WO1997017433A1 - Tissue-specific and target rna-specific ribozymes - Google Patents

Tissue-specific and target rna-specific ribozymes Download PDF

Info

Publication number
WO1997017433A1
WO1997017433A1 PCT/US1996/018088 US9618088W WO9717433A1 WO 1997017433 A1 WO1997017433 A1 WO 1997017433A1 US 9618088 W US9618088 W US 9618088W WO 9717433 A1 WO9717433 A1 WO 9717433A1
Authority
WO
WIPO (PCT)
Prior art keywords
ribozyme
nucleic acid
sequence
specific
rna
Prior art date
Application number
PCT/US1996/018088
Other languages
French (fr)
Inventor
James S. Norris
Gary A. Clawson
Original Assignee
Medical University Of South Carolina
Penn State Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medical University Of South Carolina, Penn State Research Foundation filed Critical Medical University Of South Carolina
Priority to EP96940377A priority Critical patent/EP0866852B1/en
Priority to JP51838197A priority patent/JP4033902B2/en
Priority to AU77272/96A priority patent/AU728732B2/en
Priority to AT96940377T priority patent/ATE294854T1/en
Priority to IL12438396A priority patent/IL124383A0/en
Priority to DE69634698T priority patent/DE69634698T2/en
Priority to CA002236998A priority patent/CA2236998C/en
Publication of WO1997017433A1 publication Critical patent/WO1997017433A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C12N15/1137Non-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 enzymes
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07006DNA-directed RNA polymerase (2.7.7.6)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • 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
    • C12N2310/127DNAzymes
    • 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
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid

