EP1104463A1 - COMPOSITIONS DE hKIS ET PROCEDES D'UTILISATION CORRESPONDANTS - Google Patents

COMPOSITIONS DE hKIS ET PROCEDES D'UTILISATION CORRESPONDANTS

Info

Publication number
EP1104463A1
EP1104463A1 EP99942320A EP99942320A EP1104463A1 EP 1104463 A1 EP1104463 A1 EP 1104463A1 EP 99942320 A EP99942320 A EP 99942320A EP 99942320 A EP99942320 A EP 99942320A EP 1104463 A1 EP1104463 A1 EP 1104463A1
Authority
EP
European Patent Office
Prior art keywords
nucleic acid
cell
serine
hkis
isolated nucleic
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.)
Ceased
Application number
EP99942320A
Other languages
German (de)
English (en)
Inventor
Gary J. Nabel
Elisabeth G. Nabel
Manfred Boehm
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.)
University of Michigan
Original Assignee
University of Michigan
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 University of Michigan filed Critical University of Michigan
Publication of EP1104463A1 publication Critical patent/EP1104463A1/fr
Ceased 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4738Cell cycle regulated proteins, e.g. cyclin, CDC, INK-CCR
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/022Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from an adenovirus

Definitions

  • CKIs cyclin-dependent kinase inhibitors
  • Rb retinoblastoma protein
  • CKIs directly implicated in CDK regulation are p21 cip1/Waf1 (Xiong et al., 1993; Harper et al., 1993), p27 Kip1 (Pyoshima and Hunter, 1994; Polyak et al., 1994; Coats et al., 1996), and p16/p15 INK4 (Serrano et al., 1993).
  • CKIs to arrest cells in G1 have made the proteins of particular use in gene therapy techniques for treating diseases or disorders associated with cell proliferation, such as cancer and leukemias, psoriasis, bone diseases, fibroproliferative disorders, atherosclerosis, restenosis, and chronic inflammation.
  • diseases or disorders associated with cell proliferation such as cancer and leukemias, psoriasis, bone diseases, fibroproliferative disorders, atherosclerosis, restenosis, and chronic inflammation.
  • diseases or disorders associated with cell proliferation such as cancer and leukemias, psoriasis, bone diseases, fibroproliferative disorders, atherosclerosis, restenosis, and chronic inflammation.
  • diseases or disorders associated with cell proliferation such as cancer and leukemias, psoriasis, bone diseases, fibroproliferative disorders, atherosclerosis, restenosis, and chronic inflammation.
  • very little is known about the regulation of these very important proteins in vivo.
  • the inventors have discovered a novel mechanism of regulation of CKIs. Specifically, disclosed herein are serine/threonine kinases that inhibit the ability of CKIs to arrest cells in G1. In light of this discovery, the inventors were able to construct a transdominant mutant of a serine/threonine kinase that interferes with the respective endogenous serine/threonine kinase when introduced into a cell transgenically. Furthermore, the inventors were able to construct a CKI unable to be inhibited by a serine/threonine kinase. Such constructs may be used alone, together, or in conjunction with other therapies for inhibiting or reducing cell proliferation.
  • the present invention provides isolated nucleic acid segments.
  • Such isolated nucleic acid segments may encode wild-type or mutant hKIS polypeptides.
  • the isolated nucleic acid segments encode a transdominant mutant hKIS.
  • a transdominant mutant hKIS is a polypeptide that is capable of interfering with the ability of endogenous hKIS to phosphorylate p27.
  • a transdominant mutant hKIS would lead to or enhance cell cycle arrest in a cell containing the mutant.
  • An example of a transdominant mutant hKIS is an hKIS that contains a mutation altering its serine/threonine kinase activity, such as that encoded by SEQ ID NO:3).
  • the isolated nucleic acid encodes a cyclin kinase inhibitor containing a mutation at a serine or threonine amino acid. It is preferred that the cyclin kinase inhibitor retains its ability to arrest the cell cycle. Examples of mutated cyclin dependent kinases include mutated of p16, p21 , p27, and p57.
  • the isolated nucleic acids of the present invention may be contained in an expression vector.
  • the expression vector may be a plasmid or a viral vector.
  • the viral vector may be replication deficient and includes a retroviral vector, an adenoviral vector, an adenovirus associated viral vector, or a lentivirai vector.
  • the isolated nucleic acids of the present invention may be contained in or associated with a medical device, such as a catheter.
  • kits may also include one or more medical devices for administering the nucleic acid or polypeptide to a patient or one or more cells of a patient.
  • hKIS Saccharomyces cervisiae.
  • hKIS was identified using a yeast two hybrid assay (Example 1).
  • hKIS was co-transfected with either p27 K ⁇ p1 , p27 K ⁇ p1 (1-85aa), p27 K ⁇ p1 (144-198aa), p57 K ⁇ p2 , p57 K ⁇ 2 (1-87aa), p57 K ⁇ p2 (260-335aa), and p21 C ⁇ p1 into yeast, ⁇ -galactosidase assay (Sherr, 1994) on selection plates was performed, and the intensity was categorized by visualization at the intensity of the staining.
  • CKI cyclin-dependent kinase inhibitor
  • ⁇ -gal ⁇ - galactosidase
  • CDK CS cyclin-dependent kinase consensus sequence
  • NLS nuclear localization signal
  • PCNA proliferating cell nuclear antigen
  • +++ very strong staining
  • ++ strong staining
  • (+/-) weak staining
  • - no staining. No staining was observed with the GBT9 backbone or irrelevant negative controls, pLAM5' (human laminC/GAL4 DNA binding domain) or pVA3 (murine p53/GAL4 DNA binding domain) (Clontech, Palo Alto, CA).
  • the inventors In order to identify proteins that bind to p27, the inventors first utilized the yeast two-hybrid system to identify proteins that associate with p27 in vivo (Fields and Song, 1989; Chien et al, 1991 ; Durfee et al, 1993; Harper et al, 1993). Such analyses led to the discovery a novel gene that encodes a polypeptide that binds to p27 in the yeast assay. This gene is included herein as hKIS (human KIS) DNA and protein sequences (SEQ ID NO:1 and SEQ ID NO:2, respectively).
  • hKIS human KIS
  • Prominent examples include MDM2, which binds and inhibits the tumor suppressor function of p53, and the transforming proteins encoded by certain DNA viruses (e.g., the SV40 large T antigen), that also bind and inactivate tumor suppressors such as p53 and Rb.
  • DNA viruses e.g., the SV40 large T antigen
  • the inventors have determined that the interaction between hKIS and p27 reduces the ability of p27 to arrest cells in G1.
