WO1992017211A1 - Inhibition d'un retrovirus a l'aide d'acides nucleiques non codants s'hybridant sur des sequences d'encapsidation - Google Patents

Inhibition d'un retrovirus a l'aide d'acides nucleiques non codants s'hybridant sur des sequences d'encapsidation Download PDF

Info

Publication number
WO1992017211A1
WO1992017211A1 PCT/US1992/002911 US9202911W WO9217211A1 WO 1992017211 A1 WO1992017211 A1 WO 1992017211A1 US 9202911 W US9202911 W US 9202911W WO 9217211 A1 WO9217211 A1 WO 9217211A1
Authority
WO
WIPO (PCT)
Prior art keywords
rna
antisense
dna
virus
retrovirus
Prior art date
Application number
PCT/US1992/002911
Other languages
English (en)
Inventor
Thomas E. Wagner
Lei Han
Original Assignee
Edison Animal Biotechnology Center, Ohio University
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 Edison Animal Biotechnology Center, Ohio University filed Critical Edison Animal Biotechnology Center, Ohio University
Priority to JP4510048A priority Critical patent/JPH06506599A/ja
Priority to AU17780/92A priority patent/AU655279B2/en
Publication of WO1992017211A1 publication Critical patent/WO1992017211A1/fr

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
    • 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
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; 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; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/02Animal zootechnically ameliorated
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; 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
    • A01K2267/0337Animal models for infectious diseases
    • 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

