGCCGAA AAGGUAG
1219 AAGCUCU CUGAUGAGGCCGAAAGGCCGAA AAGGUAG
1226 GUCUGGA CUGAUGAGGCCGAAAGGCCGAA AGGUCUG
1226 GUCUGGA CUGAUGAGGCCGAAAGGCCGAA AGGUCUG
1227 AGUCUGG CUGAUGAGGCCGAAAGGCCGAA AAGGUCU
1227 AGUCUGG CUGAUGAGGCCGAAAGGCCGAA AAGGUCU
1228 GAGUCUG CUGAUGAGGCCGAAAGGCCGAA AAAGGUC
1238 CCUCAGG CUGAUGAGGCCGAAAGGCCGAA AAGAGUC
1262 CUGUGAG CUGAUGAGGCCGAAAGGCCGAA AAGGCUG
1283 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AGGGGGG
1283 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AGGGGGG
1285 AUAUAAA CUGAUGAGGCCGAAAGGCCGAA AGAGGGG
1287 AAAUAUA CUGAUGAGGCCGAAAGGCCGAA AUAGAGG
1287 AAAUAUA CUGAUGAGGCCGAAAGGCCGAA AUAGAGG
1288 CAAAUAU CUGAUGAGGCCGAAAGGCCGAA AAUAGAG
1289 GCAAAUA CUGAUGAGGCCGAAAGGCCGAA AAAUAGA
1293 AAGUGCA CUGAUGAGGCCGAAAGGCCGAA AUAUAAA
1293 AAGUGCA CUGAUGAGGCCGAAAGGCCGAA AUAUAAA
1294 UAAGUGC CUGAUGAGGCCGAAAGGCCGAA AAUAUAA
1300 AAAUAAU CUGAUGAGGCCGAAAGGCCGAA AGUGCAA
1303 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAAGUG
1304 UAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUAAGU.
1306 AAUAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
1307 AAAUAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUA
1307 AAAUAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUA 1308 UAAAUAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAU
1310 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1310 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1310 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1311 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1311 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1311 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1313 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
1313 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
1313 AUAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
1314 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUA
1314 AAUAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAUA
1315 UAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAU
1317 AAUAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1318 AAAUAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1319 UAAAUAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAA
1325 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAU
1328 GCAAAUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1329 AGCAAAU CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1330 AAGCAAA CUGAUGAGGCCGAAAGGCCGAA AAAUAAA
1332 AUAAGCA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1333 CAUAAGC CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1337 CAUUCAU CUGAUGAGGCCGAAAGGCCGAA AGCAAAU
1338 ACAUUCA CUGAUGAGGCCGAAAGGCCGAA AAGCAAA
1346 AAAUAAA CUGAUGAGGCCGAAAGGCCGAA ACAUUCA
1348 CCAAAUA CUGAUGAGGCCGAAAGGCCGAA AUACAUU
1349 UCCAAAU CUGAUGAGGCCGAAAGGCCGAA AAUACAU
1350 UUCCAAA CUGAUGAGGCCGAAAGGCCGAA AAAUACA
1352 CCUUCCA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1352 CCUUCCA CUGAUGAGGCCGAAAGGCCGAA AUAAAUA
1353 GCCUUCC CUGAUGAGGCCGAAAGGCCGAA AAUAAAU
1369 CCUCCAG CUGAUGAGGCCGAAAGGCCGAA ACACCCC
1398 CUGUCUG CUGAUGAGGCCGAAAGGCCGAA AGACAGC
1398 CUGUCUG CUGAUGAGGCCGAAAGGCCGAA AGACAGC
1412 CACAGAA CUGAUGAGGCCGAAAGGCCGAA ACAUGUC
1413 UCACAGA CUGAUGAGGCCGAAAGGCCGAA AACAUGU
1414 UUCACAG CUGAUGAGGCCGAAAGGCCGAA AAACAUG
1415 UUUCACA CUGAUGAGGCCGAAAGGCCGAA AAAACAU
1415 UUUCACA CUGAUGAGGCCGAAAGGCCGAA AAAACAU
1438 AGGUGGG CUGAUGAGGCCGAAAGGCCGAA ACAGCUC
1451 AGGUAGA CUGAUGAGGCCGAAAGGCCGAA AGGGCAG
1453 CAAGGUA CUGAUGAGGCCGAAAGGCCGAA AGAGGCC
1455 AACAAGG CUGAUGAGGCCGAAAGGCCGAA AGAGAGG
1462 AGGAGGC CUGAUGAGGCCGAAAGGCCGAA ACAAGCU
1470 AGCAAAA CUGAUGAGGCCGAAAGGCCGAA AGGAGGC
1472 UAAGCAA CUGAUGAGGCCGAAAGGCCGAA AGAGGAG
1473 AUAAGCA CUGAUGAGGCCGAAAGGCCGAA AAGAGGA
1474 CAUAAGC CUGAUGAGGCCGAAAGGCCGAA AAAGAGG
1473 UAAACAU CUGAUGAGGCCGAAAGGCCGAA AGCAAAA 1479 UUAAACA CUGAUGAGGCCGAAAGGCCGAA AAGCAAA
1479 UUAAACA CUGAUGAGGCCGAAAGGCCGAA AAGCAAA
1484 UUGUUUU CUGAUGAGGCCGAAAGGCCGAA AACAUAA
1498 GUUAGAU CUGAUGAGGCCGAAAGGCCGAA AAUAUUU
1511 UUAAGAC CUGAUGAGGCCGAAAGGCCGAA AUUGGGU
1514 UUAUUAA CUGAUGAGGCCGAAAGGCCGAA ACAAUUG
1516 CGUUAUU CUGAUGAGGCCCAAAGGCCGAA AGACAAU
1529 GUCACCA CUGAUGAGGCCGAAAGGCCGAA AUCAGCG
1529 GUCACCA CUGAUGAGGCCGAAAGGCCGAA AUCAGCG
1530 GGUCACC CUGAUGAGGCCGAAAGGCCGAA AAUCAGC 1530 GGUCACC CUGAUGAGGCCGAAAGGCCGAA AAUCAGC 1563 GGGAGCA CUGAUGAGGCCGAAAGGCCGAA AGGUUCA 1563 GGGAGCA CUGAUGAGGCCGAAAGGCCGAA AGGUUCA 1563 CCGUGGG CUGAUGAGGCCGAAAGGCCGAA AGCAGAG 1589 GGGCAAU CUGAUGAGGCCGAAAGGCCGAA ACAGUCA 1592 GUAGGGC CUGAUGAGGCCGAAAGGCCGAA AUUACAG 1617 CGAUCUU CUGAUGAGGCCGAAAGGCCGAA AUUUCUC 1623 UUUAAGC CUGAUGAGGCCGAAAGGCCGAA AUCUUUA 1633 GGUUUUU CUGAUGAGGCCGAAAGGCCGAA AUUUUAA
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Table 29: Human bcr/abl HH Target Sequence
Sequence HH Target Sequence
ID NO.
b2 -a2
Junction
20 UGACCAUCA AUA AGGAAGAAGCC
21 GAAGAAGCC CUU CAGCGGGCAGU 22 AAGAAGCCC UUC AGCGGCCAGUA b3-a2
Junction
23 UAAGCAGAG UUC AAAAGCCCUUC 24 UCAAAAGCC CUU CAGCGGCCAGU 25 CAAAAGCCC UUC AGCGGCCAGUA
Table 30: Human bcr-abl HH Ribozyme Sequences
Sequence HH Ribozyme Sequence
ID No .
26 GGCUUCUUCCU CUGAUGAGGCCGAAAGGCCGAA AUUGAUGGUCA
27 ACUGGCCGCUG CUGAUGAGGCCGAAAGGCCGAA AGGGCUUCUUC
28 UACUGGCCGCU CUGAUGAGGCCGAAAGGCCGAA AAGGGCUUCUU
29 GAAGGGCUUUU CUGAUGAGGCCGAAAGGCCGAA AACUCUGCUUA
30 ACUGGCCGCUG CUGAUGAGGCCGAAAGGCCGAA AGGGCUUUUGA
31 UACUGGCCGCU CUGAUGAGGCCGAAAGGCCGAA AAGGGCUUUUG
Table 31: RSV (1B) HH Target Sequence
nt. HH Target Sequence nt. HH Target Sequence Position Position
10 GGCAAAU A AAUCAAU 276 AAAAUAU A CUGAAUA
14 AAUAAAU C AAUUCAG 283 ACUGAAU A CAACACA
18 AAUCAAU U CAGCCAA 295 ACAAAAU A UGGCACU
19 AUCAAUU C AGCCAAC 303 UGGCACU U UCCCUAU
54 CAAUGAU A AUACACC 304 GGCACUU U CCCUAUG
57 UGAUAAU A CACCACA 305 GCACUUU C CCUAUGC
77 UGAUGAU C ACAGACA 309 UUUCCCU A UGCCAAU
94 AGACCGU U GUCACUU 317 UGCCAAU A UUCAUCA
97 CCGUUGU C ACUUGAG 319 CCAAUAU U CAUCAAU
101 UGUCACU U GAGACCA 320 CAAUAUU C AUCAAUC
110 AGACCAU A AUAACAU 323 UAUUCAU C AAUCAUG
113 CCAUAAU A ACAUCAC 327 CAUCAAU C AUGAUGG
118 AUAACAU C ACUAACC 337 GAUGGGU U CUUAGAA
122 CADCACU A ACCAGAG 338 AUGGGUU C UUAGAAU
134 GAGACAU C AUAACAC 340 GGGUUCU U AGAAUGC 137 ACAUCAU A ACAGACA 341 GGUUCUU A GAAUGCA
148 CACAAAU U UAUAUAC 350 AAUGCAU U GGCAUUA 149 ACAAAUU U AUAUACU 356 UUGGCAU U AAGCCUA 150 CAAAUUU A UAUACUU 357 UGGCAUU A AGCCUAC 152 AAUUUAU A UACUUGA 363 UAAGCCU A CAAAGCA 154 UUUAUAU A CUUGAUA 372 AAAGCAU A CUCCCAU 157 AUAUACU U GAUAAAU 375 GCAUACU C CCAUAAU 161 ACUUGAU A AAUCAUG 380 CUCCCAU A AUAUACA 165 GAUAAAU C AUGAAUG 383 CCAUAAU A UACAAGU 176 AAUGCAU A GUGAGAA 385 AUAAUAU A CAAGUAU 188 GAAAACU U GAUGAAA 391 UACAAGU A UGAUCUC 208 GCCACAU U UACAUUC 396 GUAUGAU C UCAAUCC 209 CCACAUU U ACAUUCC 398 AUGAUCU C AAUCCAU 210 CACAUUU A CAUUCCU 402 UCUCAAU C CAUAAAU 214 UUUACAU U CCUGGUC 406 AAUCCAU A AAUUUCA 215 UUACAUU C CUGGUCA 410 CAUAAAU U UCAACAC 221 UCCUGGU C AACUAUG 411 AUAAAUU U CAACACA 226 GUCAACU A UGAAAUG 412 UAAAUUU C AACACAA 239 UGAAACU A UUACACA 421 ACACAAU A UUCACAC 241 AAACUAU U ACACAAA 423 ACAAUAU U CACACAA 242 AACUAUU A CACAAAG 424 CAAUAUU C ACACAAU 251 ACAAAGU A GGAAGCA 432 ACACAAU C UAAAACA 261 AAGCACU A AAUAUAA 434 ACAAUCU A AAACAAC 265 ACUAAAU A UAAAAAA 446 AACAACU C UAUGCAU 267 UAAAUAU A AAAAAUA 448 CAACUCU A UGCAUAA 274 AAAAAAU A UACUGAA 454 UAUGCAU A ACUAUAC 458 CAUAACU A UACUCCA
460 UAACUAU A CUCCAUA
463 CUAUACU C CAUAGUC
467 ACUCCAU A GUCCAGA
470 CCAUAGU C CAGAUGG
489 UGAAAAU U AUAGUAA
490 GAAAAUU A UAGUAAU
492 AAAUUAU A GUAAUUU
495 UUAUAGU A AUUUAAA
Table 32: RSV (IB) HH Ribozyme Sequence nt. HH Ribozyme Sequence
Position
10 AUUGAUU CUGAUGAGGCCGAAAGGCCGAA AUUUGCC
