CA2175703C - Alteration of sequence of a target molecule - Google Patents

Alteration of sequence of a target molecule Download PDF

Info

Publication number
CA2175703C
CA2175703C CA002175703A CA2175703A CA2175703C CA 2175703 C CA2175703 C CA 2175703C CA 002175703 A CA002175703 A CA 002175703A CA 2175703 A CA2175703 A CA 2175703A CA 2175703 C CA2175703 C CA 2175703C
Authority
CA
Canada
Prior art keywords
nucleic acid
acid molecule
rna
splicing
ribozyme
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA002175703A
Other languages
French (fr)
Other versions
CA2175703A1 (en
Inventor
Bruce A. Sullenger
Thomas R. Cech
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sirna Therapeutics Inc
Original Assignee
Ribozyme Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/152,450 external-priority patent/US5667969A/en
Application filed by Ribozyme Pharmaceuticals Inc filed Critical Ribozyme Pharmaceuticals Inc
Publication of CA2175703A1 publication Critical patent/CA2175703A1/en
Application granted granted Critical
Publication of CA2175703C publication Critical patent/CA2175703C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2310/124Type of nucleic acid catalytic nucleic acids, e.g. ribozymes based on group I or II introns
    • C12N2310/1241Tetrahymena

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Saccharide Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Method for splicing a target nucleic acid molecule with a separate nucleic acid molecule. Such splicing gen- erally causes production of a chimeric protein with ad- vantageous features over that protein naturally produced from the target nucleic acid prior to splicing. The method includes contacting the target nucleic acid molecule with a catalytic nucleic acid molecule including the separate nucleic acid molecule. Such contacting is performed un- der conditions in which at least a portion of the separate nucleic acid molecule is spliced with at least a portion of the target nucleic acid molecule to form a chimeric nu- cleic acid molecule. In this method, the catalytic nucleic molecule is chosen so that it is not naturally associated with the separate nucleic acid molecule.

Description

DESCRIPTION
Alteration of Seauence of a Target Molecule This invention relates to therapy of diseases using ribozymea.
The following is a brief history of the discovery and activity of enzymatic RNA molecules or ribozymes. This history is not meant to be complete but is provided only for understanding of the invention that follows. This summary is not an admission that all of the work described below is prior art to the claimed invention.
Prior to the 1970s it was thought that all genes were direct linear representations of the proteins that they encoded. This simplistic view implied that all genes were like ticker tape messages, with each triplet of DNA
"letters" representing one protein "word" in the transla tion. Protein synthesis occurred by first transcribing a gene from DNA into RNA (letter for letter) and then trans-lating the RNA into protein (three letters at a time). In the mid 1970s it was discovered that some genes were not exact, linear representations of the proteins that they encode. These genes were found to contain interruptions in the coding- sequence which were removed from, or "spliced out" of, the RNA before it became translated into protein. These interruptions in the coding sequence were given the name of intervening sequences (or introns) and the process of removing them from the RNA was termed splicing.--A general reference for spliceosomes and how they are- related to self-splicing introns is Guthrie, C., 253 Scier~ce 157, 1991. After the discovery of introns, two questions immediately arose: (i) why are introns present in genes in the first place, and (ii) how do they get removed from the RNA prior to protein synthesis? The first question is still being debated, with no clear answer yet available. The second question, how introns get removed from the RNA, is much better understood after a decade and a half of intense research on this question.
At least three different mechanisms have been discovered for removing-introns from RNA. Two of these splicing mechanisms involve the binding of multiple protein factors which then act to correctly cut and join the RNA. A third ' F
mechanism involves cutting-and joining of the RNA by the intron itself, in what was the first discovery of-catalyt-ic RNA molecules.
Cech and colleagues were trying to understand how RNA
splicing was accomplished in a single-celled pond organism called _T~trahvmena thermoDhs~a They had chosen Tetrahvmena thermobhila as a matter of convenience, since each individual cell contains over 10,000 copies of one intron-containing gene (the gene for ribosomal RNA). They reasoned that such a large number of intron-containing RNA
molecules would require a large amount of (protein) splic-ing factors to get the introns removed quickly. Their goal was to purify these hypothesized splicing factors and to demonstrate that the purified factors could splice the intron-containing RNA,i~ vi r . Cech rapidly succeeded in getting RNA splicing to work in vitro, but something funny was going on.- -As expected, splicing occurred when the intron-containing RNA was mixed with protein-containing extracts from Tetrahymena, but splicing also occurred when the protein extracts were left out. Cech.proved that the intervening sequence RNA was acting as its own splicing factor to snip itself out of the surrounding RNA. They published this startling discovery in 1982. Continuing studies in the early 1980's served to elucidate the complicated structure of the Tetrahvmena intron and to decipher the mechanism by which self-splicing occurs. -Many research groups helped to demonstrate that the specific folding of the Tetrahymena_intron is critical for bringing-together the parts of the RNA that will be cut and spliced. Even after- splicing is complete, the released intron maintains its catalytic structure. As a consequence, the released intron is capable of carrying 217~7~3 i out additional cleavage and splicing reactions on itself (to form intron circles). By 1986, Cech was able to show that a shortened form of the Tetrahvmena intron could carry out a variety of cutting and joining reactions on other pieces of RNA. The demonstration proved that the Tetrahvmena intron canact as_a true enzyme,: (i) each intron molecule was able to cut many substrate molecules while the intron molecule remained unchanged, and (ii) reactions were specific for RNA molecules that contained a unique sequence (CUCU) which allowed the intron to recognize and bind the RNA. Zaug and Cech coined the term "ribozyme" to describe any ribonucleic acid molecule that hae enzyme-like properties. Also in 1986, Cech showed that theRNA substrate sequence recognized by the Tetrahymena ribozyme could be changed by altering a sequence within the ribozyme itself. This property has led to the development of a number of site-specific ribozymes that have been individually designed to cleave at other RNA sequences. The Tetrahvmeria intron is the most well-studied of what is now recognized as a large class of introns, Group I -introns. The overall folded structure, including several sequence elements, is conserved among the Group I_ introns, as is the general mechanism of splicing. Like the Tetrahvmena intron, some members of this class are catalytic, i.e.. the intron itself is capable of the self-splicing reaction. Other Group I introns require additional (protein) factors, presumably to help the intron fold into and/or maintain its active structure. While the Tetrahvmena intron is relatively large, (413 nucleotides) a shortened form of at least one other catalytic intron (SunY intron of phage T4, 180 nucleotides) may prove advantageous not only because . of its smaller-size but because it undergoes self-splicing at an even faster rate than the Tetrahvmena intron.