Definitions

  • This invention which is in the field of virology and medicine, relates to DNA sequences encoding an antisense RNA molecule capable of hybridizing with a retrovirus packaging sequence and thereby inhibiting a retrovirus infection, the antisense RNA sequences, hosts transfected with the DNA sequences, and methods for rendering cells and animals resistant to retrovirus infection.
  • Retroviruses are a major threat to the health of humans and animals. This is most dramatically demonstrated by the role of HIV-1 in the worldwide AIDS epidemic. To date, no effective treatment or cure for retroviral infections has been found. It is widely accepted that the damage caused (both directly and indirectly) by retroviruses as infectious agents has become the most serious global challenge to medical science in recent times. Hence, the need for effective methods and compositions for prevention or treatment of retrovirus injections is abundantly clear.
  • Retroviruses comprise a large class of RNA viruses which have the property of "reverse transcribing" their genomic RNA into DNA which can integrate into the host cell genome. Many members of this virus class are tumorigenic. These viruses require only a limited number of genes and genetic regulatory sequences to complete their cycle of infection in host cells or organisms (1-4) . This
  • SUBSTITUTESHEET remarkable molecular efficiency partially explains the effectiveness of retroviruses, such as human immunodeficiency virus (HIV) , as pathogens.
  • retroviruses such as human immunodeficiency virus (HIV)
  • HIV human immunodeficiency virus
  • Retroviruses are small, single-stranded positive- sense RNA viruses. Their genomes contain, among other things, the sequence for the RNA-dependent DNA polymerase, reverse transcriptase. Many molecules of reverse transcriptase are found in close association with the genomic RNA in the mature viral particle. Upon entering a cell, this reverse transcriptase produces a double-stranded DNA copy of the viral genome, which is inserted into the host cell's chromatin. Once inserted, the viral sequence is called a provirus. In some ways retroviral integration resembles that of various eucaryotic mobile genetic elements such as Copia and 412 of Drosophila or Ty-1 in yeast.
  • Retroviral integration is directly dependent upon viral proteins.
  • Linear viral DNA termini (the LTRs) form the structure allowing integration of the proviral DNA.
  • the retroviral protein directly involved in inserting the viral DNA into the host DNA is called the integrase protein (IN) .
  • the sequence of the IN protein is encoded in the 3' part of the viral polymerase gene. After translation it is proteolytically processed from the larger precursor molecule to yield an active protein (of 46kd in Mo- MLV; in avian viruses and the human immunodeficiency virus it is 32kd) .
  • an active protein of 46kd in Mo- MLV; in avian viruses and the human immunodeficiency virus it is 32kd.
  • the IN protein removes bases from the 3 • hydroxyl termini of both strands of the reverse transcriptase produced viral DNA. These 3' ends are covalently attached to 5'- phosphoryl ends of the host cells' DNA.
  • the IN protein of all retroviruses is thought to have an endonuclease activity which results in the production of a staggered cut in the host DNA at the site of integration.
  • the filling in of this staggered cut by cellular enzymes after the ligation of the viral DNA to the host DNA results in the duplication of the short sequences of the host DNA.
  • Progeny viral genomes and RNAs are transcribed from the inserted proviral sequence by host cell RNA polymerase II in response to transcriptional, regulatory signals in the terminal regions of the proviral sequence, the long terminal repeats or LTRs.
  • the host cell's protein production machinery is used to produce viral proteins, many of which are inactive until processed by virally encoded proteases.
  • progeny viral particles bud from the cell surface in a non-lytic manner.
  • Retroviral infection does not necessarily interfere with the normal life cycle of an infected cell or organism. While most classes of DNA viruses may be implicated in tumorogenesis, retroviruses are the only taxonomic group of RNA viruses that are oncogenic.
  • SUBSTITUTESHEET retroviruses such as the Human Immunodeficiency Virus (HIV) , which is the etiological agent responsible for acquired immune deficiency syndrome in humans, are also responsible for several very unusual diseases of the immune systems of higher animals.
  • HIV Human Immunodeficiency Virus
  • retroviruses share common morphological characteristics. They are enveloped viruses typically around lOOnm in diameter.
  • the envelope is derived from the cytoplasmic membrane of the host cell as the maturing virus buds from that cell. It is covered by glycoprotein spikes, coded for by the viral genome.
  • glycoprotein spikes coded for by the viral genome.
  • the cytoplasmic membranes of infected cells actively transcribing the proviral sequences and processing viral proteins have viral envelope glycoproteins inserted into them. In some cases, these glycoproteins have been shown to cluster on certain regions of the cell membrane. Furthermore, viruses tend to bud preferentially from these regions.
  • the envelope encloses an icosahedral capsid, or nucleoid, composed of proteins coded for by the virus.
  • the capsid contains a ribonucleoprotein complex that includes the genomic RNA, reverse transcriptase, the integrase protein and certain other factors necessary for the production of the double-stranded DNA copy of the viral genome. It is not uncommon for the capsid to contain small amounts of non-viral RNA other than the cellular tRNAs which are always present. The cellular tRNAs are base-paired to specific regions on the viral genome and play an important role in reverse transcription as will be described later. There is also an inner coat composed of core proteins found between the nucleocapsid and the envelope.
  • retroviruses This morphological classification scheme of retroviruses is based on structural similarities apparent in electron micrographs. In this scheme, retroviruses are divided into four groups or types of particles designated as A-type, B-type, C-type and D- type.
  • the A-type particles are non-infectious and are found only within cells. They range in size from 60- 90nm. They do not have an encapsulating membrane. They may be found intracisternally or intracytoplasmically. Their classification is further subdivided on this basis. The role of the intracisternal A-type particles is unknown, but the intracytoplasmic particles appear to be immature, or precursor forms, to B-type mouse mammary tumor virions. It has also been speculated that they might be retrotransposons.
  • B-type particles exhibit very prominent spikes on their envelope surface, and their nucleoids are eccentrically located.
  • Mouse mammary tumor virus is the primary example of this type of retrovirus.
  • C-type particles represent the largest morphological class of retroviruses.
  • the envelope spikes of the C-type viral particles vary greatly in size and quantity, but all viruses of this type have a centrally located core in the mature virion.
  • Moloney murine leukemia virus is a typical C-type virus.
  • the D-type particles exhibit the same eccentrically located nucleoid as the B-type.
  • the spikes of the D-type are noticeably shorter than those of the B-type. Examples of this type have only been found in primates.
  • Mo-MLV SUBSTITUTESHEET virus
  • the sequence of the entire virus is known, see Shimnick, et al., Nature, 293:543-48 (1981) and Miller and Ver a, J. Virol., 49:214-22 (1984).
  • Mo-MLV carries two copies of its genomic RNA in the mature viral particle.
  • the diploid RNA genome of retroviruses is unique among viruses and is a necessary component of the reverse transcription process.
  • the identical subunits of the viral genomic RNA exhibit several characteristics of a eucaryotic RNA molecule.
  • the 5' end of the molecule carries a typical in RNA cap structure (m 7 G 5 'ppp 5 'Gm) .
  • a poly- A tail of about 200 residues is attached to the 3' end, and several internal adenosine residues are methylate .
  • the cap and the poly-A tail three primary coding regions and six functional regions can be identified on the viral RNA.
  • a copy of the highly conserved LTR, or long terminal repeat, region is found at both ends of the molecule. Like the diploid genome, these are needed for successful reverse transcription.
  • the 5' LTR region includes sequences having promoter and enhancer activity.
  • the LTR region also contains a poly-A addition signal.
  • the 5' LTR region is followed by the U5 region. Next, is the L, or untranslated leader,, sequence.
  • the L region includes the primer binding (PB) site for the initiation of negative strand DNA synthesis, the PB- site.
  • PB primer binding
  • a molecule of tRNA which is used as a primer in the initiation of negative strand DNA synthesis, is base-paired to the PB-site. It also includes the site, which is required for encapsidation of the viral genome.
  • sag the three major coding regions: sag, or the group-specific antigen gene, which code for the
  • SUBSTITUTESHEET viral core proteins pol. which encodes the viral polymerase (or reverse transcriptase) ; and env, which encodes the envelope proteins and glycoproteins. These are followed by the PB+ site, which binds a primer used in positive strand DNA synthesis.
  • the U3 region which contains the viral enhancer and promoter.
  • the U3 region is followed by the second copy of the LTR region.
  • the mature viral particle of MLV contains nine major proteins. These are produced by the post- translational processing of primary translational products. These proteins are typically named according to a system introduced in 1974. In this system, the symbol "p" (for protein) or "gp"
  • glycoprotein is followed by a number showing the approximate molecular weight of the protein in kDa as determined by sodium dodecyl sulfate- polyacrylamide gel electrophoresis (SDS-PAGE) (23) .
  • SDS-PAGE sodium dodecyl sulfate- polyacrylamide gel electrophoresis
  • internal structural proteins are products of the viral gag gene. They are: p30, the major capsid protein; pl5, a hydrophobic matrix protein; plO, a basic protein found in the nucleocapsid; and pl2, an acidic phosphoprotein often designated as ppl2, whose virion location and function have yet to be determined.
  • the pl5 protein is a transmembrane protein which attaches to 9P70 and anchors it to the membrane.