14 CUGAAUU CUGAUGAGGCCGAAAGGCCGAA AUUUAUU
18 UUGGCUG CUGAUGAGGCCGAAAGGCCGAA AUUGAUU
19 GUUGGCU CUGAUGAGGCCGAAAGGCCGAA AAUUGAU 54 GGUGUAU CUGAUGAGGCCGAAAGGCCGAA AUCAUUG 57 UGUGGUG CUGAUGAGGCCGAAAGGCCGAA AUUAUCA 77 UGUCUGU CUGAUGAGGCCGAAAGGCCGAA AUCAUCA 94 AAGUGAC CUGAUGAGGCCGAAAGGCCGAA ACGGUCU 97 CUCAAGU CUGAUGAGGCCGAAAGGCCGAA ACAACGG
101 UGGUCUC CUGAUGAGGCCGAAAGGCCGAA AGUGACA
110 AUGUUAU CUGAUGAGGCCGAAAGGCCGAA AUGGUCU
113 GUGAUGU CUGAUGAGGCCGAAAGGCCGAA AUUAUGG
118 GGUUAGU CUGAUGAGGCCGAAAGGCCGAA AUGUUAU
122 CUCUGGU CUGAUGAGGCCGAAAGGCCGAA AGUGAUG
134 GUGUUAU CUGAUGAGGCCGAAAGGCCGAA AUGUCUC
137 UGUGUGU CUGAUGAGGCCGAAAGGCCGAA AUGAUGU
148 GUAUAUA CUGAUGAGGCCGAAAGGCCGAA AUUUGUG
149 AGUAUAU CUGAUGAGGCCGAAAGGCCGAA AAUUUGU
150 AAGUAUA CUGAUGAGGCCGAAAGGCCGAA AAAUUUG 152 UCAAGUA CUGAUGAGGCCGAAAGGCCGAA AUAAAUU 154 UAUCAAG CUGAUGAGGCCGAAAGGCCGAA AUAUAAA 157 AUUUAUC CUGAUGAGGCCGAAAGGCCGAA AGUAUAU 161 CAUGAUU CUGAUGAGGCCGAAAGGCCGAA AUCAAGU 165 CAUUCAU CUGAUGAGGCCGAAAGGCCGAA AUUUAUC 176 UUCUCAC CUGAUGAGGCCGAAAGGCCGAA AUGCAUU 188 UUUCAUC CUGAUGAGGCCGAAAGGCCGAA AGUUUUC
208 GAAUGUA CUGAUGAGGCCGAAAGGCCGAA AUGUGGC
209 GGAAUGU CUGAUGAGGCCGAAAGGCCGAA AAUGUGG
210 AGGAAUG CUGAUGAGGCCGAAAGGCCGAA AAAUGUG
214 GACCAGG CUGAUGAGGCCGAAAGGCCGAA AUGUAAA
215 UGACCAG CUGAUGAGGCCGAAAGGCCGAA AAUGUAA 221 CAUAGUU CUGAUGAGGCCGAAAGGCCGAA ACCAGGA 226 CAUUUCA CUGAUGAGGCCGAAAGGCCGAA AGUUGAC 239 UGUGUAA CUGAUGAGGCCGAAAGGCCGAA AGUUUCA
241 UUUGUGU CUGAUGAGGCCGAAAGGCCGAA AUAGUUU
242 CUUUGUG CUGAUGAGGCCGAAAGGCCGAA AAUAGUU 251 UGCUUCC CUGAUGAGGCCGAAAGGCCGAA ACUUUGU 261 UUAUAUU CUGAUGAGGCCGAAAGGCCGAA AGUGCUU 265 UUUUUUA CUGAUGAGGCCGAAAGGCCGAA AUUUAGU 267 UAUUUUU CUGAUGAGGCCGAAAGGCCGAA AUAUUUA 74 UUCAGUA CUGAUGAGGCCGAAAGGCCGAA AUUUUUU 276 UAUUCAG CUGAUGAGGCCGAAAGGCCGAA AUAUUUU 283 UGUGUUG CUGAUGAGGCCGAAAGGCCGAA AUUCAGU
295 AGUGCCA CUGAUGAGGCCGAAAGGCCGAA AUUUUGU
303 AUAGGGA CUGAUGAGGCCGAAAGGCCGAA AGUGCCA
304 CAUAGGG CUGAUGAGGCCGAAAGGCCGAA AAGUGCC
305 GCAUAGG CUGAUGAGGCCGAAAGGCCGAA AAAGUGC 309 AUUGGCA CUGAUGAGGCCGAAAGGCCGAA AGGGAAA 317 UGAUGAA CUGAUGAGGCCGAAAGGCCGAA AUUGGCA
319 AUUGAUG CUGAUGAGGCCGAAAGGCCGAA AUAUUGG
320 GAUUGAU CUGAUGAGGCCGAAAGGCCGAA AAUAUCG 323 CAUGAUU CUGAUGAGGCCGAAAGGCCGAA AUGAAUA 327 CCAUCAU CUGAUGAGGCCGAAAGGCCGAA AUUGAUG
337 UUCUAAG CUGAUGAGGCCGAAAGGCCGAA ACCCAUC
338 AUUCUAA CUGAUGAGGCCGAAAGGCCGAA AACCCAU
340 GCAUUCU CUGAUGAGGCCGAAAGGCCGAA AGAACCC
341 UGCAUUC CUGAUGAGGCCGAAAGGCCGAA AAGAACC 350 UAAUGCC CUGAUGAGGCCGAAAGGCCGAA AUGCAUU
356 UAGGCUU CUGAUGAGGCCGAAAGGCCGAA AUGCCAA
357 GUAGGCU CUGAUGAGGCCGAAAGGCCGAA AAUGCCA 363 UGCUUUG CUGAUGAGGCCGAAAGGCCGAA AGGCUUA 372 AUGGGAG CUGAUGAGGCCGAAAGGCCGAA AUGCUUU 375 AUUAUGG CUGAUGAGGCCGAAAGGCCGAA AGUAUGC 380 UGUAUAU CUGAUGAGGCCGAAAGGCCGAA AUGGGAG 383 ACUUGUA CUGAUGAGGCCGAAAGGCCGAA AUUAUGG 385 AUACUUG CUGAUGAGGCCGAAAGGCCGAA AUAUUAU 391 GAGAUCA CUGAUGAGGCCGAAAGGCCGAA ACUUGUA 396 GGAUUGA CUGAUGAGGCCGAAAGGCCGAA AUCAUAC 398 AUGGAUU CUGAUGAGGCCGAAAGGCCGAA AGAUCAU 402 AUUUAUG CUGAUGAGGCCGAAAGGCCGAA AUUGAGA 406 UGAAAUU CUGAUGAGGCCGAAAGGCCGAA AUGGAUU
410 GUGUUGA CUGAUGAGGCCGAAAGGCCGAA AUUUAUG
411 UGUGUUG CUGAUGAGGCCGAAAGGCCGAA AAUUUAU
412 UUGUGUU CUGAUGAGGCCGAAAGGCCGAA AAAUUUA 421 GUGUGAA CUGAUGAGGCCGAAAGGCCGAA AUUGUGU
423 UUGUGUG CUGAUGAGGCCGAAAGGCCGAA AUAUUGU
424 AUUGUGU CUGAUGAGGCCGAAAGGCCGAA AAUAUUG 432 UGUUUUA CUGAUGAGGCCGAAAGGCCGAA AUUGUGU 434 GUUGUUU CUGAUGAGGCCGAAAGGCCGAA AGAUUGU 446 AUGGAUA CUGAUGAGGCCGAAAGGCCGAA AGUUGUU 448 UUAUGCA CUGAUGAGGCCGAAAGGCCGAA AGAGUUG 454 GUAUAGU CUGAUGAGGCCGAAAGGCCGAA AUGCAUA 458 UGGAGUA CUGAUGAGGCCGAAAGGCCGAA AGUUAUG 460 UAUGGAG CUGAUGAGGCCGAAAGGCCGAA AUAGUUA 463 GACUAUG CUGAUGAGGCCGAAAGGCCGAA AGUAUAG 467 UCUGGAC CUGAUGAGGCCGAAAGGCCGAA AUGGAGU 470 CCAUCUG CUGAUGAGGCCGAAAGGCCGAA ACUAUGG
489 UUACUAU CUGAUGAGGCCGAAAGGCCGAA AUUUUCA
490 AUUACUA CUGAUGAGGCCGAAAGGCCGAA AAUUUUC 492 AAAUUAC CUGAUGAGGCCGAAAGGCCGAA AUAAUUU 495 UUUAAAU CUGAUGAGGCCGAAAGGCCGAA ACUAUAA Table 33 : RSV (1C) HH target Sequence
nt. Target Sequence nt. Target Sequence Position Position
10 GGCAAAU A AGAAUUU 165 UACAUUU A ACUAACG 16 UAAGAAU U UGAUAAG 169 UUUAACU A ACGCUUU 17 AAGAAUU U GAUAAGU 175 UAACGCU U UGGCUAA 21 AUUUGAU A AGUACCA 176 AACGCUU U GGCUAAG 25 GAUAAGU A CCACUUA 131 UUUGGCU A AGGCAGU 31 UACCACU U AAAUUUA 192 CAGCGAU A CAUACAA 32 ACCACUU A AAUUUAA 196 GAUACAU A CAAUCAA 36 CUUAAAU U UAACUCC 201 AUACAAU C AAAUUGA 37 UUAAAUU U AACUCCC 206 AUCAAAU U GAAUGGC 38. UAAAUUU A ACUCCCU 216 AUGGCAU U GUGUUUG 42 UUUAACU C CCUUGGU 221 AUUGUGU U UGCGCAU 46 ACUCCCU U GGUUAGA 222 UUGUGUU U GUGCAUG 50 CCUUGGU U AGAGAUG 231 UGCAUGU U AUUACAA 51 CUUGGUU A GAGAUGG 232 GCAUGUU A UUACAAG 67 CAGCAAU U CAUUGAG 234 AUGUUAU U ACAAGUA 68 AGCAAUU C AUUGAGU 235 UGUUAUU A CAAGUAG 71 AAUUCAU U GAGUAUG 241 UACAAGU A GUGAUAU 76 AUUGAGU A UGAUAAA 247 UAGUGAU A UUUGCCC 81 GUAUGAU A AAAGUUA 249 GUGAUAU U UGCCCUA 87 UAAAAGU U AGAUUAC 250 UGAUAUU U GCCCUAA 88 AAAAGUU A GAUUACA 256 UUGCCCU A AUAAUAA 92 GUUAGAU U ACAAAAU 259 CCCUAAU AUAAUAU 93 UUAGAUU A CAAAAUU 262 UAAUAAU AUAUUGU 100 ACAAAAU U UGUUUGA 265 UAAUAAU UUGUAGU 101 CAAAAUU U GUUUGAC 267 AUAAUAU GUAGUAA 104 AAUUUGU U UGACAAU 270 AUAUUGU GUAAAAU 105 AUUUGUU U GACAAUG 273 UUGUAGU A AAAUCCA 120 AUGAAGU A GCAUUGU 278 GUAAAAU C CAAUUUC 125 GUAGCAU U GUUAAAA 283 AUCCAAU U UCACAAC 128 GCAUUGU U AAAAAUA 284 UCCAAUU U CACAACA 129 CAUUGUU A AAAAUAA 285 CCAAUUU C ACAACAA 135 UAAAAAU A ACAUGCU 300 UGCCAGU A CUACAAA 143 ACAUGCU A UACUGAU 303 CAGUACU A CAAAAUG 145 AUGCUAU A CUGAUAA 316 UGGAGGU U AUAUAUG 151 UACUGAU A AAUUAAU 317 GGAGGUU A UAUAUGG 155 GAUAAAU U AAUACAU 319 AGGUUAU A UAUGGGA 156 AUAAAUU A AUACAUU 321 GUUAUAU A UGGGAAA 159 AAUUAAU A CAUUUAA 338 AUGGAAU U AACACAU 163 AAUACAU U UAACUAA 339 UGGAAUU A ACACAUU 164 AUACAUU U AACUAAC 346 AACACAU U GCUCUCA 350 CAUUGCU C UCAACCU
352 UUGCUCU C AACCUAA
358 UCAACCU A AUGGUCU
364 UAAUGGU C UACUAGA
366 AUGGUCU A CUAGAUG
369 GUCUACU A GAUGACA
379 UGACAAU U GUGAAAU
387 GUGAAAU U AAAUUCU
388 UGAAAUU A AAUUCUC
392 AUUAAAU U CUCCAAA
393 UUAAAUU C UCCAAAA
395 AAAUUCU C CAAAAAA
405 AAAAACU A AGUGAUU
412 AAGUGAU U CAACAAU
413 AGUGAUU C AACAAUG
427 GACCAAU U AUAUGAA
428 ACCAAUU A UAUGAAU
430 CAAUUAU A UGAAUCA
436 UAUGAAU C AAUUAUC
440 AAUCAAU U AUCUGAA
441 AUCAAUU A UCUGAAU
443 CAAUUAU C UGAAUUA
449 UCUGAAU U ACUUGGA
450 CUGAAUU A CUUGGAU
453 AAUUACU U GGAUUUG
458 CUUGGAU U UGAUCUU
459 UUGGAUU U GAUCUUA
463 AUUUGAU C UUAAUCC
465 UUGAUCU U AAUCCAU
466 UGAUCUU A AUCCAUA
469 UCUUAAU C CAUAAAU
473 AAUCCAU A AAUUAUA
477 CAUAAAU U AUAAUUA
478 AUAAAUU A UAAUUAA
480 AAAUUAU A AUUAAUA
483 UUAUAAU U AAUAUCA
484 UAUAAUU A AUAUCAA
487 AAUUAAU A UCAACUA
489 UUAAUAU C AACUAGC
494 AUCAACU A GCAAAUC
501 AGCAAAU C AAUGUCA
507 UGAAUGU C ACUAACA
511 UGUCACU A ACACCAU
519 ACACCAU U AGUUAAU
520 CACCAUU A GUUAAUA
523 CAUUAGU U AAUAUAA
524 AUUAGUU A AUAUAAA Table 34: RSV (IC) HH Ribozyme Sequence
nt . HH Ribozyme Sequence Position
10 AAAUUCU CUGAUGAGGCCGAAAGGCCGAA AUUUGCC
16 CUUAUCA CUGAUGAGGCCGAAAGGCCGAA AUCCUCA
17 ACUUAUC CUGAUGAGGCCGAAAGGCCGAA AAUUCUU 21 UGGUACU CUGAUGAGGCCGAAAGGCCGAA AUCAAAU 25 UAAGUGG CUGAUGAGGCCGAAAGGCCGAA ACUUAUC
31 UAAAUUU CUGAUGAGGCCGAAAGGCCGAA AGUGGUA
32 UUAAAUU CUGAUGAGGCCGAAAGGCCGAA AAGUGGU
36 GGAGUUA CUGAUGAGGCCGAAAGGCCGAA AUUUAAG
37 GGGAGUU CUGAUGAGGCCGAAAGGCCGAA AAUUUAA
38 AGGGAGU CUGAUGAGGCCGAAAGGCCGAA AAAUUUA 42 ACCAAGG CUGAUGAGGCCGAAAGGCCGAA AGUUAAA 46 UCUAACC CUGAUGAGGCCGAAAGGCCGAA AGGGAGU
50 CAUCUCU CUGAUGAGGCCGAAAGGCCGAA ACCAAGG
51 CCAUCUC CUGAUGAGGCCGAAAGGCCGAA AACCAAG
67 CUCAAUG CUGAUGAGGCCGAAAGGCCGAA AUUGCUG
68 ACUCAAU CUGAUGAGGCCGAAAGGCCGAA AAUUGCU 71 CAUACUC CUGAUGAGGCCGAAAGGCCGAA AUGAAUU 76 UUUAUCA CUGAUGAGGCCGAAAGGCCGAA ACCCAAU 81 UAACUUU CUGAUGAGGCCGAAAGGCCGAA AUCAUAC 37 GUAAUCU CUGAUGAGGCCGAAAGGCCGAA ACUUUUA 88 UGUAAUC CUGAUGAGGCCGAAAGGCCGAA AACUUUU