  • hKIS serves as a dominant gene controlling cell proliferation.
  • inhibiting hKIS is a therapeutic approach.
  • hKIS inhibition could be achieved by providing to a hyperproliferative cell, or administering to a patient, any compound that inhibits the hKIS gene, mRNA, or protein.
  • embodiments of the present invention include assays to find compounds capable of reducing the level of transcription of the hKIS gene.
  • Such assays include contacting a cell with a compound and comparing the amount of hKIS RNA in the cell as compared to a control. This comparison may be done through any of a number of techniques including, Northern blotting, semi-quantitative PCR, or RNase protection assays.
  • Preferred compounds are hKIS antisense oligonucleotides.
  • Other embodiments of the present invention include assays to find compounds that inhibit the ability of serine/threonine kinases, such as hKIS, to phosphorylate CKIs, such as p27. Such methods may be in vitro or in vivo.
  • the assay may include contacting a cell or protein composition comprising a hKIS and p27 protein with a compound and determine the ability of the two proteins to interact with each other.
  • the ability of the two proteins to interact may be determined by a number of methods including immunoprecipitation, plasmon resonance techniques, or fluorescence energy transfer techniques.
  • the substance may permit binding of the two proteins but inhibit phosphorylation. Such instances may be determined by detecting phosphorylation of the CKI.
  • cell proliferation may be inhibited by providing to a hyperproliferative cell, or administering to a patient, a CKI comprising one or more mutations that prevent or reduce phosphorylation of the CKI's serine/threonine residues by a serine/kinase, such as hKIS.
  • a serine/kinase such as hKIS.
  • a such mutated CKI would maintain the ability to arrest cells in G1 phase, as shown in example 1 , yet not be inhibited by the expression of a serine /threonine kinase.
  • the mutation may prevent interaction of the CKI with the respective serine/threonine or prevent phosphoryation of one or more serine/threonine subsequent to interaction of the two proteins.
  • the mutated CKI is p27 comprising a serine to alanine mutation at amino acid number 10.
  • Other methods of inhibiting cell proliferation include, but are not limited to, compounds that reduce transcription of the endogenous hKIS, compounds that prevent translation of hKIS mRNA, compounds that prevent the interaction of hKIS and p27, and compounds that prevent phosphorylation of p27 by hKIS.
  • Important aspects of the present invention concern isolated DNA segments and recombinant vectors encoding wild-type, polymorphic or mutant hKIS, and the creation and use of recombinant host cells that express wild- type, polymorphic or mutant hKIS, using the sequence of SEQ ID NO:1.
  • the present invention concerns DNA segments, isolatable from mammalian and human cells, that are free from total genomic DNA and that are capable of expressing a protein or polypeptide that has p27-binding activity.
  • the DNA segments encode hKIS.
  • DNA segment refers to a DNA molecule that has been isolated free of total genomic DNA of a particular species.
  • a DNA segment encoding hKIS refers to a DNA segment that contains wild-type, polymorphic or mutant hKIS coding sequences yet is isolated away from, or purified free from, total mammalian or human genomic DNA. Included within the term "DNA segment”, are DNA segments and smaller fragments of such segments, and also recombinant vectors, including, for example, plasmids, cosmids, phage, viruses, and the like.
  • a DNA segment comprising an isolated or purified wild-type, polymorphic or mutant hKIS gene refers to a DNA segment including wild- type, polymorphic or mutant hKIS coding sequences and, in certain aspects, regulatory sequences, isolated substantially away from other naturally occurring genes or protein encoding sequences.
  • the term "gene” is used for simplicity to refer to a functional protein, polypeptide, or peptide encoding unit. As will be understood by those in the art, this functional term includes both genomic sequences, cDNA sequences and smaller engineered gene segments that express, or may be adapted to express, proteins, polypeptides, domains, peptides, fusion proteins and mutants.
  • isolated substantially away form other coding sequences means that the gene of interest, in this case the wild-type, polymorphic or mutant hKIS gene forms the significant part of the coding region of the DNA segment, and that the DNA segment does not contain large portions of naturally-occurring coding DNA, such as large chromosomal fragments or other functional genes or cDNA coding regions. Of course, this refers to the DNA segment as originally isolated, and does not exclude genes or coding regions later added to the segment by the hand of man.
  • the invention concerns isolated DNA segments and recombinant vectors incorporating DNA sequences that encode a wild-type, polymorphic or mutant hKIS protein or peptide that includes within its amino acid sequence a contiguous amino acid sequence in accordance with, or essentially as set forth in, SEQ ID NO:2 corresponding to wild-type, polymorphic or mutant human KIS.
  • the invention concerns isolated DNA segments and recombinant vectors that encode a hKIS protein or peptide that includes within its amino acid sequence the substantially full length protein sequence of SEQ ID NO:2.
  • sequence essentially as set forth in SEQ ID NO:2 means that the sequence substantially corresponds to a portion of SEQ ID NO:2 and has relatively few amino acids that are not identical to, or a biologically functional equivalent of, the amino acids of SEQ ID NO:2.
  • biologically functional equivalent is well understood in the art and is further defined in detail herein. Accordingly, sequences that have between about 70% and about 80%; or more preferably, between about 81 % and about 90%; or even more preferably, between about 91 % and about 99%; of amino acids that are identical or functionally equivalent to the amino acids of SEQ ID NO:2 will be sequences that are "essentially as set forth in SEQ ID NO:2", provided the biological activity of the protein is maintained.
  • the invention concerns isolated DNA segments and recombinant vectors that include within their sequence a nucleic acid sequence essentially as set forth in SEQ ID NO:1.
  • the term "essentially as set forth in SEQ ID NO:1 is used in the same sense as described above and means that the nucleic acid sequence substantially corresponds to a portion of SEQ ID NO:1 and has relatively few codons that are not identical, or functionally equivalent, to the codons of SEQ ID NO:1.
  • DNA segments that encode proteins exhibiting p27-binding activity will be most preferred.
  • codons that encode the same amino acid such as the six codons for arginine or serine, and also refers to codons that encode biologically equivalent amino acids (see Table 1 , below).
  • amino acid and nucleic acid sequences may include additional residues, such as additional N- or C-terminal amino acids or 5' or 3' sequences, and yet still be essentially as set forth in one of the sequences disclosed herein, so long as the sequence meets the criteria set forth above, including the maintenance of biological protein activity where protein expression is concerned.
  • the addition of terminal sequences particularly applies to nucleic acid sequences that may, for example, include various non-coding sequences flanking either of the 5' or 3' portions of the coding region or may include various internal sequences, i.e., introns, which are known to occur within genes.