  • the gp70 protein attaches the mature virion to cell surface viral receptors.
  • the viral polymerase protein is also referred to as p70.
  • this protein appears to be a dimer held together by noncovalent and disulfide bonds (26) .
  • RNA-dependent DNA polymerase Besides acting as an RNA-dependent DNA polymerase.
  • SUBSTITUTESHEET reverse transcriptase also has RNase H activity, which results in digestion of the RNA in a RNA-DNA hybrid into short oligonucleotides.
  • Two other proteins are thought to be products of the pol gene (in at least some retroviruses) .
  • a viral protease, pl4 is responsible for maturation of other viral proteins.
  • An integrase protein, p46 in MuLV, acts to join or insert the double-stranded DNA copy of the viral genome into the host cell DNA (27, 28, 29, 30).
  • retroviruses feature additional retroviral genes.
  • tax and rex The product of tax, essential for viral replication, is a transacting transcription-activating factor which enhances viral gene expression (Seiki, M. et al., Proc. Natl. Acad. Sci. USA 80:3618-3622 (1983); Sodorski, J.G. et al. r Science 225:381-385 (1984); Chen, I. et al.. Science 229:54-58 (1987)).
  • the rex gene also required for viral replication, is a posttranscriptional regulator of viral gene expression (Hidaka, M. et al.. EMBO J. 7:519-523 (1988) ; Inoue, J. et al., Proc. Natl. Acad. Sci. USA 84:3652-3657 (1988)).
  • the HIV-l retrovirus also possesses genes modulating viral replication, including vif, vpr, tat, rev, vpu. and nef (Haseltine, W.A. , J. Acguired Immune Deficiency Syndrome 1:217240 (1988)).
  • retroviral replication Another required element for retroviral replication is the cis-acting viral genomic sequences necessary for the specific encapsidation of the genomic viral RNA molecules into virus particles (4- 10) .
  • These packaging sequences termed Psi. have been identified and exploited in the construction of retroviral vectors designed for gene transfer (10-13) . Functional packaging sequences are absolutely required for retroviral replication in host cells.
  • SUBSTITUTESHEET packaging sequences were deleted from the retroviral genome (to prevent viral multiplication) and the DNA was transfected into host cells, the cells could produce all the viral proteins and viral RNA, but could not package the viral RNA genome into an infectious particle. However, such cells could serve as "helper” cells and complement a DNA vector which contained only the retroviral LTRs and the packaging sequences.
  • the "helper” cell provided: (a) the gag- encoded proteins needed to package the vector RNA; (b) the envelope protein to form the capsid; and (3) the reverse transcriptase to convert the RNA genome into a DNA copy upon arrival into the infected cell.
  • the sequences required for RNA packaging into virions have been defined and Shown to reside between the 5• LTR and the beginning of the early portion of the gag gene.
  • Gene expression involves the transcription of pre-messenger RNA from a DNA template, the processing of the pre-messenger RNA into mature messenger RNA, and the translation of the messenger RNA into one or more polypeptides.
  • the use of antisense RNA to inhibit RNA function within cells and whole organism has generated much recent interest (14-16) .
  • Antisense RNA can bind in a highly specific manner to its complementary sequences ("sense RNA") . This blocks the processing and translation of the sense RNA and may even disrupt interactions with sequence-specific RNA binding proteins (17-20) .
  • a plasmid was constructed leaving a promoter which directed the transcription of a RNA complementary to the normal thymidine kinase (TK) mRNA. When such plasmids, together with plasmids containing a normally expressed
  • SUBSTITUTESHEET TK gene were injected into mutant mouse L cells lacking TK, the presence of the antisense gene substantially reduced expression of TK from the normal plasmid rizant et al. , Cell 36:1007 (1984).
  • Antisense oligonucleotides have been shown to be inhibitory in various viral systems. Zamecnik and Stephensen, Biochemistry, 75:280-84 (1978) inhibited Rous sarcoma virus (a retrovirus) production in cultured CEF cells by adding an oligodeoxynucleotide. complementary to 13 nucleotides of the 3' and 5• LTRS, to the culture medium. The DNA was terminally blocked to reduce its susceptibility to exonucleases. They speculated that this antisense DNA might act by blocking circulatizatoin, DNA integration, DNA transcription, translation initiation or ribosomal association. Note the conspicuous absence of any reference to interference with packaging.
  • RNA northern blot analysis the expression assay that is most typically used to test antisense nucleic acid inhibition of HIV- 1 (See Thomas, PNAS, 77:3201, 1980), cannot detect useful antisense sequences that are homologous to the viral packaging sequence. Ruden and Gilboa, J. Virol., 63:677-682 (Feb.
  • HTLV-I replication in primary human T cells engineered to express an antisense RNA.
  • One antisense RNA was directed against the first kilobase of the tax gene CDNA.
  • the other was a 1.1 kilobase Hindlll-Pst I fragment from the 5' end of the proviral DNA.
  • the latter target is said to include the 5' splice site, the tRNA primer binding site, and "possibly" signals for packaging of genomic virus RNA.
  • Antisense-encoding DNAs were operably linked to either the SV40 early promoter or the cytomegalovirus immediate early promoter. Only the vectors expressing the antisense RNA under the control of the CMV promoter exerted an inhibitory effect on cell proliferation, though the SV40 early promoter/anti-tax gene also depressed viral production.
  • SUBSTITUTESHEET Large antisense molecules of the sort advocated by Ruden and Gilboa have several disadvantages. They are difficult to synthesize (particularly if abnormal bases are incorporated as discussed below) . They conceivably could recombine with the original virus, another virus, or an oncogene. They are also more liable to form secondary structures which interfere with their binding to the viral target. Additionally, they are more prone to hybridize to cellular DNA, thereby possibly blocking expression of essential genes.
  • nucleoside or nucleotide analogues have the advantageous properties of resistance to nuclease hydrolysis and improved penetration of mammalian cells in culture (Miller, P.S. et al., Biochemistry 20:1874-1880
  • an oligo(deoxyribonucleoside phosphonate) complementary to the Shine-Dalgarno sequences of 16S rRNA inhibited protein synthesis in E. coli but not mammalian cells (Jayaraman, K. et al. , Proc. Natl. Acad. Sci. USA 78:1537-1541 (1983)).
  • Such oligomers complementary to initiation codon regions of rabbit globin mRNA inhibited translation in cell-free systems.
  • SUBSTITUTESHEET selectively inhibit viral infection (Smith, C.C. et al.. Proc. Natl. Acad. Sci. USA 11:2787-2791 (1986)) .
  • An object of the present invention is to overcome the deficiencies noted above.
  • the present inventors have taken a novel approach to the inhibition of retro v irus replication based on the blockade of virus packaging through hybridization of antisense RNA essentially only to packaging sequences of the viral genome.
  • the inventors have constructed recombinant plasmids in which murine leukemia virus proviral Psi (packaging) sequences, under the transcriptional regulation of lymphotropic virus promoter/regulatory elements from the Moloney-MuLV LTR or the cytomegalovirus immediate early region, were inserted in reverse orientation. This gives rise to production of antisense RNA complementary to Psi. which achieves complete inhibition of productive virus infection.
  • St -JSTITUTESHEET antisense Psi transgenic mice developed any symptoms of leukemia.
  • the present invention is directed to an antisense molecule capable of specifically hybridizing to the packaging sequence of a retrovirus and thereby inhibiting essentially only the packaging of the genomic RNA of the retrovirus.
  • the antisense molecule is less than about 100 bases, and more preferably less than about 60 bases, and it may be DNA, RAN, or an analogue thereof.
  • the antisense molecule may be administered directly like a drug or, if an RNA, it may be generated in vivo in the subject through expression of an introduced gene. Where the antisense molecule is administered directly, it may be composed of a nuclease-resistance RNA or DAN analogue that penetrates the cell membrane.
  • the invention includes recombinant DAN molecules comprising a DNA sequence which is transcribable into such an antisense molecule and hosts transformed or transfected with the above DNA sequence, preferably a mammalian cell host, most preferably a human cell.
  • the invention further relates to a method for rendering a cell resistant to productive infection by a retrovirus comprising inserting into the genome of the cell a DNA sequence, operably linked to a promoter, wherein the DNA sequence is transcribable into an antisense RNA molecule which hybridizes to the packaging sequence and thereby inhibits the packaging of the genomic RNA of the retrovirus, thus rendering the cell resistant to productive infection.
  • the present invention includes a method for rendering a vertebrate animal, such as a mammal, resistant to productive infection by a retrovirus comprising inserting into the genome of essentially all of the germ cells and somatic cells of the mammal
  • SUBSTITUTESHEET a DNA sequence containing the packaging sequence of the retrovirus or a segment thereof in reverse orientation operably linked to a promoter and regulatory elements, wherein the DNA sequence or segment is transcribable into an antisense RNA molecule capable of inhibiting the packaging of the genomic RNA of the retrovirus, thereby rendering the mammal resistant to infection.
  • the DNA sequence is preferably introduced into the mammal or its ancestor at an embryonic stage.
  • the invention therefore relates also to a transgenic non-human mammal essentially all of whose germ cells and somatic cells contain the above DNA sequence, a transgenic in which said DNA sequence has been introduced into the mammal or its ancestor of said mammal at an embryonic stage. Also intended is a chimeric animal, including a human, at least some of whose cells contain the above DNA sequence.
  • Figure 1 is a schematic diagram showing the construction of plasmids pLPPsias (A) and pCPPsias (B)-
  • Figure 2 is a partial sequence (SEQIDN0:1) of the
  • M-MuLV Moloney-Murine leukemia virus
  • Figure 3 is a partial sequence (SEQIDNO:2) of the genomic RNA of bovine leukosis virus, with the region (341-415) expected to include the packaging signal indicated by open bold letters.