92 AUUUUGU CUGAUGAGGCCGAAAGGCCGAA AUCUAAC
93 AAUUUUG CUGAUGAGGCCGAAAGGCCGAA AAUCUAA
100 UCAAACA CUGAUGAGGCCGAAAGGCCGAA AUUUUGU
101 GUCAAAC CUGAUGAGGCCGAAAGGCCGAA AAUUUUG
104 AUUGUCA CUGAUGAGGCCGAAAGGCCGAA ACAAAUU
105 CAUUGUC CUGAUGAGGCCGAAAGGCCGAA AACAAAU 120 ACAAUGC CUGAUGAGGCCGAAAGGCCGAA ACUUCAU 125 UUUUAAC CUGAUGAGGCCGAAAGGCCGAA AUGCUAC
128 UAUUUUU CUGAUGAGGCCGAAAGGCCGAA ACAAUGC
129 UUAUUUU CUGAUGAGGCCGAAAGGCCGAA AACAAUG 135 AGCAUGU CUGAUGAGGCCGAAAGGCCGAA AUUUUUA 143 AUCAGUA CUGAUGAGGCCGAAAGGCCGAA AGCAUGU 145 UUAUCAG CUGAUGAGGCCGAAAGGCCGAA AUAGCAU 151 AUUAAUU CUGAUGAGGCCGAAAGGCCGAA AUCAGUA
155 AUGUAUU CUGAUGAGGCCGAAAGGCCGAA AUUUAUC
156 AAUGUAU CUGAUGAGGCCGAAAGGCCGAA AAUUUAU 159 UUAAAUG CUGAUGAGGCCGAAAGGCCGAA AUUAAUU
163 UUAGUUA CUGAUGAGGCCGAAAGGCCGAA AUGUAUU
164 GUUAGUU CUGAUGAGGCCGAAAGGCCGAA AAUGUAU
165 CGUUAGU CUGAUGAGGCCGAAAGGCCGAA AAAUGUA 169 AAAGCGU CUGAUGAGGCCGAAAGGCCGAA AGUUAAA
175 UUAGCCA CUGAUGAGGCCGAAAGGCCGAA AGCGUUA
176 CUUAGCC CUGAUGAGGCCGAAAGGCCGAA AAGCGUU 181 ACUGCCU CUGAUGAGGCCGAAAGGCCGAA AGCCAAA 192 UUGUAUG CUGAUGAGGCCGAAAGGCCGAA AUCACUG 196 UUGAUUG CUGAUGAGGCCGAAAGGCCGAA AUGUAUC
201 UCAAUUU CUGAUGAGGCCGAAAGGCCGAA AUUGUAU
206 GCCAUUC CUGAUGAGGCCGAAAGGCCGAA AUUUGAU
216 CAAACAC CUGAUGAGGCCGAAAGGCCGAA AUGCCAU
221 AUGCACA CUGAUGAGGCCGAAAGGCCGAA ACACAAU
222 CAUGCAC CUGAUGAGGCCGAAAGGCCGAA AACACAA
231 UUGUAAU CUGAUGAGGCCGAAAGGCCGAA ACAUGCA
232 CUUGUAA CUGAUGAGGCCGAAAGGCCGAA AACAUGC
234 UACUUGU CUGAUGAGGCCGAAAGGCCGAA AUAACAU
235 CUACUUG CUGAUGAGGCCGAAAGGCCGAA AAUAACA 241 AUAUCAC CUGAUGAGGCCGAAAGGCCGAA ACUUGUA 247 GGGCAAA CUGAUGAGGCCGAAAGGCCGAA AUCACUA
249 UAGGGCA CUGAUGAGGCCGAAAGGCCGAA AUAUCAC
250 UUAGGGC CUGAUGAGGCCGAAAGGCCGAA AAUAUCA 256 UUAUUAU CUGAUGAGGCCGAAAGGCCGAA AGGGCAA 259 AUAUUAU CUGAUGAGGCCGAAAGGCCGAA AUUAGGG 262 ACAAUAU CUGAUGAGGCCGAAAGGCCGAA AUUAUUA 265 ACUACAA CUGAUGAGGCCGAAAGGCCGAA AUUAUUA 267 UUACUAC CUGAUGAGGCCGAAAGGCCGAA AUAUUAU 270 AUUUUAC CUGAUGAGGCCGAAAGGCCGAA ACAAUAU 273 UGGAUUU CUGAUGAGGCCGAAAGGCCGAA ACUACAA 278 GAAAUUG CUGAUGAGGCCGAAAGGCCGAA AUUUUAC
283 GUUGUGA CUGAUGAGGCCGAAAGGCCGAA AUUGGAU
284 UGUUGUG CUGAUGAGGCCGAAAGGCCGAA AAUUGGA
285 UUGUUGU CUGAUGAGGCCGAAAGGCCGAA AAAUUGG 300 UUUGUAG CUGAUGAGGCCGAAAGGCCGAA ACUGGCA 303 CAUUUUG CUGAUGAGGCCGAAAGGCCGAA AGUACUG
316 CAUAUAU CUGAUGAGGCCGAAAGGCCGAA ACCUCCA
317 CCAUAUA CUGAUGAGGCCGAAAGGCCGAA AACCUCC 319 UCCCAUA CUGAUGAGGCCGAAAGGCCGAA AUAACCU 321 UUCCCCA CUGAUGAGGCCGAAAGGCCGAA AUAUAAC
338 ACGUGUU CUGAUGAGGCCGAAAGGCCGAA AUUCCAU
339 AAUGUGU CUGAUGAGGCCGAAAGGCCGAA AAUUCCA 346 UGAGAGC CUGAUGAGGCCGAAAGGCCGAA AUGUGUU 350 AGGUUGA CUGAUGAGGCCGAAAGGCCGAA AGCAAUG 352 UUAGGUU CUGAUGAGGCCGAAAGGCCGAA AGAGCAA 358 AGACCAU CUGAUGAGGCCGAAAGGCCGAA AGGUUGA 364 UCUAGUA CUGAUGAGGCCGAAAGGCCGAA ACCAUUA 366 CAUCUAG CUGAUGAGGCCGAAAGGCCGAA AGACCAU 369 UGUCAUC CUGAUGAGGCCGAAAGGCCGAA AGUAGAC 379 AUUUCAC CUGAUGAGGCCGAAAGGCCGAA AUUGUCA
387 AGAAUUU CUGAUGAGGCCGAAAGGCCGAA AUUUCAC
388 GAGAAUU CUGAUGAGGCCGAAAGGCCGAA AAUUUCA 392 UUUGGAG CUGAUGAGGCCGAAAGGCCGAA AUUUAAU 393 UUUUGGA CUGAUGAGGCCGAAAGGCCGAA AAUUUAA
395 UUUUUUG CUGAUGAGGCCGAAAGGCCGAA AGAAUUU
405 AAUCACU CUGAUGAGGCCGAAAGGCCGAA AGUUUUU
412 AUUGUUG CUGAUGAGGCCGAAAGGCCGAA AUCACUU
413 CAUUGUU CUGAUGAGGCCGAAAGGCCGAA AAUCACU
427 UUCAUAU CUGAUGAGGCCGAAAGGCCGAA AUUGGUC
428 AUUCAUA CUGAUGAGGCCGAAAGGCCGAA AAUUGGU 430 UGAUUCA CUGAUGAGGCCGAAAGGCCGAA AUAAUOG 436 GAUAAUU CUGAUGAGGCCGAAAGGCCGAA AUCCAUA
440 CUCAGAU CUGAUGAGGCCGAAAGGCCGAA AUUGAUU
441 AUUCAGA CUGAUGAGGCCGAAAGGCCGAA AAUUGAU 443 UAAUUCA CUGAUGAGGCCGAAAGGCCGAA AUAAUUG
449 UCCAAGU CUGAUGAGGCCGAAAGGCCGAA AUUCAGA
450 AUCCAAG CUGAUGAGGCCGAAAGGCCGAA AAUUCAG 453 CAAAUCC CUGAUGAGGCCGAAAGGCCGAA AGUAAUU
458 AAGAUCA CUGAUGAGGCCGAAAGGCCGAA AUCCAAG
459 UAAGAUC CUGAUGAGGCCGAAAGGCCGAA AAUCCAA 463 GGAUUAA CUGAUGAGGCCGAAAGGCCGAA AUCAAAU
465 AUGGAUU CUGAUGAGGCCGAAAGGCCGAA AGAUCAA
466 UAUGGAU CUGAUGAGGCCGAAAGGCCGAA AAGAUCA 469 AUUUAUG CUGAUGAGGCCGAAAGGCCGAA AUUAAGA 473 UAUAAUU CUGAUGAGGCCGAAAGGCCGAA AUGGAUU
477 UAAUUAU CUGAUGAGGCCGAAAGGCCGAA AUUUAUG
478 UUAAUUA CUGAUGAGGCCGAAAGGCCGAA AAUUUAU 480 UAUUAAU CUGAUGAGGCCGAAAGGCCGAA AUAAUUU
483 UGAUAUU CUGAUGAGGCCGAAAGGCCGAA AUUAUAA
484 UUGAUAU CUGAUGAGGCCGAAAGGCCGAA AAUUAUA 487 UAGUUGA CUGAUGAGGCCGAAAGGCCGAA AUUAAUU 489 GCUAGUU CUGAUGAGGCCGAAAGGCCGAA AUAUUAA 494 GAUUUGC CUGAUGAGGCCGAAAGGCCGAA AGUUGAU 501 UGACAUU CUGAUGAGGCCGAAAGGCCGAA AUUUGCU 507 UGUUAGU CUGAUGAGGCCGAAAGGCCGAA ACAUOGA 511 AUGGUGU CUGAUGAGGCCGAAAGGCCGAA AGUGACA
519 AUUAACU CUGAUGAGGCCGAAAGGCCGAA AUGGUGU
520 UAUUAAC CUGAUGAGGCCGAAAGGCCGAA AAUGGUG
523 UUAUAUU CUGAUGAGGCCGAAAGGCCGAA ACUAAUG
524 UUUAUAU CUGAUGAGGCCGAAAGGCCGAA AACUAAU
Table 35: RSV (N) HH Target Sequence
nt. HH Target Sequence nt. HH Target Sequence Position Position
9 GGCAAAU A CAAAGAU 217 GGUAUGU U AUAUGCG
21 GAUGGCU C UUAGCAA 218 GUAUGUU A UAUGCGA 23 UGGCUCU U AGCAAAG 220 AUGUUAU A UGCGAUG 24 GGCUCUU A GCAAAGU 229 GCGAUGU C UAGGCCA 32 GCAAAGU C AAGUUGA 231 GAUGUCU A GGUUAGG 37 GUCAAGU U GAAUGAU 235 UCUAGGU U AGGAAGA 45 GAAUGAU A CACUCAA 236 CUAGGUU A GGAAGAG 50 AUACACU C AACAAAG 254 ACACCAU A AAAAUAC 60 CAAAGAU C AACUUCU 260 UAAAAAU A CUCAGAG 65 AUCAACU U CUGUCAU 263 AAAUACU C AGAGAUG 66 UCAACUU C UGUCAUC 277 GCGGGAU A UCAUGUA 70 CUUCUGU C AUCCAGC 279 GGGAUAU C AUGUAAA 73 CUGUCAU C CAGCAAA 284 AUCAUGU A AAAGCAA 82 AGCAAAU A CACCAUC 299 AUGGAGU A GAUGUAA 89 ACACCAU C CAACGGA 305 UAGAUGU A ACAACAC 108 AGGAGAU A GUAUUGA 315 AACACAU C GUCAAGA 111 AGAUAGU A UUGAUAC 318 ACAUCGU AAGACAU 113 AUAGUAU U GAUACUC 326 AAGACAU AAUGGAA 117 UAUUGAU A CUCCUAA 327 AGAGAUU AUGGAAA 120 UGAUACU C CUAAUUA 346 AUGAAAU UGAAGUG 123 UACUCCU A AUUAUGA 347 UGAAAUU GAAGUGU 126 UCCUAAU U AUGAUGU 355 GAAGUGU U AACA 27 UUG 1 CCUAAUU A UGAUGUG 356 AAGUGUU A ACA 146 UUGG
AACACAU C AAUAAGU 361 UUAACAU U GGCAAGC 150 CAUCAAU A AGCUAUG 370 GCAAGCU U AACAACU 154 AAUAAGU U AUGUGGC 371 CAAGCUU A ACAACCG 155 AUAAGUU A UGUGGCA 383 CUGAAAU U CAAAUCA 166 GGCAUGU U AUUAAUC 384 UGAAAUU C AAA
167 UCAA
GCAUGUU A UUAAUCA 389 UUCAAAU C AACAUUG 169 AUGUUAU U AAUCACA 395 UCAACAU U GAGAUAG 170 UGUUAUU A AUCACAG 401 UUGAGAU A GAA
173 UCUA
UAUUAAU C ACAGAAG 406 AUAGAAU UAGAAAA 186 AGAUGCU A AUCAUAA 408 AGAAUCU GAAAAUC 189 UGCUAAU C AUAAAUU 415 AGAAAAU CUACAAA 192 UAAUCAU A AAUUCAC 418 AAAUCCU CAAAAAA 196 CAUAAAU U CACUGGG 431 AAAUGCU AAAGAAA 197 AUAAAUU C ACUGGGU 449 GAGAGGU GCUCCAG 205 ACUGGGU U AAUAGGU 453 GGUAGCU CAGAAUA 206 CUGGGUU A AUAGGUA 460 CCAGAAU CAGGCAU 209 GGUUAAU A GGUAUGU 472 CAUGACU UCCUGAU 213 AAUAGGU A UGUUAUA 474 UGACUCU CUGAUUG 480 UCCUGAU U GUGGGAU 696 UUUUGGU A UAGCACA
491 GGAUGAU A AUAUUAU 698 UUGGUAU A GCACAAU
494 UGAUAAU A UUAUGUA 706 GCACAAU C UUCUACC
496 AUAAUAU U AUGUAUA 708 ACAAUCU U CUACCAG
497 UAAUAUU A UGUAUAG 709 CAAUCUU C UACCAGA
501 AUUAUGU A UAGCAGC 711 AUCUUCU A CCAGAGG
503 UAUGUAU A GCAGCAU 726 UGGCAGU A GAGUUGA
511 GCAGCAU U AGUAAUA 731 GUAGAGU U GAAGGGA
512 CAGCAUU A GUAAUAA 740 AAGGGAU U UUUGCAG
515 CAUUAGU A AUAACUA 741 AGGGAUU U UUGCAGG
518 UAGUAAU A ACUAAAU 742 GGGAUUU U UGCAGGA
522 AAUAACU A AAUUAGC 743 GGAUUUU U GCAGGAU
526 ACUAAAU U AGGAGCA 751 GCAGGAU U GUUUAUG
527 CUAAAUU A GCAGCAG 754 GGAUUGU U UAUGAAU
544 GACAGAU C UGGUCUU 755 GAUUGUU U AUGAAUG
549 AUCUGGU C UUACAGC 756 AUUGUUU A UGAAUGC
551 CUGGUCU U ACAGCCG 766 AAUGCCU A UGGUGCA
552 UGGUCUU A CAGCCGU 787 GUGAUGU U ACGGUGG
563 CCGUGAU U AGGAGAG 788 UGAUGUU A CGGUGGG
564 CGUGAUU A GGAGAGC 800 GGGGAGU C UUAGCAA
573 GAGAGCU A AUAAUGU 802 GGAGUCU U AGCAAAA
576 AGCUAAU A AUGUCCU 803 GAGUCUU A GCAAAAU
581 AUAAUGU C CUAAAAA 811 GCAAAAU C AGUUAAA