Ribonuclease P (RNAseP) is an enzyme comprised of both RNA and protein components which are responsible for con-verting precursor tRNA molecules into their final form by 2~ 7 5~ ~3 trimming extra RNA off one of their ends. RNAseP activity has been found in all organisms tested, but the bacterial enzymes have been the most studied. The function of RNAseP has been studied since the mid-1970s by many labs.
In the late 1970x, Sidney Altman and his colleagues showed that the RNA component of _RNAaeP is essential for its processing activity; however, they also showed that the protein component also was required for processing under their experimental conditions. After Cech's discovery of self-splicing by the Tetrahvmena intron, the requirement for both protein and RNA components in RNAseP was reex-amined. In 1983, Altman and Pace showed that the RNA was the enzymatic component of- the RNAaeP complex. This demonstrated that an RNA molecule was capable of acting as a true enzyme, processing numerous tRNA molecules without itself undergoing any change. The folded structure of RNAseP RNA has been determined, and while the sequence is not strictly conserved between RNAs from different organ-isms, this higher order structure is. It is thought that the protein component of the BNAseP complex may serve to stabilize the folded RNA in 'vo. At.least one RNA posi-tion important both to substrate recognition arid to determination of the cleavage site has been identified, however little else is known about the active site.
Because tRNA sequence recognition is minimal, it is clear that some aspects) of the tRNA structure must also be involved in substrate recognition and cleavage activity.
The size of RNAseP RNA (>350 nucleotides), and the com-plexity of the substrate recognition, may limit the potential for the use of an RNAseP-like RNA in thera-peutics. However, the size--of RNAaeP is being trimmed ' down (a molecule of only 290 nucleotides functions reasonably well). In addition, substrate recognition has been simplified by the recent discovery that RNAseP RNA
can cleave small RNAs lacking the natural tRNA secondary structure if an additional RNA (containing a "guide's sequence and a sequence element naturally present at the end of all tRNAs) is present-as well.

Symons and colleagues identified two examples of a self-cleaving RNA that differed from other forms of 5 catalytic RNA already reported- Symons was studying the propagation of the avocado sunblotch viroid (ASV), an RNA

virus that infects avocado plants. Symons demonstrated that as little- as 55 nucleotides of the ASV RNA was capable of folding in such a way as to cut itself into two pieces. It is thought that j,~ vivo self-cleavage of these RNAs is responsible- for cutting the RNA into single a=nome-length pieces during viral propagation. Symons discovered- that variations on the minimal catalytic sequence from ASV could be found in a number of other plant pathogenic RNAa as well. Comparison of these sequences revealed a common structural design consisting of three. stems and loops connected by central loop con-taining many conserved (invariant from one RNA to the next) nucleotides. The predict-~d secondary structure for this catalytic RNA reminded the researchers of the head of-a hammer; thus it was named as such. Uhl: peck was successful in separating the catalytic region of the ribozyme from that of the substrate. Thus, it became possible to assemble a hammerhead ribozyme from 2 (or 3) small synthetic RNAs. A-19-nucleotide catalytic region and a 24-nucleotide -substrate were sufficient to support specific cleavage. The catalytic domain of numerous hammerhead ribozymes have now been studied by both the Uhlenbeck and Symons groups with regard to defining the nucleoides required for specific assembly and catalytic activity and determining the rates of cleavage under various conditions.

Haseloff and Gerlach showed it was possible to divide the domains of the hammerhead ribozyme in a different manner. By doing so, they placed most of the required sequences in the strand that didn't get cut (the ribozyme) and only a required UH where H = C, A, or B in the strand 21 ~ 5103 that did get cut (the substrate). This resulted in a catalytic ribozyme that could be designed to cleave any UH
RNA sequence embedded within a longer "substrate recogni-tion" sequence. The specific cleavage of a long mRNA, in a predictable manner using several such hammerhead ribozymes, was reported in 1988.
One plant pathogen RNA (from the negative strand of the tobacco ringspot virus) undergoes self-cleavage but cannot be folded into the consensus hammerhead structure described above. Bruening and colleagues have indepen-dently identified a 50-nucleotide catalytic domain for this RNA. In 1990, Hampel and Tritz succeeded in dividing the catalytic domain into--two partsthat could act as substrate and ribozyme in a-multiple-turnover, cutting reaction. As with the hammerhead ribozyme, the hairpin catalytic portion contains most of the sequences required for catalytic activity while only a short.sequence (GUC in this case) is required in the target. Hampel and Tritz described the folded structure of this RNA as consisting of a single hairpin and coined the term "hairpin" ribozyme (Bruening and colleagues use the term "paper clip" for this ribozyme motif). -Continuing experiments suggest an increasing number of similarities between the hairpin and hammerhead ribozymes in respect to both binding of target.
RNA and mechanism of cleavage. At the same time, the minimal size of the hairpin ribozyme is still 50-60%
larger than the minimal hammerhead ribozyme.
Hepatitis Delta Virus (HDV) is a virus whose genome consists of single-stranded RNA. A small region (-.80 nucleotides) in both the genomic RNA, and in the comple mentary anti-genomic RNA, is sufficient to support self-cleavage. As the most recently discovered ribozyme, HDV's ability to self-cleave has only been studied for a few years, but is interesting because of its connection to a human disease. In 1991, Been and Perrotta proposed a secondary structure for the I3DV RNAs that is conserved betweenthe genomic and anti-genomic RNAa and is necessary W O 95113379 ~ , ~ J ~ ~ ~ PCTIUS94112976 for catalytic activity. Separation of the HDV RNA into "ribozyme" and "substrate" portions has recently been achieved by Been, but the rules for targeting different substrate RNAs have not yet been determined fully. Been has also succeeded in reducing the size of the HDV
ribozyme to -.60 nucleotides.
The table below lists some of the characteristics of the ribozymes discussed above:

rh-,-acteristics of ribozvmes Group I Introns Size: -300 to >1000 nucleotides.
Requires a U in the target sequence immediately 5' of the cleavage site.
Binds 4-6 nucleotides at 5' side of cleavage site.
Over 75 known members of this class. Found in Tetrahvmena thermobhila rRNA, fungal mitochondria, chloroplasts, phage T4, blue-green algae, and others.
RNAseP RNA (M1 RNA) Size: -290 to 400 nucleotides.
RNA portion of a ribonucleoprotein enzyme. Cleaves tRNA
precursors to form mature tRNA.
Roughly 10 known members of this group all are bacterial in origin.
Hammerhead R~bozvme ~ Size: ~30 to 40 nucleotides.
Requires the target sequence UH immediately 5' of the ~ cleavage site.
Binds a variable number nucleotides on both sides of the cleavage site.
14 known members of this class. Found in a number of plant pathogens (virusoids) that use RNA as the infectious agent.