  • sequences that have between about 70% and about 79%; or more preferably, between about 80% and about 89%; or even more preferably, between about 90%, 92%, 93%, 94% and about 99%; of nucleotides that are identical to the nucleotides of SEQ ID NO:1 will be sequences that are "essentially set forth in SEQ ID NO:1.”
  • Sequences that are essentially the same as those set forth in SEQ ID NO:1 may also be functionally defined as sequences that are capable of hybridizing to a nucleic acid segment containing the complement of SEQ ID NO:1 under relatively stringent conditions. Suitable relatively stringent hybridization conditions will be well known to those of skill in the art, as disclosed herein. Naturally, the present invention also encompasses DNA segments that are complementary, or essentially complementary, to the sequence set forth in SEQ ID NO:1. Nucleic acid sequences that are "complementary" are those that are capable of base-pairing according to the standard Watson-Crick complementarily rules.
  • the term "complementary sequences” means nucleic acid sequences that are substantially complementary, as may be assessed by the same nucleotide comparison set forth above, or as defined as being capable of hybridizing to the nucleic acid segment of SEQ ID NO:1 under relatively stringent conditions such as those described herein.
  • the nuclear acid segments of the present invention regardless of the length of the coding sequence itself, may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may very considerably.
  • nucleic acid fragments of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol.
  • nucleic acid fragments may be prepared that include a short contiguous stretch identical to or complementary to SEQ ID NO:1 , such as about 8, about 10 to about 14, or about 15 to about 20 nucleotides.
  • DNA segments with total lengths of about 1 ,000, about 500, about 200, about 100 and about 50 base pairs in length (including all intermediate lengths) are also contemplated to be useful.
  • intermediate lengths means any length between the quoted ranges, such as 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, etc.; 21 , 22, 23, etc.; 30, 31 , 32, etc.; 50, 51 , 52, 53, etc.; 60, 61 , 62, 63, 64, etc.; 100, 101 , 102, 103, etc.; 150, 151 , 152,
  • the various probes and primers designed around the disclosed nucleotide sequences of the present invention may be of any length.
  • an algorithm defining all primers can be proposed;
  • n is an integer from 1 to the last number of the sequence and y is the length of the primer minus one, where n + y does not exceed the last number of the sequence.
  • the probes correspond to bases 1 to 10, 2 to 11 , 3 to 12 ... and so on.
  • the probes correspond to bases 1 to 15, 2 to 16, 3 to 17... and so on.
  • the probes correspond to bases 1 to 20, 2 to 21 , 3 to 22... and so on.
  • this invention is not limited to the particular nucleic acid and amino acid sequences of SEQ ID NO:1.
  • Recombinant vectors and isolated DNA segments may therefore variously include these coding regions themselves, coding regions bearing selected alterations or modifications in the basic coding region, or they may encode larger polypeptides that nevertheless include such coding regions or may encode biologically functional equivalent proteins or peptides that have variant amino acids sequences.
  • DNA segments of the present invention encompass biologically functional equivalent hKIS proteins and peptides. Such sequences may arise as a consequence of codon redundancy and functional equivalency that are known to occur naturally with nucleic acid sequences and the proteins thus encoded.
  • functionally proteins or peptides may be created via the application of recombinant DNA technology, in which changes in the protein structure may be engineered, based on considerations of the properties of the amino acids being exchanged. Changes designed by man may be introduced through the application of site-directed mutagenesis techniques, e.g., to disrupt the kinase properties of hKIS or to alter the domain of hKIS responsible for interaction with p27.
  • DNA segments encoding relatively small peptides such as, for example, peptides of from about 15 to about 50 amino acids in lengths, and more preferably, of from about 15 to about 30 amino acids in length; and also larger polypeptides up to and including proteins corresponding to the full-length sequences set forth in SEQ ID NO:2. It is contemplated that such DNA segments encoding polypeptides or mutants thereof may be useful in methods of interring with the biological function of the endogenous hKIS protein. In a preferred embodiment, the DNA segment is that of SEQ ID NO: 3.
  • the present invention concerns isolated DNA segments and recombinant vectors encoding mutant CKIs and the creation and use of recombinant host cells that express mutant CKIs.
  • CKIs have the ability to arrest the cell cycle and are useful in therapies designed to inhibit or otherwise limit cell proliferation.
  • Examples of cell proliferation disorders that can be affected by the modified CKIs described herein include restenosis, atherosclerosis, cancer, smooth muscle cell proliferative diseases or disorders of any vessel in the body, and the like.
  • Examples of CKI include p16, p21 , p27, p57.
  • the present invention includes a DNA segment encoding a CKI modified such that it containing a mutation of one or more serine or threonine residues.
  • Such mutants maintain the ability to arrest cells under going proliferation and are no longer able to be phosphorylated/inhibited by a serine/threonine kinase.
  • the serine or threonine residue is changed to alanine.
  • the serine or threonine may be changed to essentially any other amino acid as long as the change allows the CKI protein to maintain its functional activity and is no longer phosporylatable/inhibited by a serine/threonine kinase.
  • Alanine generally is preferred because it tends to have the least amount of effect on the conformation or function of the protein possessing the mutation.
  • Other amino acids, such as glycine also can be used to advantage.
  • One or more nucleotides of a serine or threonine codon can be modified to create an alanine codon.
  • the codons for alanine are listed in table 1.
  • One of ordinary skill in the art would be able to create the mutation and test for a decrease in the ability of the CKI to be phosphorylated/inhibited by a serine/threonine kinase and maintain its ability to arrest the cell cycle.
  • the serine or threonine residue/codon to be modified occurs within residues 1-20 of the encoded protein. In another embodiment the serine/threonine residue to modified occurs within residues
  • p16 is modified at S4 or T10; p21 is modified at S2 or S15; p27 is modified at S2, S7, S10, or S12; p57 is modified at S2, S5, S8, T9, S10 or T11.
  • expression vector or construct means any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed.
  • the transcript may be translated into a protein, but it need not be.
  • expression includes both transcription of a gene and translation of a RNA into a gene product.
  • expression only includes transcription of the nucleic acid, for example, to generate antisense constructs.
  • vectors are contemplated to be those vectors in which the coding portion of the DNA segment, whether encoding a full length protein or smaller peptide, is positioned under the transcriptional control of a promoter.
  • a “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene.
  • the phrases “operatively positioned", “under control” or “under transcriptional control” means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene.
  • the promoter may be in the form of the promoter that is naturally associated with the expressed gene, as may be obtained by isolating the 5' non-coding sequences located upstream of the coding segment or exon, for example, using recombinant cloning and/or PCR technology, in connection with the compositions disclosed herein (PCR technology is disclosed in U.S.