584 AUGUCCU A AAAAAUG 815 AAUCAGU U AAAAAUA
603 GAAACGU U AGAAAGG 816 AUCAGUU A AAAAUAU
604 AAACGUU A CAAAGGC 822 UAAAAAU A UUAUGUU
613 AAAGGCU U ACUACCC 824 AAAAUAU U AUGUUAG
614 AAGGCUU A CUACCCA 825 AAAUAUU A UGUUAGG
617 GCUUACU A CCCAAGG 829 AUUAUGU U AGGACAU
629 AGGACAU A GCCAACA 830 UUAUGUU A GGACAUG
640 AACAGCU U CUAUGAA 840 ACAUGCU A GUGUGCA
641 ACAGCUU C UAUGAAG 866 AACAAGU U GUUGAGG
643 AGCUCCU A UGAAGUG 869 AAGUUGU U GAGGUUU
652 GAAGUGU U UGAAAAA 875 UUGAGGU U UAUGAAU
653 AAGUGUU U GAAAAAC 876 UGAGGUU U AUGAAUA
663 AAAACAU C CCCACUU 877 GAGGUUU A UGAAUAU
670 CCCCACU U UAUAGAU 883 UAUGAAU A UGCCCAA
671 CCCACUU U AUAGAUG 895 CAAAAAU U GGGUGGU
672 CCACUUU A UAGAUGU 913 GCAGGAU U CUACCAU
674 ACUUUAU A GAUGUUU 914 CAGGAUU C UACCAUA
680 UAGAUGU U UUUGUUC 916 GGAUUCU A CCAUAUA
681 AGAUGUU U UUGUUCA 921 CUACCAU A UAUUGAA
682 GAUGUUU U UGUUCAU 923 ACCAUAU A UUGAACA
683 AUGUUUU U GUUCAUU 925 CAUAUAU U GAACAAC
686 UUUUUGU U CAUUUUG 943 AAAGCAU C AUUAUUA
687 UUUUGUU C AUUUUGG 946 GCAUCAU U AUUAUCU
690 UGUUCAU U UUGGUAU 947 CAUCAUU A UUAUCUU
691 GUUCAUU U UGGUAUA 949 UCAUUAU U AUCUUUG
692 UUCAUUU U GGUAUAG 950 CAUUAUU A UCUUUGA 952 UUAUUAU C UUUGACU
954 AUUAUCU U UGACUCA
955 UUAUCUU U GACUCAA
960 UUUGACU C AAUUUCC
964 ACUCAAU U UCCUCAC
965 CUCAAUU U CCUCACU
966 UCAAUUU C CUCACUU
969 AUUUCCU C ACUUCUC
973 CCUCACU U CUCCAGU
974 CUCACUU C UCCAGUG
976 CACUUCU C CAGUGUA
983 CCAGUGU A GUAUCAG
986 GUGUAGU A UUAGGCA
988 GUAGUAU U AGGCAAU
989 UAGUAUU A GGCAAUG
1007 CUGGCCU A GGCAUAA
1013 UAGGCAU A AUGGGAG
1024 GGAGAGU A CAGAGGU
1032 CAGAGGU A CACCGAG
1044 GAGGAAU C AAGAUCU
1050 UCAAGAU C UAUAUGA
1052 AAGAUCU A UAUGAUG
1054 GAUCUAU A UGAUGCA
1072 AAGGCAU A UGCUGAA
1085 AACAACU C AAAGAAA
1103 GOGUGAU U AACUACA
1104 UGUGAUU A ACUACAG
1108 AUUAACU A CAGUGUA
1115 ACAGUGU A CUAGACU
1118 GUGUACU A GACUCGA
1123 CUAGACU U GACAGCA
1139 AAGAACU A GAGGCUA
1146 AGAGGCU A UCAAACA
1148 AGGCUAU C AAACAUC
1155 CAAACAU C AGCUUAA
1160 AUCAGCU U AAUCCAA
1151 UCAGCUU A AUCCAAA
1154 GCUUAAU C CAAAAGA
1173 AAAAGAU A AUGAUGU
1181 AUGAUGU A GAGCUUU
1187 UAGAGCU U UGAGUUA
1188 AGAGCUU U GAGUUAA
1193 UUUGAGU U AAUAAAA
1194 UUGAGUU A AUAAAAA Table 36: RSV (N) HH Ribozyme Sequence
nt . HH Ribozyme Sequence
Position
9 AUCUUUG CUGAUGAGGCCGAAAGGCCGAA AUUUGCC
21 UUGCUAA CUGAUGAGGCCGAAAGGCCGAA AGCCAUC
23 CUUUGCU CUGAUGAGGCCGAAAGGCCGAA AGAGCCA
24 ACUUUGC CUGAUGAGGCCGAAAGGCCGAA AAGAGCC 32 UCAACUU CUGAUGAGGCCGAAAGGCCGAA ACUUUGC 37 AUCAUUC CUGAUGAGGCCGAAAGGCCGAA ACUUGAC 45 UUGAGUG CUGAUGAGGCCGAAAGGCCGAA AUCAUUC 50 CUUUGUU CUGAUGAGGCCGAAAGGCCGAA AGUGUAU 60 AGAAGUC CUGAUGAGGCCGAAAGGCCGAA AUCUUUG
65 AUGACAG CUGAUGAGGCCGAAAGGCCGAA AGUUGAU
66 GAUGACA CUGAUGAGGCCGAAAGGCCGAA AAGUUGA 70 GCUGGAU CUGAUGAGGCCGAAAGGCCGAA ACAGAAG 73 UUUGCUG CUGAUGAGGCCGAAAGGCCGAA AUGACAG 82 GAUGGUG CUGAUGAGGCCGAAAGGCCGAA AUUUGCU 89 UCCGUUG CUGAUGAGGCCGAAAGGCCGAA AUGGUGU
108 UCAAUAC CUGAUGAGGCCGAAAGGCCGAA AUCUCCU
111 GUAUCAA CUGAUGAGGCCGAAAGGCCGAA ACUAUCU
113 GAGUAUC CUGAUGAGGCCGAAAGGCCGAA AUACUAU
117 UUAGGAG CUGAUGAGGCCGAAAGGCCGAA AUCAAUA
120 UAAUUAG CUGAUGAGGCCGAAAGGCCGAA AGUAUCA
123 UCAUAAU CUGAUGAGGCCGAAAGGCCGAA AGGAGUA
126 ACAUCAU CUGAUGAGGCCGAAAGGCCGAA AUUAGGA
127 CACAUCA CUGAUGAGGCCGAAAGGCCGAA AAUUAGG 146 ACUUAUU CUGAUGAGGCCGAAAGGCCGAA AUGUGUU 150 CAUAACU CUGAUGAGGCCGAAAGGCCGAA AUUGAUG
154 GCCACAU CUGAUGAGGCCGAAAGGCCGAA ACUUAUU
155 UGCCACA CUGAUGAGGCCGAAAGGCCGAA AACUUAU
166 GAUUAAU CUGAUGAGGCCGAAAGGCCGAA ACAUGCC
167 UGAUUAA CUGAUGAGGCCGAAAGGCCGAA AACAUGC
169 UGUGAUU CUGAUGAGGCCGAAAGGCCGAA AUAACAU
170 CUGUGAU CUGAUGAGGCCGAAAGGCCGAA AAUAACA 173 CUUCUGU CUGAUGAGGCCGAAAGGCCGAA AUUAAUA 186 UUAUGAU CUGAUGAGGCCGAAAGGCCGAA AGCAUCU 189 AAUUUAU CUGAUGAGGCCGAAAGGCCGAA AUUAGCA 192 GUGAAUU CUGAUGAGGCCGAAAGGCCGAA AUGAUUA
196 CCCAGUG CUGAUGAGGCCGAAAGGCCGAA AUUUAUG
197 ACCCAGC CCGAUGAGGCCGAAAGGCCGAA AAUUUAU
205 ACCUAUU CUGAUGAGGCCGAAAGGCCGAA ACCCAGU
206 UACCUAU CUGAUGAGGCCGAAAGGCCGAA AACCCAG 209 ACAUACC CUGAUGAGGCCGAAAGGCCGAA AUUAACC 213 UAUAACA CUGAUGAGGCCGAAAGGCCGAA ACCUAUU 217 CGCAUAU CUGAUGAGGCCGAAAGGCCGAA ACAUACC
218 UCGCAUA CUGAUGAGGCCGAAAGGCCGAA AACAUAC 220 CAUCGCA CUGAUGAGGCCGAAAGGCCGAA AUAACAU 229 UAACCUA CUGAUGAGGCCGAAAGGCCGAA ACAUCGC 231 CCUAACC CUGAUGAGGCCGAAAGGCCGAA AGACAUC
235 UCUUCCU CUGAUGAGGCCGAAAGGCCGAA ACCUAGA
236 CUCUUCC CUGAUGAGGCCGAAAGGCCGAA AACCUAG 254 GUAUUUU CUGAUGAGGCCGAAAGGCCGAA AUGGUGU 260 CUCUGAG CUGAUGAGGCCGAAAGGCCGAA AUUUUUA 263 CACCUCU CUGAUGAGGCCGAAAGGCCGAA AGUAUUU 277 UACAUGA CUGAUGAGGCCGAAAGGCCGAA AUCCCGC 279 UUUACAU CUGAUGAGGCCGAAAGGCCGAA AUAUCCC 284 UUGCUUU CUGAUGAGGCCGAAAGGCCGAA AGAUGAU 299 UUACAUC CUGAUGAGGCCGAAAGGCCGAA ACUCCAU 305 GUGUUGU CUGAUGAGGCCGAAAGGCCGAA ACAUCUA 315 UCUUGAC CUGAUGAGGCCGAAAGGCCGAA AUGUGUU 318 AUGUCUU CUGAUGAGGCCGAAAGGCCGAA ACGAUGU
326 UUCCAUU CUGAUGAGGCCGAAAGGCCGAA AUGUCUU
327 UUUCCAU CUGAUGAGGCCGAAAGGCCGAA AAUGUCU
346 CACCUCA CUGAUGAGGCCGAAAGGCCGAA AUUUCAU
347 ACACUUC CUGAUGAGGCCGAAAGGCCGAA AAUUUCA
355 CAAUGUU CUGAUGAGGCCGAAAGGCCGAA ACACUUC
356 CCAAUGU CUGAUGAGGCCGAAAGGCCGAA AACACUU 361 GCUUGCC CUGAUGAGGCCGAAAGGCCGAA AUGUUAA
370 AGUUGUU CUGAUGAGGCCGAAAGGCCGAA AGCUUGC
371 CAGUUGU CUGAUGAGGCCGAAAGGCCGAA AAGCUUG
383 UGAUUUG CUGAUGAGGCCGAAAGGCCGAA AUUUCAG
384 UUGAUUU CUGAUGAGGCCGAAAGGCCGAA AAUUUCA 389 CAAUGUU CUGAUGAGGCCGAAAGGCCGAA AUUUGAA 395 CUAUCUC CUGAUGAGGCCGAAAGGCCGAA AUGUUGA 401 UAGAUUC CUGAUGAGGCCGAAAGGCCGAA AUCUCAA 406 UUUUCUA CUGAUGAGGCCGAAAGGCCGAA AUUCUAU 408 GAUUUUC CUGAUGAGGCCGAAAGGCCGAA AGAUUCU 415 UUUGUAG CUGAUGAGGCCGAAAGGCCGAA AUUUUCU 418 UCUUUUG CUGAUGAGGCCGAAAGGCCGAA AGGAUUU 431 UUUCUUU CUGAUGAGGCCGAAAGGCCGAA AGCAUUU 449 CCGGAGC CUGAUGAGGCCGAAAGGCCGAA ACCUCUC 453 UAUUCUG CUGAUGAGGCCGAAAGGCCGAA AGCUACC 460 AUGCCUG CUGAUGAGGCCGAAAGGCCGAA AUUCUGG 472 AUCAGGA CUGAUGAGGCCGAAAGGCCGAA AGUCAUG 474 CAAUCAG CUGAUGAGGCCGAAAGGCCGAA AGAGUCA 480 AUCCCAC CUGAUGAGGCCGAAAGGCCGAA AUCAGGA 491 AUAAUAU CUGAUGAGGCCGAAAGGCCGAA AUCAUCC 494 UACAUAA CUGAUGAGGCCGAAAGGCCGAA AUUACCA
496 UAUACAU CUGAUGAGGCCGAAAGGCCGAA AUAUUAU
497 CUAUACA CCGAUGAGGCCGAAAGGCCGAA AAUAUUA 501 GCUGCUA CUGAUGAGGCCGAAAGGCCGAA ACAUAAU 503 AUGCUGC CUGAUGAGGCCGAAAGGCCGAA AUACAUA 511 UAUUACU CUGAUGAGGCCGAAAGGCCGAA AUGCUGC 512 UUAUUAC CUGAUGAGGCCGAAAGGCCGAA AAUGCUG 515 UAGUUAU CUGAUGAGGCCGAAAGGCCGAA ACUAAUG 518 AUUUAGU CUGAUGAGGCCGAAAGGCCGAA AUUACUA 522 GCUAAUU CUGAUGAGGCCGAAAGGCCGAA AGUUAUU 526 UGCUGCU CUGAUGAGGCCGAAAGGCCGAA AUUUAGU 527 CUGCUGC CUGAUGAGGCCGAAAGGCCGAA AAUUUAG 544 AAGACCA CUGAUGAGGCCGAAAGGCCGAA AUCUGUC 549 GCUGUAA CUGAUGAGGCCGAAAGGCCGAA ACCAGAU 551 CGGCUGU CCGAUGAGGCCGAAAGGCCGAA AGACCAG 552 ACGGCUG CUGAUGAGGCCGAAAGGCCGAA AAGACCA 563 CUCUCCU CUGAUGAGGCCGAAAGGCCGAA AUCACGG 564 GCUCUCC CUGAUGAGGCCGAAAGGCCGAA AAUCACG 573 ACACUAU CUGAUGAGGCCGAAAGGCCGAA AGCUCUC 576 AGGACAU CUGAUGAGGCCGAAAGGCCGAA AUUAGCU 581 UUUUUAG CUGAUGAGGCCGAAAGGCCGAA ACAUUAU 584 CAUUUUU CUGAUGAGGCCGAAAGGCCGAA AGGACAU 603 CCUUUGU CUGAUGAGGCCGAAAGGCCGAA ACGUUUC 604 GCCUUUG CUGAUGAGGCCGAAAGGCCGAA AACGUUU 613 GGGUAGU CUGAUGAGGCCGAAAGGCCGAA AGCCUCU 614 UGGGCAG CUGAUGAGGCCGAAAGGCCGAA AAGCCUU 617 CCUUGGG CUGAUGAGGCCGAAAGGCCGAA AGUAAGC 629 UGUUGGC CUGAUGAGGCCGAAAGGCCGAA AUGUCCU 640 UUCAUAG CUGAUGAGGCCGAAAGGCCGAA AGCUGUU 641 CUUCAUA CUGAUGAGGCCGAAAGGCCGAA AAGCUGU 643 CACUUCA CUGAUGAGGCCGAAAGGCCGAA AGAAGCU 652 UUUUUCA CUGAUGAGGCCGAAAGGCCGAA ACACUUC 653 GUUUUUC CUGAUGAGGCCGAAAGGCCGAA AACACUU 663 AAGUGGG CUGAUGAGGCCGAAAGGCCGAA AUGUUUU 670 AUCUAUA CUGAUGAGGCCGAAAGGCCGAA AGUGGGG 671 CAUCUAU CUGAUGAGGCCGAAAGGCCGAA AAGUGGG 672 ACAUCUA CUGAUGAGGCCGAAAGGCCGAA AAAGUGG 674 AAACAUC CUGAUGAGGCCGAAAGGCCGAA AUAAAGU 680 GAACAAA CUGAUGAGGCCGAAAGGCCGAA ACAUCUA 681 UGAACAA CUGAUGAGGCCGAAAGGCCGAA AAGAUCU 682 AUGAACA CUGAUGAGGCCGAAAGGCCGAA AAACAUC 683 AAUGAAC CUGAUGAGGCCGAAAGGCCGAA AAAACAU 686 CAAAAUG CUGAUGAGGCCGAAAGGCCGAA ACAAAAA 687 CCAAAAU CUGAUGAGGCCGAAAGGCCGAA AACAAAA 690 AUACCAA CUGAUGAGGCCGAAAGGCCGAA AUGAACA 691 UAUACCA CUGAUGAGGCCGAAAGGCCGAA AAUGAAC 692 CUAUACC CUGAUGAGGCCGAAAGGCCGAA AAAUGAA 696 UGUGCUA CUGAUGAGGCCGAAAGGCCGAA ACCAAAA 698 AUUGCGC CUGAUGAGGCCGAAAGGCCGAA AUACCAA 706 GGUAGAA CCGACGAGGCCGAAAGGCCGAA ACCGCGC 708 CUGGUAG CUGAUGAGGCCGAAAGGCCGAA AGAUUGU 709 UCUGGUA CUGAUGAGGCCGAAAGGCCGAA AAGAUCG 711 CCUCUGG CUGAUGAGGCCGAAAGGCCGAA AGAAGAU 726 UCAACUC CUGAUGAGGCCGAAAGGCCGAA ACUGCCA 731 UCCCUUC CUGAUGAGGCCGAAAGGCCGAA ACUCUAC 740 CUGCAAA CUGAUGAGGCCGAAAGGCCGAA AUCCCUU