R'O 95/13379 1151 ~3 Fia~ Yp~ n R~ bozvm Size: -50 nucleotides.
Requires the target sequence GUC immediately 3' of the cleavage site.
Binds 4 nucleotides at 5' side of the cleavage site and a variable number to the 3' side of the cleavage site.
Only 1 known member of this class. Found in one plant pathogen (satellite RNA of- thetobacco ringspot virus) which uses RNA as the infectious agent.
Hepatit;a Dei a Vim (HDTT1 17; hnv _ Size: -60 nucleotides (at present).
Cleavage of target RNAs recently demonstrated.
Sequence requirements not fully determined.
Binding sites and structural requirements not -fully determined, although no sequences 5' of cleavage site are required.
Only 1 known member of this class. Found in human HDV.
As the term is used in this application, ribozymes are RNA molecules having an enzymatic activity which is able to cleave and splice other separate RNA mol-ecules in a nucleotide base sequence specific manner. Such enzymatic RNA molecules can be targeted- to virtually any RNA
transcript, and efficient cleavage and splicing achieved .~ vitro. Kim et al., 84 Proc. Nat Pcad of ~r.; rrer 8788, 1987, Hazeloff et al., 234 Na ,r 585, 1988, Cech, 260 AJ N~ 3030, 1988, and Jefferies et al., I7 Nu ~ ; p ;d Research 1371, 1989.
Ribozymes act by first binding to a target RNA. Such binding occurs through the target RNA binding portion of a ribozyme which is held in close proximity to an enzymatic portion of the RNA which acts to cleave the target RNA. Thus, the ribozyme first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts to cut and splice the target RNA. Strategic cleavage and splicing of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After a ribozyme has bound, cleaved and spliced its RNA target it is released from that RNA.
By the phrase "catalytic" or "enzymatic RNA molecule"
is meant an RNA molecule which has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity wh:~~h is active to .specifically cleave and splice RNA in shat target. That is, ._~e. $.nzymaGic-RNA-mo~.ecule-- is-- able to iritermolecularly cleave and splice RNA and thereby alter a target RNA
molecule. This complementarity functions to allow sufficient hybridization of the enzymatic RNA molecule to the target RNA to allow the cleavage to occur. 100%
complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention..
In preferred embodiments of this invention, the enzymatic RNA molecule is formed in a hammerhead motif, but may also be formed in the motif of a hairpin, hepatitis delta virus, group I intron or RNAsE~P RNA (in association with an RNA guide s,equence). Examples of such hammerhead motifs are described by Rossi et al., 8 AIDS
RESEARCH AND HUMAN RETROVIRUSES 183, 1992, Hampel and Tritz, 28 Biochemistrv 4929, 1989 and Hampel et 'al. , 18 Nucl,e~c Acids Research 299, 1990, and an example of the hepatitis delta virus motif is described by Perrotta and Been, 31 Biochemistry 16, 1992, of the RNAseP motif by Guerrier-Takada et al., Cell 849, 1983, and of the group I intron by Cech et al., U.S. Patent 4,987,071. These specific motifs are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic 35 RNA molecule of this invention is that it has a specific substrate binding site which is complementary to one or more of the target gene RNA regions, and that it have a C ~ ~ '~ Q3 1~
nucleotide sequences within or-surrounding that substrate binding site which impart an RNA cleaving activity to the molecule.
The invention provides a method for designing a class _ of enzymatic cleaving and splicing agents which exhibit a high degree of specificity for the RNA of a desired target. The ribozyme molecule is preferably targeted to a highly conserved sequence region of a-target such that specific treatment of a disease or condition can be provided with a single ribozyme. Such enzymatic RNA
molecules can be delivered exogenously to specific cells as required.
Synthesis of ribozymes greater than 100 nucleotides in length is very difficult using automated-methods, and the therapeutic cost of - such molecules is prohibitive.
However, delivery of such ribozymes by expression vectors is primarily feasible using-~x v'vo treatments.
moue et al., 43 Cell -431, 1985, state that short oligonucleotides of 2-6 nucleotides can undergo inter molecular exon ligation or splicing in traps, It indi cates that "long 5' exons should be reactive provided that three conditions are met: the exonmust have-a 3' hydroxyl group, it must terminate in a sequence similar to that of the 3' -end of the 5' exon, and the 3' terminal sequence must be available as opposed to being tied up in some secondary structure. Thus, it appears that exon switching is possible in this system, though limited by the availability of alternative 5' exons that meet the above criteria.- These could-include transcripts that are not 5' exons from other precursors, since RNA polymerases always leave 3' hydroxyl ends". .
~ummarv of the Invention This invention features a method in which natural transcripts are altered by use of a splicing reaction ~
v'vo or ~ v' ro. It involves the manipulation of genetic information to ensure that a useful transcript is provided within a cellular system or extract.
In a first aspect, the invention features a method for splicing a target nucleic acid molecule with a separate nucleic acid molecule. Such splicing-generally causes production of a chimerie protein with advantageous features over that protein naturally produced from the target nucleic acid prior 'to splicing. The method includes contacting the target nu._-cleic. acid mole~u~.e -w-ith a catalytic nucleic acid molecule including the separate nucleic acid molecule. Such contacting is performed under conditions in which at least a portion of the separate nucleic acid molecule is spliced with at least a portion of the target nucleic acid molecule to form a chimeric nucleic acid molecule. In this method, the catalytic nucleic acid molecule is chosen so that it is not naturally associated with the separate nucleic acid molecule.
The target nucleic acid molecule can be~any desired molecule with which a splicing reaction can occur.
Generally, this will be an RNA molecule, preferably a messenger RNA molecule, but it may also include molecules that have one or more non-ribonucleotides substituents, such as deoxyribonucleotides or other analogs as described by Eckstein et al. EP90j01731.
Generally, the target nucleic acid molecule is present within a cell and is chosen or targeted because it encodes a defective protein or is deleterious to that cell.
Splicing of the separate nucleic acid molecule with such a target nucleic acid molecule is designed to alter the protein product of that nucleic acid molecule. Such alteration causes production of a useful protein which will allow that cell to either survive or die, as desired.
Thus, for example, in a gene therapy setting, the target nucleic acid molecule may encode a non-functional protein necessary for normal life. This molecule can be spliced 2~~~~03 i with a separate nucleic acid molecule to allow appropriate expression of a functional protein. Alternatively, the splicing may cause production of a more stable protein, or of a protein which acts as an agonist or antagonist of a function, e-g., a viral ,or bacterial replication function.
The separate nucleic acid molecule is generally chosen such that it encodes a 3' exon which it is desirable to express within a cell. This exon will generally not include control sequences such as promoter regions, but may include poly(A) tails and other stabilizing or enhancing functions well known in the art. As with the target nucleic acid molecule, the separate nucleic acid molecule generally is a ribonucleic acid molecule but may be substituted as described above.
By "enzymatic" or "catalytic nucleic acid molecule" is meant a molecule having a motif generally as described above in the Background of the Invention, which and is preferably selected from the moti~ of a group I or group II intron having a cleavage and splicing activity.
Alternatively, the splicing or cleavage activity may be provided by a different nucleic-acid molecule, or may supplement the catalytic nucleic acid molecule. Those of ordinary skill in the art will recognize that other motifs than those of the group I and group II introns may also be manipulated to provide useful splicing activity.
The conditions chosen for the contacting step may be those naturally occurring within a cell, or may be manipu-lated ~n v'troto ensure that the splicing reaction will occur. These conditions are well known to those in the art, for example, as described by Inoue et al., sur~ra. , By at least a portion of the respective nucleic acid molecules is meant that the 5' end of the target nucleic.
acid molecule will be spliced with the 3' end of the separate nucleic acid molecule. Such a portion may be only a few nucleotides (10-500 nucleotides) or may be significantly greater and may represent almost all of a 1. 3 molecule encoding a gene product (i.e., at least 1 to 5 kbases) .
Tre chimeric nucleic acid molecule is one which may occur naturally in nature but is not present prior to the splicing reaction. Alternatively, it may be a completely novel structure which does not occur in nature, but which is useful in gene therapeutic treatment of an organism.
The catalytic nucleic acid molecule is not naturally assoc3at-ed-w3t~ the separate nucleic acid tttoie~ule s~.nce it is not generally desired to splice the 3' end of a naturally occurring catalytic nucleic acid molecule with a target nucleic acid molecule. Rather, the separate nucleic acid molecule is chosen or selected to have a beneficial function once spliced with the target nucleic molecule.
In a related aspect, the invention features a method f or splicing a target nucleic acid molecule with a separ-ate nucleic acid molecule by contacting those molecules in the presence of one or more splicing factors or 20. spliceosomes under splicing conditions. Such molecules are not naturally spliced together in nature, although the final splice product may be a natural product.
The various splicing factors and spliceosomes are well known in the art, and this activity is generally described by Bruzik and Maniatis in 360 Nature 692, 1:992.
The invention concerns splicing of target nucleic acid molecules and separate nucleic acid molecules which are not normally spliced together within a cell as described by Bruzik and Maniatis, supra. Rather, as described above, a separate nucleic acid molecule is selected such that a useful function can be achieved in a gene therapeutic fashion.
In preferred embodiments, the catalytic nucleic acid is able to cleave and splice, e~ct. , it has a group I or group II intron motif; the method is performed sn vitro or in vivo with an RNA target; and the method can be used to treat g?netic disease in a gene therapy type manner, for 2175?03 example, by correcting an abnormal transcript, or by providing antiviral activity such as a dominant negative allele to a viral RNA.
In other aspects, the invention features catalytic nucleic acid molecules having a selected separate nucleic acid molecule as a 3' exon encoding at least a portion of -a useful gene which can be..used in gene therapy. Such a molecule can be spliced with and thereby correct or modify the expression of- other target RNA molecules. The invention also features vectors encoding such catalytic nucleic acid molecules.
The observation that ribozymes can specifically cleave targeted RNAs in vitro hassled to much speculation about their potential usefulness as gene inhibitors. By cleaving targeted mRNAs ~ ivo, ribozymes can be used to atop the flow of genetic information. Here we describe a different application of ribozymes. For example, a group I intron ribozyme can be used to manipulate the flow of genetic information by targeted trans-splicing. Defective cellular transcripts may be-repaired, or pathogen-derived transcripts may be altered=to encode antagonists to the pathogen using such technology.
In nature the group I intron ribozyme from Tetrahvmena thermophila self-splices itself from precursor ribosomal RNAs (7.R. Cech, A.J. Zaug, P.J. Grabowski, Cell 27 487 (1981); K. Kruger et al., Cell 31, 147 (1982)). This process is accomplished in two successive steps. First the phosphodieater bond at the 5' exon-intron border is cleaved. Then the 3' hydroxyl group on the 5' exon is covalently attached to the3' exon, and the intron is -removed (Fig. 1A). It has been previously demonstrated _in ' vitro that the 5' exon in this reaction-can be mimicked by RNA molecules supplied ~ traps; the minimum active unit is the dinucleotide substrate rCU (Fig. 1B) (T. moue, F.X. Sullivan, T.R. Cech, Cell 43, 431 (1985)). We propose use of-traps-splicing reactions to ligate foreign sequences onto targeted transcripts after cleavage (Fig.