  • a recombinant or heterologous promoter is intended to refer to a promoter that is not normally associated with the gene in its natural environment.
  • Such promoters may include promoters normally associated with other genes, and/or promoters isolated from any other bacterial, viral, eukaryotic, or mammalian cell.
  • promoters normally associated with other genes, and/or promoters isolated from any other bacterial, viral, eukaryotic, or mammalian cell.
  • promoter and cell type combinations for protein expression is generally known to those of skill in the art of molecular biology, for example, see Sambrook et al. (1989), incorporated herein by reference.
  • the promoters employed may be constitutive, or inducible, and can be used under the appropriate conditions to direct high level expression of the introduced DNA segment, such as is advantageous in the large-scale production of recombinant proteins or peptides.
  • the particular promoter that is employed to control the expression of a nucleic acid is not believed to be critical, so long as it is capable of expressing the nucleic acid in the targeted cell.
  • a human cell it is preferable to position the nucleic acid coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell.
  • a promoter might include a human or viral promoter.
  • the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter and the Rous sarcoma virus long terminal repeat can be used to obtain high-level expression of transgenes.
  • CMV human cytomegalovirus
  • SV40 early promoter the Rous sarcoma virus long terminal repeat
  • the use of other viral or mammalian cellular or bacterial phage promoters which are well-known in the art to achieve expression of a transgene is contemplated as well, provided that the levels of expression are sufficient for a given purpose.
  • Tables 3 and 4 below list several elements/promoters which may be employed, in the context of the present invention, to regulate the expression of a heterologous gene. This list is not intended to be exhaustive of all the possible elements involved in the promotion of transgene expression but, merely, to be exemplary thereof. Any promoter/enhancer combination (as per the Eukaryotic Promoter
  • Eukaryotic cells can support cytoplasmic transcription from certain bacterial and viral promoters if the appropriate bacterial or viral polymerase is provided, either as part of the delivery complex or as an additional genetic expression construct.
  • Tumor Necrosis Factor FMA Hensel ef a/., 1989 Thyroid Stimulating Thyroid Hormone Chatterjee ef al, 1989 Hormone A Gene
  • the added sequences may include additional enhancers, promoters or even other genes.
  • one may, for example prepare a DNA fragment that contains the native regulatory elements positioned to regulate one or more copies of the native gene and/or another gene or prepare a DNA fragment which contains not one but multiple copies of the promoter region such that transcription levels of the desired gene are relatively increased.
  • a transgene to produce a protein once a suitable clone or clones have been obtained, whether they be cDNA based or genomic, one may proceed to prepare an expression system.
  • cDNA and genomic sequences are suitable for eukaryotic expression, as the host cell will generally process the genomic transcripts to yield functional mRNA for translation into protein.
  • cDNA version will provide advantages in that the size of the gene will generally be much smaller and more readily employed to transfect the targeted cell than will be a genomic gene, which will typically be up to an order of magnitude larger than the cDNA gene.
  • the inventors do not exclude the possibility of employing a genomic version of a particular gene where desired.
  • polyadenylation signal In expression, one will typically include a polyadenylation signal to effect proper polyadenylation of the transcript.
  • the nature of the polyadenylation signal is not believed to be crucial to the successful practice of the invention, and any such sequence may be employed.
  • Preferred embodiments include the SV40 polyadenylation signal and the bovine growth hormone polyadenylation signal, convenient and known to function well in various target cells.
  • a terminator also contemplated as an element of the expression cassette. These elements can serve to enhance message levels and to minimize read through from the cassette into other sequences.
  • a specific initiation signal also may be required for efficient translation of coding sequences. These signals include the ATG initiation codon and adjacent sequences. Exogenous translational control signals, including the ATG initiation codon, may need to be provided.
  • initiation codon must be "in-frame" with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • the exogenous translational control signals and initiation codons can be either natural or synthetic.
  • the efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements. It is proposed that a wild-type, polymorphic or mutant hKIS gene may be co-expressed with wild-type or mutant p27, wherein the proteins may be co-expressed in the same cell or wherein wild-type, polymorphic or mutant hKIS genes may be provided to a cell that already has wild-type or mutant p27.
  • Co-expression may be achieved by co-transfecting the cell with two distinct recombinant vectors, each bearing a copy of either the respective DNA.
  • a single recombinant vector may be constructed to include the coding regions for both of the proteins, which could then be expressed in cells transfected with the single vector.
  • the term "co-expression" herein refers to the expression of both the wild-type, polymorphic or mutant hKIS and wild-type or mutant p27 proteins in the same recombinant cell.
  • the wild-type, polymorphic or mutant hKIS gene may be co-expressed with genes encoding other CKI or tumor suppressor proteins or polypeptides.
  • Tumor suppressor proteins contemplated for use include, but are not limited to, the retinoblastoma, p53, Wilms tumor (WT-1), DCC, neurofibromatosis type 1 (NF-1), von Hippel-Lindau (VHL) disease tumor suppressor, Maspin, Brush-1 ,
  • BRCA-2 BRCA-2
  • MTS multiple tumor suppressor
  • contemplated is co-expression with a selected wild-type version of a selected oncogene.
  • Wild-type oncogenes contemplated for use include, but are not limited to, tyrosine kinases, both membrane-associated and cytoplasmic forms, such as members of the Src family, serine/threonine kinases, such as
  • Mos, growth factor and receptors such as platelet derived growth factor (PDDG), SMALL GTPases (G proteins) including the ras family, cyclin- dependent protein kinases (cdk), members of the myc family members including c-myc, N-myc, and L-myc and bcl-2 and family members.
  • PDDG platelet derived growth factor
  • G proteins G proteins
  • cdk cyclin- dependent protein kinases
  • members of the myc family members including c-myc, N-myc, and L-myc and bcl-2 and family members.
  • engineered cells are intended to refer to a cell into which an exogenous DNA segment or gene, such as a cDNA or gene encoding a hKIS has been introduced. Therefore, engineered cells are distinguishable from naturally occurring cells which do not contain a recombinantly introduced exogenous DNA segment or gene. Engineered cells are thus cells having a gene or genes introduced through the hand of man
  • Expression vectors for use in mammalian cells may include an origin of replication (as necessary), a promoter located in front of the gene to be expressed, along with any necessary ribosome binding sites, RNA splice sites, polyadenylation site, and transcriptional terminator sequences.
  • the origin of replication may be provided either by construction of the vector to include exogenous origin, such as may be derived from SV40 or other viral (e.g., polyoma, adenovirus, VSV, or BPV) source, or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the later is often is sufficient.