741 CCUGCAA CUGAUGAGGCCGAAAGGCCGAA AAUCCCU
742 UCCUGCA CUGAUGAGGCCGAAAGGCCGAA AAAUCCC
743 AUCCUGC CUGAUGAGGCCGAAAGGCCGAA AAAAUCC 751 CAUAAAC CUGAUGAGGCCGAAAGGCCGAA AUCCUGC
754 AUUCAUA CUGAUGAGGCCGAAAGGCCGAA ACAAUCC
755 CAUUCAU CCGAUGAGGCCGAAAGGCCGAA AACAAUC
756 GCAUUCA CUGAUGAGGCCGAAAGGCCGAA AAACAAU 766 UGCACCA CUGAUGAGGCCGAAAGGCCGAA AGGCAUU
787 CCACCGU CUGAUGAGGCCGAAAGGCCGAA ACAUCAC
788 CCCACCG CUGAUGAGGCCGAAAGGCCGAA AACAUCA 800 UUGCUAA CUGAUGAGGCCGAAAGGCCGAA ACUCCCC
802 UUCCGCU CUGAUGAGGCCGAAAGGCCGAA AGACUCC
803 AUUCUGC CUGAUGAGGCCGAAAGGCCGAA AAGACUC 811 UUUAACU CUGAUGAGGCCGAAAGGCCGAA AUUUUGC
815 UAUUUUU CUGAUGAGGCCGAAAGGCCGAA ACUGAUU
816 AUAUUUU CUGAUGAGGCCGAAAGGCCGAA AACUGAU 822 AACAUAA CUGAUGAGGCCGAAAGGCCGAA AUUUUUA
824 CUAACAU CCGAUGAGGCCGAAAGGCCGAA AUAUUUU
825 CCUAACA CUGAUGAGGCCGAAAGGCCGAA AAUAUUU
829 AUGUCCU CUGAUGAGGCCGAAAGGCCGAA ACAUAAU
830 CAUGUCC CUGAUGAGGCCGAAAGGCCGAA AACAUAA 840 UGCACAC CUGAUGAGGCCGAAAGGCCGAA AGCAUGU 866 CCUCAAC CCGAUGAGGCCGAAAGGCCGAA ACUUGUU 869 AAACCUC CCGAUGAGGCCGAAAGGCCGAA ACAACUU
875 AUUCAUA CUGAUGAGGCCGAAAGGCCGAA ACCUCAA
876 UAUUCAU CUGAUGAGGCCGAAAGGCCGAA AACCUCA
877 AUAUUCA CUGAUGAGGCCGAAAGGCCGAA AAACCUC 883 UUGGGCA CUGAUGAGGCCGAAAGGCCGAA AUUCAUA 895 ACCACCC CUGAUGAGGCCGAAAGGCCGAA AUUUUUG
913 AUGGUAG CUGAUGAGGCCGAAAGGCCGAA AUCCUGC
914 UAUGGUA CUGAUGAGGCCGAAAGGCCGAA AAUCCUG 916 UAUAUGG CUGAUGAGGCCGAAAGGCCGAA AGAAUCC 921 UUCAAUA CUGAUGAGGCCGAAAGGCCGAA AUGGUAG 923 UGUUCAA CUGAUGAGGCCGAAAGGCCGAA AUAUGGU 925 GUUGUUC CUGAUGAGGCCGAAAGGCCGAA AUAUAUG 943 UAAUAAU CUGAUGAGGCCGAAAGGCCGAA AUGCUUU
946 AGAUAAU CUGAUGAGGCCGAAAGGCCGAA AUGAUGC
947 AAGAUAA CUGAUGAGGCCGAAAGGCCGAA AAUGAUG
949 CAAAGAU CUGAUGAGGCCGAAAGGCCGAA AUAAUGA
950 UCAAAGA CUGAUGAGGCCGAAAGGCCGAA AAUAAUG 952 AGUCAAA CUGAUGAGGCCGAAAGGCCGAA AUAAUAA
954 UGAGUCA CUGAUGAGGCCGAAAGGCCGAA AGAUAAU
955 UUGAGUC CUGAUGAGGCCGAAAGGCCGAA AAGAUAA 960 GGAAAUU CUGAUGAGGCCGAAAGGCCGAA AGUCAAA
964 GUGAGGA CUGAUGAGGCCGAAAGGCCGAA AUUGAGU
965 AGUGAGG CUGAUGAGGCCGAAAGGCCGAA AAUUGAG
966 AAGUGAG CUGAUGAGGCCGAAAGGCCGAA AAAUUGA 969 GAGAAGU CUGAUGAGGCCGAAAGGCCGAA AGGAAAU 973 ACUGGAG CUGAUGAGGCCGAAAGGCCGAA AGUGAGG
974 CACUGGA CUGAUGAGGCCGAAAGGCCGAA AAGUGAG 976 UACACUG CUGAUGAGGCCGAAAGGCCGAA AGAAGUG 983 CUAAUAC CUGAUGAGGCCGAAAGGCCGAA AGACUGG 986 UGCCUAA CUGAUGAGGCCGAAAGGCCGAA ACUACAC
988 AUUGCCU CUGAUGAGGCCGAAAGGCCGAA AUACUAC
989 CAUUGCC CUGAUGAGGCCGAAAGGCCGAA AAUACUA 1007 UUAUGCC CUGAUGAGGCCGAAAGGCCGAA AGGCCUG 1013 CUCCCAU CUGAUGAGGCCGAAAGGCCGAA AUGCCUA 1024 ACCUCUG CUGAUGAGGCCGAAAGGCCGAA ACUCUCC 1032 CUCGGUG CUGAUGAGGCCGAAAGGCCGAA ACCUCUG 1044 AGAUCUU CUGAUGAGGCCGAAAGGCCGAA AUUCCUC 1050 UCAUAUA CUGAUGAGGCCGAAAGGCCGAA AUCUUGA 1052 CAUCAUA CUGAUGAGGCCGAAAGGCCGAA AGAUCCU 1054 UGCAUCA CUGAUGAGGCCGAAAGGCCGAA AUAGAUC 1072 UUCAGCA CUGAUGAGGCCGAAAGGCCGAA AUGCCUU 1085 UUUCUUU CUGAUGAGGCCGAAAGGCCGAA AGUUGUU
1103 UGUAGUU CUGAUGAGGCCGAAAGGCCGAA AUCACAC
1104 CUGUAGU CUGAUGAGGCCGAAAGGCCGAA AAUCACA 1108 UACACUG CUGAUGAGGCCGAAAGGCCGAA AGUUAAU 1115 AGUCUAG CUGAUGAGGCCGAAAGGCCGAA ACACUGU 1118 UCAAGUC CUGAUGAGGCCGAAAGGCCGAA AGUACAC 1123 UGCUGUC CUGAUGAGGCCGAAAGGCCGAA AGUCUAG 1139 UAGCCUC CUGAUGAGGCCGAAAGGCCGAA AGUUCUU 1146 UGUUUGA CUGAUGAGGCCGAAAGGCCGAA AGCCUCU 1148 GAUGUUU CUGAUGAGGCCGAAAGGCCGAA AUAGCCU 1155 UUAAGCU CUGAUGAGGCCGAAAGGCCGAA AUGUUUG
1160 UUGGAUU CUGAUGAGGCCGAAAGGCCGAA AGCUGAU
1161 UUUGGAU CUGAUGAGGCCGAAAGGCCGAA AAGCUGA 1154 UCUUUUG CUGAUGAGGCCGAAAGGCCGAA AUUAAGC 1173 ACAUCAU CUGAUGAGGCCGAAAGGCCGAA AUCUUUU 1181 AAAGCUC CUGAUGAGGCCGAAAGGCCGAA ACAUCAU
1187 UAACUCA CUGAUGAGGCCGAAAGGCCGAA AGCCCUA
1188 CUAACUC CUGAUGAGGCCGAAAGGCCGAA AAGCUCU
1193 UUUUAUU CUGAUGAGGCCGAAAGGCCGAA ACUCAAA
1194 UUUUUAU CUGAUGAGGCCGAAAGGCCGAA AACUCAA
Figure imgf000284_0001
Figure imgf000285_0001
Table 39: Large-Scale Synthesis
Sequence Activator Amidite Time* % Full
[Added/Final] [Added/Final] Length
(min) (min) Product
A9T T [0.50/0.33] [0.1/0.02] 15 m 85 A9T S [0.25/0.17] [0.1/0.02] 15 m 89
(GGU)3GGT T [0.50/0.33] [0.1/0.02] 15 m 78 (GGU)3GGT S [0.25/0.17] [0.1/0.02] 15 m 81
C9T T [0.50/0.33] [0.1/0.02] 15 m 90 C9T S [0.25/0.17] [0.1/0.02] 15 m 97
U9T T [0.50/0.33] [0.1/0.02] 15 m 80 U9T S [0.25/0.17] [0.1/0.02] 15 m 85
A (36-mer) T [0.50/0.33] [0.1/0.02] 15/15m 21 A (36-mer) S [0.25/0.17] [0.1/0.02] 15/15 m 25 A (36-mer) S [0.50/0.24] [0.1/0.03] 15/15 m 25 A (36-mer) S [0.50/0.18] [0.1/0.05] 15/15 m 38 A (36-mer) S [0.50/0.18] [0.1/0.05] 10/5 m 42
*Where two coupling times are indicated the first refers to RNA coupling and the second to 2'-O-methyl coupling. S = 5-S-Ethyltetrazole, T = tetrazole activator. A is 5' -ucu ccA UCU GAU GAG GCC GAA AGG CCG AAA Auc ccu -3' where lowerecase represents 2'-O-methylnucleotides. Table 40: Base Deprotection
Sequence Deprotection Time T °C % Full
Reagent (min) Length
Product iBu(GGU)4 NH4OH/EtOH 16h 55 62.5
MA 10m 65 62.7
AMA 10m 65 74.8
MA 10m 55 75.0
AMA 10m 55 77.2 iPrP(GGU)4 NH4OH/EtOH 4h 65 44.8
MA 10m 65 65.9
AMA 10 m 65 59.8
MA 10 m 55 61.3
AMA 10m 55 60.1
C9U NH4OH/EtOH 4h 65 75.2
MA 10m 65 79.1
AMA 10m 65 77.1
MA 10 m 55 79.8
AMA 10m 55 75.5 A (36-mer) NH4OH/EtOH 4h 65 22.7
MA 10m 65 28.9
Table 41 : 2'-O-AIkylsiIyI Deprotection
Sequence Deprotection Time T °C % Full
Reagent (min) Length
Product
AgT TBAF 24 h 20 84.5
1.4 MHF 0.5 h 65 81.0
(GGU)4 TBAF 24 h 20 60.9
1.4 MHF 0.5 h 65 67.8
C10 TBAF 24 h 20 86.2
1.4 MHF 0.5 h 65 86.1
U10 TBAF 24 h 20 84.8
1.4 MHF 0.5 h 65 84.5
B (36-mer) TBAF 24 h 20 25.2
1.4 MHF 1.5 h 65 30.6
A (36-mer) TBAF 24 h 20 29.7
1.4 M HF 1.5 h 65 30.4
B is 5'- UCU CCA UCU GAU GAG GCC GAA AGG CCG AAA AUC CCU
-3'.
Figure imgf000289_0001
Figure imgf000290_0001
Table 44. Kinetics of Self-Processing In Vitro
Figure imgf000291_0002
* k represents the unimolecular rate constant for ribozyme self-cleavage determined from a non-linear, least-squares fit (KaleidaGraph, Synergy Software, Reeding, PA) to the equation:
Figure imgf000291_0001
The equation describes the extent of ribozyme processing in the presense of ongoing transcription (Long & Uhlenbeck, 1994 Proc. Natl. Acad. Sci. USA 91 , 6977) as a function of time (t) and the unimolecular rate constant for cleavage (k). Each value of k represents the average (± range) of values determined from two experiments.
Table 45
Entry Modification t1/2 (m ) t1/2 (m) β = tS/tA
Activity Stability x 10 (tA) (ts) 1 U4 & U7 = U 1 0.1 1 2 U4 & U7 = 2'-O-Me-U 4 260 650 3 U4 = 2'=CH2-U 6.5 120 180 4 U7 = 2'=CH2-U 8 280 350 5 U4 & U7 = 2'=CH2-U 9.5 120 130 6 U4 = 2'=CF2-U 5 320 640 7 U7 = 2'=CF2-U 4 220 550 8 U4 & U7 = 2'=CF2-U 20 320 160 9 U4 = 2'-F-U 4 320 800 10 U7 = 2'-F-U 8 400 500 11 U4 & U7 = 2'-F-U 4 300 750 12 U4 = 2'-C-Allyl-U 3 >500 >1700 13 U7 = 2'-C-Allyl-U 3 220 730 14 U4 & U7 = 2'-C-Allyl-U 3 120 400 15 U4 = 2'-araF-U 5 >500 >1000 16 U7 = 2'-araF-U 4 350 875
17 U4 & U7 = 2'-araF-U 15 500 330
18 U4 = 2'-NH2-U 10 500 500
19 U7 = 2'-NH2-U 5 500 1000 20 U4 & U7 = 2'-NH2-U 2 300 1500 21 U4 = dU 6 100 170
22 U4 & U7 = dU 4 240 600