R'O 95143379 PCTfUS94112976 1C). In this manner, ribozymes can be employed to manipulate the flow of genetic information inside cells by changing what a-targeted RNA encodes.
Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims.
nPacrip ion of the Preferred Embodiments The drawings will briefly be described Drawings FIGS. 1A, 1B, and 1C are diagrammatic representations showing reactions of the group I intron from Tetrahymena for targeted trans-splicing.

FIGS. 2A and 2B are a comparison of cis- and trans-splicing reactions for LacZ transcripts.

FIG. 3 is a copy of an autoradiogram showing targeted trans-splicing to correct truncated transcripts from the alpha complement of LacZ 39 nucleotides long. L-21 (or L-21 4e1) ribozyme-3' exon chimeric RNAs (see Fig. 2B) (;2P-body-labeled) (200nM) were preheated in reaction buffer [50mM Hepes (pH 7.0), 150mM NaCl, and 5mM MgCl,] at 50C

for 5 minutes and then equilibrated at 37C for 2 minutes.

The 13 (5'-A5: GGCCCUCUAS) or 39 (5'L-A2: see Fig. 2B) nucleotide substrate RNAs (1~M) and GTP (100~M) were preheated to 37C and added to the ribozymes to start the reactions which proceeded at 37C. Portions containing one fifteenth of the reactions were removed at 0, 2, 10, 60; and 180 minutes and added to an equal volume of lOmM

EDTA to stop the reactions. Reaction products were analyzed upon a 4g polyacrylamide gel with 8M urea. The inactive L-21 del ribozyme was generated by deleting 93 nucleotide of--the ribozyme (nucleotides 237-330 comprising L6b to P9).