Definitions

  • the invention pertains to the use of tissue-specific and target RNA-specific ribozymes for treatment of cancers and bacterial, parasitic and viral infections More specifically, the invention relates to a ribozyme targeted to the RNA polymerase 1(A) under the control ofthe probasin promoter
  • a ribozyme is a catalytic RNA molecule that cleaves RNA in a sequence specific manner
  • ribozyme as potential gene regulators in mammalian cells and antiviral agents has been suggested, but subject to serious questions regarding technical feasibility
  • Other questions concern the effects of target RNA secondary structure on ribozymal activity The art has not been successful in answering any of these questions
  • ribozymes are a form of antisense technology
  • the problems encountered in applying antisense technology to disease treatment are also encountered in the use of ribozyme technology
  • tissue-specific cancers or other tissue-specific disease can be treated by delivering to that tissue a ribozyme having a tissue-specific promoter, and that it is targeted to an RNA that is essential for cell survival
  • the invention provides such a ribozyme capable of treating tissue-specific cancers and other tissue-specific diseases
  • the recently developed probasin promoter provides target target-specificity enabling systemic delivery ofthe present ribozyme-encoding vector (Greenburg et al Mol. Endocrinol. 8 230-239, 1994)
  • the present vector consists of a tandem array of 3 hammerhead ribozymes, the 5' and 3 ' of which are designed to autocatalytically cleave themselves from the primary transcript
  • This novel construct eliminates problems inherent with extensive residual flanking sequences which might otherwise be present to compromise catalytic activity and specificity
  • the present constructs couple the prostate specific probasin promoter to triple-ribozymes targeted at mRNAs critical for prostate cell growth
  • RNA polymerase I produces large amounts of cellular ribosomal RNA
  • a common problem in gene therapy is the difficulty in delivering the ribozyme to the correct tissue
  • the present invention avoids this difficulty by targeting the ribozyme to non-cellular RNAs in cells to which ribozyme constructs can be efficiently delivered IV liposome delivery will be effective for treatment of HBV hepatitis IV and/or extracorporeal treatment will effectively delivery construct to erythrocytes for treatment of malarial infection And topical (with or without iv) administration will effectively deliver ribozyme construct to cervical epithelium in dysplastic/precancerous/cancerous HPV 16 cervical lesions This latter example is of extreme importance for treatment of dysplastic/carcinoma in situ lesions diagnosed via abnormal Pap smears
  • a second advantageous facet ofthe non-cellular target ribozymes is that even if promoter leakiness and/or extraneous delivery and/or expression ofthe ribozyme occurs in unintended cells, the ribozymes should not cleave any cellular RNAs
  • the invention provides tissue-specific and target RNA-specific ribozymes These ribozymes can be used to destroy target-specific neoplasms and to treat viral infections, among other uses
  • the ribozymes ofthe present invention comprise a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme
  • the invention also provides nucleic acids which encode the ribozymes ofthe invention These nucleic acids can be used to express the ribozymes ofthe invention at the selected site
  • the nucleic acids ofthe invention comprise a tissue-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
  • a method of treating a subject having a proliferative disease ofa specific tissue by inhibiting cell proliferation in the tissue comprising administering to the subject the nucleic acid of claim 5, wherein the target-specific promoter binding sequence is specific for the diseased tissue, whereby the ribozyme encoded by the nucleic acid is expressed, ribosomal RNA production in the tissue is inhibited, cell proliferation is inhibited, and the proliferative disease treated is provided
  • a method for treating a subject having prostate cancer comprising administering to the subject the nucleic acid of claim 7, whereby the ribozyme encoded by the nucleic acid is expressed in the prostate and the prostate cancer is treated
  • a method of treating an infection in a subject comprising administering to the subject the nucleic acid of claim 1, wherein the encoded target RNA-specific binding site is specific for an RNA unique to the infectious agent, whereby the ribozyme encoded by the nucleic acid is expressed and the infectious agent is killed is also provided BRIEF DESCRIPTION OF THE FIGURES
  • Fig 1 shows DNA encoding the parental double ribozyme starting at Not I site Underlined sequences are Not I sites (GCGGCCGC) and Bgl II (AGATCT) Sequence of parental double ribozyme starting at Not I site Underlined sequences are Not I sites (GCGGCCGC) and Bgl II (AGATCT)
  • Fig 2 shows DNA encoding the entire sequence with the internal Pol I targeted triple ribozyme in bold Underlined sequences are Not I (GCGGCCGC) and Bgl II (AGATCT) This is the entire sequence with the internal Pol I targeted triple ribozyme in bold Underlined sequences are Not I (GCGGCCGC) and Bgl II (AGATCT)
  • Fig 3 shows the two dimensional structure of parent double ribozyme into which the core ribozyme was cloned, as its reverse complement DNA
  • the invention provides tissue-specific and target RNA-specific ribozymes These ribozymes can be used to destroy tissue-specific neoplasms and to treat viral, bacterial or parasitic infections, among other uses
  • the ribozymes ofthe present invention comprise a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme
  • One example ofthe present ribozyme is shown in by its DNA coding sequence in Fig 2 and in SEQ ID NO 1
  • the nucleotides numbered 1-164 encode the ribozyme, including the 5' and 3' autocatalytic ribozyme sequences
  • the 5' autocatalytically cleaving ribozyme, catalytic ribozyme, and 3 ' autocatalytically cleaving ribozyme of this exemplary ribozyme are
  • the construct In this RNA, the first 20-30 nt (or longer) are followed by a sequence which represents the reverse complement ofthe initial 20-30 nt. This way, the construct would presumably still be capped at the 5 ' end the way pol II transcripts are, but the initial nucleotides should not alter the specificity ofthe nucleotides on the 5 ' side ofthe targeted middle ribozyme. Based upon the fact that the transcript would be capped, it should be exported efficiently to the cytoplasm. For present triple ribozyme construct having both 5' and 3' autocatalytic ribozymes, it is expected that there will be some diffusion mediated transport to the cytoplasm ofthe internal targeted ribozyme, although this alternative 5 ' end should increase the cytoplasmic proportion.
  • the invention provides ribozymes that have the unique characteristic of being both target RNA-specific in their catalytic action, and subject to tissue-specific expression.
  • the target RNA specificity is conferred by an RNA binding site that specifically binds a sequence that is unique to RNA polymerase 1(A) (ribosomal RNA polymerase).
  • RNA polymerase 1(A) ribosomal RNA polymerase
  • an RNA sequence unique for any RNA can be the target ofthe present target RNA- specific ribozyme.
  • the determination of unique sequences is routine given the availability of numerous computer databases (GenBank) and computer programs
  • catalytic sequence ofthe present ribozymes is also shown as its DNA coding sequence in Fig. 1 and SEQ ID NO: 1.
  • Other catalytic sequences include those known in the art. A number of sequence variation have defined permissible nucleotide alteration in "stem” regions (Fedor and Uhlenbeck Proc. Nat. Acad. Sci. 87: 1668-1672, 1990). Those skilled in the art will understand that any catalytic sequence, even those not yet discovered, can be used to construct a ribozyme ofthe invention when it is routinely combined with the autocatalytically cleaving ribozymes and RNA binding site as described herein
  • Fig 1 and SEQ ID NO 1 One example ofthe 5' and 3 ' autocatalytically cleaving ribozymes that are expressed with the catalytic ribozyme ofthe invention are shown in Fig 1 and SEQ ID NO 1, and also in Figs 2 and 3 As further described below, these ribozymes are important for the expression ofthe catalytic ribozyme, because they cleave off of the ribozyme transcript as soon as they are transcribed to produce a catalytic ribozyme having minimal extraneous 5' or 3' sequences
  • the target-specific binding site and the catalytic sequence that comprise the catalytic ribozyme are in the correct configuration to bind and cleave the target RNA
  • the extraneous sequences in the exemplified construct are the result ofthe cloning procedure It is understood that with the selection of an alternative cloning scheme some or all of these extraneous nucleotides can be eliminated
  • the invention also provides nucleic acids which encode the ribozymes ofthe invention. These nucleic acids can be used to express the ribozymes ofthe invention at the selected site
  • the site can be tissue-specific in the case of treating tissue-specific cancers, or it can be target-specific in the case of ribozymes that prevent replication of infectious agents to treat infection (e g hepatitis, herpes, malaria, tuberculosis, etc )
  • the nucleic acids ofthe invention comprise a tissue-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
  • tissue-specific promoter binding site in the ribozyme-producing construct results in tissue-specific expression ofthe ribozyme in tissue(s) that actively transcribe RNA from the selected promoter
  • tissue-specific promoter binding site uses the binding site for the probasin promoter, a promoter-specific for prostate epithelium (Greenburg et al , 1994))
  • This tissue-specific promoter binding site has the sequence shown in (Greenburg et al , 1994)
  • tissue-specific promoters can be used in the present nucleic acid constructs
  • these promoters include the binding sites for prostate-specific antigen (prostate), albumin (liver), fatty acid binding protein (ilium), whey acidic protein (breast), smooth muscle action (smooth muscle), etc
  • target-specific promoters not yet identified can be used to target expression ofthe present ribozymes to the selected tissue(s)
  • Once a target-specific promoter is identified its binding sequence can be routinely determined by routine methods such as sequence analysis
  • the promoter is defined by deletion analysis, mutagenesis, footprinting, gel shifts and transfection analyses (Sambrook et al , Molecular Cloning: A Laboratory Manual, 2nd Ed , Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989)
  • the nucleic acid encoding the 5' autocatalytically cleaving ribozyme can have the sequence of nucleotides 1-54 shown in SEQ ED NO 1
  • the nucleic acid encoding the 3 ' autocatalytically cleaving ribozyme can have the sequence of nucleotides 1 11-164 shown in SEQ ID NO 1
  • the present nucleic acid encodes a catalytic ribozyme that contains two separable functional regions a highly conserved catalytic sequence which cleaves the target RNA (also known as the "catalytic core"), and flanking regions which include a target RNA-specific binding site By nucleic acid complementarity, the binding site directs the ribozyme core to cleave a specific site on the target RNA molecule
  • the length of flanking sequences have implications not only for specificity, but also for the cleavage efficiency ofthe individual ribozyme molecules
  • the flanking sequences are highly specific for the target RNA, yet allow ready dissociation from the target RNA once cleavage occurs This permits cycling ofthe ribozyme (with an expected K at of about 1 cleavage per minute) and reduces the amount of ribozyme required to be effective
  • a range of binding/dissociation values from 16-21 Kcal should be effective
  • RNA-RNA duplex The complexity of human RNA is about 100 fold lower than that for human DNA, and specificity can be achieved with as few as 12-15 base pairs
  • the stability of the RNA-RNA duplex is effected by several factors, such as GC content, temperature, pH, ionic concentration, and structure
  • the nearest neighbor rules can provide a useful estimate ofthe stability ofthe duplex (Castanotto et al "Antisense Catalatic RNAs as Therapeutic Agents" Advances in Pharmacol. 25.289-317, 1994)
  • RNA binding site is unique, so the encoding nucleic acid sequence will be the corresponding unique DNA sequence
  • the RNA binding site can comprise a sequence that binds to an RNA sequence unique to ribosomal RNA polymerase 1(A) subunit
  • the ribosomal RNA polymerase binding site encoding DNA can have the sequence shown in Fig 3 This is a sequence from the RNA polymerase 1(A) subunit
  • the catalytic ribozyme ofthe invention also includes a catalytic sequence, which cleaves the target RNA near the middle ofthe site to which the target RNA-specific binding site binds
  • the catalytic sequence is generally highly conserved
  • the conserved catalytic core residues are 5' CUGANGA 3 ' and 5' GAAA 3' linked by an evolutionarily conserved stem-loop structure
  • the most conserved and probably most efficiently cleaved sequence on the target RNA is 5 ' GUC 3 '
  • Such cleavage sites are ubiquitous in most RNAs allowing essentially all RNA's to be targeted (Whitton, J Lindsay "Antisense Treatment of Viral Infection” Adv. in Virus Res. Vol 44, 1994)
  • target site secondary structure can have an effect on cleavage in vitro (Whitton, 1994)
  • the selected target molecule's sequence can be routinely screened for potential secondary structure, using the program RNAFOLD (from the PCGENE group of programs or available on the Internet)
  • RNAFOLD from the PCGENE group of programs or available on the Internet
  • reasonable predictions of target accessibility can be made Computer assisted RNA folding (Castanotto et al , 1994), along with computational analysis for 3 -dimensional modeling of RNA (Major et al , Science 253 1255-1260, 1991 and Castanotto et al , 1994) is certainly effective in guiding the choice of cleavage sites
  • the internal ribozyme can be targeted to noncellular RNAs necessary for growth of parasites, virus life cycles, etc , and expression can be driven with tissue-specific or virus-specific promoters Three important examples which are specifically presented in the application are
  • nucleic acid of the invention has the nucleotides in the sequence shown in the Sequence Listing as SEQ ID NO 1
  • This exemplary nucleic acid includes a probasin promoter, upstream from a sequence that encodes the 5 ' autocatalytically cleaving ribozyme having the sequence shown in SEQ ID NO 1, the ribosomal RNA binding site encoding DNA having the sequence shown in the Sequence Listing as SEQ ID NO 1 and the 3 ' autocatalytically cleaving ribozyme having the sequence shown in SEQ ID NO 1
  • nucleic acid having substantially the nucleotide sequence shown in SEQ ID NO 1, which encodes the ribozyme shown in SEQ ID NO 1, is provided
  • the nucleic acid can vary based on the characteristics/definition ofthe promoter chosen, and will have 80%-99% sequence identity with SEQ ID NO 1 , more preferably, it will have 90%-99% sequence identity with SEQ ID NO 1
  • Other modifications could include for example, changes (or deletion) of nucleotides inserted for cloning purposes (Fig 2), which include -1 to -8, +69 to +76
  • the box includes extraneous nucleotides that are a function of cloning choices and, thus, can be modified
  • the unpaired bases can be any base, determined only by the cloning scheme chosen If one ofthe bases of a pair is changed, the other must be changed in a complementary fashion
  • RNA binding and core sequences are synthesized as reverse complementary oligonucleotides and are cloned into a vector that will allow production ofthe relevant RNA containing the ribozyme
  • the present ribozymes are prepared by synthesis of an oligonucleotide (5' GGA AGA TCT TTC AAA GAC TGA TGA CTC CGT GAG GAC GAA ACG AGG ATC AGA TCT TCC 3 ') and its reverse complement
  • the Bgl II site used in cloning is underlined Following appropriate restriction digestion, in this particular case Bgl II, the double-stranded DNA oligonucleotide is cloned into the multiple cloning site within the parent vector ( Figure 1)