Claims

CLAIMS What is claimed is:
1. An enzymatic nucleic acid molecule which cleaves ICAM-1 mRNA, IL- 5 mRNA, rel A mRNA, TNF-α mRNA sites shown in Table 23, 25, 27, or 28, CML associated mRNA selected from those identified as
SEQ. ID NOS 1-25, or RSV mRNA or RSV genomic RNA in a region selected from the group consisting of 1C, 1B and N.
2. The enzymatic nucleic acid molecule of claim 1, the binding arms of which contain sequences complementary to any one of the sequences defined in any of those in Tables 2, 3, 6-9, 11, 13, 15- 23, 27, 28, 31 , 33, 34, 36, and 37.
3. The enzymatic nucleic acid molecule of claim 1 or 2, wherein said nucleic acid molecule is in a hammerhead motif.
4. The enzymatic nucleic acid molecule of claim 1 or 2, wherein said RNA molecule is in a hairpin, hepatitis delta virus, group 1 intron,
Neurospora VS RNA or RNaseP RNA motif.
5. The enzymatic nucleic acid molecule of claim 1 or 2, comprising between 12 and 100 bases complementary to said mRNA or genomic RNA.
6. The enzymatic nucleic acid molecule of claim 5 comprising between
14 and 24 bases complementary to said mRNA or genomic RNA.
7. The enzymatic nucleic acid molecule of claim 1 or 2, comprising between 5 and 23 bases complementary to said mRNA or genomic RNA.
8. The enzymatic nucleic acid molecule of claim 7 comprising between
10 and 18 bases complementary to said mRNA or genomic RNA.
9. An enzymatic nucleic acid molecule consisting essentially of a sequence selected from the group of those shown in Tables 4-8, 10, 12, 14-16, 19-22, 24, 26-28, 30, 32, 34 and 36-38.
10. A mammalian cell including an enzymatic nucleic acid molecule of claims 1 or 2.
11. The ceil of claim 10, wherein said cell is a human cell.
12. An expression vector including nucleic acid encoding an enzymatic nucleic acid molecule or multiple enzymatic molecules of claims 1 or 2 in a manner which allows expression of that enzymatic RNA molecule(s) within a mammalian cell.
13. A mammalian cell including an expression vector of claim 12.
14. The cell of claim 13, wherein said cell is a human cell.
15. A method for treatment of a pathological condition related to the mRNA level of ICAM-1 , IL-5, rel A, TNF-α, or RSV by administering to a patient an enzymatic nucleic acid molecule of claim 1 or 2.
16. A method for treatment of a pathological condition related to the mRNA level of ICAM-1 , IL-5, rel A, TNF-α, or RSV by administering to a patient an expression vector of claim 12.
17. The method of claims 15 or 16, wherein said patient is a human.
18. The method of claim 17 wherein said condition is selected from the group consisting of atherosclerosis, myocardial infraction, stroke, restenosis, heart diseases, cancer, rheumatoid arthritis, asthma, reperfusion injury, inflammatory or autoimmune disorders, transplant rejection, myocardial ischemia, stroke, psoriasis, Kawasaki disease, HIV and AIDS, and septic shock.
19. A nucleoside selected from the group consisting of 5'-C- alkylnucleoside, 2'-deoxy-2'-alkylnucleoside, nucleoside 5'-deoxy- 5'-dihalo-methylphosphonate, nucleoside 5'-deoxy-5'-difluoromethylphosphonate, nucleoside 3'-deoxy-3'-dihalomethylphosphonate, and 5',3'-dideoxy-5',3'-bis(dihalo)- methylphosphonate.
20. A nucleotide selected from the group consisting of 5'-C- alkylnucleotide, 2'-deoxy-2'-alkylnucleotide, 5'-deoxy-5'-dihalomethylnucleotide, 5'-deoxy-5'-difluoro-methylnucleotide, 3'-deoxy3'-dihalo-methylnucleotide, and 5',3,-dideoxy-5',3'-bis(dihalo)- methylphosphonate.
21. A nucleotide triphosphate comprising a nucleotide selected from the group consisting of 5'-C-alkylnucleotide, 2'-deoxy-2'- alkylnucleotide, 5'-deoxy-5'-dihalo-methylnucleotide, 5'-deoxy-5'- difluoro-methylnucleotide, 3'-deoxy-3'-dihalo-methylnucleotide, and 5',3'-dideoxy-5',3'-bis(dihalo)-methylphosphonate.
22. The 5'-C-alkylnucleoside of claim 19, wherein the sugar portion is in a talo configuration.
23. The 5'-C-alkylnucleoside of claim 19, wherein the sugar portion is in an allo configuration.
24. An oligonucleotide comprising a nucleotide selected from the group consisting of 5'-C-alkylnucleotide, 2'-deoxy-2'-alkylnucleotide, 5'- deoxy-5'-dihalo-methylnucleotide, 5'-deoxy-5'-difluoromethylnucleotide, 3'-deoxy-3'-dihalo-methylnucleotide, and 5',3'- dideoxy-5',3'-bis(dihalo)-methylphosphonate.
25. An oligonucleotide comprising a moiety having the formula: wherein B is a nucleotide base or hydrogen; R1 , R2 and R3 independently is selected from the group consisting of hydrogen, an alkyl group containing between 2 and 10 carbon atoms inclusive, an amine, an amino acid, and a peptide containing between 2 and 5 amino acids inclusive; and the zigzag lines are independently hydrogen or a bond.
26. An oligonucleotide comprising a 3'-amido or peptido group.
27. An oligonucleotide comprising a 5'-amido or peptido group.
28. The oligonucleotide of claim 24, 25, 26, or 27 having enzymatic activity.
29. Method for producing an enzymatic nucleic acid molecule having activity to cleave an RNA or single-stranded DNA molecule, comprising the step of forming said enzymatic molecule with at least one nucleotide having an alkyl group at its 5'-position or 2'- position.
30. Method for conversion of a protected allo sugar to a protected talo sugar, comprising the step of contacting said protected allo sugar with triphenyl phosphine, diethylazodicarboxylate, p-nitrobenzoic acid under inversion causing conditions to provide said protected talo sugar.
31. Method for the synthesis of a nucleoside 5' or a 3'-dihalomethylphosphonate comprising the step of condensing a difluoromethylphosphonate-containing sugar with a pyrimidine or purine under conditions suitable for forming a nucleoside 5'- or 3'- difluoromethylphosphonate.
32. The oligonucleotide of claim 3, wherein the normal hammerhead U4 and/or U7 positions are substituted with 2'-NH-amino acid.
33. A method for the synthesis of RNA comprising the step of providing 5-S-alkyitetrazole at a delivered 0.1-1.0 M concentration for the activation of a RNA amidite during a coupling step for less than or equal to 10 minutes.
34. A method for the synthesis of RNA comprising the step of providing 5-S-alkyltetrazole at 0.15-0.35 M effective, or final, concentration for the activation of a RNA amidite during a coupling step for less than or equal to 10 minutes.
35. A method for the deprotection of RNA comprising the step of providing alkylamine (MA) or NH4OH/alkylamine (AMA) at between 60°C - 70°C for 5 to 15 minutes to remove any exocyclic amino protecting groups from protected RNA; wherein said alkyl is selected from the group consisting of methyl, ethyl, propyl and butyl.
36. A method for the deprotection of RNA alkylsilyl protecting groups comprising, contacting said groups with anhydrous triethylamine-hydrogen fluoride (aHF•TEA) trimethylamine or disopropylethylamine at between 60 °C-70 °C for 0.25-24 h.
37. A method for the purification of an RNA molecule by passing said enzymatic RNA molecule over an HPLC column, wherein said HPCC column is an anion exchange chromatography column.
38. Method for one pot deprotection of RNA comprising, contacting a protected base with anhydrous methyl amine at between 60 °C-70 °C for at least 5 min, cooling the resulting mixture and contacting said mixture with TEA-3HF reagents under conditions which remove a protecting group of the 2'-hydroxyl position.
39. Method for synthesizing RNA containing a phosphorothioate linkage comprising the step of contacting 6-10 equivalents of 3H-1 ,2- benzodithiole-3-one 1 ,1 -dioxide (Beaucage reagent) with the growing RNA chain for 5 seconds with a reaction time of at least 300 seconds.
40. Method of synthesizing RNA containing a phosphorothioate linkage comprising the step of achieving coupling with 5-S-ethyltetrazole or 5-S-methyltetrazole prior to sulfurization.
41. Method of claims 38, 39 or 40 wherein said RNA is enzymatically active.
42. Method for synthesizing 2'-deoxy-2'-amino-nucleoside phosphoramidite, comprising the step of protecting the 2'-amino group with a N-phtaloyl group.
43. The method of claim 42 wherein the said nucleoside lacks a base.
44. Method for synthesis of RNA comprising the step of: protecting the
2'-position of a nucleotide during said synthesis with a (trimethylsilyl)ethoxymethyl (SEM) group.
45. Method for covalently linking a SEM group to the 2'-position of a nucleotide, comprising the step of: contacting a nucleoside with an SEM-containing molecule under SEM bonding conditions.
46. The method of claim 45, wherein said conditions comprise dibutyltin oxide and tetrabutylammonium fluoride and SEM-Cl.
47. Method for removal of an SEM group from a nucleoside molecule or an oligonucleotide, comprising the step of: contacting said molecule or oligonucleotide with boron trifluoride etherate
(BF3•OEt2) under SEM removing conditions.
48. The method of claim 57 wherein said (BF3•OEt2) is provided in acetonitrile.
49. One or more vectors comprising a first nucleic acid sequence encoding a first ribozyme having intramolecular or intermolecular cleaving activity, said first ribozyme being selected from the group consisting of a hammerhead, hairpin, hepatitis delta virus, Neurospora VS RNA,
Group I, and RNaseP motif; and a second nucleic acid sequence encoding a second ribozyme having intermolecular cleaving activity, said Second ribozyme being selected from the group consisting of a hammerhead, hairpin, hepatitis delta virus, Neurospora VS RNA, Group I, and RNaseP motif and said second nucleic acid being flanked by other nucleic acid sequences encoding RNA which is cleaved by said first ribozyme to release said second ribozyme from RNA encoded by said vector; wherein said first and second nucleic acid sequences may be on the same or separate nucleic acid molecules, and said vector encodes mRNA or comprises RNA which lacks secondary structure which reduces release of said second ribozyme by more than 20%.
50. Cell comprising the vector of claim 49.
51. A transcribed non-naturally occurring RNA molecule, comprising a desired therapeutic RNA portion, wherein said molecule comprises an intramolecular stem formed by base-pairing interactions between a 3' region and 5' complementary nucleotides in said
RNA, wherein said stem comprises at least 8 base pairs.
52. The RNA molecule of claim 51, wherein said molecule is transcribed by a RNA polymerase III based promoter system.
53. The RNA molecule of claim 51, wherein said molecule is transcribed by a type 2 pol III promoter system.
54. The RNA molecule of claim 51 , wherein said molecule is a chimeric tRNA.
55. The RNA molecule of claim 53, said RNA having A and B boxes of a type 2 pol III promoter separated by between 0 and 300 bases.
56. The RNA molecule of claim 53, wherein said desired RNA molecule is at the 3' end of said B box.
57. The RNA molecule of claim 53, wherein said desired RNA molecule is in between the said A and the B box.
58. The RNA molecule of claim 53, wherein said desired RNA molecule includes said B box.
59. The RNA molecule of claim 51 , wherein said desired RNA molecule is selected from the group consisting of antisense RNA, decoy RNA, therapeutic editing RNA, enzymatic RNA, agonist RNA and antagonist RNA.
60. The RNA molecule of claim 51 , wherein said 5' terminus is able to base-pair with at least 12 bases of said 3' region.
61. The RNA molecule of claim 51 , wherein said 5' terminus is able to base-pair with at least 15 bases of said 3' region.
62. DNA vector encoding the RNA molecule of claim 51
63. The vector of claim 62, wherein said vector is derived from an AAV or adeno virus.
64. RNA vector encoding the RNA molecule of claim 51.
65. The vector of claim 64, wherein said vector is derived from an alpha virus or retro virus.
66. The vector of claim 62 wherein the portions of the vector encoding said RNA function as a RNA pol III promoter.
67. Cell comprising the vector of claim 62.
68. Cell comprising the vector of claim 53.
69. Cell comprising the RNA of claim 51 .
70. Method to provide a desired RNA molecule in a cell, comprising introducing said molecule into said cell a RNA comprising a desired RNA molecule, having a 5' terminus able to base pair with at least 8 bases of a 3' region of said RNA molecule.
71. The method of claim 70, wherein said introducing comprises providing a vector encoding said RNA molecule.
72. Hammerhead ribozyme having 2 or 3 base pairs in stem II with an interconnecting loop of 4 or more bases between said base pairs.
73. Hairpin ribozyme lacking a substrate moiety, comprising at least six bases in helix 2 and able to base-pair with a separate substrate
RNA, wherein the said ribozyme comprises one or more bases 3' of helix 3 able to base-pair with the said substrate RNA to form a helix 5 and wherein the said ribozyme can cleave and/or ligate said separate RNA(s) in trans.
74. The ribozyme of claim 73, wherein said ribozyme comprises six bases in helix 2.
75. The ribozyme of claim 73, having the structure of Fig. 3, wherein each N and N' is independently any base and each dash may represent a hydrogen bond, r is 1-20, q is 2-20, o is 0 - 20, n is 1 - 4, and m is 1 - 20.
76. Method for increasing the activity of a hairpin ribozyme by providing one or more bases 3' of helix 3 able to base-pair with a substrate RNA to form a helix 5.
77. Trans-cleaving Hairpin ribozyme comprising at least 6 base pairs in helix 2 lacking a substrate RNA moiety.
78. Trans-ligating Hairpin ribozyme comprising at least 6 base pairs in helix 2 lacking a substrate RNA moiety.
79. The ribozyme of claim 73 having the structure of Fig. 73.
80. The ribozyme of claim 73 having the structure of Fig. 74.
81. A cell including the ribozyme of any of claims 73-80.
82. An expression vector comprising nucleic acid encoding the ribozyme of any of claims 73-80, in a manner which allows expression of that ribozyme within a cell.
83. A cell including an expression vector of claim 82.
84. Method for altering in vivo the nucleotide base sequence of a naturally occurring mutant nucleic acid molecule, comprising the steps of: contacting said nucleic acid molecule in vivo with an oligonucleotide or peptide nucleic acid able to form a duplex or triplex molecule with said nucleic acid molecule, wherein formation of said duplex or triplex molecule directly, or after nucleic acid repair in vivo, causes at least one base in said nucleic acid molecule to be chemically modified to functionally alter the nucleotide base sequence of said nucleic acid sequence.
85. The method of claim 84, wherein said oligonucleotide is of a length sufficient to activate dsRNA deaminase in vivo to cause conversion of an adenine base to inosine in an RNA molecule.
86. The method of claim 84, wherein said oligonucleotide comprises an enzymatic nucleic acid molecule which is active to chemically modify a base.
87. The method claim 84, wherein said nucleic acid molecule is DNA or
RNA.
88. The method of claim 84, wherein said oligonucleotide comprises a chemical mutagen.
89. The method of claim 88, wherein said mutagen is nitrous acid.
90. The method of claim 84 wherein said oligonucleotide causes deamination of 5-methylcytosine to thymidine, cytosine to uracil, or adenine to inosine, or methylation of cytosine to 5-methylcytosine.
91. The method of claim 84, wherein an endogenous mammalian editing system is co-opted to cause said chemical modification.
92. Method for introduction of enzymatic nucleic acid into a cell or tissue, comprising the steps of; providing a complex of a first nucleic acid molecule encoding said enzymatic nucleic acid associated with a second nucleic acid molecule having sufficient complementarity with said first nucleic acid molecule so that it is able to form an R-loop base-paired structure under physiological conditions with said first nucleic acid molecule; wherein said R-loop is formed in a region of said first nucleic acid molecule at a location which promotes expression of RNA from said first nucleic acid under said conditions; and contacting said complex with said cell or tissue under conditions in which said enzymatic nucleic acid molecule is produced in said cell or tissue.
93. Method for introduction of a desired nucleic acid into a cell or tissue, comprising the steps of; providing a complex of a first nucleic acid molecule encoding said desired nucleic acid associated with a second nucleic acid molecule having sufficient complementarity with said first nucleic acid molecule so that it is able to form an R-loop base-paired structure under physiological conditions with said first nucleic acid molecule; wherein said first nucleic acid molecule lacks a promoter region and said R-loop is formed in a region of said first nucleic acid molecule at a location which promotes expression of RNA from said first nucleic acid under said conditions; and contacting said complex with said cell or tissue under conditions in which said desired acid molecule is produced in said cell or tissue.
94 Method for introduction of a desired nucleic acid into a cell or tissue, comprising the steps of; providing a complex of a first nucleic acid molecule encoding said enzymatic nucleic acid associated with a second nucleic acid molecule having sufficient complementarity with said first nucleic acid molecule so that it is able to form an R-loop base-paired structure under physiological conditions with said first nucleic acid molecule; wherein said R-loop is formed in a region of said first nucleic acid molecule at a location which promotes expression of RNA from said first nucleic acid under said conditions; and wherein said second nucleic acid further comprises a localization factor; and contacting said complex with said cell or tissue under conditions in which said desired nucleic acid molecule is produced in said cell or tissue.
95. Complex of a first nucleic acid molecule encoding an enzymatic nucleic acid associated with a second nucleic acid molecule having sufficient complementarity with said first nucleic acid molecule so that it is able to form an R-loop base-paired structure under physiological conditions with said first nucleic acid molecule; wherein said R-loop is formed in a region of said first nucleic acid molecule at a location which promotes expression of RNA from said first nucleic acid under said conditions.
96. Complex of a first nucleic acid molecule encoding a desired nucleic acid associated with a second nucleic acid molecule having sufficient complementarity with said first nucleic acid molecule so that it is able to form an R-loop base-paired structure under physiological conditions with said first nucleic acid molecule; wherein said first nucleic acid molecule lacks a promoter region and said R-loop is formed in a region of said first nucleic acid molecule at a location which promotes expression of RNA from said first nucleic acid under said conditions.
97. Complex of a first nucleic acid molecule encoding an enzymatic nucleic acid associated with a second nucleic acid molecule having sufficient complementarity with said first nucleic acid molecule so that it is able to form an R-loop base-paired structure under physiological conditions with said first nucleic acid molecule; wherein said R-loop is formed in a region of said first nucleic acid molecule at a location which promotes expression of RNA from said first nucleic acid under said conditions, and wherein said second nucleic acid further comprises a localization factor.
PCT/IB1995/000156 1994-02-23 1995-02-23 Method and reagent for inhibiting the expression of disease related genes WO1995023225A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
MX9603638A MX9603638A (en) 1994-02-23 1995-02-23 Method and reagent for inhibiting the expression of disease related genes.
EP95909920A EP0746614A1 (en) 1994-02-23 1995-02-23 Method and reagent for inhibiting the expression of disease related genes
AU18214/95A AU706417B2 (en) 1994-02-23 1995-02-23 Method and reagent for inhibiting the expression of disease related genes
JP7522236A JPH09509323A (en) 1994-02-23 1995-02-23 Methods and reagents for inhibiting expression of disease-related genes