FIG. 4 is a graphical representation of the targeted traps-splicing rate for- correcting the 39 nucleotide truncated LacZ transcript. The products from the trans-splicing reaction time course containing the action L-21 ribozyme and the 39 nucleotide substrate shown in figure 2 were quantified with an AMBIS* Image Acquisition and Analysis System (AMBTS, Tnc., San Diego, CA). The percentage of the ribozyme-3' exon RNA remaining is plotted versus time.
FTG. 5 is a copy of an autoradiogram showing hydrolysis of the 3' LaeZ exon attached to the L-21 ribozyme .. L-21 _ (.0r_.. L-_21.. del ) ribQZ~rme_ 3 ~_ _ exon. .chimeric RNAs ('2P-body-labeled) (100nM) wee incubated at 37°C in reaction buffer (50mM Hepes (pH 7.0), 150mM NaCl, 5mM
MgCl2, and 100~.M GTPl . A portion of the reaction was removed after 0, 2, 10, and 60 minutes and added to an equal, volume of lOmM EDTA to stop the reaction. Products were analyzed upon a.4% polyacrylamide gel containing 8M
urea.
FIGS. 6A and 6B are representations of trans-splicing to recreate an entire 3074 nucleotide LacZ messenger RNA
from a 1106 nucleotide truncated trans,cri.pt. A. Scheme for correcting transcript. 8. Trans-splicing reaction.
L-21 (or L-21 del) ribozyme-3' exon chimeric RNAs (20nM) were preheated in reactian buffer (50mM Hepes (pH 7.0) , 150mM NaCl, and 5mM MgClz] at 50°C for 5 minutes and then equilibrated at 37°C for 2 minutes. The 1106 nucleotide substrate RNA ('2P-end labeled) (200nM) and GTP (100uM) were preheated at 37°C and added to the ribozymes to start the reactions which proceeded at 37°C. One sixth of each reaction was removed at 0, 2, 10, 60; and 1B0 minutes and added to an equal volume of lOmM EDTA to stop the reaction. Reaction products were analyzed upon a 1.2%
agarose gel containing 1.1% formaldehyde. rRNAs from mouse NIH 3T3 cells were used to as 5100 and 1900 nt molecular weight markers: The remaining sixth of the reactions, which had proceeded for 120 minutes, were in vitro translated.
FIG. 7 is a scheme for correcting genetic mutations using targeted trans-splicing.
*Trade-mark W095/13379 2 i ~ 5 7 0 3 pCT~s94112976 FIG. S is a scheme for mutating HIV transcripts using targeted trans-splicing.
Taraeted Trans-splicina The general scheme for a targeted trana-splicing is shown in Fig. 1 using ,the group I intron of Tetrahvmena thermophila assn example. Those in theart will recog nize that this example is not limiting in the invention and that other enzymatic RNA molecules having the appro priate splicing activity can be used in the invention.
Alternatively, as discussed above, these molecules can be supplemented by other molecules having a suitable splicing activity, or by spliceosomes or splicing factors.
Generally, the reaction involves base pairing of the catalytic nucleic acid molecule with the targeted transcript, cleavage of the targeted transcript, and then ligation of the 3' exon (separate nucleic acid molecule) with this targeted 5' exon. The catalytic nucleic acid is removed in the reaction. As will be noted, the specificity of-the reaction can be changed by alteration of the substrate binding site in the catalytic nucleic acid molecule by methods well known in the art.
The following is an example of various constructs used to show the operability of the claimed invention. Those in the art will recognize that this example indicates the utility of the invention for both in vitro and in vivo splicing reactions. While significant utility will be attained in vivo by use of the present invention, those in the art will also recognize that in vitro utility is important and can be used to create chimeric transcripts for use in laboratory situations or in a clinical setting.
Example l LaCZ Fusion _ To assess the feasibility of- the targeted trans-splicing approach, we tested the ability of the Tetrahvmena ribozyme to correct truncated LacZ transcripts with targeted trans-splicing. It has previously been shown that in ~. coli the Tetrahvmena self-splicing group WO 95/13379 PCTlU594/12976 2~ ~ ~ 03 I intron can efficiently splice itself from transcripts encoding the alpha-complement of,Q-galactosidase ((3-gal) , (Fig. 2A) (J. V. Price, T.R. Cech, Science 228, 719 (1985);
blaring et al., Cell 40, 371 (1985)). Since this reaction proceeded very efficiently in cis, we decided to determine if the ribozyme could perform a similar reaction in traps.
This system consists oft RNA molecules (Fig. 2B): a ribozyme-3' exon RNA and a 5' exon RNA. The group I ribo-zyme used in this study lacks the first 21 nucleotides present in the full length intron from which it is derived (A.J. Zaug, T.R. Cech, Science 231, 470 (1986)). The first 23 nucleotides of the=3' exon arederived from the pre-rRNA 3' exon sequence from Tetrahymena (M. D. Been, T.R. Cech, Cell 47, 207 (1986)). This 23 nucleotide sequence is fused in-frame to 200 nucleotides of the alpha-complement of the LacZ gene (Been and Cech, supra).
The 39 nucleotide 5' exon =contains a ribosome binding site, the first 21 coding nucleotides of an alpha-complement LacZ transcript, the ribozyme recognition sequence CCCUCU, and two adenosines. These adenosines must be removed if traps-splicing is to correct these LacZ
transcripts. (Fig. 2B). (Previous studies have shown that the sequence and length of the RNA following the CCCUCU is not critical for Tetrahvmena ribozyme action (A.J. Zaug, M.D. Been, T.C. Cech, Nature 324, 429 (1986))1.
In vitro, the ribozyme can quickly and accurately traps-splice this LacZ 3' exon onto the truncated 39 nucleotide LacZ 5' exon to generate an RNA product which encodes the alpha-complement of f~-galactosidase (Fig. 3).
The reaction proceeds with speed and efficiency similar to ' those seen in a reaction with a short 13 nucleotide substrate. The t1~2 for the arans-splicing reaction with the 39 nucleotide substrate was determined to be 13 minutes under conditions of substrate excess (Fig. 4).
In these experiments, traps-splicing (production of 5'-3' or 5'L-3') occurred faster than hydrolysis (produc-tion of free 3' exon;-see Fig. 2). The rate of hydrolysis of the 3' exon from the ribozyme was determined to be tl/z -60 minutes in a separate experiment (Fig. 5). An inactive version of the L-21 ribozyme (L-21 del) was not able to perform either- she traps-splicing or the hydrolysis reaction (Fig. 3 and 5}. Sequencing of the traps-splicing product confirmed that the ultimate and penultimate 3' adenosine nucleotide were correctly removed from the 5' exon-substrate RNA, and this cleaved 5' exon was accurately spliced onto the 3' exon (data not shown). The splice junction gave the proper reading frame for ,Q-gal expression.
Example 2: mRNA Solicins To determine if targeted traps-splicing could be 1.. employed to correct mRNA-size RNA fragments, a transcript which contained the first 1106 nucleotides of the LacZ
coding sequence as well as signals for in vitro translation was created and targeted for alteration by traps-splicing. The L-21 ribozyme was directed to cleave 2D the truncated LacZ transcript 19 nucleotides from its 3' end and traps-splice a 3' exon brought in by the ribozyme onto the cleaved LacZ target RNA (Fig. 6A). The 3' exon sequence attached to the ribozyme encoded the last 1987 nucleotides of the LacZ coding sequence and no sequences 25 from the Tetrahymena pre rRNA. Accurate traps-splicing of the 3' exon sequences onto the truncated transcript resulted in a 3074 nucleotide product which encoded the entire LacZ coding sequence (Fig. 6B).
Once again the inactive version of the ribozyme (L-21 30 del) was unable to perform this reaction, confirming its expected dependence of the catalytic--activity of the RNA
itself. The traps-splicing products from the 120 minute time points of the reactions shown in figure 6 were vitro translated in wheat germ extract, and the in vitro 35 translated proteins were assayed for-;Q-gal activity using R'O 95/13379 PC."f/US94/12976 2~~~~03 a standard ONPG assay (C. Smith et al., Leukemia 7, 310 (1993)).
Proteins from traps-splicing reactions containing active ribozymes were shown.to Contain 1500 units [1000 x 5 OD920/(ml-min)] of (3-gal activity, while no activity was found in proteins translated from reactions containing the inactive ribozyme. Therefore, traps-splicing can be employed to correct the coding sequence of large defective transcripts.
10 In the reaction shown in -figure- 6, the labeled substrate RNA is in a 10 fold excess to the ribozyme-3' exon RNA. Therefore, only 10% of the labeled substrate RNAa could at best be converted to traps-spliced products.
In this reaction however, we roughly estimate (by 15 comparing different X-ray film exposures of the gel) that at most 1% of the truncated RNAs are corrected: This lack of efficiency is probably a result of the targeted RNAs adopting conformations which inhibit the ribozyme from correctly interacting with them. To improve the 20 efficiency of this traps-splicing reaction, alternative sites for cleavage and splicing which are more accessible to the ribosome can be targeted by standard manipulation of this experiment. In vivo, cellular proteins may improve the efficiency of formation of the-correct RNA
interaction (Z. Tsuchihashi-, M. Khosla, D. Herschlag, Science 262, 99 (1993)).
Uses Gene mapping and human genome sequencing provides the genetic basis for an increasing number .of inherited diseases. With each discovery or identification of a new disease-related gene there is an opportunity to develop gene therapy based treatments. Conventional gene therapy approaches attempt to correct a genetic deficiency by transferring a wild-type cDNA copy of a gene under the control of a heterologous promoter to cells harboring a defective copy of the gene. One obstacle for implementing such treatments is an inability to Faithfully recapitulate the normal expression pattern of endogenous genes after gene transfer (R. A. Morgan, W.F. Anderson, Ann. Rev. Bio-chem. 62, 191 (1993); E.A. Dzierzak, T. Papayannopoulou, R.C. Mulligan, Nature 331, 35 (1989)). This may limit the number of genetic diseases treatable by gene therapy.
Targeted trans-splicing offers a solution to this problem.
Ribozymes can be -used to correct the defective transcripts issuing from mutant genes: This approach will be valuable for the treatment of the many genetic diseases caused by a common set of specific mutations which do not affect the expression of the mutant gene. For example, the genetic basis of- many globin diseases is well understood. However, gene therapy based treatments for such diseases have been slow in coming, perhaps, because the expression -patterns of the globin genes cannot be recapitulated after gene transfer: Targeted trans-aplicing can potentially repair or correct globin transcripts that are either truncated or contain pout mutations. In the process, the cellular expression pattern of these genes is maintained (Fig. 7). Therefore, targeted trans-splicing represents an important, novel strategy for the treatment of many genetic diseases.
Trans-splicing ribozymes based on any of the self splicing group I introns can be designed to cleave a targeted transcript upstream of a specific mutation or upstream of a premature 3' end at essentially any uridine residue -(F. L. Murphy, T.R. Cech, Proc. Natl. Acad. Sci, USA 86, 9218 (1989)). One simply changes the sequence of the internal -guide sequence within the ribozyme (5'-GNNNNN) to match the sequence preceding the site of target RNA cleavage (5'-N'N'N'N'N'U), where N-N' represent any allowable base pair. -The 3' exons attached to the ends of these ribozymes are comprised of a sequence designed to correct the mutant transcripts being targeted. The ribo-zyme will both cleave the mutant transcript and replace the mutant 3' region by a functional sequence. There is very little sequence requirement for a 3' exon in these reactions, so virtually any sequence can serve (J. V.
Price, T.R. Cech, Genes and-Development 2, 1439 (1988)).
Thus, traps-splicing ribozymea.can be made to correct es-sentially any mutant transcript because sequence require-ments for 5' cleavage sites and 3' exons are minimal.
Traps-splicing -ribozymes are also be effective antiviral agents. Several groups have employed trana cleaving ribozymes to inhibit viral replication. Use of such ribozymes results in the destruction ofthe targeted viral RNA inside cells (N. Sarver et al., Science 247, 1222 (1990)). Thus, the effectiveness of these trans-cleavage ribozymes rests upon their ability to destroy the vast majority of the targeted viral RNAS. We propose employing traps-splicing ribozymes not to destroy viral RNAs, but to change the sequence of the viral RNAs to give them antiviral activity. For example, the HIV transcripts that encode the aaa protein can be changed to encode a dominant negative version of this protein via targeted traps-splicing (Fig. 8) (M.H. Malim, E. Bohniein, J.
Hauber, B.R. Culien, Cell 58, 205 (1989); D. Trono, M.B.
Feinberg, D. Baltimore, Cell 59, 1I3 (1989)) or to contain a large number of TAR or RRE decoy RNAs (B. A. Sullenger, H.F. Gallardo, G.E. Ungers, E. Gilboa, Cell 63, 601 (1990)).
In contrast to traps-cleaving ribozymes, such antiviral traps-splicing ribozymes would have to affect only a small percentage of the targeted HIV transcripts to be effective at inhibiting viral replication. In general, the ability to change the information encoded by targeted transcripts by traps-splicing represents a broad new approach to gene inhibition-because now transcripts can be altered to encode proteins or RNAs which can inhibit the function of the targeted gene. In other words, with targeted traps-splicing, deleterious transcripts can be turned against themselves.-WO 95113379 ~ ~ 7 J 7 ~ ~ PCTfUS94112976 As noted above, trans-splicing may also be accomplished without the use of ribozymes. It has been demonstrated that spliced leader sequences from lower eucaryotes can be trana-spliced onto mammalian 3' splice sites in tissue culture cells (J. P. Bruzik, T. Maniatis, Nature 360, 692 (1992)). Trans-splicing in this case is mediated by the spliceosome or splicing factors. Thus, it is possible to employ spliceosomes to alter the sequence of targeted transcripts for some desired end via targeted trans-splicing.
Thus, this invention provides a means for performing molecular reconstructive surgery. A defective part of a useful RNA molecule can be cut away from the rest of the -molecule and subsequently replaced by a functional part.
Alternatively, a functional portion of a disease-causing or -deleterious RNA can be replaced by an inhibitory portion.
Administration The above trans-splicing factors or agents can be administered by standard techniques, some of which are discussed below. They may be administered as RNA or expressed from expression vectors. Selected agents, e-a., oligonucleotides or ribozymes can be administered prophylactically, or to patients suffering from a target disease, e-a., by exogenous delivery of the agent to an infected -tissue by means of an appropriate delivery vehicle, e--a., a liposome, a controlled release vehicle, by use of iontophoreais, electroporation or ion paired molecules, or covalently attached adducts, and other pharmacologically approved methods of delivery. Routes of administration include intramuscular, aerosol, oral (tablet or pill form), topical, systemic, ocular, intra peritoneal and/or intrathecal. Expression vectors for immunization with ribozymes and/or delivery of oligo nucleotides are also suitable.