  • the test can involve transcription ofthe ribozyme using one ofthe two possible bacterial promoters, in this case SP-6 or T-7, (in the presence of trace amounts of radioactivity) followed by evaluating the autocatalytic cleavage ofthe ribozyme by electrophoresis Data from these tests are provided in the Examples
  • RNAs are examined by standard Northern blot analyses to verify specific degradation of target RNA transcripts
  • the triple-ribozyme that has been constructed can be further tested by subcloning it behind one ofthe tissue-specific promoters that will drive expression ofthe vector in a tissue-specific manner in the target Data from these tests are provided in the Examples
  • the nucleic acids ofthe invention can be in a vector for delivering the nucleic acid to the site for expression ofthe ribozyme
  • the vector can be one ofthe commercially available preparations, such as the pGM plasmid (Promega)
  • Vector delivery can be by liposome, using commercially available liposome preparations or newly developed liposomes having the features ofthe present liposomes
  • Other delivery methods can be adopted routinely tested in methods taught herein An example of a delivery method using liposomes is further described in the Examples
  • the modes of administration of the liposome will vary predictably according to the disease being treated and the tissue being targeted For lung (e.g , tuberculosis, cancer) and liver (e g , hepatitis and cancer) which are both sinks for liposomes, intravenous administration is reasonable
  • lung e.g , tuberculosis, cancer
  • liver e g , hepatitis and cancer
  • intravenous administration is reasonable
  • intravenous administration is reasonable
  • catheterization of an artery upstream from the organ is a preferred mode of delivery, because it avoids significant clearance ofthe liposome by the lung and liver
  • lesions at a number of other sites e g , skin cancer, human papilloma virus infection, herpes (oral or genital) and precancerous cervical dysplasia
  • topical delivery is expected to be effective and may be preferred, because of its convenience
  • Leukemias and other conditions such as malaria may also be more readily treated by ex vivo administration ofthe ribozyme
  • the liposomes may be administered topically, parenterally (e.g , intravenously), by intramuscular injection, by intraperitoneal injection, transdermally, excorporeally or the like, although IV or topical administration is typically preferred
  • the exact amount ofthe liposomes required will vary from subject to subject, depending on the species, age, weight and general condition ofthe subject, the severity ofthe disease that is being treated, the particular compound used, its mode of administration, and the like Thus, it is not possible to specify an exact amount However, an appropriate amount may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein Generally, dosage will approximate that which is typical given in the Examples
  • Parenteral administration if used, is generally characterized by injection Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions
  • injection Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions
  • a more recently revised approach for parenteral administration involves use of a slow release or sustained release system, such that a constant level of dosage is maintained See, e.g., U S Patent No 3,710,795, which is inco ⁇ orated by reference herein
  • Topical administration can be by creams, gels, suppositories and the like
  • Ex vivo (exco ⁇ oreal) delivery can be as typically used in other contexts
  • the invention provides a transgenic non-human animal, containing, in a germ or somatic cell, a nucleic acid comprising a target-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence, wherein the animal expresses a ribozyme comprising a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3' autocatalytically cleaving ribozyme sequence
  • the nucleic acid can be the nucleic acid shown in Fig 1 and SEQ ID NO.1 Alternatively, silent base substitutions in the promoter binding site and ribozyme encoding sequence can be made that express the same ribozyme in
  • the transgenic non-human animal ofthe invention is useful, because the animal does not express a phenotype associated with the target RNA (e.g , with the protein it encodes)
  • a phenotype includes mo ⁇ hology, biochemical profiles (e g , changes in amounts of RNA or protein expressed, etc ) and other parameters that are affected by the knockout For example, cell death of otherwise healthy cells can be a measure of altered phenotype resulting from ribozyme expression
  • the present ribozymes can be expressed in a transformed cell line
  • the transformed cell can be used to validate both the specificity ofthe ribozyme's expression and the specificity and cleavage activity against the target RNA An example of such a screening function is described in the Examples
  • the transgenic animals and transformed host cells ofthe invention can be used in a method of screening a compound for its ability to cause the animal or host cell to express a phenotype associated with the target RNA
  • the method requires administering the compound to the animal/cell and assessing the compounds ability to cause expression ofthe phenotype If the phenotype is restored, the compound is considered to be effective
  • an L-dopa functional knockout transgenic animal can be made and used to screen for drugs that restore an L-dopa associated phenotype Treating Proliferative Diseases
  • a method of treating a subject having a proliferative disease of a specific tissue is provided.
  • the treatment is carried out by inhibiting cell proliferation in the specific tissue, and this is accomplished by administering to the subject a nucleic acid encoding a ribozyme that is targeted to an RNA that is essential to cell survival or replication, and containing a target-specific promoter binding sequence that is specific for the diseased tissue.
  • the ribozyme encoded by the nucleic acid is expressed in the diseased tissue, production of an essential RNA in the tissue is inhibited, cell proliferation is inhibited in the tissue, cell death ensues and the proliferative disease treated
  • the proliferative diseases that can be treated by the present method include almost all cancers for which a target-specific promoter exists, including, prostate, breast, colon, pancreatic, lung and liver.
  • the invention provides a method of treating a subject having prostate cancer, comprising administering to the subject the nucleic acid shown in SEQ ID NO: 1, whereby the ribozyme encoded by the nucleic acid is expressed in the prostate and the prostate cancer is treated
  • a method of treating a viral infection in a subject comprising administering to the subject a nucleic acid ofthe invention, wherein the encoded target RNA-specific binding site is specific for an RNA unique to the infectious agent, whereby the ribozyme encoded by the nucleic acid is expressed and the infectious agent is killed Transcription can be driven using a virus specific promoter or a tissue-specific promoter which will selectively express the targeted ribozyme in virus-infected tissue, i.e., using the liver-specific albumin promoter for expression of a targeted ribozyme directed against hepatitis B virus.
  • ribozyme expressing cell lines can be compared with their ribozyme negative counte ⁇ arts for their ability to support viral infection/replication/yield In a manner similar to that described above, ribozyme expressing cell lines can be obtained and assayed, and in all cases the abilities ofthe ribozyme to prevent infection can be determined
  • This example presents the vectors and a novel strategy to utilizing prostate targeted expression of a hammerhead ribozyme to kill normal and neoplastic prostate epithelium
  • the ribozyme is a highly innovative, triple-ribozyme targeted to destroy cells by attacking essential RNA(s)
  • the 5 ' and 3 ' ribozymes have been designed to undergo autocatalytic cleavage during transcription, freeing the internal ribozyme (at high levels) within the cells
  • RNA polymerase 1(A) Intracellular expression of a hammerhead ribozyme, targeted towards an essential cellular RNA (such as RNA polymerase 1(A)), results in the death ofthe cell
  • RNA polymerase 1(A) an essential cellular RNA
  • the ribozyme is targeted to a specific tissue in a constrained manner, then only cells in that tissue will be affected by expression ofthe ribozyme Targeting selectively to prostate can be achieved via the rat probasin promoter (pb) (or the prostate specific antigen promoter)
  • pb rat probasin promoter
  • tissue-specific prostate targeting exists using the probasin promotor, delivery of vectors systemically or by direct introduction into the prostate will result in death of transfected prostate cancer cells with some collateral damage also being observed in the remaining normal prostate epithelium Because ofthe rather unique specificity ofthe probasin promoter, no additional collateral damage is expected to be observed elsewhere in the body This is also expected to be true for the prostate-
  • targets include the 1(A) subunit of RNA polymerase I and II
  • Other intemal targeted ribozymes are tested for in vitro and in vivo activity by the methods described
  • the primary double ribozyme vector depicted in Figure 1 was constructed The two flanking ribozymes (bases 1 to 54 and 66 to 120) are capable of self-cleavage A third ribozyme (Table 2 ) (bases 64 to 105) targeted to pol I mRNA is cloned between the flanking ribozymes This intemal ribozyme has 19 bases within two regions (TTCAAAGA-catalytic core-ACGAGGATCAG) that are anti-sense to the pol I message and interact by base pairing in regions with minimal secondary structures to effect cleavage
  • the intemal pol I ribozyme was prepared by synthesis of an oligonucleotide (5 ' GGA AGA TCT TTC AAA GAC TGA TGA CTC CGT GAG GAC GAA ACG AGG ATC AGA TCT TCC 3 )(only the ribozyme sense strand is shown in its reverse complement)
  • the Bgl II site used in cloning is underlined Following appropriate restriction digestion with Bgl II the double-stranded oligonucleotide was cloned into the
  • the prostate specific promoter when coupled to the triple ribozyme construct, will be delivered to the prostate systemically via liposomes
  • Various routes of introduction mto the blood vascular system are evaluated as described Orthotopic routes can also be utilized
  • the liposome vehicle is expected to be efficient enough to deliver the molecule to prostate cancer cells, because of a high degree of vascularization)
  • a lipofectamine reagent (GIBCO BRL, Gaithersburg, MD) is a polycationic lipid composed of a positively charged lipid, DOSP A, and the neutral lipid, DOPE, in a 3 1 molar ration
  • the cationic lipid, DDAB used in combination with DOPE at 2 1 or 0 6 1 0 ratios (Brunette et al Mol.
  • the test mechanism involves transcription ofthe triple ribozyme using one ofthe two possible bacterial promoters, in this case SP-6 or T-7 present in the pCRII vector (Invitrogen, San Diego, Ca), (in the presence of trace amounts of radioactivity) followed by evaluating the autocatalytic cleavage ofthe ribozyme by electrophoresis This was carried out with the pol I ribozyme and cleavage was observed, i e , first a 113 bp fragment was produced that included the intemal targeting ribozyme and the 3 ' ribozyme, followed by the appearance of a 74 bp fragment containing the intemal pol I ribozyme
  • the ribozyme was subsequently tested by transient transfection it into C3H10T
  • a subsequent step is to introduce the triple-ribozyme into a vector under the control of a tissue-specific promoter such as probasin or prostate specific antigen that will target expression in a tissue-specific manner
  • a tissue-specific promoter such as probasin or prostate specific antigen that will target expression in a tissue-specific manner
  • Transgenic mice are generated by standard pronuclear injection as described in Hogan (Manipulating the mouse embryo: a laboratory manual, Cold Spring Harbor, NY 1986) Prior to injection, constructs are separated from vector DNA by restriction digestion (Hind III and Sac II) ofthe plasmid, followed by sucrose gradient fractionation Isolated constructs are dialyzed against 10mm tris ph 8.0, 0 1mm EDTA before injection In these mice, once probasin expression becomes apparent at the 3-5 week stage of post natal development destruction of prostate epithelium is expected to occur The efficiency of generating transgenic mice is 10-15% with twenty-six mice delivered, i e.
  • tn vivo studies are conducted in transgenic mice bearing prostate tumors Tumors are induced by probasin directed expression of genes such as EcoRI (a restriction enzyme), cfos (a proto-oncogene), or a modified version of lamin (a nuclear mat ⁇ x molecule) Administration ofthe ribozyme is as described above
  • RNA polymerase 1(A) This represents an excellent target, because it is an abundant RNA If there is a leakiness (low levels of transcription) ofthe probasin promoter in other than prostate cells, they are predicted to survive the presence of limited levels ofthe Pol I ribozyme
  • Other potential targets will be examined including phosphofructokinase (ribozyme targets nt 178, 121, or 162 depending on tissue involved), RNA pol II subunit 14 4 kd (ribozyme targets nt 83 or 884), mRNA pol II 140 kd subunit (ribozyme targets nt 204), and RNA pol II 23 kd subunit (ribozyme targets nt 143)
  • Another potential target of interest is the 70 kD subunit of replication protein A It is needed for formation of DNA replication centers/foci, so it should disrupt DNA replication without danger of introducing errors
  • the sequence reference is Kim, et al , Mol. Cell. Biol. 12 3050-3059, 1992
  • the methods described above are applicable to the present context, except where specified For example, The administration mode will be different for parasitic infection than for prostate cancer and will depend upon tissue site For malaria (Plasmodium falciparum) the EMP-1 proteins, which are necessary for cytoadherence and present a problem because they cause rapid antigenic variation, are targeted Specifically, highly conserved GTCs in exon II are targeted Pertinent EMBL accession numbers are L42246, L42244, L42245, L42247, L40600-L40609, L42636 See Smith, J et al Cell 82 101-110, 1995, and Su, X et al Cell 82 89-100, 1995 A promoter active in red blood cells can be used and treatment also could be extraco ⁇ oreal
  • the methods described above are applicable to the present context, except where specified For example, The administration mode will be different for bacterial infection than for prostate cancer, and will depend on the targeted tissue
  • a transcribed fragment which is essential for cell entry will be targeted
  • the EMBL accession number is X70901 See Arruda, S et al Science 261 1454-1457, 1993 The target will be near the N-terminus of the open reading frame coding for the 52 kD protein, from the 1535 bp fragment
  • the administration mode will be different for viral infection than for prostate cancer
  • E6 and E7 proteins are translated from a bicistronic mRNA Antisense oligonucleotides to the translational start site of E6 inhibit synthesis of both E6 and E7
  • the target will be a GTT, cleaving at nt 108 (Tan, et al , J. Gen. Virol. 75.2663-2670, 1994) Original numbering is from Seedort, et al , Virology 145: 181-185, 1985.
  • IV and topical administration should be an effective combination. Topical administration should be effective for dysplastic/precancerous lesions.
  • Hepatitis B virus is a partially single-stranded DNA virus, and it is now thought that integration ofthe viral genome is not the critical incident. Rather, the viral genome is made into an extended RNA intermediate, which is then reverse transcribed into DNA A target with multiple advantage has been chosen. It will cut the viral RNA pregenome in the first place, and its located in both the S (envelope) and polymerase/reverse transcriptase domains. The cut is at a GTC, after nt 438 in the HBV subtype ayw
  • the EMBL accession number is X02496, and the original sequence reference is Galibert, et al , Nature 281, 646-650, 1979. Expression could be driven by the albuming promoter and IV administration should be highly effective
  • NAME Spratt, Gwendolyn D.
  • MOLECULE TYPE oligonucleotide (XI) SEQUENCE DESCRIPTION: SEQ ID N ⁇ :l:

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides tissue-specific and target RNA-specific ribozymes. These ribozymes can be used to destroy target-specific neoplasms and to treat viral infections, among other uses. The ribozymes of the present invention comprise a 5' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3' autocatalytically cleaving ribozyme. The invention also provides nucleic acids which encode the ribozymes of the invention. These nucleic acids can be used to express the ribozymes of the invention at the selected site. Methods of treating disease by administering the ribozymes are provided.

Description

TISSUE-SPECIFIC AND TARGET RNA-SPECIFIC RIBOZYMES
BACKGROUND OF THE INVENTION
Field of the Invention
The invention pertains to the use of tissue-specific and target RNA-specific ribozymes for treatment of cancers and bacterial, parasitic and viral infections More specifically, the invention relates to a ribozyme targeted to the RNA polymerase 1(A) under the control ofthe probasin promoter
Background Art
A ribozyme is a catalytic RNA molecule that cleaves RNA in a sequence specific manner The use of ribozyme as potential gene regulators in mammalian cells and antiviral agents has been suggested, but subject to serious questions regarding technical feasibility For example, it is not known how ribozymes can be introduced to target cells or how they can be directed to the same subcellular compartments as their target RNAs Other questions concern the effects of target RNA secondary structure on ribozymal activity The art has not been successful in answering any of these questions
Furthermore, because ribozymes are a form of antisense technology, the problems encountered in applying antisense technology to disease treatment are also encountered in the use of ribozyme technology For example, it has been shown that the expression of antisense RNA in transgenic mice did not invariably lead to a reduction in target RNA molecules, and when reduction in target RNA molecules did occur, it was not predictably paralleled by a reduction in protein Even when protein levels were reduced sometimes no biological effect was detected (Whitton, J Lindsay "Antisense Treatment of Viral Infection" Adv. in Virus Res. Vol 44, 1994)
The experience in the art suggests that it is also not clear whether ribozymes work best when free, or when embedded in an unrelated large RNA molecule (Whitton, 1994) At present, sufficient data are not available, either in vitro or in cell culture to allow systematic comparison ofthe transactivities of free ribozymes with their embedded equivalents
There have been some studies that focus on the potential use of ribozyme technology in the treatment of cancer In these studies, ribozymes have been directed against both c-fos and c-ras oncogenes in cell culture and showed some suppression of the malignant potential of cells when transplanted into mice Nevertheless, these ribozymes specifically target an oncogene
There has been no suggestion in the literature that tissue-specific cancers or other tissue-specific disease can be treated by delivering to that tissue a ribozyme having a tissue-specific promoter, and that it is targeted to an RNA that is essential for cell survival The invention provides such a ribozyme capable of treating tissue-specific cancers and other tissue-specific diseases
The magnitude ofthe prostate cancer problem requires little introduction Approximately 44,000 men die each year of prostate cancer and about 10,000,000 men have precancerous conditions ofthe prostate It is clear that new approaches to therapy are needed Animal models for testing therapeutic approaches are just becoming available, and will require a number of years for validation However, the present invention provides important reagents to address this problem
One ofthe difficulties in using gene therapy to treat prostate cancer is the long standing problem of target-specific delivery However, the recently developed probasin promoter provides target target-specificity enabling systemic delivery ofthe present ribozyme-encoding vector (Greenburg et al Mol. Endocrinol. 8 230-239, 1994) The present vector consists ofa tandem array of 3 hammerhead ribozymes, the 5' and 3 ' of which are designed to autocatalytically cleave themselves from the primary transcript This novel construct eliminates problems inherent with extensive residual flanking sequences which might otherwise be present to compromise catalytic activity and specificity The present constructs couple the prostate specific probasin promoter to triple-ribozymes targeted at mRNAs critical for prostate cell growth
Endogenous delivery ofa ribozyme under the control of a tissue-specific or other promoter can be complicated by "leakiness", where low levels of transcription occur in extraneous tissues This could present a considerable therapeutic problem, depending upon the cellular target chosen The present ribozyme compensates for this problem by targeting a cellular target which is associated with high levels of product (that is, RNA polymerase I produces large amounts of cellular ribosomal RNA) Thus, in the event promoter leakiness occurs in unintended tissues, it is not likely that cell death would occur This choice, therefore, provides a needed level of safety, and targeting of pol I would be applicable to many selected tissues using other promoters
A common problem in gene therapy is the difficulty in delivering the ribozyme to the correct tissue The present invention avoids this difficulty by targeting the ribozyme to non-cellular RNAs in cells to which ribozyme constructs can be efficiently delivered IV liposome delivery will be effective for treatment of HBV hepatitis IV and/or extracorporeal treatment will effectively delivery construct to erythrocytes for treatment of malarial infection And topical (with or without iv) administration will effectively deliver ribozyme construct to cervical epithelium in dysplastic/precancerous/cancerous HPV 16 cervical lesions This latter example is of extreme importance for treatment of dysplastic/carcinoma in situ lesions diagnosed via abnormal Pap smears A second advantageous facet ofthe non-cellular target ribozymes is that even if promoter leakiness and/or extraneous delivery and/or expression ofthe ribozyme occurs in unintended cells, the ribozymes should not cleave any cellular RNAs
SUMMARY OF THE INVENTION
The invention provides tissue-specific and target RNA-specific ribozymes These ribozymes can be used to destroy target-specific neoplasms and to treat viral infections, among other uses The ribozymes ofthe present invention comprise a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme
The invention also provides nucleic acids which encode the ribozymes ofthe invention These nucleic acids can be used to express the ribozymes ofthe invention at the selected site The nucleic acids ofthe invention comprise a tissue-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
A method of treating a subject having a proliferative disease ofa specific tissue by inhibiting cell proliferation in the tissue, comprising administering to the subject the nucleic acid of claim 5, wherein the target-specific promoter binding sequence is specific for the diseased tissue, whereby the ribozyme encoded by the nucleic acid is expressed, ribosomal RNA production in the tissue is inhibited, cell proliferation is inhibited, and the proliferative disease treated is provided
A method is provided for treating a subject having prostate cancer, comprising administering to the subject the nucleic acid of claim 7, whereby the ribozyme encoded by the nucleic acid is expressed in the prostate and the prostate cancer is treated
A method of treating an infection in a subject, comprising administering to the subject the nucleic acid of claim 1, wherein the encoded target RNA-specific binding site is specific for an RNA unique to the infectious agent, whereby the ribozyme encoded by the nucleic acid is expressed and the infectious agent is killed is also provided BRIEF DESCRIPTION OF THE FIGURES
Fig 1 shows DNA encoding the parental double ribozyme starting at Not I site Underlined sequences are Not I sites (GCGGCCGC) and Bgl II (AGATCT) Sequence of parental double ribozyme starting at Not I site Underlined sequences are Not I sites (GCGGCCGC) and Bgl II (AGATCT)
Fig 2 shows DNA encoding the entire sequence with the internal Pol I targeted triple ribozyme in bold Underlined sequences are Not I (GCGGCCGC) and Bgl II (AGATCT) This is the entire sequence with the internal Pol I targeted triple ribozyme in bold Underlined sequences are Not I (GCGGCCGC) and Bgl II (AGATCT)
Fig 3 shows the two dimensional structure of parent double ribozyme into which the core ribozyme was cloned, as its reverse complement DNA
DETAILED DESCRIPTION OF THE INVENTION
Ribozymes
The invention provides tissue-specific and target RNA-specific ribozymes These ribozymes can be used to destroy tissue-specific neoplasms and to treat viral, bacterial or parasitic infections, among other uses The ribozymes ofthe present invention comprise a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme One example ofthe present ribozyme is shown in by its DNA coding sequence in Fig 2 and in SEQ ID NO 1 The nucleotides numbered 1-164 encode the ribozyme, including the 5' and 3' autocatalytic ribozyme sequences The 5' autocatalytically cleaving ribozyme, catalytic ribozyme, and 3 ' autocatalytically cleaving ribozyme of this exemplary ribozyme are shown separately in SEQ ED No 1 Alternatively, the 5' autocatalytically cleaving ribozyme can be replaced with another stretch of transcribed RNA. In this RNA, the first 20-30 nt (or longer) are followed by a sequence which represents the reverse complement ofthe initial 20-30 nt. This way, the construct would presumably still be capped at the 5 ' end the way pol II transcripts are, but the initial nucleotides should not alter the specificity ofthe nucleotides on the 5' side ofthe targeted middle ribozyme. Based upon the fact that the transcript would be capped, it should be exported efficiently to the cytoplasm. For present triple ribozyme construct having both 5' and 3' autocatalytic ribozymes, it is expected that there will be some diffusion mediated transport to the cytoplasm ofthe internal targeted ribozyme, although this alternative 5 ' end should increase the cytoplasmic proportion.
The invention provides ribozymes that have the unique characteristic of being both target RNA-specific in their catalytic action, and subject to tissue-specific expression. In the example shown in Fig 1 and SEQ ID NO: 1 , the target RNA specificity is conferred by an RNA binding site that specifically binds a sequence that is unique to RNA polymerase 1(A) (ribosomal RNA polymerase). It will be understood that an RNA sequence unique for any RNA can be the target ofthe present target RNA- specific ribozyme. The determination of unique sequences is routine given the availability of numerous computer databases (GenBank) and computer programs
(Genetics Computer Group, PCGENE and BLAST) which can search for and find any matches between nucleic acid sequences. A unique DNA sequence located on one of the databases will have a corresponding unique RNA sequence.
One example ofthe catalytic sequence ofthe present ribozymes is also shown as its DNA coding sequence in Fig. 1 and SEQ ID NO: 1. Other catalytic sequences include those known in the art. A number of sequence variation have defined permissible nucleotide alteration in "stem" regions (Fedor and Uhlenbeck Proc. Nat. Acad. Sci. 87: 1668-1672, 1990). Those skilled in the art will understand that any catalytic sequence, even those not yet discovered, can be used to construct a ribozyme ofthe invention when it is routinely combined with the autocatalytically cleaving ribozymes and RNA binding site as described herein
One example ofthe 5' and 3 ' autocatalytically cleaving ribozymes that are expressed with the catalytic ribozyme ofthe invention are shown in Fig 1 and SEQ ID NO 1, and also in Figs 2 and 3 As further described below, these ribozymes are important for the expression ofthe catalytic ribozyme, because they cleave off of the ribozyme transcript as soon as they are transcribed to produce a catalytic ribozyme having minimal extraneous 5' or 3' sequences Thus, the target-specific binding site and the catalytic sequence that comprise the catalytic ribozyme are in the correct configuration to bind and cleave the target RNA The extraneous sequences in the exemplified construct are the result ofthe cloning procedure It is understood that with the selection of an alternative cloning scheme some or all of these extraneous nucleotides can be eliminated
Ribozyme Encoding Nucleic Acids
The invention also provides nucleic acids which encode the ribozymes ofthe invention These nucleic acids can be used to express the ribozymes ofthe invention at the selected site The site can be tissue-specific in the case of treating tissue-specific cancers, or it can be target-specific in the case of ribozymes that prevent replication of infectious agents to treat infection (e g hepatitis, herpes, malaria, tuberculosis, etc )
The nucleic acids ofthe invention comprise a tissue-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
The tissue-specific promoter binding site in the ribozyme-producing construct results in tissue-specific expression ofthe ribozyme in tissue(s) that actively transcribe RNA from the selected promoter Thus, only the target RNA in tissue that utilizes the promoter will be cleaved by the ribozyme The exemplary ribozyme shown in Fig 1 and SEQ ID NO 1 uses the binding site for the probasin promoter, a promoter-specific for prostate epithelium (Greenburg et al , 1994)) This tissue-specific promoter binding site has the sequence shown in (Greenburg et al , 1994)
As expected, other tissue-specific promoters can be used in the present nucleic acid constructs Examples of these promoters include the binding sites for prostate- specific antigen (prostate), albumin (liver), fatty acid binding protein (ilium), whey acidic protein (breast), smooth muscle action (smooth muscle), etc It will also be clear that target-specific promoters not yet identified can be used to target expression ofthe present ribozymes to the selected tissue(s) Once a target-specific promoter is identified its binding sequence can be routinely determined by routine methods such as sequence analysis The promoter is defined by deletion analysis, mutagenesis, footprinting, gel shifts and transfection analyses (Sambrook et al , Molecular Cloning: A Laboratory Manual, 2nd Ed , Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989)