  • the promoters may be from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., CMV immediate early, the adenovirus late promoter; the vaccinia virus 7.5K promoter). Further, it is also possible, and may be desirable, to utilize promoter or control sequences normally associated with the desired gene sequence, provided such control sequences are compatible with the host cell systems.
  • mammalian cells e.g., metallothionein promoter
  • mammalian viruses e.g., CMV immediate early, the adenovirus late promoter; the vaccinia virus 7.5K promoter.
  • a number of viral based expression systems may be utilized, for example, commonly used promoters are derived from polyoma, Adenovirus 2, and most frequently Simian Virus 40 (SV40).
  • the early and late promoters of SV40 virus are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 origin of replication. Smaller or larger SV40 fragments may also be used, providing there is included the approximately 250 bp sequence extending from the Hind ⁇ site toward the Bgl ⁇ site located in the viral origin of replication.
  • the coding sequence may be ligated to an adenovirus transcription/ translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the transgene in infected hosts.
  • a non-essential region of the viral genome e.g., region E1 or E3
  • one will also typically desire to incorporate into the transcriptional unit an appropriate polyadenylation site (e.g., 5'- AATAAA-3') if one was not contained within the original cloned segment.
  • the poly A addition site is placed about 30 to 2000 nucleotides "downstream" of the termination site of the protein at a position prior to transcription termination.
  • selection systems including, but not limited, to the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase and adenine phosphoribosyltransferase genes.
  • antimetabolite resistance can be used as the basis of selection for dhfr, that confers resistance to methotrexate; gpt, that confers resistance to mycophenolic acid; neo, that confers resistance to the amonoglycoside G- 418; and hygro, that confers resistance to hygromycin.
  • a gene of the present invention may be "overexpressed", i.e., expressed in increasing levels of relative to its natural expression in cells.
  • overexpression may be assessed by a variety of methods, including semi-quanitative PCR, Northern blotting, RNase protection assays, radio- and Immuno- assays.
  • simple and direct methods are preferred, for example, those involving SDS/PAGE and protein staining or western blotting, followed by quantitative analyses, such as densitometric scanning of the resultant gel or blot.
  • a specific increase in the level of the mRNA, recombinant protein or peptide in comparison to the level in natural cells is indicative of overexpression.
  • Certain embodiments of the present invention may require the introduction of mutations into hKIS or a CKI gene.
  • Site-specific mutagenesis provides a ready ability to prepare and test sequence variants, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA.
  • Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed.
  • a primer of about 17 to 25 nucleotides in length is preferred, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
  • site-specific mutagenesis is well known in the art and commercial kits are available (ALTERED SITES® ii in vitro Mutagenesis Systems; Promega Corp., Madison, Wis.).
  • the technique employs a bacteriophage vector that exists in both a single stranded and double stranded form.
  • Typical vectors useful in site-directed mutagenesis include vectors such as the M13 phage. These phage vectors are commercially available and their use is generally well known to those skilled in the art.
  • Double stranded plasmids are also routinely employed in site directed mutagenesis, which eliminates the step of transferring the gene of interest from a phage to a plasmid.
  • site-directed mutagenesis is performed by first obtaining a single-stranded vector, or melting of two strands of a double stranded vector which includes within its sequence a DNA sequence encoding the desired protein.
  • An oligonucleotide primer bearing the desired mutated sequence is synthetically prepared. This primer is then annealed with the single-stranded DNA preparation, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand.
  • a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation.
  • This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
  • appropriate cells such as E. coli cells
  • clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
  • sequence variants of the selected gene using site- directed mutagenesis is provided as a means of producing potentially useful species and is not meant to be limiting, as there are other ways in which sequence variants of genes may be obtained.
  • recombinant vectors encoding the desired gene may be treated with mutagenic agents, such s hydroxylamine, to obtain sequence variants.
  • the present invention provides purified, and in preferred embodiments, substantially purified proteins and peptides.
  • purified protein or peptide as used herein, is intended to refer to an aqueous composition, isolatable from mammalian cells or recombinant host cells, wherein the protein or peptide is purified to any degree relative to its naturally-obtainable state, i.e., relative to its purity within a cellular extract.
  • a purified protein or peptide therefore also refers to a protein or peptide free from the environment in which it naturally occurs.
  • the proteins or polypeptides may be full length proteins or may also be less then full length proteins, such as individual domains, regions or even epitopic peptides. Where less than full length proteins are concerned the most preferred will be those containing the functional domains.
  • purified will refer to protein or peptide composition that has been subjected to fractionation to remove various other protein or peptide components, and which composition substantially retains the biological activity of the desired protein. For example, a purified wild-type hKIS protein would still maintain biological activity, as may be assessed by binding to p27, forming complexes with p27, or phosphorylating p27.
  • substantially purified will refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50% of the proteins in the composition or more.
  • a substantially purified protein will constitute more than 60%, 70%, 80%, 90%, 95%, 99% or even more of the proteins in the composition.
  • a polypeptide or protein that is "purified to homogeneity," as applied to the present invention means that he polypeptide or protein has a level or purity where the polypeptide or protein is substantially free from other proteins and biological components. For example, a purified polypeptide or protein will often be sufficiently free of other protein components so that degradative sequencing may be performed successfully.
  • a natural or recombinant composition comprising at least some proteins or peptides of interest will be subjected to fractionation to remove various other polypeptide or protein components from the composition.
  • Various techniques suitable for use in protein purification will be well known to those of skill in the art. These include, for example, precipitation with ammonium sulfate, PEG, antibodies and the like or by heat denaturation, followed by centrifugation; chromatography steps such as ion exchange, gel filtration, reverse phase, hydroxylapatite, lectin affinity and other affinity chromatography steps; isoelectric focusing; gel electrophoresis; and combinations of such and other techniques.
  • the general approach to the cell proliferation suppression aspect of the present invention is to provide a cell with a transdominant hKIS protein, a CKI mutated such that it is no longer inhibited by a serine/threonine kinase, or both.
  • transdominant hKIS it is meant that the mutated hKIS or hKIS polypeptide fragment interferes the ability of endogenous hKIS protein to inhibit p27 mediated G1 arrest. While it is conceivable that a protein may be delivered directly to a cell, a preferred embodiment involves providing a nucleic acid encoding a protein to the cell.
  • the polypeptide is synthesized by the transcriptional and translational machinery of the cell, as well as any that may be provided by the expression construct.
  • the preferred mode is also to provide a nucleic acid encoding the construct to the cell. All such approaches are herein encompassed within the term "gene therapy”.
  • DNA can be directly transferred to somatic target cells by viral vectors, such as retroviruses and adenoviruses, and non-viral methods, such as cationic liposomes, liposome viral conjugates, and polymers.