2~~ ~~~3 The specific delivery route of any selected agent will depend on the use of the agent. Generally, a specific delivery program for each agent will focus on naked agent uptake with regard to intracellular localization, followed by demonstration of efficacy. Alternatively, delivery to these same cells in an organor tissue of an animal can be pursued. Uptake studies will include uptake assays to evaluate, e-g., cellular oligonucleotide uptake, regard-less of the delivery vehicle or strategy. Such assays will also determine the intracellular localization of the agent following uptake, ultimately establishing the requirements for maintenance of steady-state concentra-tions within the cellular compartment containing the target sequence (nucleus and/or cytoplasm). Efficacy and cytotoxicity can then be tested. Toxicity will not only include cell viability but also cell function.
Some methods of delivery-that may be used include:
a. encapsulation in liposomes, b. traneduction by retroviral vectors, c. conjugation with cholesterol, d. localization to nuclear compartment utilizing antigen binding site found on most snRNAs, e. neutralization of charge of ribozyme by using nucleotide derivatives, and f. use of blood stem cells to distribute ribozymea throughout the body.
At least three types of delivery strategies are useful in the present invention, including: ribozyme modifications,--particle carrier drug delivery vehicles, and retroviral expression vectors. Unmodified ribozymes and antisenae oligonucleotides, like most small molecules, are taken up by cells, albeit slowly. To enhance cellular uptake, the ribozyme may be modified essentially at random, in ways which reduce its charge but maintain specific functional groups required for RNA cleavage and splicing activity. This results in a molecule which is WO 95/d3379 PCTIU5941d2976 1 2?75703 able to diffuse across the cell membrane, thus removing the permeability barrier.
Modification of ribozymes to reduce charge is just one approach to enhance the cellular uptake of these larger -5 molecules. The random approach, however, is not advisable since- ribozymes are structurally and functionally more complex than small drug molecules. The structural requirements necessary to maintain- ribozyme catalytic activity are well understood by those in the art. (See, 10 Cech, Curr. Op. Structural Biol., 1992) These requirements are taken into consideration when designing modifications to enhance- cellular delivery. The modifications are also designed to reduce susceptibility to nuclease degradation. Both of these characteristics 15 -should greatly improve the efficacy of the ribozyme.
Cellular -uptake can be increased by several orders of magnitude without .having to alter the. phosphodiester linkages necessary for ribozyme cleavage activity.
Chemical modifications of the phosphate backbone will 20 reduce the negative charge thereby facilitating diffusion across the membrane. This principle has been successfully demonstrated for antisenae DNA technology. The similarities in chemical composition between DNA and RNA
make this a feasible approach. In the body, maintenance 25 of an external concentration will be necessary to drive the diffusion of the modified ribozyme into the cells of the tissue. Administration routes which allow the diseased tissue to be exposed to a transient high concentration of the drug, which is slowly dissipated by systemic adsorption are preferred. Intravenous adminis-tration with a drug carrier designed to increase the circulation half-life of the ribozyme can be used. The size and composition of the drug carrier restricts rapid clearance from the blood stream. The carrier, made to accumulate at -the site of infection, can protect the ribozyme from degradative processes.