In the ribozyme-encoding nucleic acid ofthe invention, the nucleic acid encoding the 5' autocatalytically cleaving ribozyme can have the sequence of nucleotides 1-54 shown in SEQ ED NO 1 In the ribozyme-encoding nucleic acid ofthe invention, the nucleic acid encoding the 3 ' autocatalytically cleaving ribozyme can have the sequence of nucleotides 1 11-164 shown in SEQ ID NO 1
It is understood that other 5' and 3' autocatalytically cleaving ribozymes may be developed that can be encoded by the present nucleic acids These ribozymes can be developed according to the methods of Taira et al (Nuc. Acids Res. 19(19) 5125-5130, 1991)
The present nucleic acid encodes a catalytic ribozyme that contains two separable functional regions a highly conserved catalytic sequence which cleaves the target RNA (also known as the "catalytic core"), and flanking regions which include a target RNA-specific binding site By nucleic acid complementarity, the binding site directs the ribozyme core to cleave a specific site on the target RNA molecule The length of flanking sequences have implications not only for specificity, but also for the cleavage efficiency ofthe individual ribozyme molecules In the present catalytic ribozyme, the flanking sequences are highly specific for the target RNA, yet allow ready dissociation from the target RNA once cleavage occurs This permits cycling ofthe ribozyme (with an expected Kat of about 1 cleavage per minute) and reduces the amount of ribozyme required to be effective A range of binding/dissociation values from 16-21 Kcal should be effective
The complexity of human RNA is about 100 fold lower than that for human DNA, and specificity can be achieved with as few as 12-15 base pairs The stability of the RNA-RNA duplex is effected by several factors, such as GC content, temperature, pH, ionic concentration, and structure The nearest neighbor rules can provide a useful estimate ofthe stability ofthe duplex (Castanotto et al "Antisense Catalatic RNAs as Therapeutic Agents" Advances in Pharmacol. 25.289-317, 1994)
As described above, the encoded RNA binding site is unique, so the encoding nucleic acid sequence will be the corresponding unique DNA sequence The RNA binding site can comprise a sequence that binds to an RNA sequence unique to ribosomal RNA polymerase 1(A) subunit The ribosomal RNA polymerase binding site encoding DNA can have the sequence shown in Fig 3 This is a sequence from the RNA polymerase 1(A) subunit
The catalytic ribozyme ofthe invention also includes a catalytic sequence, which cleaves the target RNA near the middle ofthe site to which the target RNA-specific binding site binds In the hammerhead type of ribozyme, the catalytic sequence is generally highly conserved The conserved catalytic core residues are 5' CUGANGA 3 ' and 5' GAAA 3' linked by an evolutionarily conserved stem-loop structure The most conserved and probably most efficiently cleaved sequence on the target RNA is 5 ' GUC 3 ' However, XUN (N = A, U or C) can also be cleaved efficiently Such cleavage sites are ubiquitous in most RNAs allowing essentially all RNA's to be targeted (Whitton, J Lindsay "Antisense Treatment of Viral Infection" Adv. in Virus Res. Vol 44, 1994)
With regard to the selection ofthe appropriate sites on target RNA, it is known that target site secondary structure can have an effect on cleavage in vitro (Whitton, 1994) Thus, the selected target molecule's sequence can be routinely screened for potential secondary structure, using the program RNAFOLD (from the PCGENE group of programs or available on the Internet) Thus, reasonable predictions of target accessibility can be made Computer assisted RNA folding (Castanotto et al , 1994), along with computational analysis for 3 -dimensional modeling of RNA (Major et al , Science 253 1255-1260, 1991 and Castanotto et al , 1994) is certainly effective in guiding the choice of cleavage sites
The internal ribozyme can be targeted to noncellular RNAs necessary for growth of parasites, virus life cycles, etc , and expression can be driven with tissue-specific or virus-specific promoters Three important examples which are specifically presented in the application are
A) Use ofthe albumin promoter with a Hepatitis B virus target (chosen to cleave the viral RNA pregenome, S protein, and polymerase/reverse transcriptase transcripts using the same ribozyme target site),
B) Use of generic promoters active in erythrocytes, using a ribozyme targeted to highly conserved regions ofthe EMP-1 protein family from P. falciparum, which are necessary for cytoadherence and antigenic variation in malaria, and
C) Use ofthe HPV promoter, with a ribozyme targeted to a specific site near the translational start site ofthe E6 protein, a site known to be critical for expression of both the E6 and E7 proteins which are intimately involved in cervical carcinogenesis One example ofthe nucleic acid ofthe invention has the nucleotides in the sequence shown in the Sequence Listing as SEQ ID NO 1 This exemplary nucleic acid includes a probasin promoter, upstream from a sequence that encodes the 5 ' autocatalytically cleaving ribozyme having the sequence shown in SEQ ID NO 1, the ribosomal RNA binding site encoding DNA having the sequence shown in the Sequence Listing as SEQ ID NO 1 and the 3 ' autocatalytically cleaving ribozyme having the sequence shown in SEQ ID NO 1
Alternatively, silent base substitutions in the promoter binding site and ribozyme encoding sequence can be made that express the same ribozyme in the same tissue Thus, a nucleic acid having substantially the nucleotide sequence shown in SEQ ID NO 1, which encodes the ribozyme shown in SEQ ID NO 1, is provided The nucleic acid can vary based on the characteristics/definition ofthe promoter chosen, and will have 80%-99% sequence identity with SEQ ID NO 1 , more preferably, it will have 90%-99% sequence identity with SEQ ID NO 1 Other modifications could include for example, changes (or deletion) of nucleotides inserted for cloning purposes (Fig 2), which include -1 to -8, +69 to +76 In Figure 3, the box includes extraneous nucleotides that are a function of cloning choices and, thus, can be modified The unpaired bases can be any base, determined only by the cloning scheme chosen If one ofthe bases of a pair is changed, the other must be changed in a complementary fashion Furthermore, the ribozyme-coding sequence can be altered in ways that modify the ribozyme sequence, but do not effect the ribozyme's target RNA-specificity or negate its cleavage activity For example, changes in the stem loop regions of the 5 ', 3 ', and internal ribozyme (Fig 2) could be incoφorated into other constructs while maintaining catalytic activity (Fedor and Uhlenbeck, 1990)
Synthesis of the Ribozyme Producing Construct
Typically, the RNA binding and core sequences are synthesized as reverse complementary oligonucleotides and are cloned into a vector that will allow production ofthe relevant RNA containing the ribozyme The present ribozymes are prepared by synthesis of an oligonucleotide (5' GGA AGA TCT TTC AAA GAC TGA TGA CTC CGT GAG GAC GAA ACG AGG ATC AGA TCT TCC 3 ') and its reverse complement The Bgl II site used in cloning is underlined Following appropriate restriction digestion, in this particular case Bgl II, the double-stranded DNA oligonucleotide is cloned into the multiple cloning site within the parent vector (Figure 1)
Functional Testing
Once sequenced, these ribozymes are functionally tested The test can involve transcription ofthe ribozyme using one ofthe two possible bacterial promoters, in this case SP-6 or T-7, (in the presence of trace amounts of radioactivity) followed by evaluating the autocatalytic cleavage ofthe ribozyme by electrophoresis Data from these tests are provided in the Examples
Additional testing procedures encompass incubation of in vitro transcribed ribozymes with in vitro synthesized target RNA transcript or with cytoplasmic RNA preparations Following incubations, RNAs are examined by standard Northern blot analyses to verify specific degradation of target RNA transcripts
The triple-ribozyme that has been constructed can be further tested by subcloning it behind one ofthe tissue-specific promoters that will drive expression ofthe vector in a tissue-specific manner in the target Data from these tests are provided in the Examples
Finally, the triple-ribozyme experimental approach is further validated by doing in vivo studies in mice Two such studies have been performed as described in the Examples The first case used a control vector more easily monitored than the pol I ribozyme consisting ofthe probasin promoter driving expression of algal green fluorescent protein (pBGFT) This test vector is used to ascertain the effectiveness of our in vivo delivery system A second experiment has also been carried out in which the pol I ribozyme is introduced In both cases where either the triple ribozyme or the green fluorescent protein was introduced, the animals were euthanized at various time post operatively, autopsied, and various tissue were examined for activity ofthe vector by immunohistochemistry
Delivery
The nucleic acids ofthe invention can be in a vector for delivering the nucleic acid to the site for expression ofthe ribozyme The vector can be one ofthe commercially available preparations, such as the pGM plasmid (Promega) Vector delivery can be by liposome, using commercially available liposome preparations or newly developed liposomes having the features ofthe present liposomes Other delivery methods can be adopted routinely tested in methods taught herein An example of a delivery method using liposomes is further described in the Examples
The modes of administration of the liposome will vary predictably according to the disease being treated and the tissue being targeted For lung (e.g , tuberculosis, cancer) and liver (e g , hepatitis and cancer) which are both sinks for liposomes, intravenous administration is reasonable For many other localized pathologic conditions including cancers, infections (e g , hepatitis, cystitis, proctitis, cervicitis, etc ) as well as precancerous conditions, catheterization of an artery upstream from the organ is a preferred mode of delivery, because it avoids significant clearance ofthe liposome by the lung and liver For lesions at a number of other sites (e g , skin cancer, human papilloma virus infection, herpes (oral or genital) and precancerous cervical dysplasia), topical delivery is expected to be effective and may be preferred, because of its convenience
Leukemias and other conditions such as malaria, may also be more readily treated by ex vivo administration ofthe ribozyme The liposomes may be administered topically, parenterally (e.g , intravenously), by intramuscular injection, by intraperitoneal injection, transdermally, excorporeally or the like, although IV or topical administration is typically preferred The exact amount ofthe liposomes required will vary from subject to subject, depending on the species, age, weight and general condition ofthe subject, the severity ofthe disease that is being treated, the particular compound used, its mode of administration, and the like Thus, it is not possible to specify an exact amount However, an appropriate amount may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein Generally, dosage will approximate that which is typical given in the Examples
Parenteral administration, if used, is generally characterized by injection Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions A more recently revised approach for parenteral administration involves use of a slow release or sustained release system, such that a constant level of dosage is maintained See, e.g., U S Patent No 3,710,795, which is incoφorated by reference herein
Topical administration can be by creams, gels, suppositories and the like Ex vivo (excoφoreal) delivery can be as typically used in other contexts
Transgenic Animals
The invention provides a transgenic non-human animal, containing, in a germ or somatic cell, a nucleic acid comprising a target-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence, wherein the animal expresses a ribozyme comprising a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3' autocatalytically cleaving ribozyme sequence The nucleic acid can be the nucleic acid shown in Fig 1 and SEQ ID NO.1 Alternatively, silent base substitutions in the promoter binding site and ribozyme encoding sequence can be made that express the same ribozyme in the same tissue For example, these substitutions can be as described above
The transgenic non-human animal ofthe invention is useful, because the animal does not express a phenotype associated with the target RNA (e.g , with the protein it encodes) As used herein the term "phenotype" includes moφhology, biochemical profiles (e g , changes in amounts of RNA or protein expressed, etc ) and other parameters that are affected by the knockout For example, cell death of otherwise healthy cells can be a measure of altered phenotype resulting from ribozyme expression
Transformed Host Cells
The present ribozymes can be expressed in a transformed cell line The transformed cell can be used to validate both the specificity ofthe ribozyme's expression and the specificity and cleavage activity against the target RNA An example of such a screening function is described in the Examples
Screening Methods The transgenic animals and transformed host cells ofthe invention can be used in a method of screening a compound for its ability to cause the animal or host cell to express a phenotype associated with the target RNA The method requires administering the compound to the animal/cell and assessing the compounds ability to cause expression ofthe phenotype If the phenotype is restored, the compound is considered to be effective For example an L-dopa functional knockout transgenic animal can be made and used to screen for drugs that restore an L-dopa associated phenotype Treating Proliferative Diseases
A method of treating a subject having a proliferative disease ofa specific tissue is provided. The treatment is carried out by inhibiting cell proliferation in the specific tissue, and this is accomplished by administering to the subject a nucleic acid encoding a ribozyme that is targeted to an RNA that is essential to cell survival or replication, and containing a target-specific promoter binding sequence that is specific for the diseased tissue. The ribozyme encoded by the nucleic acid is expressed in the diseased tissue, production of an essential RNA in the tissue is inhibited, cell proliferation is inhibited in the tissue, cell death ensues and the proliferative disease treated
The proliferative diseases that can be treated by the present method include almost all cancers for which a target-specific promoter exists, including, prostate, breast, colon, pancreatic, lung and liver.
For example, the invention provides a method of treating a subject having prostate cancer, comprising administering to the subject the nucleic acid shown in SEQ ID NO: 1, whereby the ribozyme encoded by the nucleic acid is expressed in the prostate and the prostate cancer is treated