  • viral vectors such as retroviruses and adenoviruses
  • non-viral methods such as cationic liposomes, liposome viral conjugates, and polymers.
  • Viruses naturally infect mammalian cells and introduce their viral DNA to convert the host biosynthetic pathway to produce viral DNA, RNA, and protein. Molecular biologists have been able to modify these viruses so that they deliver foreign DNA to the target cell but cannot replicate in the host cell nor express viral proteins necessary for encapsulation. In general, early response virai sequences, involved in viral transcription, translocation or capsid synthesis, have been removed from the viral genome and are replaced by the foreign gene of interest. Therefore, these recombinant viruses can only propagate in specific packaging cell lines which express the deleted viral proteins. Replication- deficient retroviruses, adenoviruses, adeno-associated viruses and adenoviral conjugates are now used in gene transfer techniques.
  • Retroviruses are RNA viruses that require vector integration into the host genome for expression of the transgene thus limiting their use to dividing cells. As most of the vascular and myocardial cellular components are non-replicating cells, retroviruses are of limited use in cardiovascular gene transfer. In addition, integration at random locations may lead to insertional mutagenesis and transformation. However, there have been no reported short- or long-term toxicity associated with their use in human gene therapy trials. Retrovirus-mediated gene transfer has been used for cell-mediated gene transfer using endothelial cells and for direct gene transfer into porcine arteries. The long-term, high-level expression renders retroviral vectors in particular ideal for ex Vo, cell-mediated gene transfer.
  • endothelial cells or vascular smooth muscle cells may be isolated, expanded and transduced in the laboratory and reseeded on to an artery in vivo.
  • the technique of ex vivo gene transfer is however fairly cumbersome since it requires cell expansion.
  • ex wVo gene transfer of endothelial cells and smooth muscle cells may be useful in seeding stents, grafts or injured arteries during vascular procedures to treat thrombotic disorders or graft hyperplasia.
  • Recombinant gutted lentiviruses may represent an attractive alternative to retroviruses.
  • Lentiviruses have not been directly implicated in any malignancies and, in contrast to retroviral based vector systems, human, simian and bovine immunodeficiency viral (HIV, BIV, SIV) vector systems have been shown to mediate stable gene transfer in terminally differentiated neurons and macrophages in culture. In vivo, transgene expression is detected for up to 6 months in liver, muscle, retinal tissue, and brain of immune-competent rats in vivo and does not appear to evoke an immune response or local inflammation, permitting repeated viral challenge.
  • HIV immunodeficiency viral
  • Recombinant adenoviruses efficiently transfect proliferating and non-proliferating cells, but lack mutagenicity since the transgenic genome is not integrated into the host chromosome but remains episomal.
  • rAAV Recombinant adeno-associated viruses
  • rAAV vectors are described in U.S. Pat. No. 5,139,941.
  • rAAV has not been associated with disease in any host and has not been associated with malignancies despite integration of the transgene into the host genome.
  • rAAV integrates viral and transgenic DNA preferentially but not exclusively at chromosome 19q locus.
  • Adeno-associated viruses are incapable of replication and depend on co-infection with adenovirus or a herpes virus for replication.
  • ⁇ -galactosidase and tyrosine hydroxylase have been achieved in non-dividing neurons in the rat CNS by rAAV, and intravenous delivery of rAAV encoding human clotting factor IX resulted intransduction of 3% of all hepatocytes over a 5 month observation period.
  • intraluminal and periadventitial vascular delivery of rAAV in atherosclerotic carotid arteries of cynomolgus monkeys results in efficient transgenic expression.
  • transgenic expression is predominantly found in adventitial endothelial cells of microvessels.
  • Other viral vectors that may be used for gene therapy include herpes simplex virus (U.S. Pat. No. 5,288,641 ) and cytomegalovirus (Miller, 1992).
  • Non-viral gene transfer can be performed by microinjection, DEAE-dextran transfection, calcium phosphate precipitation, electroporation, liposomes, and particle-mediated gene transfer (i.e. introducing DNA-coated particles).
  • the most common non-viral gene transfer vectors are DNA-liposomes.
  • Cationic liposomes condense and entrap the DNA through electrostatic interaction. They are prepared by sonification and remain stable in aqueous solution for months. The positively charged liposome complex fuses with the negatively charged cell surface to release the DNA into the cytoplasm of target cells, bypassing the lysosomal compartment and degradation by serum.
  • plasmid DNA is subsequently incorporated in the nucleus as an episome.
  • the relatively safe profile of liposomes, the lack of vector size or target cell constraints, as well as the relative ease of liposome-DNA complex preparation favors this gene transfer technique.
  • Preclinical studies using different forms of these lipids DOTMA, DC-
  • Lipofection-mediated gene transfer results in site-specific expression of foreign recombinant genes in vascular endothelial and smooth muscle cells and alters the biology of the vessel wall.
  • Cationic liposomes are well tolerated in vivo and do not induce any biochemical, hemodynamic or cardiac intoxications. Additional advances in lipid chemistry are developing newer generations of cationic liposomes, which permit higher transfection with minimal toxicity.
  • the transfection efficacy and specificity of lipofection may be further augmented by coupling of ligands or viral particles (Ad, HVJ, VSVG) to the liposomes.
  • Ad, HVJ, VSVG ligands or viral particles
  • plasmid DNA or RNA may be injected directly into tissue such as skeletal muscle or myocardium.
  • anti-sense oligonucleotides are used for gene therapy (Morishita et al, 1993). Anti-sense oligonucleotides do not require a vector for cell transduction and can be directly injected in the target tissue. Anti-sense oligonucleotides are short DNA sequences complementary to the RNA message of interest, which are chemically modified to resist nuclease degradation. The oligonucleotide may be modified at the 5; end to prevent nuclease degradation or may made up of ribonucleotide bases attached to a peptide backbone (protein nucleic acid).
  • anti-sense oligonucleotides are able to efficiently modify intracellular expression of factors involved in smooth muscle cell and endothelial cell migration and proliferation, including by use of anti-sense oligonucleotides against c-myc, c-myb, cdc2, and PCNA.
  • the nucleotide sequence hybridizes to target RNA, which prevents translation of RNA, targets the message for degradation by ribonuclease H, and interferes with cytosolic translocation.
  • gene therapy is used to treat or prevent cell proliferation.
  • vascular cell proliferation such as that associated with restenosis or atherosclerosis is prevented using gene therapy.
  • a gene therapy vector or composition of the present invention may be tested in an animal model. Studies in animal models of cardiovascular disease have demonstrated that transgenes can be expressed at high levels at local sites in the vasculature.