2~-~ 5~ 03 Drug delivery vehicles are effective for both systemic and topical administration. They can be designed to serve as a slow release reservoir, onto deliver their contents directly to the. target cell. ,Aw advantage o~-using direct delivery drug vehicles is..that multiple molecules are delivered per uptake. Such vehicles have been shown to increase thecirculation half-life of drugs which would otherwise be rapidly cleared from the blood stream. Some examples ofsuch specialized drug delivery vehicles which fall into this category are liposomes, hydrogels, cyclo-dextrins, biodegradable nanocapsules, and bioadhesive microspheres.
From this category of delivery systems, liposomes are preferred. Liposomes -increase .intracellular stability, increase uptake efficiency and improve biological activity. Liposomes are hollow spherical vesicles composed of lipids arranged in a, similar fashion as those lipids which make up the cell membrane. They have an internal aqueous space for entrapping water soluble compounds and range in size from 0.05 to several microns in diameter. Several studies have shown that liposomes can deliver RNA to cells and that the RNA remains biologically active.
For example, a liposome delivery vehicle originally designed-as a research tool, Lipofectin, has been shown to deliver intact mRNA molecules to cells yielding production of the corresponding protein.
Liposomes offer several advantages: They are non- -toxic and biodegradable in composition; they display long circulation half-lives; and-recognition molecules can be readily attached to theirsurface. for targeting to tissues. Finally, cost effective manufacture of lipoaome-based pharmaceuticals, either in a liquid suspension or lyophilized product, has demonstrated the viability of this technology as an acceptable drug delivery system.
Other controlled release drug delivery systems, such as nonoparticles and hydrogels may be potential delivery W O 95113379 PCTIfi594112976 ~1~3703 vehicles for a ribozyme. These carriers have been developed for chemotherapeutic .agents and protein-based pharmaceuticals, and consequently, can be adapted for ribozyme delivery.
Topical administration of traps-splicing ribozymes is advantageous since it allows localized concentration at the site of administration with minimal systemic adsorption. This simplifies the delivery strategy of the ribozyme to the disease site and reduces the extent of toxicological characterization. Furthermore, the amount of material to be applied is far less than that required for other administration routes. Effective delivery requires the-ribozyme to diffuse into the infected cells.
Chemical modification of the ribozyme to neutralize negative charge may be all that is required for penetration. However, in the event that charge neutralization is insufficient, the modified ribozyme can be co-formulated with permeability enhancexs, such as Azone or oleic acid, in a liposome. The liposomes can either represent a slow release presentation vehicle in which the modified ribozyme and permeability enhancer transfer from the liposome into the infected cell, or the liposome phospholipids can participate directly with the modified ribozyme and permeability enhancer in facilitating cellular delivery. In some cases, both the ribozyme and permeability enhancer can be formulated into a suppository formulation for slow release.
Such ribozymes may also be systemically administered.
Systemic absorption refers to the accumulation of drugs in the blood stream followed by distribution throughout the entire body. Administration routes which lead to systemic absorption include: intravenous, subcutaneous=, intra-peritoneal, intranasal, intrathecal and ophthalmic. Each of these administration routes expose the-ribozyme to an accessible diseased tissue. Subcutaneous administration drains into a localized lymph node which proceeds through the lymphatic network into-the circulation. The rate of R'O 95/13379 PCT/US94112976 21?~~03 entry into the circulation has been shown to be a function of-molecular weight or size. The use of a liposome or other drug carrier localizes the ribozyme at the lymph node. The ribozyme can bemodified to diffuse into the cell, or the liposome can- directly participate in the delivery of either the unmodified or modified ribozyme to the cell.
A liposome formulation which can associate ribozymes with the surface of lymphocytes and macrophages is also useful. This will provide enhanced delivery to HIV
infected cells by taking advantage of the specificity of macrophage and lymphocyte immune recognition of infected cells. Whole blood studies show that the formulation is taken up by 90% of the lymphocytes after 8 hours at 37°C.
Preliminary biodistribution and pharmacokinetic studies yielded 70% of the injected dose/gm of tissue in the spleen after one hour - following intravenous administration.
Intraperitoneal administration also leads to entry into the circulation with the molecular weight or size of the ribozyme-delivery vehicle complex controlling the rate of entry.
Liposomes injected intravenously show accumulation in the liver, lung and spleeri.- The composition and size can be adjusted so that this accumulation represents 30% to 40% of the injected dose. The rest is left to circulate in the blood-stream for up to 24 hours.
The chosen method of delivery will result in cytoplasmic accumulation in the afflicted cells and molecules should have some nuclease-resistance for pptimal dosing. Nuclear delivery may be used but is less prefer-able. Most preferred delivery methods include liposomes (10-400 nm?, hydrogels, controlled-release polymers, microinjection or electroporation-(for ex vivo treatments) and other pharmaceutically applicable vehicles. The dosage will depend upon the disease indication and the route of administration but should be between 100-200 WO 95/fi3379 PCTlUS94112976 2 J 7~~03 mg/kg of- body weight/day. The duration of treatment will extend through the course of the disease symptoms, usually at least 14-16 days and possibly continuously. Multiple daily doses are anticipated for topical applications, ocular applications and vaginal applications. The number of doses will depend upon disease delivery vehicle and efficacy data from clinical trials.
Establishment of therapeutic levels of ribozyme within the cell is dependent upon the rate of uptake and degrada tion. Decreasing the degree of degradation will prolong the intracellular half-life of the ribozyme. Thus, chemically modified ribozymes, e.a., with modification of the phosphate backbone, or capping of the 5' and 3' ends of the ribozyme with nucleotide analogs may require different dosaging. Descriptions of useful systems are provided in the art cited above, all of which is hereby incorporated by reference herein.
Particular diseases that may be treated in this manner include any disease which can be treated by such RNAs, for example, HSV, HBV, EBV, and HIV infection; as well as various carriers (where-the target molecule is located in a known cellular compartment).
Any disease -caused by a specific set of mutations in a given genes RNA is potentially treatable by using target trans-splicing to correct such defective RNAs. Such diseases would include:
A. ,Q-globin diseases (such as sickle cell anemia), cystic fibrosis, as well as any other genetic diseases caused by a point mutations or deletions in RNA.
B. Cancers caused by specific mutant oncogene encoding RNAs (e-a. bcr-abl mRNAs, mutant p53 mRNAs).
C. Genetic diseases caused by unstable trinucleotide repeats in RNAs (e.a. Huntington's disease, fragile X
syndrome).
Other embodiments are within the following claims.
'"'r,.. ~_,_. t ; ;~;:~t:=,'-a