Treating Viral Infection
A method of treating a viral infection in a subject, comprising administering to the subject a nucleic acid ofthe invention, wherein the encoded target RNA-specific binding site is specific for an RNA unique to the infectious agent, whereby the ribozyme encoded by the nucleic acid is expressed and the infectious agent is killed Transcription can be driven using a virus specific promoter or a tissue-specific promoter which will selectively express the targeted ribozyme in virus-infected tissue, i.e., using the liver-specific albumin promoter for expression of a targeted ribozyme directed against hepatitis B virus. In the context of determining anti-viral efficacy, ribozyme expressing cell lines can be compared with their ribozyme negative counteφarts for their ability to support viral infection/replication/yield In a manner similar to that described above, ribozyme expressing cell lines can be obtained and assayed, and in all cases the abilities ofthe ribozyme to prevent infection can be determined
The present invention will be illustrated in further detail in the following non¬ limiting examples
EXAMPLES
ANALYSISOFRIBOZYMEGENETHERAPYINPROSTATECANCER
This example presents the vectors and a novel strategy to utilizing prostate targeted expression of a hammerhead ribozyme to kill normal and neoplastic prostate epithelium The ribozyme is a highly innovative, triple-ribozyme targeted to destroy cells by attacking essential RNA(s) The 5 ' and 3 ' ribozymes have been designed to undergo autocatalytic cleavage during transcription, freeing the internal ribozyme (at high levels) within the cells
Intracellular expression ofa hammerhead ribozyme, targeted towards an essential cellular RNA (such as RNA polymerase 1(A)), results in the death ofthe cell If the ribozyme is targeted to a specific tissue in a constrained manner, then only cells in that tissue will be affected by expression ofthe ribozyme Targeting selectively to prostate can be achieved via the rat probasin promoter (pb) (or the prostate specific antigen promoter) Because tissue-specific prostate targeting exists using the probasin promotor, delivery of vectors systemically or by direct introduction into the prostate will result in death of transfected prostate cancer cells with some collateral damage also being observed in the remaining normal prostate epithelium Because ofthe rather unique specificity ofthe probasin promoter, no additional collateral damage is expected to be observed elsewhere in the body This is also expected to be true for the prostate- specific antigen
Examples of targets include the 1(A) subunit of RNA polymerase I and II Other intemal targeted ribozymes are tested for in vitro and in vivo activity by the methods described
Synthesis
The primary double ribozyme vector depicted in Figure 1 was constructed The two flanking ribozymes (bases 1 to 54 and 66 to 120) are capable of self-cleavage A third ribozyme (Figure 2 ) (bases 64 to 105) targeted to pol I mRNA is cloned between the flanking ribozymes This intemal ribozyme has 19 bases within two regions (TTCAAAGA-catalytic core-ACGAGGATCAG) that are anti-sense to the pol I message and interact by base pairing in regions with minimal secondary structures to effect cleavage
The intemal pol I ribozyme was prepared by synthesis of an oligonucleotide (5' GGA AGA TCT TTC AAA GAC TGA TGA CTC CGT GAG GAC GAA ACG AGG ATC AGA TCT TCC 3 )(only the ribozyme sense strand is shown in its reverse complement) The Bgl II site used in cloning is underlined Following appropriate restriction digestion with Bgl II the double-stranded oligonucleotide was cloned into the
Bgl II site within the parent vector (Figure 1 )
Delivery
The prostate specific promoter, when coupled to the triple ribozyme construct, will be delivered to the prostate systemically via liposomes Various routes of introduction mto the blood vascular system (some bypassing the lung and liver) are evaluated as described Orthotopic routes can also be utilized The liposome vehicle is expected to be efficient enough to deliver the molecule to prostate cancer cells, because of a high degree of vascularization) Thus, curing or at least reducing tumor burden by this gene therapy approach is reasonably expected The following liposome preparations were used in these studies (a) lipofectamine reagent (GIBCO BRL, Gaithersburg, MD) is a polycationic lipid composed of a positively charged lipid, DOSP A, and the neutral lipid, DOPE, in a 3 1 molar ration, (b) the cationic lipid, DDAB, used in combination with DOPE at 2 1 or 0 6 1 0 ratios (Brunette et al Mol. Cell. Biol. 14 2411-2418, 1994), and (c) DMRIE in combination with DOPE in a 1 1 molar ratio (Feigner et al Methods (Orlando) 5 67-75, 1995), obtained from VICAL Coφ (San Diego, CA) Liposome reagents were stored at 4°C prior to transfection
Testing
Once sequenced, these ribozymes are functionally tested The test mechanism involves transcription ofthe triple ribozyme using one ofthe two possible bacterial promoters, in this case SP-6 or T-7 present in the pCRII vector (Invitrogen, San Diego, Ca), (in the presence of trace amounts of radioactivity) followed by evaluating the autocatalytic cleavage ofthe ribozyme by electrophoresis This was carried out with the pol I ribozyme and cleavage was observed, i e , first a 113 bp fragment was produced that included the intemal targeting ribozyme and the 3 ' ribozyme, followed by the appearance of a 74 bp fragment containing the intemal pol I ribozyme
The ribozyme was subsequently tested by transient transfection it into C3H10T
1/2 mouse fibroblast cells This demonstrated that pol 1 RNA was degraded when triple ribozyme expression was induced Thus, two phases ofthe activity ofthe molecule have been examined and demonstrated to occur Autocatalytic cleavage ofthe cis ribozymes and functional degradation of pol I mRNA in trans occur as expected
A subsequent step is to introduce the triple-ribozyme into a vector under the control ofa tissue-specific promoter such as probasin or prostate specific antigen that will target expression in a tissue-specific manner This was done by taking the Not 1 fragment containing the entire 3', 5' and intemal ribozymes (Fig 2) and subcloning it into a vector containing the probasin promoter region (-426 to +28 (Greenburg et al , 1994)) This promoter has been demonstrated to target gene expression to the prostate
Vectors are tested in vivo in transgenic mice expressing the triple ribozyme anti- pol I construct Transgenic mice are generated by standard pronuclear injection as described in Hogan (Manipulating the mouse embryo: a laboratory manual, Cold Spring Harbor, NY 1986) Prior to injection, constructs are separated from vector DNA by restriction digestion (Hind III and Sac II) ofthe plasmid, followed by sucrose gradient fractionation Isolated constructs are dialyzed against 10mm tris ph 8.0, 0 1mm EDTA before injection In these mice, once probasin expression becomes apparent at the 3-5 week stage of post natal development destruction of prostate epithelium is expected to occur The efficiency of generating transgenic mice is 10-15% with twenty-six mice delivered, i e. 2-3 transgenic mice are predicted Another way to verify the present ribozymes' functionality is to introduce the vector into tissue culture, e.g , human PC3 prostate cancer cells, and observe cell death in response to activation ofthe ribozyme C3H10T 1/2 mouse fibroblasts have been confirmed as sensitive to the pol I triple ribozyme
Further validation of this gene therapy approach is obtained through in vivo studies Two such studies have been performed In the first case, a control vector, more easily monitored than the pol I ribozyme, which consists ofthe probasin promoter driving expression ofthe algal green fluorescent protein was used to ascertain the effectiveness ofthe in vivo liposomal delivery system The procedure involves mice that are anesthetized and prepared by a surgical procedure to expose the descending aorta A 30 gauge catheter is placed in the descending aorta followed by introduction of 3 OOμg vector/kilogram body weight (vector liposome ratio of 10μg/40μl) The results verify that expression ofthe green fluorescent protein occurs in dorsolateral prostate epithelium but not in the lung (a normal target of liposomes) Further studies determine if expression is observed in liver and other tissue including the kidney, adrenal gland and the brain. A second experiment has also been carried out in which the pol I ribozyme was introduced in vivo using the same method described above In both cases where either the triple ribozyme or the green fluorescent protein was introduced, animals were euthanized at various time post operatively, autopsied, and tissues were examined for activity ofthe vector by immunohistochemistry There was evidence of some apoptotic cell death in prostate epithelium at seven days following administration
tn vivo studies are conducted in transgenic mice bearing prostate tumors Tumors are induced by probasin directed expression of genes such as EcoRI (a restriction enzyme), cfos (a proto-oncogene), or a modified version of lamin (a nuclear matπx molecule) Administration ofthe ribozyme is as described above
Target Choice
Various molecules have been chosen for targeting The first is RNA polymerase 1(A) This represents an excellent target, because it is an abundant RNA If there is a leakiness (low levels of transcription) ofthe probasin promoter in other than prostate cells, they are predicted to survive the presence of limited levels ofthe Pol I ribozyme Other potential targets will be examined including phosphofructokinase (ribozyme targets nt 178, 121, or 162 depending on tissue involved), RNA pol II subunit 14 4 kd (ribozyme targets nt 83 or 884), mRNA pol II 140 kd subunit (ribozyme targets nt 204), and RNA pol II 23 kd subunit (ribozyme targets nt 143)
Another potential target of interest is the 70 kD subunit of replication protein A It is needed for formation of DNA replication centers/foci, so it should disrupt DNA replication without danger of introducing errors The sequence reference is Kim, et al , Mol. Cell. Biol. 12 3050-3059, 1992
RIBOZYME GENE THERAPY IN PARASITIC INFECTION
The methods described above are applicable to the present context, except where specified For example, The administration mode will be different for parasitic infection than for prostate cancer and will depend upon tissue site For malaria (Plasmodium falciparum) the EMP-1 proteins, which are necessary for cytoadherence and present a problem because they cause rapid antigenic variation, are targeted Specifically, highly conserved GTCs in exon II are targeted Pertinent EMBL accession numbers are L42246, L42244, L42245, L42247, L40600-L40609, L42636 See Smith, J et al Cell 82 101-110, 1995, and Su, X et al Cell 82 89-100, 1995 A promoter active in red blood cells can be used and treatment also could be extracoφoreal
RIBOZYME GENE THERAPY IN BACTERIAL INFECTION
The methods described above are applicable to the present context, except where specified For example, The administration mode will be different for bacterial infection than for prostate cancer, and will depend on the targeted tissue
For Mycobacterium tuberculosis, a transcribed fragment which is essential for cell entry will be targeted The EMBL accession number is X70901 See Arruda, S et al Science 261 1454-1457, 1993 The target will be near the N-terminus of the open reading frame coding for the 52 kD protein, from the 1535 bp fragment
RIBOZYME GENE THERAPY IN VTRAL INFECTION
The methods described above are applicable to the present context, except where specified For example, the administration mode will be different for viral infection than for prostate cancer
Human papillomavirus type 16 E6 and E7 proteins are translated from a bicistronic mRNA Antisense oligonucleotides to the translational start site of E6 inhibit synthesis of both E6 and E7 The target will be a GTT, cleaving at nt 108 (Tan, et al , J. Gen. Virol. 75.2663-2670, 1994) Original numbering is from Seedort, et al , Virology 145: 181-185, 1985. IV and topical administration should be an effective combination. Topical administration should be effective for dysplastic/precancerous lesions.
Hepatitis B virus is a partially single-stranded DNA virus, and it is now thought that integration ofthe viral genome is not the critical incident. Rather, the viral genome is made into an extended RNA intermediate, which is then reverse transcribed into DNA A target with multiple advantage has been chosen. It will cut the viral RNA pregenome in the first place, and its located in both the S (envelope) and polymerase/reverse transcriptase domains. The cut is at a GTC, after nt 438 in the HBV subtype ayw The EMBL accession number is X02496, and the original sequence reference is Galibert, et al , Nature 281, 646-650, 1979. Expression could be driven by the albuming promoter and IV administration should be highly effective
While the foregoing invention has been described in some detail for puφoses of clarity and understanding, it will be appreciated by one skilled in the art from a reading of this disclosure that various changes in form and detail can be made without departing from the true scope ofthe invention and appended claims
Throughout this application various publications are referenced within parentheses. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(1) APPLICANT: MEDICAL UNIVERSITY OF SOUTH CAROLINA
171 Ashley Avenue Charleston, South Carolina 29464
(ll) TITLE OF INVENTION: TISSUE SPECIFIC AND TARGET RNA SPECIFIC RIBOZYMES
(111) NUMBER OF SEQUENCES: 1
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: NEEDLE & ROSENBERG, P.C.
(B) STREET: 127 Peachtree Street, Suite 1200
(C) CITY: Atlanta
(D) STATE: Georgia
(E) COUNTRY: USA
(F) ZIP: 30303
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy d sk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.30
( i) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: U.S. Serial No. 08/554,369
(B) FILING DATE: 8 NOV 1995
(C) CLASSIFICATION:
( m) ATTORNEY/AGENT INFORMATION:
(A) NAME: Spratt, Gwendolyn D.
(B) REGISTRATION NUMBER: 36,016
(C) REFERENCE/DOCKET NUMBER: 19070.0030/P
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 404/688-0770
(B) TELEFAX: 404/688-9880
(2) INFORMATION FOR SEQ ID Nθ:l:
(1) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 184 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ll) MOLECULE TYPE: oligonucleotide (XI) SEQUENCE DESCRIPTION: SEQ ID Nθ:l:
GCGGCCGCTC GAGCTCTGAT GAGTCCGTGA GGACGAAACG GTACCCGGTA CCGTCAGCTC 60
GAGCTCAGAT CTTTCAAAGA CTGATGACTC GCTGAGGACG AAACGAGGAT CAGATCTGGA 120
TCCGTCGACG GATCTAGATC CGTCCTGATG AGTCGTGAGG ACGAAACGGA TCTGCAGCGG 180
CCGC 184