Applications Claiming Priority (52)

Application Number Priority Date Filing Date Title
US20110994A 1994-02-23 1994-02-23
US08/201,109 1994-02-23
US08/218,934 1994-03-29
US08/218,934 US5639647A (en) 1994-03-29 1994-03-29 2'-deoxy-2'alkylnucleotide containing nucleic acid
US22279594A 1994-04-04 1994-04-04
US08/222,795 1994-04-04
US22448394A 1994-04-07 1994-04-07
US08/224,483 1994-04-07
US22804194A 1994-04-15 1994-04-15
US22795894A 1994-04-15 1994-04-15
US08/228,041 1994-04-15
US08/227,958 1994-04-15
US24573694A 1994-05-18 1994-05-18
US08/245,736 1994-05-18
US27128094A 1994-07-06 1994-07-06
US08/271,280 1994-07-06
US08/291,932 US5658780A (en) 1992-12-07 1994-08-15 Rel a targeted ribozymes
US08/291,932 1994-08-15
US29143394A 1994-08-16 1994-08-16
US08/291,433 1994-08-16
US08/292,620 1994-08-17
US08/292,620 US5837542A (en) 1992-12-07 1994-08-17 Intercellular adhesion molecule-1 (ICAM-1) ribozymes
US29352094A 1994-08-19 1994-08-19
US08/293,520 1994-08-19
US30000094A 1994-09-02 1994-09-02
US08/300,000 1994-09-02
US30303994A 1994-09-08 1994-09-08
US08/303,039 1994-09-08
US31174994A 1994-09-23 1994-09-23
US08/311,486 US5811300A (en) 1992-12-07 1994-09-23 TNF-α ribozymes
US08/311,749 1994-09-23
US08/311,486 1994-09-23
US31439794A 1994-09-28 1994-09-28
US08/314,397 1994-09-28
US31677194A 1994-10-03 1994-10-03
US08/316,771 1994-10-03
US08/319,492 1994-10-07
US08/319,492 US5616488A (en) 1992-12-07 1994-10-07 IL-5 targeted ribozymes
US08/321,993 1994-10-11
US08/321,993 US5631359A (en) 1994-10-11 1994-10-11 Hairpin ribozymes
US08/334,847 US5693532A (en) 1994-11-04 1994-11-04 Respiratory syncytial virus ribozymes
US08/334,847 1994-11-04
US08/337,608 1994-11-10
US08/337,608 US5902880A (en) 1994-08-19 1994-11-10 RNA polymerase III-based expression of therapeutic RNAs
US34551694A 1994-11-28 1994-11-28
US08/345,516 1994-11-28
US08/357,577 US5783425A (en) 1993-10-27 1994-12-16 Amino and peptido modified enzymatic nucleic acid
US08/357,577 1994-12-16
US08/363,233 US5714383A (en) 1992-05-14 1994-12-23 Method and reagent for treating chronic myelogenous leukemia
US08/363,233 1994-12-23
US38073495A 1995-01-30 1995-01-30
US08/380,734 1995-01-30

Publications (2)

Publication Number Publication Date
WO1995023225A2 true WO1995023225A2 (en) 1995-08-31
WO1995023225A3 WO1995023225A3 (en) 1996-02-01

Family

ID=27586798

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB1995/000156 WO1995023225A2 (en) 1994-02-23 1995-02-23 Method and reagent for inhibiting the expression of disease related genes

Country Status (4)

Country Link
EP (1) EP0746614A1 (en)
AU (1) AU706417B2 (en)
CA (1) CA2183992A1 (en)
WO (1) WO1995023225A2 (en)

Cited By (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996018736A2 (en) * 1994-12-13 1996-06-20 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of arthritic conditions, induction of graft tolerance and reversal of immune responses
WO1996018733A2 (en) * 1994-12-14 1996-06-20 Innovir Laboratories, Inc. Ribozyme-mediated inactivation of leukemia-associated rna
WO1997010334A2 (en) * 1995-09-12 1997-03-20 Immusol, Inc. Ribozyme therapy for the treatment and/or prevention of restenosis
US5672501A (en) * 1994-12-23 1997-09-30 Ribozyme Pharmaceuticals, Inc. Base-modified enzymatic nucleic acid
US5807743A (en) * 1996-12-03 1998-09-15 Ribozyme Pharmaceuticals, Inc. Interleukin-2 receptor gamma-chain ribozymes
WO1998050530A2 (en) * 1997-05-09 1998-11-12 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids: synthesis, selection and use
US5869248A (en) * 1994-03-07 1999-02-09 Yale University Targeted cleavage of RNA using ribonuclease P targeting and cleavage sequences
US5877162A (en) * 1996-03-14 1999-03-02 Innovir Laboratories, Inc. Short external guide sequences
US6013447A (en) * 1997-11-21 2000-01-11 Innovir Laboratories, Inc. Random intracellular method for obtaining optimally active nucleic acid molecules
WO2000020645A1 (en) * 1998-10-05 2000-04-13 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE MODULATION OF TUMOR NECROSIS FACTOR-α (TNF-α) EXPRESSION
US6054576A (en) * 1997-10-02 2000-04-25 Ribozyme Pharmaceuticals, Inc. Deprotection of RNA
US6057153A (en) * 1995-01-13 2000-05-02 Yale University Stabilized external guide sequences
US6069008A (en) * 1998-11-25 2000-05-30 Isis Pharmaceuticals Inc. Antisense modulation of NF-kappa-B p65 subunit expression
US6080580A (en) * 1998-10-05 2000-06-27 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of tumor necrosis factor-α (TNF-α) expression
NL1010917C2 (en) * 1998-12-30 2000-07-03 Stichting Dienst Landbouwkundi DNA oligonucleotide specific for Meloidogyne species, DNA vector and host cell containing the oligonucleotide, use and kit.
US6127535A (en) * 1997-11-05 2000-10-03 Ribozyme Pharmaceuticals, Inc. Nucleoside triphosphates and their incorporation into oligonucleotides
US6136603A (en) * 1999-03-26 2000-10-24 Isis Pharmaceuticals Inc. Antisense modulation of interleukin-5 signal transduction
US6159951A (en) * 1997-02-13 2000-12-12 Ribozyme Pharmaceuticals Inc. 2'-O-amino-containing nucleoside analogs and polynucleotides
US6183959B1 (en) 1997-07-03 2001-02-06 Ribozyme Pharmaceuticals, Inc. Method for target site selection and discovery
US6248878B1 (en) 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
WO2001047530A1 (en) * 1999-12-28 2001-07-05 Murdoch Childrens Research Institute A method of therapy and prophylaxis of inflammation
US6262251B1 (en) 1995-10-19 2001-07-17 Proligo Llc Method for solution phase synthesis of oligonucleotides
US6280936B1 (en) 1997-06-09 2001-08-28 Ribozyme Pharmaceuticals, Inc. Method for screening nucleic acid catalysts
US6316612B1 (en) 1997-08-22 2001-11-13 Ribozyme Pharmaceuticals, Inc. Xylofuranosly-containing nucleoside phosphoramidites and polynucleotides
US6346398B1 (en) 1995-10-26 2002-02-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
EP1251170A2 (en) * 1992-07-17 2002-10-23 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of NF-kappaB dependent animal diseases
US6528639B2 (en) 1992-10-15 2003-03-04 Ribozyme Pharmaceuticals, Inc. Base-modified enzymatic nucleic acid
US6548657B1 (en) 1997-06-09 2003-04-15 Ribozyme Pharmaceuticals, Inc. Method for screening nucleic acid catalysts
WO2003050241A2 (en) 2001-12-12 2003-06-19 The Government Of The United States Of America As Represented By The Secretary, Department Of Healthand Human Services Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation
US6610478B1 (en) 1996-08-16 2003-08-26 Yale University Phenotypic conversion of cells mediated by external guide sequences
US6617438B1 (en) 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US6620805B1 (en) 1996-03-14 2003-09-16 Yale University Delivery of nucleic acids by porphyrins
US6969589B2 (en) 2001-03-30 2005-11-29 Perlegen Sciences, Inc. Methods for genomic analysis
US6989442B2 (en) 2002-07-12 2006-01-24 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US6995259B1 (en) 1998-10-23 2006-02-07 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
US7022828B2 (en) 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
US7034009B2 (en) 1995-10-26 2006-04-25 Sirna Therapeutics, Inc. Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R)
US7115726B2 (en) 2001-03-30 2006-10-03 Perlegen Sciences, Inc. Haplotype structures of chromosome 21
US7125660B2 (en) 2000-09-13 2006-10-24 Archemix Corp. Nucleic acid sensor molecules and methods of using same
US7205399B1 (en) 2001-07-06 2007-04-17 Sirna Therapeutics, Inc. Methods and reagents for oligonucleotide synthesis
EP1958964A2 (en) 2004-02-24 2008-08-20 The Government of the United States of America, as represented by The Secretary, Department of Health and Human Services RAB9A, RAB11A, and modulators thereof related to infectious disease
EP1970058A1 (en) 1998-08-18 2008-09-17 The Regents of the University of California Office of Technology Transfer Epidermal growth factor receptor antagonists for treating hypersecretion of mucus in the lungs
EP1982720A1 (en) 2002-05-20 2008-10-22 The Regents of the University of California Ribaviren for use to treat cancer
WO2009042686A1 (en) 2007-09-27 2009-04-02 Perlegen Sciences, Inc. Methods for genetic analysis
EP2121921A2 (en) * 2006-12-13 2009-11-25 Idiverse, Inc. Novel genes and rna molecules that confer stress tolerance
US7655790B2 (en) 2002-07-12 2010-02-02 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US7888325B2 (en) 1999-01-28 2011-02-15 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US8063198B2 (en) 2004-04-05 2011-11-22 Alnylam Pharmaceuticals, Inc. Processes and reagents for desilylation of oligonucleotides
US8106013B2 (en) 2006-05-19 2012-01-31 Georgia Tech Research Corporation ABC transporter ligand GATX1
US8324158B2 (en) 2006-07-14 2012-12-04 Georgia Tech Research Corporation Methods for inhibiting CLC-2 channel with GATX2
EP2628799A3 (en) * 2005-01-07 2013-08-28 Alnylam Pharmaceuticals Inc. RNAi modulation of RSV and therapeutic uses thereof
WO2014165814A1 (en) 2013-04-04 2014-10-09 Georgia State University Research Foundation, Inc. Rna microchip detection using nanoparticle-assisted signal amplification
US9029527B2 (en) 1998-03-20 2015-05-12 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
US9051566B2 (en) 2001-01-31 2015-06-09 Alnylam Pharmaceuticals, Inc. Post-transcriptional gene silencing using expressed double stranded RNA
WO2016011381A1 (en) 2014-07-18 2016-01-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Reducing cxcr4 expression and/or function to enhance engraftment of hematopoietic stem cells
WO2016187217A2 (en) 2015-05-18 2016-11-24 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated impairments
US9708621B2 (en) 1999-08-13 2017-07-18 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US9963698B2 (en) 1998-03-20 2018-05-08 Commonwealth Scientific And Industrial Research Organisation Control of gene expression
US10265372B2 (en) 2014-08-12 2019-04-23 The Regents Of The University Of California Molecular composition for enhancing and rejuvenating maintenance and repair of mammalian tissues
WO2021158789A1 (en) * 2020-02-07 2021-08-12 Ultragenyx Pharmaceutical Inc. Chaotropic agents for reducing formation of double-stranded rna
CN114544821A (en) * 2020-11-24 2022-05-27 重庆医科大学 Application of reagent for detecting phosphatidylethanolamine (36:4) in blood plasma in preparation of depression detection kit

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111851128A (en) * 2020-07-28 2020-10-30 太和县昌达工贸有限公司 Air-permeable uniform anti-fouling non-woven fabric mesh belt and preparation method thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992007065A1 (en) * 1990-10-12 1992-04-30 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Modified ribozymes
WO1994002595A1 (en) * 1992-07-17 1994-02-03 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of animal diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992007065A1 (en) * 1990-10-12 1992-04-30 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Modified ribozymes
WO1994002595A1 (en) * 1992-07-17 1994-02-03 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of animal diseases