Claims (11)

CLAIMS:
1. A method for splicing a target nucleic acid molecule within a cell in culture with a separate nucleic acid molecule, wherein the target nucleic acid molecule encodes a protein that is deleterious to the cell in which it is located, and wherein the separate nucleic acid molecule is adapted to encode a non-deleterious chimeric protein when spliced with at least a part of the target nucleic acid molecule, which method comprises the step of:
contacting the target nucleic acid molecule with a catalytic nucleic acid molecule comprising the separate nucleic acid molecule under conditions in which at least a portion of the separate nucleic acid molecule is spliced with at least a portion of the target nucleic acid molecule to form a nucleic acid molecule encoding the non-deleterious chimeric protein.
2. The method of claim 1, wherein the catalytic nucleic acid molecule is active to cleave the target nucleic acid molecule and to splice the separate nucleic acid molecule with the target nucleic acid molecule.
3. The method of claim 1 or 2, wherein the contacting step is conducted in vitro.
4. The method of claim 1, 2 or 3, wherein the separate nucleic acid molecule is an RNA molecule.
5. The method of claim 1, 2, 3 or 4, wherein the contacting step comprises providing a vector encoding the catalytic nucleic acid molecule comprising the separate nucleic acid molecule.
6. A method for splicing a target nucleic acid molecule within a cell culture with a separate nucleic acid molecule, wherein the target nucleic acid molecule encodes a protein that is deleterious to the cell in which it is located, and wherein the separate nucleic acid molecule is adapted to encode a non-deleterious chimeric protein when spliced with at least a part of the target nucleic acid molecule, which method comprises the step of:
contacting the target nucleic acid molecule with the separate nucleic acid molecule in the presence of one or more spliceosomes or splicing factors under conditions in which at least a portion of the separate nucleic acid molecule is spliced with at least a portion of the target nucleic acid molecule to form a nucleic acid molecule encoding the non-deleterious chimeric protein.
7. The method of claim 6, wherein the separate nucleic acid molecule comprises a catalytic nucleic acid molecule.
8. The method of claim 7, wherein the catalytic nucleic acid molecule is active to cleave the target nucleic acid molecule and to splice the separate nucleic acid molecule with the target nucleic acid molecule.
9. The method of claim 1, 2, 3, 4, 5, 7 or 8, wherein the catalytic nucleic acid molecule is a group I intron molecule.
10. The method of claim 1, 6 or 8, wherein the target nucleic acid molecule is an RNA molecule.
11. The method of claim 6 or 8, wherein the contacting step comprises providing an expression vector encoding the separate nucleic acid molecule.
CA002175703A 1993-11-12 1994-11-09 Alteration of sequence of a target molecule Expired - Lifetime CA2175703C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/152,450 1993-11-12
US08/152,450 US5667969A (en) 1993-11-12 1993-11-12 Alteration of sequence of a deleterious target molecule by ribozyme catalyzed trans-splicing
PCT/US1994/012976 WO1995013379A1 (en) 1993-11-12 1994-11-09 Alteration of sequence of a target molecule

Publications (2)

Publication Number Publication Date
CA2175703A1 CA2175703A1 (en) 1995-05-18
CA2175703C true CA2175703C (en) 2006-10-10

Family

ID=37111082

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002175703A Expired - Lifetime CA2175703C (en) 1993-11-12 1994-11-09 Alteration of sequence of a target molecule

Country Status (1)

Country Link
CA (1) CA2175703C (en)

Also Published As

Publication number Publication date
CA2175703A1 (en) 1995-05-18

Similar Documents

Publication Publication Date Title
US5667969A (en) Alteration of sequence of a deleterious target molecule by ribozyme catalyzed trans-splicing
JP3015463B2 (en) Targeted cleavage of RNA using eukaryotic ribonuclease P and external guide sequences
US6368802B1 (en) Circular DNA vectors for synthesis of RNA and DNA
EP0748382B1 (en) Non-nucleotide containing enzymatic nucleic acid
US7135312B2 (en) Circular DNA vectors for synthesis of RNA and DNA
US5861288A (en) Catalytic DNA
US5989906A (en) Method and reagent for inhibiting P-glycoprotein (mdr-1-gene)
EP1251170A2 (en) Method and reagent for treatment of NF-kappaB dependent animal diseases
US5750390A (en) Method and reagent for treatment of diseases caused by expression of the bcl-2 gene
JPH10500309A (en) Methods and compositions for treating restenosis and cancer using ribozymes
US5616490A (en) Ribozymes targeted to TNF-α RNA
AU763325B2 (en) Ribozymal nucleic acids cleaving CCR5 or CXCR4
US5801158A (en) Enzymatic RNA with activity to RAS
US6080851A (en) Ribozymes with linked anchor sequences
US5714383A (en) Method and reagent for treating chronic myelogenous leukemia
US6897016B1 (en) Alteration of sequence of a target molecule
CA2398750A1 (en) Nucleozymes with endonuclease activity
CA2175703C (en) Alteration of sequence of a target molecule
WO1994029452A2 (en) Enzymatic rna molecules and their application in the treatment of fibrosis and fibrous tissue disease
JPH10510715A (en) VS ribozyme
US6492512B1 (en) Method and reagent for treatment of lung cancer and other malignancies caused by the deregulation of L-MYC gene expression
AU5661700A (en) Method and reagent for treatment of animal diseases
WO2001034793A1 (en) Method of effecting dna splicing

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20141110