  • Figure 4 shows the results of the plaque assay.
  • Figure 5 is a partial sequence (SEQIDNO:3) of HIV-1 in the region including the packaging sequence.
  • Interference with the specific interactions between the Psi sequences of viral genomic RNA 5 necessary for packaging and virion capsid proteins will block the retroviral replication cycle. This interference may be accomplished through the use of antisense nucleic acids (DNA or RNA) complementary to a part of the packaging sequences, which hybridizes o thereto and thereby inhibits replication.
  • DNA or RNA antisense nucleic acids
  • Antisense sequence directed against a retroviral gene would not block replication as effectively as the antisense molecules of the present invention.
  • An antisense RNA molecule directed against a retroviral 5 gene competes with normal messenger RNA for binding to ribosomal RNA, while one directed against the packaging sequence competes with a gag-encoded core protein for binding to genomic RNA.
  • RNA-RNA interactions are stronger than RNA-protein o interactions.
  • the present inventors constructed transgenic mice expressing RNA sequences complementary to the Psi sequences of Moloney murine leukemia virus (M-MuLV) . It was discovered that such animals completely resisted challenge with this leukemia virus.
  • M-MuLV Moloney murine leukemia virus
  • the present invention is therefore directed to the use of antisense RNA and DNA molecules complementary to retroviral packaging sequences as agents for the prevention and treatment of diseases in which the causative agent is a retrovirus.
  • the antisense RNA and genes coding therefor are intended to encompass sequences capable of hybridizing to the packaging sequence of a retrovirus.
  • SUBSTITUTESHEET preferred retroviruses of the present invention include both human and other animal retroviruses.
  • Preferred human retroviruses include Human Immunodeficiency Virus-1 (HIV-1) (or Human T-Cell Lymphotropic Virus-3 or Ly phadenopathy Associated Virus) , Human Immunodeficiency Virus-2 (HIV-2) , Human T-Cell Lymphotropic Virus-I (HTLV-l) , and Human T-Cell Lymphotropic Virus-2 (HTLV2) .
  • HIV-1 Human Immunodeficiency Virus-1
  • HAV-2 Human Immunodeficiency Virus-2
  • HTLV-l Human T-Cell Lymphotropic Virus-I
  • HTLV2 Human T-Cell Lymphotropic Virus-2
  • retroviruses of other animal species most particularly agriculturally important animals such as cows and chickens, and pets such as dogs and cats.
  • Retroviruses are described in detail in Weiss, R. et al. (eds) , RNA Tumor Viruses. "Molecular Biology of Tumor Viruses," Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1984, which is hereby incorporated by reference.
  • the target packaging sequence to which the antisense RNA of the present invention is to hybridize in order to inhibit retrovirus infection is be selected based on examination of the sequence of the retroviral genome.
  • sequences within this region are preferred candidates for antisense targeting. Because of the homology in this region among various retroviruses, an antisense sequence designed for one particular virus may be used successfully to inhibit replication of another virus.
  • preferred antisense aligonucleotides are specific for the region between bases 280-330 of the Mo-MuLV sequence since it is homologous to HIV-1. Since different isolates of HIV- 1 have the same packaging sequences, but at slightly different positions within the genome, the same sequences would be effective in all HIV-1 strains.
  • One preferred anti-sense sequence is one which is substantially complementary to all retrovirus packaging sequences, thus serving as a "universal” inhibitor.
  • a sequence complementary to the core packaging sequence from HIV-I may be worth considering in this regard.
  • oligonucleotide sequences may be produced that are "consensus" packaging sequences having a high degree of complementarity to the packaging sequences of a set of retroviruses, or which are specific to a particular retroviruses. Shown below is an exemplary oligonucleotide sequence according to the present invention, useful for inhibition of HIV-l, which are complementary to viral genomic RNA, viral mRNA, as well as the viral DNA sequences.
  • SEQ ID NO: 4 is a 44mer sequence shown below, which is complementary to the HIV-l packaging sequence.
  • the invention is not limited to any of these packaging site-targeting sequences, but rather includes shorter and longer sequences.
  • the antisense molecule e.g., RNA
  • the antisense molecule of the present invention preferably has 100% complementarity to at least a significant subsequence of the packaging sequence (on one strand of the viral DNA) for which it is targeted.
  • the DNA strand encoding this RNA should be 100% homologous to the DNA strand which is complementary to the packaging sequence.
  • the sequence may have a lower degree of homology, such as at least about 60 or 80%. The homology must be sufficient such that the antisense RNA hybridizes to the target packaging sequences with sufficient affinity to achieve its purpose, i.e. inhibition of viral packaging.
  • the efficiency of such hybridization is a function of the length and structure of the hybridizing sequences. The longer the seguence and the closer the complementarity to perfection, the stronger the interaction. As the number of base pair mismatches increases, the hybridization efficiency will fall off. Furthermore, the GC content of the packaging sequence DNA or the antisense RNA will also affect the hybridization efficiency due to the additional hydrogen bond present in a GC base pair compared to an AT (or AU) base pair. Thus, a target subsequence richer in GC content is preferable as a target.
  • SUBSTITUTESHEET It is desirable to avoid sequences of antisense RNA which would form secondary structure due to intramolecular hybridization, since this would render 5 the antisense RNA less active or inactive for its intended purpose.
  • One of ordinary skill in the art will readily appreciate whether a sequence has a tendency to form a secondary structure. Secondary structures may be avoided by selecting a different o target subsequence within the packaging site.
  • An oligonucleotide between about 15 and about 100 bases in length and complementary to the target subsequence of the retroviral packaging region may be synthesized from natural mononucleosides or, 5 alternatively, from mononucleosides having substitutions at the non-bridging phosphorous bound oxygens.
  • a preferred analogue is a methylphosphonate analogue of the naturally occurring mononucleosides. More generally, the mononucleoside is any analogue o whose use results in oligonucleotides which have the advantages of (a) an improved ability to diffuse through cell membranes and/or (b) resistance to nuclease digestion within the body of a subject (Miller, P.S. et al..
  • nucleoside analogues are well-known in the art, and their use in the inhibition of gene expression are detailed, in a number of references (Miller, P.S. et al.. supra: Jayaraman, K. et al.. supra: Blake, K.R. et al.. supra: Miller, P. et al.. feder. Proc. 43. abstr. 1811 (1984); Smith, C.C. et al.. supra) .
  • Oligonucleotide molecules having a strand which encodes antisense RNA complementary to the target retrovirus packaging sequences can be prepared using procedures which are well known to those of ordinary skill in the art (Belagaje, R. , et al. , J. Biol. Chem. 254:5765-5780 (1979); Maniatis, T., et al.. In: Molecular Mechanisms in the Control of Gene
  • DNA synthesis may be achieved through the use of automated synthesizers. Techniques of nucleic acid hybridization are disclosed by Sambrook et al. (supra) r and by Haymes, B.D., et al. (In: Nucleic Acid Hybridization. A Practical Approach. IRL Press, Washington, DC (1985)), which references are herein incorporated by reference.
  • an "expression vector” is a vector which (due to the presence of appropriate transcriptional and/or translational control sequences) is capable of expressing a DNA (or cDNA) molecule which has been cloned into the vector and of thereby producing an RNA or protein product. Expression of the cloned sequences occurs when the expression vector is introduced into an appropriate host cell. If a prokaryotic expression vector is employed, then the appropriate host cell would be any prokaryotic cell capable of expressing the cloned sequences. Similarly, when a eukaryotic expression vector is employed, then the appropriate host cell would be any eukaryotic cell capable of expressing the cloned sequences.
  • a DNA sequence encoding the antisense RNA of the present invention may be recombined with vector DNA in accordance with conventional techniques, including blunt-ended or staggered-ended termini for ligation, restriction enzyme digestion to provide appropriate termini, filling in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and ligation with appropriate ligases. Techniques for such manipulations are disclosed by
  • a nucleic acid molecule such as DNA, is said to be "capable of expressing" a mRNA if it contains nucleotide sequences which contain transcriptional regulatory information and such sequences are
  • RNA Ribonucleic acid sequences which encode the RNA.
  • the precise nature of the regulatory regions needed for gene expression may vary from organism to organism, but in general include a promoter which directs the initiation of RNA transcription. Such regions may include those 5•-non-coding sequences involved with initiation of transcription such as the TATA box.
  • the non-coding region 3' to the gene sequence coding for the desired RNA product may be obtained by the above-described methods.
  • This region may be retained for its transcriptional termination regulatory sequences, such as those which provide for termination and polyadenylation.
  • the transcriptional termination signals may be provided. Where the transcriptional termination signals are not satisfactorily functional in the expression host cell, then a 3' region functional in the host cell may be substituted.
  • DNA sequences are said to be operably linked if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-shift mutation in the region sequence to direct the transcription of the desired gene sequence, or (3) interfere with the ability of the gene sequence to be transcribed by the promoter region sequence.
  • a promoter region would be operably linked to a DNA sequence if the promoter were capable of effecting transcription of that DNA sequence. In order to be "operably linked" it is not necessary that two sequences be immediately adjacent to one another.