  • Medical devices that are suitable for use in the present invention include known devices for the localized delivery of therapeutic agents.
  • Such devices include, for example, catheters such as injection catheters, balloon catheters, double balloon catheters, microporous balloon catheters, channel balloon catheters, infusion catheters, perfusion catheters, etc., which are, for example, coated with the therapeutic agents or through which the agents are administered; needle injection devices such as hypodermic needles and needle injection catheters; needleless injection devices such as jet injectors; coated stents, bifurcated stents, vascular grafts, stent grafts, etc.; and coated vaso-occlusive devices such as wire coils.
  • stents that are commercially available and may be used in the present application include the RADIUSTM (Scimed Life Systems, Inc.), the SYMPHONY® (Boston Scientific Corporation), the Wallstent (Schneider Inc.), the Precedent IITM (Boston Scientific
  • Such devices are delivered to and/or implanted at target locations within the body by known techniques.
  • the double balloon catheter was an initial catheter employed in animal model studies and was useful to demonstrate the basic principles of gene transfer.
  • the catheter consists of two balloons placed about 1.5 cm apart with an inner protected space.
  • the genetic vector is instilled into the isolated arterial segment between the balloons.
  • Adenoviral-mediated recombinant gene expression is detected in endothelial cells, vascular smooth muscle cells and adventitial cells for several weeks following infection and is not found downstream to the arterial segment or in other tissues by PCR.
  • Retroviral-mediated gene expression can be detected for up to 6 months.
  • a disadvantage to this catheter is the possibility of distal ischemia due to occlusion of blood flow. Alternate delivery devices permit flow distal to the isolated segment allowing a prolonged instillation time period without compromising distal perfusion.
  • Porous and microporous balloons infuse the vector directly into the juxtapositioned arterial wall through small pores in the catheter.
  • the depth of delivery is directly related to the perfusion pressure.
  • Channel balloon catheters combine two separate inflatable compartments for balloon angioplasty and drug infusion, allowing separate control of balloon inflation pressure for positioning and drug infusion pressure.
  • a hydrogel coated balloon catheter has a hydrophilic polyacrylic acid polymer coating of the balloon. This polymer absorbs the DNA suspension and when the balloon is inflated, the DNA coating is pressed against the vessel wall.
  • the iontophoretic balloon uses a local current between the balloon and the skin of the subject to drive the negatively charged DNA into the arterial wall.
  • Other delivery devices include stents coated with a DNA- impregnated polymer or cells comprising a nucleic acid of the present invention (ex wVo gene transfer) into arterial and venous grafts.
  • tissue may be selectively targeted for gene therapy by use of tissue specific promoters and enhancers.
  • the method of the present invention can be combined with other methods for treating cell proliferation.
  • other genes such as thymidine kinase, cytosine deaminase, wild-type or mutated p21 , p27, and p53 and combinations thereof can be concomitantly transformed into cells and expressed.
  • HS-tK herpes simplex-thymidine kinase
  • mutant hKIS or CKI gene expression could be used similarly in conjunction with other gene therapy approaches.
  • the genes may be encoded on a single nucleic acid but separately transcribed. Alternatively, the genes may be operably linked such that they are contranscribed. In preferred embodiments, the genes are operably linked to encode a fusion protein. In other embodiments the co-transcribed genes are separated by an internal ribosome binding site allowing the proteins to be translated separately. Such combination therapies are described in WO 99/03508 (incorporated herein by reference in its entirety).
  • nucleic acid or protein compositions of the present invention may be introduced into the myocardium.
  • Myocardial gene transfer requires tranfection of terminally differentiated myocytes.
  • Adenoviral gene transfer by intracoronary or intramyocardial delivery results in transient gene expression for several weeks in a limited number of cells.
  • Adeno-associated viral vectors have been shown to induce stable transgene expression in up to 50% of murine, rat and porcine cardiomyocytes after ex vivo intracoronary infusion and myocardial injections for at least 6 months. These vectors may be useful for gene delivery to treat human myocardial diseases.
  • vascular diseases are characterized by abnormalities of cell proliferation.
  • One approach to therapies is to express genes that inhibit cell proliferation within vascular lesions, for example, after angioplasty or in a by-pass graft. Most approaches regulate the cell cycle in vascular smooth muscle, endothelial or macrophage cells.
  • CDKs cyclin/cyclin-dependent kinase complexes
  • Endogenous inhibitors of the cyclin-CDKs termed the cyclin-dependent kinase inhibitors (CKIs) result in cell cycle arrest and cessation of cell proliferation.
  • CKIs cyclin-dependent kinase inhibitors
  • Prodrug-enzyme therapies using thymidine kinase or cytosine deaminase, constitute a form of local therapy in which an enzyme is expressed locally that converts a prodrug into an active form.
  • Gene transfer of DNA encoding these converting enzymes to the injured arterial wall combined with systemic prodrugs administration produces high levels of growth inhibitory drugs in the target tissue.
  • the therapeutic effect of transgene expression can be regulated by administration of the prodrug and can be initiated independently of the gene transfer.
  • Herpes simplex virus thymidine kinase converts an inert nucleoside analog, ganciclovir into a phosphorylated, toxic form in transduced cells. Its subsequent incorporation into the host DNA induces chain termination and cell death in dividing cells, while non-dividing cells remain unaffected. Local delivery of recombinant adenovirus encoding for
  • HSV-tk at the time of the balloon injury and systemic administration to ganciclovir inhibited smooth muscle cell proliferation in vivo, and decreased intimal formation in balloon-injured porcine and rat arteries and atherosclerotic rabbit arteries.
  • a similar reduction of neointimal hyperplasia was observed in arterial interposition grafts which overexpress
  • Cytosine deaminase catalyzes the hydrolytic deamination of non-toxic cytosine and 5-fluorocytosine (5-FC) into uracil and 5-fluorouracil, which inhibits thymidilate synthase and hence DNA and RNA synthesis.
  • CD/5- FC does not induce significant necrosis or apoptosis but results in cytostatic effects on vascular smooth muscle cells.
  • CD gene transfer in the rabbit femoral injury model followed by systemic 5-FC treatment resulted in a decrease of the intima to media area ratio, comparable to the efficacy of HSV-tk/ganciclovir in a rat and pig model of vascular injury.
  • the Fas/FasL death-signaling pathway mediates cellular immunocytotoxicity in activated lymphocytes. Binding of the Fas receptor to FasL activates the caspase pathway leading to apoptosis. FasL is expressed in intimal smooth muscle cells and immune competent cells in atherosclerotic plaques.
  • FasL may function to protect the vessel from leukocyte extravasation to the subendothelial space during arterial repair by inducing T lymphocyte apoptosis.