Claims

What is claimed is
1 A nucleic acid comprising a target-specific promoter binding site upstream from a sequence encoding a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
2 The nucleic acid of claim 1 , wherein the target-specific promoter binding site is the binding site for the probasin promoter
3 The nucleic acid of claim 1, wherein the nucleic acid encoding the 5' autocatalytically cleaving ribozyme has the sequence of nucleotides 9-62 set forth in the Sequence Listing as SEQ ID NO 1
4 The nucleic acid of claim 1, wherein the nucleic acid encoding the 3 ' autocatalytically cleaving ribozyme has the sequence of nucleotides 119-172 set forth in the Sequence Listing as SEQ ID NO 1
5 The nucleic acid of claim 1, wherein the nucleic acid encodes a catalytic ribozyme comprising, a target RNA-specific binding site that binds to an RNA sequence unique to ribosomal RNA (polymerase 1(A)
6 The nucleic acid of claim 5, wherein the nucleic acid has the sequence of nucleotides 72-113 set forth in the Sequence Listing as SEQ ID NO 1
7 The nucleic acid of claim 1, consisting essentially ofthe nucleotides in the sequence set forth in the Sequence Listing as SEQ ID NO 1
8 The nucleic acid of claim 1, having the sequence set forth in the Sequence Listing as SEQ ID NO 1
9. The nucleic acid of claim 1 in a vector
10 The nucleic acid of claim 2 in a vector.
11. The nucleic acid of claim 3 in a vector.
12 The nucleic acid of claim 4 in a vector
13. The nucleic acid of claim 5 in a vector.
14. The nucleic acid of claim 6 in a vector
15 The nucleic acid of claim 7 in a vector
16 The nucleic acid of claim 8 in a vector
17. A transgenic non-human animal, containing, in a germ or somatic cell, the nucleic acid of claim 1, wherein the animal expresses a ribozyme comprising a 5' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence.
18. A transgenic non-human animal, containing, in a germ or somatic cell, the nucleic acid of claim 7, wherein the animal expresses a ribozyme comprising a 5' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
19. A transgenic non-human animal, containing, in a germ or somatic cell, the nucleic acid of claim 8, wherein the animal expresses a ribozyme comprising a 5 ' autocatalytically cleaving ribozyme sequence, a catalytic ribozyme comprising a target RNA-specific binding site and a 3 ' autocatalytically cleaving ribozyme sequence
20. The transgenic non-human animal of claim 17, wherein the animal does not express a phenotype associated with the target RNA.
21. A method of screening a compound for its ability to cause the animal of claim 20 to express a phenotype associated with the target RNA, comprising administering the compound to the animal and assessing its ability to cause expression of the phenotype.
22. A method of treating a subject having a proliferative disease ofa specific tissue by inhibiting cell proliferation in the tissue, comprising administering to the subject the nucleic acid of claim 5, wherein the target-specific promoter binding sequence is specific for the diseased tissue, whereby the ribozyme encoded by the nucleic acid is expressed, ribosomal RNA production in the tissue is inhibited, cell proliferation is inhibited, and the proliferative disease treated.
23. A method of treating a subject having prostate cancer, comprising administering to the subject the nucleic acid of claim 7, whereby the ribozyme encoded by the nucleic acid is expressed in the prostate and the prostate cancer is treated.
24. A method of treating an infection in a subject, comprising administering to the subject the nucleic acid of claim 1, wherein the encoded target RNA-specific binding site is specific for an RNA unique to the infectious agent, whereby the ribozyme encoded by the nucleic acid is expressed and the infectious agent is killed.
PCT/US1996/018088 1995-11-08 1996-11-08 Tissue-specific and target rna-specific ribozymes WO1997017433A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP96940377A EP0866852B1 (en) 1995-11-08 1996-11-08 Tissue-specific and target rna-specific ribozymes
JP51838197A JP4033902B2 (en) 1995-11-08 1996-11-08 Tissue specific and target RNA specific ribozymes
AU77272/96A AU728732B2 (en) 1995-11-08 1996-11-08 Tissue-specific and target RNA-specific ribozymes
AT96940377T ATE294854T1 (en) 1995-11-08 1996-11-08 TISSUE-SPECIFIC AND TARGET RNA-SPECIFIC RIBOZYMES
IL12438396A IL124383A0 (en) 1995-11-08 1996-11-08 Tissue-specific and target rna-specific ribozymes
DE69634698T DE69634698T2 (en) 1995-11-08 1996-11-08 TISSUE SPECIFIC AND TARGET RNA SPECIFIC RIBOZYMES
CA002236998A CA2236998C (en) 1995-11-08 1996-11-08 Tissue-specific and target rna-specific ribozymes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/554,369 1995-11-08
US08/554,369 US5824519A (en) 1995-11-08 1995-11-08 Tissue-specific and target RNA-specific ribozymes