  • the promoter sequences of the present invention may be either prokaryotic, eukaryotic or viral. Suitable promoters are inducible, repressible, or, more preferably, constitutive. Examples of suitable prokaryotic promoters include promoters capable of recognizing the T4 polymerases (Malik, S. et al.. J. Biol. Chem. 263:1174-1181 (1984); Rosenberg, A.H. et al., Gene .59.:191-200 (1987) Shinedling, S. et al. , J. Molec. Biol. 195:471-480 (1987) Hu, M.
  • Eukaryotic promoters include the promoter of the mouse metallothionein I gene (Hamer, D. , et al.. J. Mol. APPI. Gen. 1:273-288 (1982)); the TK promoter of Herpes virus (McKnight, S., Cell 31:355-365 (1982)); the SV40 early promoter (Benoist, C. , et al.. Nature (London) 290: 304-310 (1981) and the yeast ⁇ al4 gene promoter (Johnston, S.A. , et al.. Proc. Natl. Acad. Sci. (USA) 79: 6971-6975 (1982); Silver, P.A. , et al.. Proc.
  • Preferred promoters and additional regulatory elements, such as polyadenylation signals, are those which should yield maximum expression in the cell type which the retrovirus to be inhibited infects.
  • HIV-l, HIV-2, HTLV-l and HTLV-2 infect lymphoid cells, and in order to efficiently express an antisense RNA complementary to the packaging sequence of one (or more) of these viruses, a transcriptional control unit (promoter and polyadenylation signal) are selected which provide efficient expression in lymphoid cells (or tissues) .
  • a transcriptional control unit promoter and polyadenylation signal
  • preferred promoters are the cytomegalovirus immediate
  • SUBSTITUTESHEET early promoter (32) , optionally used in conjunction with the bovine growth hormone polyadenylation signals (33) , and the promoter of the Moloney-MuLV LTR, for 5 use with a lymphotropic retrovirus.
  • a desirable feature of the Moloney-MuLV LTR promoter is that it has the same tissue tropism as does the retrovirus.
  • the CMV promoter is likewise expressed primarily in lymphocyte.
  • the metallothionein promoter has the o advantage of inducibility.
  • the SV40 early promoter exhibits high level expression in vitro in bone marrow cells.
  • An antisense RNA molecule may be injected into the human or other animal subject to be protected or 5 treated by any compatible route of administration, e.g., intravenously, intramuscularly, subcutaneously or intraperitoneally, or administered by ingestion or inhalation. Special dosage forms, such as slow release capsules or implants, may be used when o appropriate. Alternatively an antisense DNA molecule may be provided. DNA is more readily synthesized in vitro than RNA.
  • the antisense molecule may be an analogue of DNA or RHA.
  • the present invention is not limited to use of any particular DNA or RNA analogue, provided it is capable of adequate hybridization to the complementary genomic DNA of a packaging sequence, has adequate resistance to nucleases, and adequate bioavailability and cell take-up.
  • DNA or RNA may be made more resistant to in vivo degradation by enzymes, e.g., nucleases, by modifying internucleoside linkages (e.g. , methylphosphonates or phosphorothioates) or by incorporating modified nucleosides (e.g., 2'-0- methylribose or l'-alpha-anomers) .
  • the naturally occurring linkage is
  • the entire antisense molecule may be formed of such modified linkages, or only certain portions, such as the 5' and 3 » ends, may be so affected, thereby providing resistance to exonucleases.
  • Antisense molecules suitable for use in the present invention include but are not limited to dideoxyribonucleoside methylphosphonates, see Mill, et al.. Biochemistry, 18:5134-43 (1979),
  • SUBSTITUTESHEET oligodeoxynucleotide phosphorothioates, see Matsukura, et al., Proc. Nat. Acad. Sci., 84:7706-10 (1987), oligodeoxynucleotides covalently linked to an 5 intercalating agent, see Zerial, et al. , Nucleic Acids Res., 15:9909-19 (1987), oligodeoxynucleotide conjugated with poly(L-lysine) , see Leonetti, et al.. Gene, 72:32-33 (1988), and carbamatelinked oligomers assembled from ribose-derived subunits, see Summerton, o J- , Antisense Nucleic Acids Conference, 37:44 (New York 1989) .
  • antisense nucleic acid drugs While direct administration of antisense nucleic acid drugs provides acute protection, the protective period is dependent on the half-life of the molecule. 5 Moreover, the supply of antisense nucleic acid can be increased only by further administrations. It may therefore be desirable to provide a self-renewing source of antisense RNA by introducing a recombinant DNA molecule, capable of transcribing said antisense o RNA, into one or more cells of the human or animal subject, thus creating a transgenic or chimeric animal having enhanced resistance to retroviral infection.
  • the recombinant DNA may be delivered to the animal by, e.g., microinjection of the expression cassette into the animal at the oocyte stage, retroviral vector transfection of the embryo, or intravenous injection of the retroviral vector into the fetal or postnatal animal.
  • the present invention is also directed to a transgenic eukaryotic animal (preferably a vertebrate, and more preferably a mammal) the germ cells and somatic cells of which contain recombinant genomic DNA according to the present invention which encodes an antisense RNA capable of hybridizing to a retroviral packaging sequence.
  • "Antisense" DNA is introduced into the animal to be made transgenic, or
  • SUBSTITUTESHEET an ancestor of the animal, at an embryonic stage, preferably the one-cell, or fertilized oocyte, stage, and generally not later than about the 8-cell stage.
  • transgene means a gene which is incorporated into the genome of the animal and is expressed in the animal, resulting in the presence of the RNA transcript of the transgene in the transgenic animal. There are several means by which such a gene can be introduced into the genome of the animal embryo so as to be chromosomally incorporated and expressed.
  • the DNA may be microinjected into the male or female pronucleus of fertilized eggs. It may also be microinjected into the cytoplasm of the embryonic cells.
  • the cells may be transfected by a retrovirus carrying the transgene. The use of retroviral transfection is not limited to the embryonic stage; the vector may be intravenously or intraperitoneally introduced into the fetal or postnatal animal.
  • transgene is present in all of the germ cells and somatic cells of the transgenic animal and has the potential to be expressed in all such cells.
  • the presence of the transgene in the germ cells of the transgenic "founder" animal in turn means that all its progeny will carry the transgene in all of their germ cells and somatic cells.
  • Introduction of the transgene at a later embryonic stage in a founder animal may result in limited presence of the transgene in some somatic cell lineages of the founder; however, all the progeny of this founder animal that inherit the transgene conventionally, from the founder's germ cells, will carry the transgene in all of their germ cells and somatic cells.
  • SUBSTITUTESHEET Chimeric mammals in which fewer than all of the somatic and germ cells contain the antisense DNA of the present invention, such as animals produced when fewer than all of the cells of the morula or blastula are transfected in the process of producing the transgenic mammal, are also intended to be within the scope of the present invention.
  • Chimeric animals may be created by "gene therapy", in which the transgene is typically introduced after birth.
  • Antisense RNA might be delivered to the lymphocytes of AIDS patients by gene therapy methods.
  • bone marrow cells may be treated with a recombinant, replicatipn deficient, retroviral vector containing DNA sequences encoding and expressing anti ⁇ sense RNA complementary to the packaging sequences of HIV.
  • bone marrow cells would be removed from the patient, treated with the recombinant retroviral vector and cells in which the DNA genome of the vector had integrated in their chromosomes selected using FACS cell sorting based upon a light visualization marker also incorporated into the retorviral vector (i.e., the ⁇ -galactosidase gene).
  • DNA can be introduced into various tissues by attaching it to proteins which are the ligands for cellular receptors (Wu, et al., J. Biol. Chem., 263:14621; 1988), or by unassisted introduction into muscle cells (Wolff, et al., Science, 247:1465; 1990).
  • proteins which are the ligands for cellular receptors
  • muscle cells Wang, et al., J. Biol. Chem., 263:14621; 1988
  • This approach would be very appealing both for animals and for humans, because it would involve simply the injection of a DNA molecule coding for the anti-sense RNA, or this DNA molecule bound to a receptor targeted ligand. This injection might have to be given several times a year (for example, when an outbreak of a retroviral disease occurs in a given region) but would provide protection against retroviral infection when needed.
  • the antisense-RNA encoding DNA may be introduced when the animal (including human) already is infected, or prior to infection, as a prophylactic measure. In the latter case, use of a regulatable promoter may be desirable.
  • Plasmids Recombinant plasmids pLPPsias and pCPPsias were constructed as shown in Fig. 1. Plasmids pLJ(21) was cleaved with Smal and the 540 bp Smal fragment containing the M-MuLV sequences isolated. Plasmid p3'-LTR-2, containing the 3'M-MuLV LTR (22), was linearized with Smal and fused to the 540 bp fragment from pLJ in both orientations using DNA ligase. Clones of pLPPsi with the antisense orientation were selected on the basis of their Kpnl digestion patterns.
  • the "sticky ends" of this fragment were filled in by incubation with deoxynucleoside triphosphates (A,T,G and C) in the presence of the DNA polymerase I Klenow fragment and blunt-end ligated to the 540 bp Smal fragment from pLJ in both orientations.
  • EXAMPLE III Cell Culture Mouse NIH 3T3 cells were maintained in Dulbecco's Modified Eagle's medium (DMEM) containing 10% Nu- serum. Stable cell lines expressing antisense Psi sequences were established by co-transformation with pCPPsias (36 ⁇ g) and pSV40neo (0.36 ⁇ g) (ATCC 33964) using a modified DEAE dextran-dimethyl sulfoxide shock procedure (26) .
  • DMEM Dulbecco's Modified Eagle's medium
  • RNA and RNA Analysis and Enzyme Assays The integrity of DNA sequences in transgenic mice was analyzed by the method of Southern (27) .
  • RNA was prepared from lymphocytes by a single step isolation method (28) using Ficoll-Hypaque gradient isolation procedure (29) .