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BIOCHIMICA ET BIOPHYSICA ACTA, vol.1131, pages 108 - 110 KITA, Y. ET AL. 'Sequence and expression of rat ICAM-1' *
BIOTECHNOLOGY, vol.10, 2, NEW YORK US pages 256 - 262 EDGINGTON, S.M. 'Ribozymes: stop making sense' *
NATURE, vol.331, 18 February 1988, LONDON GB pages 624 - 627 SIMMONS, D. ET AL. 'ICAM, an adhesion ligand of LFA-1, is homologous to the neural cell adhesion molecule NCAM' cited in the application *
NUCLEIC ACIDS RESEARCH, vol.17, no.14, , OXFORD GB page 5853 BALLANTYNE, C. ET AL. 'Nucleotide sequence of the cDNA for murine intercellular adhesion molecule-1 (ICAM-1)' *
SCIENCE, vol.262, 3 December 1993, LANCASTER, PA US pages 1512 - 1514 BARINAGA, M. 'ribozymes: killing the messenger' *
TIBS TRENDS IN BIOCHEMICAL SCIENCES, vol.17, 2, CAMBRIDGE GB pages 334 - 339 USMAN, N. & CEDERGREN, R. 'Exploiting the chemical synthesis of RNA' cited in the application *
TRENDS IN BIOTECHNOLOGY, vol.8, no.7, 1 July 1990 pages 174 - 178, XP000133091 COTTEN, M. 'THE IN VIVO APPLICATION OF RIBOZYMES' *

Cited By (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1251170A2 (en) * 1992-07-17 2002-10-23 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of NF-kappaB dependent animal diseases
EP1251170A3 (en) * 1992-07-17 2002-10-30 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of NF-kappaB dependent animal diseases
US6528639B2 (en) 1992-10-15 2003-03-04 Ribozyme Pharmaceuticals, Inc. Base-modified enzymatic nucleic acid
US5869248A (en) * 1994-03-07 1999-02-09 Yale University Targeted cleavage of RNA using ribonuclease P targeting and cleavage sequences
US5834440A (en) * 1994-03-07 1998-11-10 Immusol Incorporated Ribozyme therapy for the inhibition of restenosis
WO1996018736A3 (en) * 1994-12-13 1996-09-19 Ribozyme Pharm Inc Method and reagent for treatment of arthritic conditions, induction of graft tolerance and reversal of immune responses
WO1996018736A2 (en) * 1994-12-13 1996-06-20 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of arthritic conditions, induction of graft tolerance and reversal of immune responses
WO1996018733A2 (en) * 1994-12-14 1996-06-20 Innovir Laboratories, Inc. Ribozyme-mediated inactivation of leukemia-associated rna
WO1996018733A3 (en) * 1994-12-14 1996-12-12 Innovir Lab Inc Ribozyme-mediated inactivation of leukemia-associated rna
US5672501A (en) * 1994-12-23 1997-09-30 Ribozyme Pharmaceuticals, Inc. Base-modified enzymatic nucleic acid
US6057153A (en) * 1995-01-13 2000-05-02 Yale University Stabilized external guide sequences
WO1997010334A2 (en) * 1995-09-12 1997-03-20 Immusol, Inc. Ribozyme therapy for the treatment and/or prevention of restenosis
WO1997010334A3 (en) * 1995-09-12 1997-06-05 Immusol Inc Ribozyme therapy for the treatment and/or prevention of restenosis
US6262251B1 (en) 1995-10-19 2001-07-17 Proligo Llc Method for solution phase synthesis of oligonucleotides
US6566127B1 (en) 1995-10-26 2003-05-20 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
US6346398B1 (en) 1995-10-26 2002-02-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
US6818447B1 (en) 1995-10-26 2004-11-16 Sirna Therapeutics, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
US7034009B2 (en) 1995-10-26 2006-04-25 Sirna Therapeutics, Inc. Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R)
US5877162A (en) * 1996-03-14 1999-03-02 Innovir Laboratories, Inc. Short external guide sequences
US6620805B1 (en) 1996-03-14 2003-09-16 Yale University Delivery of nucleic acids by porphyrins
US6610478B1 (en) 1996-08-16 2003-08-26 Yale University Phenotypic conversion of cells mediated by external guide sequences
US5807743A (en) * 1996-12-03 1998-09-15 Ribozyme Pharmaceuticals, Inc. Interleukin-2 receptor gamma-chain ribozymes
US6248878B1 (en) 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
US6506888B1 (en) 1997-02-13 2003-01-14 Ribozyme Pharmaceuticals, Inc. 2′-O-amino-containing nucleoside analogs and polynucleotides
US6159951A (en) * 1997-02-13 2000-12-12 Ribozyme Pharmaceuticals Inc. 2'-O-amino-containing nucleoside analogs and polynucleotides
WO1998050530A2 (en) * 1997-05-09 1998-11-12 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids: synthesis, selection and use
WO1998050530A3 (en) * 1997-05-09 1999-07-29 Ribozyme Pharm Inc Enzymatic nucleic acids: synthesis, selection and use
US6548657B1 (en) 1997-06-09 2003-04-15 Ribozyme Pharmaceuticals, Inc. Method for screening nucleic acid catalysts
US6280936B1 (en) 1997-06-09 2001-08-28 Ribozyme Pharmaceuticals, Inc. Method for screening nucleic acid catalysts
US6183959B1 (en) 1997-07-03 2001-02-06 Ribozyme Pharmaceuticals, Inc. Method for target site selection and discovery
US6448009B1 (en) 1997-07-03 2002-09-10 Ribozyme Pharmaceuticals, Inc. Method for target site selection and discovery
US6489465B2 (en) 1997-08-22 2002-12-03 Ribozyme Pharmaceuticals, Inc. Xylofuranosly-containing nucleoside phosphoramidites and polynucleotides
US6316612B1 (en) 1997-08-22 2001-11-13 Ribozyme Pharmaceuticals, Inc. Xylofuranosly-containing nucleoside phosphoramidites and polynucleotides
US6797815B2 (en) 1997-08-22 2004-09-28 Sirna Therapeutics, Inc. Xylofuranosly-containing nucleoside phosphoramidites and polynucleotides
US6673918B2 (en) 1997-10-02 2004-01-06 Sirna Therapeutics, Inc. Deprotection of RNA
US6162909A (en) * 1997-10-02 2000-12-19 Ribozyme Pharmaceuticals, Inc. Deprotection of RNA
US6054576A (en) * 1997-10-02 2000-04-25 Ribozyme Pharmaceuticals, Inc. Deprotection of RNA
US6617438B1 (en) 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US6509460B1 (en) 1997-11-05 2003-01-21 Ribozyme Pharmaceuticals, Inc. Nucleoside triphosphates and their incorporation into oligonucleotides
US6127535A (en) * 1997-11-05 2000-10-03 Ribozyme Pharmaceuticals, Inc. Nucleoside triphosphates and their incorporation into oligonucleotides
US6013447A (en) * 1997-11-21 2000-01-11 Innovir Laboratories, Inc. Random intracellular method for obtaining optimally active nucleic acid molecules
US9029527B2 (en) 1998-03-20 2015-05-12 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
US9963698B2 (en) 1998-03-20 2018-05-08 Commonwealth Scientific And Industrial Research Organisation Control of gene expression
EP1970058A1 (en) 1998-08-18 2008-09-17 The Regents of the University of California Office of Technology Transfer Epidermal growth factor receptor antagonists for treating hypersecretion of mucus in the lungs
US6080580A (en) * 1998-10-05 2000-06-27 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of tumor necrosis factor-α (TNF-α) expression
WO2000020645A1 (en) * 1998-10-05 2000-04-13 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE MODULATION OF TUMOR NECROSIS FACTOR-α (TNF-α) EXPRESSION
US6228642B1 (en) 1998-10-05 2001-05-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of tumor necrosis factor-(α) (TNF-α) expression
US7777023B2 (en) 1998-10-23 2010-08-17 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
US6995259B1 (en) 1998-10-23 2006-02-07 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
US6069008A (en) * 1998-11-25 2000-05-30 Isis Pharmaceuticals Inc. Antisense modulation of NF-kappa-B p65 subunit expression
NL1010917C2 (en) * 1998-12-30 2000-07-03 Stichting Dienst Landbouwkundi DNA oligonucleotide specific for Meloidogyne species, DNA vector and host cell containing the oligonucleotide, use and kit.
US8148345B2 (en) 1999-01-28 2012-04-03 Georgia Health Sciences University Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US7888325B2 (en) 1999-01-28 2011-02-15 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US6136603A (en) * 1999-03-26 2000-10-24 Isis Pharmaceuticals Inc. Antisense modulation of interleukin-5 signal transduction
US9708621B2 (en) 1999-08-13 2017-07-18 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US10190127B2 (en) 1999-08-13 2019-01-29 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
WO2001047530A1 (en) * 1999-12-28 2001-07-05 Murdoch Childrens Research Institute A method of therapy and prophylaxis of inflammation
US7125660B2 (en) 2000-09-13 2006-10-24 Archemix Corp. Nucleic acid sensor molecules and methods of using same
US9051566B2 (en) 2001-01-31 2015-06-09 Alnylam Pharmaceuticals, Inc. Post-transcriptional gene silencing using expressed double stranded RNA
US7115726B2 (en) 2001-03-30 2006-10-03 Perlegen Sciences, Inc. Haplotype structures of chromosome 21
US6969589B2 (en) 2001-03-30 2005-11-29 Perlegen Sciences, Inc. Methods for genomic analysis
US11031098B2 (en) 2001-03-30 2021-06-08 Genetic Technologies Limited Computer systems and methods for genomic analysis
US7022828B2 (en) 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
US7205399B1 (en) 2001-07-06 2007-04-17 Sirna Therapeutics, Inc. Methods and reagents for oligonucleotide synthesis
WO2003050241A2 (en) 2001-12-12 2003-06-19 The Government Of The United States Of America As Represented By The Secretary, Department Of Healthand Human Services Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation
EP1982720A1 (en) 2002-05-20 2008-10-22 The Regents of the University of California Ribaviren for use to treat cancer
US7655790B2 (en) 2002-07-12 2010-02-02 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US7893249B2 (en) 2002-07-12 2011-02-22 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US6989442B2 (en) 2002-07-12 2006-01-24 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
EP1958964A2 (en) 2004-02-24 2008-08-20 The Government of the United States of America, as represented by The Secretary, Department of Health and Human Services RAB9A, RAB11A, and modulators thereof related to infectious disease
US8063198B2 (en) 2004-04-05 2011-11-22 Alnylam Pharmaceuticals, Inc. Processes and reagents for desilylation of oligonucleotides
US8431693B2 (en) 2004-04-05 2013-04-30 Alnylam Pharmaceuticals, Inc. Process for desilylation of oligonucleotides
AU2010224450B2 (en) * 2005-01-07 2014-04-10 Alnylam Pharmaceuticals, Inc. RNAi modulation of RSV and therapeutic uses thereof
EP2487243A3 (en) * 2005-01-07 2013-08-28 Alnylam Pharmaceuticals Inc. RNAI modulation of RSV and therapeutic uses thereof
EP2628799A3 (en) * 2005-01-07 2013-08-28 Alnylam Pharmaceuticals Inc. RNAi modulation of RSV and therapeutic uses thereof
US8106013B2 (en) 2006-05-19 2012-01-31 Georgia Tech Research Corporation ABC transporter ligand GATX1
US8324158B2 (en) 2006-07-14 2012-12-04 Georgia Tech Research Corporation Methods for inhibiting CLC-2 channel with GATX2
EP2121921A2 (en) * 2006-12-13 2009-11-25 Idiverse, Inc. Novel genes and rna molecules that confer stress tolerance
EP2121921A4 (en) * 2006-12-13 2010-08-04 Idiverse Inc Novel genes and rna molecules that confer stress tolerance
EP2772553A1 (en) 2007-09-27 2014-09-03 Genetic Technologies Limited Methods for genetic analysis
WO2009042686A1 (en) 2007-09-27 2009-04-02 Perlegen Sciences, Inc. Methods for genetic analysis
WO2014165814A1 (en) 2013-04-04 2014-10-09 Georgia State University Research Foundation, Inc. Rna microchip detection using nanoparticle-assisted signal amplification
WO2016011381A1 (en) 2014-07-18 2016-01-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Reducing cxcr4 expression and/or function to enhance engraftment of hematopoietic stem cells
US10265372B2 (en) 2014-08-12 2019-04-23 The Regents Of The University Of California Molecular composition for enhancing and rejuvenating maintenance and repair of mammalian tissues
WO2016187217A2 (en) 2015-05-18 2016-11-24 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated impairments
EP3892315A1 (en) 2015-05-18 2021-10-13 The Board of Trustees of the Leland Stanford Junior University Methods and compositions for treating aging-associated impairments
WO2021158789A1 (en) * 2020-02-07 2021-08-12 Ultragenyx Pharmaceutical Inc. Chaotropic agents for reducing formation of double-stranded rna
CN114544821A (en) * 2020-11-24 2022-05-27 重庆医科大学 Application of reagent for detecting phosphatidylethanolamine (36:4) in blood plasma in preparation of depression detection kit
CN114544821B (en) * 2020-11-24 2023-10-24 重庆医科大学 Application of reagent for detecting phosphatidylethanolamine (36:4) in blood plasma in preparation of depression detection kit

Also Published As

Publication number Publication date
EP0746614A1 (en) 1996-12-11
CA2183992A1 (en) 1995-08-31
AU1821495A (en) 1995-06-01
AU706417B2 (en) 1998-06-17
WO1995023225A3 (en) 1996-02-01

Similar Documents

Publication Publication Date Title
AU706417B2 (en) Method and reagent for inhibiting the expression of disease related genes
US6365374B1 (en) 2′-deoxy-2′-alkylnucleotide containing nucleic acid
US6022962A (en) Hairpin ribozymes
US5891683A (en) Non-nucleotide containing enzymatic nucleic acid
US6107078A (en) Ribozymes with optimized hybridizing arms, stems, and loops, tRNA embedded ribozymes and compositions thereof
US6017756A (en) Method and reagent for inhibiting hepatitis B virus replication
US5716824A (en) 2&#39;-O-alkylthioalkyl and 2-C-alkylthioalkyl-containing enzymatic nucleic acids (ribozymes)
US5545729A (en) Stabilized ribozyme analogs
US6057153A (en) Stabilized external guide sequences
US5616490A (en) Ribozymes targeted to TNF-α RNA
JP2005118056A (en) Method and reagent for inhibiting expression of disease related genes
US5714383A (en) Method and reagent for treating chronic myelogenous leukemia
EP1502950A2 (en) Method for purifying chemically modified RNA
US6656731B1 (en) Nucleic acid catalysts with endonuclease activity
AU744191B2 (en) Method and reagent for inhibiting the expression of disease related genes
CA2468048A1 (en) Process for purifying chemically synthesized rna
AU2774202A (en) Method and reagent for inhibiting the expression of disease related genes
JPH10510715A (en) VS ribozyme
US6492512B1 (en) Method and reagent for treatment of lung cancer and other malignancies caused by the deregulation of L-MYC gene expression
EP1073731A2 (en) Nucleic acid enzyme for rna cleavage
Been et al. Circular RNAs: Generation of Small RNAs with Unique Properties by Splicing Permuted Intron-Exon Sequences

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP KR MX

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

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: A3

Designated state(s): AU CA JP KR MX

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

WWE Wipo information: entry into national phase

Ref document number: 2183992

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995909920

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995909920

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995909920

Country of ref document: EP