Publications (1)

Publication Number Publication Date
WO1997017433A1 true WO1997017433A1 (en) 1997-05-15

Family

ID=24213065

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/018088 WO1997017433A1 (en) 1995-11-08 1996-11-08 Tissue-specific and target rna-specific ribozymes

Country Status (9)

Country Link
US (1) US5824519A (en)
EP (2) EP0866852B1 (en)
JP (2) JP4033902B2 (en)
CN (1) CN1207769A (en)
AT (1) ATE294854T1 (en)
AU (1) AU728732B2 (en)
DE (1) DE69634698T2 (en)
IL (1) IL124383A0 (en)
WO (1) WO1997017433A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998024925A1 (en) * 1995-11-08 1998-06-11 Medical University Of South Carolina Tissue specific and target rna-specific ribozymes as antimicrobial therapeutics against microbial pathogens
WO1999036518A1 (en) * 1998-01-15 1999-07-22 Btg International Limited Ribozymal nucleic acids cleaving ccr5 or cxcr4
WO2000043781A2 (en) * 1999-01-21 2000-07-27 Metamorphix, Inc. Growth differentiation factor inhibitors and uses therefor
EP1088086A1 (en) * 1998-06-24 2001-04-04 MUSC Foundation For Research Development Tissue-specific and target rna-specific ribozymes
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
AU2004201285B8 (en) * 1998-06-24 2004-04-29 Musc Foundation For Research Development Tissue-specific and target RNA-specific ribozymes
AU778737B2 (en) * 1999-04-14 2004-12-16 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
EP1611231A2 (en) * 2003-02-21 2006-01-04 The Penn State Research Foundation Rna interference compositions and methods
EP1702983A2 (en) 2000-04-13 2006-09-20 Medical University of South Carolina Tissue-specific and pathogen-specific toxic agents, ribozymes, DNAzymes and antisense oligonucleotides and methods of use thereof
WO2007052046A1 (en) * 2005-11-04 2007-05-10 King's College London Anti-hepatitis b virus ribozymal nucleic acid

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030125280A1 (en) * 1996-12-03 2003-07-03 Medical University Of South Carolina Tissue specific and target RNA-specific ribozymes as antimicrobial therapeutics against microbial pathogens
US6242589B1 (en) * 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
DE19956568A1 (en) * 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
US7829693B2 (en) * 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
US20040209263A1 (en) * 2000-12-07 2004-10-21 Clawson Gary A. Selection of catalytic nucleic acids targeted to infectious agents
US8546143B2 (en) 2001-01-09 2013-10-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
JP2005537028A (en) * 2002-06-26 2005-12-08 ザ ペン ステート リサーチ ファウンデーション Methods and materials for treating human papillomavirus infections
US7892563B2 (en) * 2003-05-20 2011-02-22 Wyeth Holdings Corporation Methods for treatment of severe acute respiratory syndrome (SARS)
US7267960B2 (en) 2003-07-25 2007-09-11 Amgen Inc. Antagonists and agonists of LDCAM and methods of use
US8129334B2 (en) 2006-03-31 2012-03-06 The Regents Of The University Of California Methods and compositions for treating neurodegenerative disorders and Alzheimer'S disease and improving normal memory
EP2671891A3 (en) 2008-06-27 2014-03-05 Amgen Inc. Ang-2 inhibition to treat multiple sclerosis
US9186370B2 (en) 2010-03-19 2015-11-17 University Of South Alabama Methods and compositions for the treatment of cancer
WO2012135696A2 (en) 2011-04-01 2012-10-04 University Of South Alabama Methods and compositions for the diagnosis, classification, and treatment of cancer
WO2013103401A1 (en) 2012-01-06 2013-07-11 University Of South Alabama Methods and compositions for the treatment of cancer

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992010590A1 (en) * 1990-12-10 1992-06-25 Gilead Sciences, Inc. Inhibition of transcription by formation of triple helixes
JPH05192151A (en) * 1990-11-02 1993-08-03 Agency Of Ind Science & Technol New rna transcription system using ribozyme
WO1994003594A1 (en) * 1992-08-07 1994-02-17 University Of Manitoba Androgen regulation with dna sequences of rat probasin gene
EP0640688A1 (en) * 1987-12-15 1995-03-01 Gene Shears Pty. Limited Ribozymes
US5436330A (en) * 1989-12-19 1995-07-25 Agency Of Industrial Science & Technology Hammerhead ribozymes with enhanced stability provided by an additional 3' ha
US5500357A (en) * 1990-11-02 1996-03-19 Agency Of Industrial Science & Technology, Ministry Of International Trade & Industry RNA transcription system using novel ribozyme

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3933384A1 (en) * 1989-10-06 1991-04-18 Hoechst Ag MULTIFUNCTIONAL RNA WITH SELF-PROCESSING ACTIVITY, THEIR PRODUCTION AND THEIR USE

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0640688A1 (en) * 1987-12-15 1995-03-01 Gene Shears Pty. Limited Ribozymes
US5436330A (en) * 1989-12-19 1995-07-25 Agency Of Industrial Science & Technology Hammerhead ribozymes with enhanced stability provided by an additional 3' ha
JPH05192151A (en) * 1990-11-02 1993-08-03 Agency Of Ind Science & Technol New rna transcription system using ribozyme
US5500357A (en) * 1990-11-02 1996-03-19 Agency Of Industrial Science & Technology, Ministry Of International Trade & Industry RNA transcription system using novel ribozyme
WO1992010590A1 (en) * 1990-12-10 1992-06-25 Gilead Sciences, Inc. Inhibition of transcription by formation of triple helixes
WO1994003594A1 (en) * 1992-08-07 1994-02-17 University Of Manitoba Androgen regulation with dna sequences of rat probasin gene

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch Week 9335, Derwent World Patents Index; Class B04, AN 93-277467, XP002026580 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998024925A1 (en) * 1995-11-08 1998-06-11 Medical University Of South Carolina Tissue specific and target rna-specific ribozymes as antimicrobial therapeutics against microbial pathogens
WO1999036518A1 (en) * 1998-01-15 1999-07-22 Btg International Limited Ribozymal nucleic acids cleaving ccr5 or cxcr4
US6916653B2 (en) 1998-01-15 2005-07-12 King's College London Ribozymal nucleic acid
AU2004201285B2 (en) * 1998-06-24 2007-07-05 Musc Foundation For Research Development Tissue-specific and target RNA-specific ribozymes
EP1088086A1 (en) * 1998-06-24 2001-04-04 MUSC Foundation For Research Development Tissue-specific and target rna-specific ribozymes
US7732197B2 (en) 1998-06-24 2010-06-08 The Penn State Research Foundation Tissue-specific and target RNA-specific ribozymes
EP1088086A4 (en) * 1998-06-24 2003-01-08 Musc Found For Res Dev Tissue-specific and target rna-specific ribozymes
AU2004201285B8 (en) * 1998-06-24 2004-04-29 Musc Foundation For Research Development Tissue-specific and target RNA-specific ribozymes
WO2000043781A2 (en) * 1999-01-21 2000-07-27 Metamorphix, Inc. Growth differentiation factor inhibitors and uses therefor
WO2000043781A3 (en) * 1999-01-21 2001-02-01 Metamorphix Inc Growth differentiation factor inhibitors and uses therefor
AU778737B2 (en) * 1999-04-14 2004-12-16 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
EP1702983A2 (en) 2000-04-13 2006-09-20 Medical University of South Carolina Tissue-specific and pathogen-specific toxic agents, ribozymes, DNAzymes and antisense oligonucleotides and methods of use thereof
EP1611231A2 (en) * 2003-02-21 2006-01-04 The Penn State Research Foundation Rna interference compositions and methods
EP1611231A4 (en) * 2003-02-21 2008-08-13 Penn State Res Found Rna interference compositions and methods
WO2007052046A1 (en) * 2005-11-04 2007-05-10 King's College London Anti-hepatitis b virus ribozymal nucleic acid

Also Published As

Publication number Publication date
CN1207769A (en) 1999-02-10
ATE294854T1 (en) 2005-05-15
JP2007084550A (en) 2007-04-05
EP1561815A2 (en) 2005-08-10
DE69634698D1 (en) 2005-06-09
JP2000500967A (en) 2000-02-02
EP0866852A1 (en) 1998-09-30
IL124383A0 (en) 1998-12-06
AU7727296A (en) 1997-05-29
JP4033902B2 (en) 2008-01-16
US5824519A (en) 1998-10-20
EP1561815A3 (en) 2007-11-07
AU728732B2 (en) 2001-01-18
DE69634698T2 (en) 2006-01-19
EP0866852B1 (en) 2005-05-04

Similar Documents

Publication Publication Date Title
AU728732B2 (en) Tissue-specific and target RNA-specific ribozymes
AU702373B2 (en) Stabilized external guide sequences
US5814500A (en) Delivery construct for antisense nucleic acids and methods of use
US5877162A (en) Short external guide sequences
JP3330930B2 (en) Gene units to inhibit RNA function
Sullivan Development of ribozymes for gene therapy
US20140220676A1 (en) Method and Medicament for Inhibiting the Expression of a Given Gene
MXPA06012605A (en) Compositions and methods for enhancing delivery of nucleic acids into cells and for modifying expression of target genes in cells.
WO1995031552A2 (en) Human papilloma virus inhibition by a hairpin ribozyme
CA2086105A1 (en) Method for treating leukemias
WO1996018733A2 (en) Ribozyme-mediated inactivation of leukemia-associated rna
CA2236998C (en) Tissue-specific and target rna-specific ribozymes
JP2005510204A (en) Nucleic acids for inhibiting hairless protein expression and methods of use thereof
CA2578402A1 (en) Human papilloma virus inhibition by a hairpin ribozyme
AU2588795A (en) Human papilloma virus inhibition by a hairpin ribozyme
MXPA96005514A (en) Horqui medianteribozima human papilloma virus inhibition

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 96198727.8

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN

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

Ref document number: 2236998

Country of ref document: CA

Ref document number: 2236998

Country of ref document: CA

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 1997 518381

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1996940377

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1996940377

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1996940377

Country of ref document: EP