  • RNA was then subjected to electrophoresis at 70V for 4.5h in a 1.2% agarose/formaldehyde gel and transferred onto nitrocellulose paper for hybridization and radioautography as described (30) .
  • Reverse Transcriptase assays were performed as described previously (31) .
  • a strand specific RNA probe was used for Northern hybridization assays of lymphocyte
  • SUBSTITUTESHEET RNA This probe was synthesized from a pSP65 vector containing the 540 bp Smal Psi sequence containing fragment inserted in the sense orientation using SP6 RNA polymerase and labeled guanosine 5*-triphosphates (Promega Riboprobe System, Promega Corp., Madison Wisconsin) .
  • Linear DNA fragments containing the M-MuLV packaging sequences in inverted orientation under the regulation of the M-MuLV LTR or the Cytomegalovirus immediate-early promoter were respectively prepared from pLPPsias as a 2.2kb Hindlll fragment or from pCPPsias as a 2.3 kb EcoRI-Clal fragment.
  • LPPsias transgenics trace to a single founder mouse resulting from the microinjection of the 2.2 kb Hindlll fragment from pLPPsias and CPPsias transgenics trace to a single founder from the microinjection of the 2.3 kb EcoRI- Clal fragment from pCPPsias.
  • SUBSTITUTESHEET was digested with Kpn I and then subjected to electrophoresis at 50V for 9h in a 0.8% agarose gel.
  • the DNAs were transferred onto nitrocellulose paper by the technique of Southern (27) .
  • the nitrocellulose paper was hybridized to a nick translated DNA probe (5 x 10 8 to 10 X 10 8 cpm/ ⁇ g) prepared from pBR322 for LPPsias DNA (this probe hybridizes to the LPPsias sequences because of the presence of pBR322 sequences in this construction but does not crosshybridize with endogenous retroviral sequences present in the animal) or a 1.3 kb Sac I- EcoRI fragment from pCPPsi for CPPsias DNA.
  • the transcriptional units introduced into these two lines of mice were constructed to provide the appropriate tissue tropism for the transcription of the antisense RNA within the lymphoid target tissue for M-MuLV.
  • the transcriptional unit including the U3R promoter/enhancer elements and the RU5 polyadenylation signals from the M-MuLV LTR, is essentially identical to the transcriptional control sequences of the intact
  • the transcriptional control unit within the CPPsias mice including the cytomegalovirus immediately-early promoter (32) and the bovine growth hormone polyadenylation signals (33) , would also be expected to have lymphoid tissue tropism (34) .
  • lymphoid tissue tropism 314
  • Northern hybridization analysis of RNA from lymphocytes isolated from LPPsias and CPPsias mice was performed.
  • RNA from both CPPsias and LPPsias mice was hybridized to a strand specific RNA probe complementary to the M-
  • MuLV antisense Psi sequence with a specific activity of 1-5 x 10 8 cpm/ ⁇ g.
  • RNA fragments found in all LPPsias lymphocyte RNA and the 750b hybridizing RNA in CPPsias mice confirmed the production of the antisense Psi sequences of M-MuLV in the white blood cells of LPPsias and CPPsias transgenic mice.
  • the different lengths of the antisense RNA in LPPsias and CPPsias mice is the result of the different gene constructions introduced into these two lines of mice.
  • the CPPsias mice contain a gene construction including a small portion of exon 5 from the bovine growth hormone gene and the bovine growth hormone poly A addition signals resulting in a longer antisense RNA product.
  • a stable cell line expressing antisense Psi sequences was established by co-transformation of mouse NIH 3T3 cells with linearized pCPPsias (36 ⁇ g) and pSV40neo (0.36 ⁇ g) (ATCC 33694) using a modified DEAE-dextran dimethyl-sulfoxide-shock procedure (26) .
  • Stable transformants were selected in G418DMEM medium, Southern analysis performed to establish the presence of presence of integrated CPPsias sequences, and Northern analysis carried out to confirm the presence of antisense M-MuLV RNA production by cloned cell lines.
  • Both control mouse NIH 3T3 cells and the CPPsias transformed cell line were challenged with M- MuLV (4X10 6 PFU/75 cm plate) for 24 hrs., washed free of virus, cultured for an additional 48 hrs, filtered through 0.45 urn filters to remove cells and cellular material from the medium and virus particles concentrated from the medium by sucrose gradient centrifugation (35) .
  • RNA was prepared from the viral pellet by a phenol-chloroform extraction procedure (35) and the presence of M- MuLV genomic RNA detected by Northern Analysis using a random primer labeled 540 bp Sma I DNA fragment from pCPPsias (1 x 10 8 - 5 x 10 8 cpm/ ⁇ g) as a M-MuLV virus specific probe.
  • No viral RNA was produced from antisense Psi RNA expressing NIH 3T3 cells after challenge with M-MuLV, while a substantial amount of viral genomic RNA (8.3 Kb) was produced from control NIH 3T3 cells challenged with M- MuLV.
  • Reverse transcriptase activity in the supernatant from the stable cell line expressing antisense Psi RNA after infection with M-MuLV was measured and compared to normal mouse NIH 3T3 cells infected with M-MuLV (31) . Substantial reverse transcriptase activity was
  • mice In order to test the level of inhibition of M- MuLV induced leukemia in antisense Psi transgenic mice, littermate control and tranegenic mice were challenged with M-MuLV at birth.
  • Transgenic male LPsias and CPsias mice Fl hybrids of C57B6 and SJL
  • C57B6/SJL non-transgenic mice
  • each offspring from these matings were injected intraperitoneally with 0.1 ml containing 1 X 10 5 M-MuLV infectious virions.
  • a DNA sample from the tail of each mouse pup was analyzed by slot blot hybridization and each mouse ear notched to code transgenic and non- transgenic offspring.
  • SUBSTITUTESHEET percentage (33%) of the control mouse showed the symptoms of leukemia, none of the LPPsi as or CPPsi as mice were judged to be leukemic. Numerous of the control mice were obviously severely impaired, showing typical leukemia lymphocyte morphology, containing spleens with weights in excess of 0.5g and several had spleens larger than l.Og (Fig. 6) while no antisense Psi transgenic mice appeared abnormal prior to sacrifice or showed enlarged spleens, low abnormal prior to sacrifice or showed enlarged spleens, low hematocrit values or leukemic lymphocyte morphology. The difference in spleen weight between control and transgenic mice could be as large as 22 times (#13/#17) .
  • SUBSTITUTESHEET were produced and challenged with M-MuLV. While control cells showed substantial quantities of M-MuLV viral RNA in RNA preparations from the cell supernatant, supernatants from cell lines expressing antisense Psi RNA were devoid of M-MuLV genomic viral RNA suggesting the inability of these cells to produce functional virus containing packaged viral RNA. In spite of the lack of viral RNA in the cell supernatant, significant reverse transcriptase activity was measured by a sensitive reverse transcriptase assay (31) (Table II) suggesting that the antisense Psi RNA expressing cells are producing empty viral particles because of antisense blockage of the packaging process.
  • the packaging sequence of HIV-l, isolate ELI is located within bases 200-340. There is a significant homology between bases 280-330 of this virus and the packaging sequence of the M-MuLV virus.
  • Oligonucleotides complementary to the packaging sequence of the HIV-l region between pase pairs 280— 330 (in the proviral genome of HIV-l,isolate ELI) are synthesized using standard techniques. The largest of such oligonucleotides is 50 bases in length and is complementary to the entire 280-330 sequence. Alternatively, a 50 base oligonucleotide containing methylphosphonate analogues of the natural mononucleosides is synthesized according to known methods.
  • SUBSTITUTESHEET natural or analogue nucleotides are injected IV into AIDS patients each day for several weeks.
  • Oligonucleotides complementary to the packaging sequence of the bovine leukosis virus (BLV) between base pairs 341 and 417 are synthesized using standard techniques. The largest of such oligonucleotides is
  • oligonucleotide containing methylphosphonate analogues of the natural mononucleosides is synthesized according to known methods.
  • a plaque assay was conducted to determine the relative inhibitory effect of various antisense molecules (30 mer, 38 mer, 40 mer, 50 mer and 60 mer) directed against the packaging sequences ⁇ + (69-106) and *• (216-570) of MO-MLV.
  • the target sequences of these molecules are described below:
  • the cells were irradiated with UV for 30-45 sees, to limit their growth, and then XC cells were laid atop the NIH 3T3 cells, 0.5 x 10 5 cells/well. After 3-4 days, the cells were fixed with 2ml/well of methanol (lmin.) and stained with lml/well hematoxylin (304 mins) . Plaques were counted under low magnification with a dissecting microscope. Results are shown in Figure 4. The result of the viral plaque assay with different oligonucleotides has shown that the 50mer oligonucleotide has the best inhibition effect. This means that in comparison with the control, the 50mer oligo reduced the plaque number by 6-7 times.
  • the 50mer sequence (RNA 3OOb-350b, 3'GACAT AGACC GCCTG GGCAC CACCT TGACT GCTCA AGCCT TGTGG GCCGG 5') (SEQIDNO:5) comprises a sequence complementary to the core of the Mo-MuLV packaging sequence. Significantly, it also comprises a sequence complementary to the core portion (19bp) of the HIV-l packaging sequence.
  • SUBSTITUTESHEET as for cells treated with the 50 mer, clearly demonstrating viral inhibition.
  • the later convergence of plaque contents was expected, as the cells were treated only once with the antisense molecules, which were gradually degraded by cell nucleases.
  • Use of resistant analogues, or replenishing the supply of antisense molecules by subsequent administrations or by providing for intracellular expression of the antisense molecule would overcome this problem.
  • Each entry includes both a reference for the published genomic sequence generally and a reference for the location of the packaging sequence within the genome.
  • Packaging sequence ( ) 150 base pairs between 300 and 600 bases from the left (gag-pol) end of the provirus. P. Shank and M. Linial, J. Virol. 36(2) :450-456 (1980).
  • Genome Couez, et al. J. Virol. 49:615-620, 1984, bases 1-341; Rice, et al. Virology 142:357-377, 1985, bases 1-4680; Sagata, et al. Proc. Natl. Acad. Sci. 82:677-681, 1985, complete BLV provirus.
  • Packaging sequence the present inventors predict that
  • SUBSTITUTESHEET it lies between the end of the primer binding site at about base 340 and the initiation codon for gag at about base 41-8.
  • SUBSTITUTESHEET transcript (micRNA) . Proc. Natl. Acad. Sci. USA 81:1966-1970.

Abstract

Cette invention concerne des molécules d'acide nucléique non codantes qui ne s'hybrident pratiquement que sur la séquence d'encapsidation d'un retrovirus et qui inhibent par conséquent l'encapsidation de l'ARN du génome du rétrovirus. Les molecules non codantes peuvent être de l'ADN, de l'ARN, ou des analogues de ces derniers. On peut administrer les molécules non codantes sous forme médicamenteuse ou bien l'individu devant être protégé peut être soumis à des manipulations génétiques afin d'exprimer un ARN non codant.
PCT/US1992/002911 1991-04-05 1992-04-03 Inhibition d'un retrovirus a l'aide d'acides nucleiques non codants s'hybridant sur des sequences d'encapsidation WO1992017211A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP4510048A JPH06506599A (ja) 1991-04-05 1992-04-03 パッケージング配列に相補的なアンチセンス核酸によるレトロウイルスの抑制
AU17780/92A AU655279B2 (en) 1991-04-05 1992-04-03 Retrovirus inhibition with antisense nucleic acids complementary to packaging sequences

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68087391A 1991-04-05 1991-04-05
US680,873 1991-04-05

Publications (1)

Publication Number Publication Date
WO1992017211A1 true WO1992017211A1 (fr) 1992-10-15

Family

ID=24732881

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1992/002911 WO1992017211A1 (fr) 1991-04-05 1992-04-03 Inhibition d'un retrovirus a l'aide d'acides nucleiques non codants s'hybridant sur des sequences d'encapsidation

Country Status (5)

Country Link
EP (1) EP0578776A4 (fr)
JP (1) JPH06506599A (fr)
AU (1) AU655279B2 (fr)
CA (1) CA2107789A1 (fr)
WO (1) WO1992017211A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4225094A1 (de) * 1992-04-28 1993-11-04 Frank Andreas Harald Meyer Medikament zur gentherapeutischen behandlung von menschen, tieren und pflanzen, insbesondere zur blockierung der virenvermehrung und der tumorgene sowie verfahren zur herstellung des medikaments
EP0594881A1 (fr) * 1991-08-10 1994-05-04 Bayer Ag Vecteurs d'expression et leur utilisation pour la production de cellules humaines résistantes au HIV pour les applications thérapeutiques
EP0598935A1 (fr) * 1992-11-24 1994-06-01 Bayer Ag Vecteurs d'expression et leur utilisation pour la production de cellules humaines résistantes à HIV pour utilisation thérapeutique
FR2702152A1 (fr) * 1993-03-03 1994-09-09 Inst Nat Sante Rech Med Virus recombinants et leur utilisation en thérapie génique.
WO1995018854A1 (fr) * 1994-01-05 1995-07-13 Gene Shears Pty., Ltd. Ribozymes ciblant les produits de recombinaison d'expression de sequences d'encapsidation retrovirales et retrovirus recombines contenant lesdits produits de recombinaison
WO1995027783A1 (fr) * 1994-04-06 1995-10-19 Joshi Sukhwal Sadna Inhibition de la multiplication du vih-1 dans des cellules de mammiferes
WO1996022368A1 (fr) * 1995-01-18 1996-07-25 Gene Shears Pty. Ltd. Ribozymes de recombinaison cibles contre des sequences exprimant la capside retrovirale et retrovirus contenant de tels produits recombines
US5583035A (en) * 1992-12-07 1996-12-10 Bayer Aktiengesellschaft HIV antisense expression vectors
US5712384A (en) * 1994-01-05 1998-01-27 Gene Shears Pty Ltd. Ribozymes targeting retroviral packaging sequence expression constructs and recombinant retroviruses containing such constructs
US6458529B1 (en) 1992-09-25 2002-10-01 Aventis Pharma S.A. Assays for promoter operability in central nervous system cells
US7345025B2 (en) 2001-07-10 2008-03-18 Johnson & Johnson Research Pty. Limited Methods for genetic modification of hematopoietic progenitor cells and uses of the modified cells
KR100696410B1 (ko) * 1994-01-05 2008-11-07 진 쉬어즈 피티와이., 엘티디. 레트로바이러스 패키징 서열 발현 제작물을 표적으로 하는 리보자임 및 그러한 제작물을 가진 재조합 레트로바이러스
US7994144B2 (en) 2001-07-10 2011-08-09 Johnson & Johnson Research Pty, Limited Process for the preparation of a composition of genetically modified hematopoietic progenitor cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4757055A (en) * 1980-11-12 1988-07-12 The Johns Hopkins University Method for selectively controlling unwanted expression or function of foreign nucleic acids in animal or mammalian cells

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4740463A (en) * 1984-04-13 1988-04-26 Massachusetts Institute Of Technology Methods and artificial genes for antagonizing the function of an oncogene
ATE118822T1 (de) * 1987-06-26 1995-03-15 Syntro Corp Rekombinanter, fremde gene enthaltender menschlicher cytomegalovirus und seine verwendung.
EP0331939B1 (fr) * 1988-02-16 2001-11-14 Greatbatch Gen-Aid, Ltd Cellules modifiées résistantes aux infections rétrovirales
DE68925278T2 (de) * 1988-02-26 1996-09-19 Worcester Found Ex Biology Hemmung von htlv-iii durch exogene oligonukleotide

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4757055A (en) * 1980-11-12 1988-07-12 The Johns Hopkins University Method for selectively controlling unwanted expression or function of foreign nucleic acids in animal or mammalian cells

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Journal of Virology, Volume 61, No. 5, issued May 1987, BENDER et al., "Evidence that the Packaging Signal of Moloney Murine Leukemia Virus Extends into the gag Region", pages 1639-1646, see the entire document. *
Journal of Virology, Volume 61, No. 9, issued September 1987, EMBRETSON et al., "Lack of Competition Results in Efficient Packaging of Heterologous Murine Retroviral RNAs and Reticuloendotheliosis Virus Encapsidation-Minus RNAs by the Reticuloendotheliosis Virus Helper Cell Line", pages 2675-2683, see the entire *
Journal of Virology, Volume 63, No. 2, issued February 1989, VON RUDEN et al., "Inhibition of Human T-Cell Leukemia Virus Type I Replication in Primary Human T Cells that Express Antisense RNA", pages 677-682, see the entire document. *
Journal of Virology, Volume 63, No. 9, issued September 1989, LEVER et al., "Identification of a Sequence Required for Efficient Packaging of Human Immunodeficiency Virus Type 1 RNA into Virions", pages 4085-4087, see the entire document. *
Proc. Natl. Acad. Sci. USA, Volume 84, issued August 1987, LEDLEY et al., "Retroviral gene transfer into primary hepatocytes: Implications for genetic therapy of liver-specific functions", pages 5335-5339, see the entire document. *
Proc. Natl. Acad. Sci. USA, Volume 85, issued August 1988, GOODSCHILD et al., "Inhibition of Human Immunodeficiency Virus replication by antisense oligodeoxynucleotides", pages 5507-5511, see the entire document. *
Science, Volume 240, issued 10 June 1988, JAENISCH, "Transgenic Animals", pages 1468-1474, see the entire document. *
See also references of EP0578776A4 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0594881A1 (fr) * 1991-08-10 1994-05-04 Bayer Ag Vecteurs d'expression et leur utilisation pour la production de cellules humaines résistantes au HIV pour les applications thérapeutiques
DE4225094A1 (de) * 1992-04-28 1993-11-04 Frank Andreas Harald Meyer Medikament zur gentherapeutischen behandlung von menschen, tieren und pflanzen, insbesondere zur blockierung der virenvermehrung und der tumorgene sowie verfahren zur herstellung des medikaments
US6458529B1 (en) 1992-09-25 2002-10-01 Aventis Pharma S.A. Assays for promoter operability in central nervous system cells
EP0598935A1 (fr) * 1992-11-24 1994-06-01 Bayer Ag Vecteurs d'expression et leur utilisation pour la production de cellules humaines résistantes à HIV pour utilisation thérapeutique
US5583035A (en) * 1992-12-07 1996-12-10 Bayer Aktiengesellschaft HIV antisense expression vectors
WO1994020146A1 (fr) * 1993-03-03 1994-09-15 Rhone-Poulenc Rorer S.A. Adenovirus recombinants et leur utilisation en therapie genique pour le traitement des pathologies oculaires
FR2702152A1 (fr) * 1993-03-03 1994-09-09 Inst Nat Sante Rech Med Virus recombinants et leur utilisation en thérapie génique.
WO1995018854A1 (fr) * 1994-01-05 1995-07-13 Gene Shears Pty., Ltd. Ribozymes ciblant les produits de recombinaison d'expression de sequences d'encapsidation retrovirales et retrovirus recombines contenant lesdits produits de recombinaison
US5712384A (en) * 1994-01-05 1998-01-27 Gene Shears Pty Ltd. Ribozymes targeting retroviral packaging sequence expression constructs and recombinant retroviruses containing such constructs
US6114167A (en) * 1994-01-05 2000-09-05 Gene Shears Pty., Ltd. Ribozymes targeting the MoMLV PSI packaging sequence and the HIV tat sequence
KR100696410B1 (ko) * 1994-01-05 2008-11-07 진 쉬어즈 피티와이., 엘티디. 레트로바이러스 패키징 서열 발현 제작물을 표적으로 하는 리보자임 및 그러한 제작물을 가진 재조합 레트로바이러스
WO1995027783A1 (fr) * 1994-04-06 1995-10-19 Joshi Sukhwal Sadna Inhibition de la multiplication du vih-1 dans des cellules de mammiferes
WO1996022368A1 (fr) * 1995-01-18 1996-07-25 Gene Shears Pty. Ltd. Ribozymes de recombinaison cibles contre des sequences exprimant la capside retrovirale et retrovirus contenant de tels produits recombines
AU703964B2 (en) * 1995-01-18 1999-04-01 Gene Shears Pty. Limited Ribozymes targeting the retroviral packaging sequence expression constructs and recombinant retroviruses containing such constructs
US7345025B2 (en) 2001-07-10 2008-03-18 Johnson & Johnson Research Pty. Limited Methods for genetic modification of hematopoietic progenitor cells and uses of the modified cells
US7776595B2 (en) 2001-07-10 2010-08-17 Johnson & Johnson Research Pty, Limited Methods for genetic modification of hematopoietic progenitor cells and uses of the modified cells
US7994144B2 (en) 2001-07-10 2011-08-09 Johnson & Johnson Research Pty, Limited Process for the preparation of a composition of genetically modified hematopoietic progenitor cells

Also Published As

Publication number Publication date
JPH06506599A (ja) 1994-07-28
AU1778092A (en) 1992-11-02
AU655279B2 (en) 1994-12-15
CA2107789A1 (fr) 1992-10-06
EP0578776A4 (fr) 1995-04-12
EP0578776A1 (fr) 1994-01-19

Similar Documents

Publication Publication Date Title
EP0702084B1 (fr) Rétrovirus recombinants
AU699706B2 (en) Improved vectors for gene therapy
Han et al. Inhibition of Moloney murine leukemia virus-induced leukemia in transgenic mice expressing antisense RNA complementary to the retroviral packaging sequences.
WO1990013641A1 (fr) Cellules eucaryotes transformees de maniere stable comprenant un adn etranger susceptible d'etre transcrit sous la direction d'un promoteur pol iii
AU655279B2 (en) Retrovirus inhibition with antisense nucleic acids complementary to packaging sequences
JP2004147659A (ja) テナガザル白血病ウイルスベースのレトロウイルスベクター
JP2001501441A (ja) 複製能力ウイルスの産生を阻害するための方法および組成物
AU696399B2 (en) Delivery system controlled through factors associated with HIV and cell
AU693317B2 (en) Retroviral vectors for expression in embryonic cells
CA2099268C (fr) Fusions d'une proteine virale et d'un enzyme destructeur
AU697095B2 (en) Expression of viral decoy proteins under the control of a locus control region and uses thereof
EP0986649A1 (fr) Particules de vecteur de retrovirus produites dans un systeme d'expression de baculovirus
Kaptein et al. Expression pattern of CD2 locus control region containing retroviral vectors in hemopoietic cells in vitro and in vivo
JP2003501070A (ja) 阻害性RNA分子を決定するためのinvivoでの選択方法
Hodgson et al. Overview: Retroviral Vectors for Gene Therapy and Transgenics
Lang et al. Development of transforming function during transduction of proto-ras into Harvey sarcoma virus.
Matesanz et al. In-vivo and in-vitro analysis of retroviral vectors carrying the N-ras oncogene
Zhang et al. MOLECULAR TARGETING OF CANCER: RETROVIRAL VECTOR-MEDIATED ANTISENSE NUCLEIC ACID THERAPY
Majumdar The Soybean Retroelement, Sire-1, Encodes an Envelope-Like Protein-Sire-1 is an Endogenous, Proretrovirus-Like Genomic Element

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP KR

AL Designated countries for regional patents

Kind code of ref document: A1

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1992910993

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2107789

Country of ref document: CA

WWP Wipo information: published in national office

Ref document number: 1992910993

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1992910993

Country of ref document: EP