  • Targeting of cell cycle regulatory proteins promotes inhibition of cell proliferation, and cell differentiation.
  • Cell cycle arrest prevents vsmc proliferation and migration and endothelial dysfunction, shown by improved vasoreactivity and NO production, rendering the vessel less susceptible to inflammatory infiltration and free radical formation. Progression through the cell cycle is controlled by the assembly and disassembly of the different cyclin-cyclin dependent kinase complexes. These complexes phosphorylate retinoblastoma protein leading to the release of the sequestered transcription factors, E2F and Elf 1.
  • the cyclin dependent kinase inhibitors modulate the enzymatic activity of cyclin/CDK complexes necessary for Gi progression.
  • Ad-p21 infection of porcine iliofemoral and rat carotid arteries following balloon injury reduces BrdU incorporation by 35% and l/M area ratio by 37%.
  • Gax homeobox gene overexpression, as an upstream regulator of p21 in the rat carotid artery injury model inhibited neointimal formation and luminal narrowing by 59 and 56 percent, respectively.
  • Adenovirus-mediated overexpression of p27 in balloon-injured rat and porcine arteries significantly attenuated intimal lesion formation.
  • Rb retinoblastoma gene product
  • E2F decoy oligodeoxynucleotide reduces not only graft susceptibility to atherosclerosis, and enhances medial hypertrophy, which renders the graft more resistant to increased hemodynamic stress and improves vein graft patency.
  • Metalloproteinases degrade the extracellular matrix, promote growth factor release and cell activation and are therefor essential for cell migration.
  • Overexpression of tissue inhibitor of metalloproteinases (TIMP) was shown to inhibit invasive and metastatic behavior of tumor cells.
  • TIMP protein expression has been evaluated in an organ culture model of neointimal formation, which lends itself for the study of smc migration rather than proliferation.
  • Overexpression of TIMPi and 2 reduced neointima formation and neointimal cell numbers by 54-79% and 71 % respectively, but did not alter smc proliferation and viability.
  • nucleic acids and proteins or polypeptides of the present invention may be particularly useful in methods of treating cardiovascular disease.
  • a nucleic acid encoding a transdominant hKIS or mutated CKI may be introduced locally into an injured artery to prevent restenosis.
  • a vector comprising both a trandominant hKIS and a mutated p27 is used to treat restenosis.
  • both genes may be introduced together but in separate vectors.
  • gene therapy using a nucleic acid of the present invention may be combined with other gene and non-gene therapies to treat a cardiovascular disease.
  • Potential molecular targets for cardiovascular disease are shown in Table 5. Table 5 Potential molecular targets for cardiovascular disease
  • Aqueous compositions of the present invention comprise an effective amount of a compound dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
  • Aqueous compositions of gene therapy vectors are also contemplated.
  • pharmaceutically or pharmacologically acceptable refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
  • the biological material should be extensively dialyzed to remove undesired small molecular weight molecules and/or lyophilized for more ready formulation into a desired vehicle, where appropriate.
  • the active compounds will then generally be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, subcutaneous, intralesional, or even intraperitoneal routes.
  • compositions suitable for local administration of a composition of the present invention include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions of the active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • a composition of the present invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, an liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the technology of U.S. Patents 4,601 ,903; 4,559,231 ; 4,559,230; 4,596,792; and 4,578,770 each incorporated herein by reference, may be used.
  • Kits may comprise compositions comprising various components made using the present invention such as for example, cells, expression vectors, virus stocks, proteins, antibodies, catheters, coated stents, and drugs. These components may be in a form appropriate for the intended application. Generally, this will entail preparing compositions that are essentially free of pyrogens, as well as other impurities that could be harmful to humans or animals.
  • kits may include a nucleic acid encoding a CKI serine/threonine mutant and/or a trandominant hKIS mutant.
  • Example 1 Identification of hKIS a Serine/Threonine Kinase that interacts with p27 Cyc ⁇ n-Dependent Kinase Inhibitor Using a yeast two-hybrid screen, cDNAs from a human B-cell library
  • E/Cdk2 (Polyak et al, 1994; Sheaff et al, 1997; Matsuoka, 1995).
  • the COOH-terminal domain is inactive as a CDK inhibitor (CKI), but it has been assumed to play a role in protein-protein interactions (Maucuer, 1997).
  • CKI CDK inhibitor
  • NCBI Center for Biotechnology Information
  • the yeast two-hybrid screen yielded several cDNAs that interacted with the p27 K ⁇ p1 COOH-terminal domain, but not the NH 2 -terminal region of p27 K ⁇ p1 , p57 K ⁇ p2 , or p21 C ⁇ p1 (Fig. I).
  • the clones interacted with full-length p27 K ⁇ p1 as well as the COOH-terminal domain of p27 K ⁇ p1 in yeast.
  • the entire coding sequence of one clone (SEQ ID NO:1 ), C21 was 99% similar to the rat serine/threonine protein kinase KIS (Zamore et al, 1992).
  • hKIS human homologue of rat KIS
  • SEQ ID NO:2 The 46.5 kDa hKIS (SEQ ID NO:2) protein consists of an NH 2 -terminal serine/threonine kinase consensus region and a COOH terminal region with 42% sequence similarity to hU2AF65, a 65kDa subunit of the splicing factor U2AF (Zamore, 1992).
  • hKIS binding was specific for COOH-terminal p27 K ⁇ p1 , because it failed to interact in the two hybrid assay with NH 2 -terminal p27 K ⁇ p1 , p57 K ⁇ p2 , p21 C ⁇ p1 and several negative controls.
  • hKIS The specificity of the interaction between hKIS and p27 K ⁇ p1 was analyzed further biochemically.
  • In vitro translated 35 S-methionine-labeled hKIS was incubated with glutathione S-transferase (GST), GST-p16 lnM , GST-p27 K,p1 , GST-p21 C ⁇ p1 , or GST-p57 K ⁇ p2 (Morgan, 1995).
  • GST glutathione S-transferase
  • GST-p16 lnM GST-p16 lnM
  • GST-p27 K,p1 GST-p21 C ⁇ p1
  • GST-p57 K ⁇ p2 GST-p57 K ⁇ p2
  • hKIS To determine the function of hKIS, epitope-tagged in vitro translated hKIS was immunoprecipitated and incubated with purified recombinant p27 K ⁇ p1 in the presence of 32 P-ATP (Zamore, 1992). In contrast to a negative control extract, hKIS readily phosphorylated bacterial-produced p27 K ⁇ p1 . Under the same conditions, hKIS did not phosphorylate p57 K ⁇ p2 , p21 C ⁇ p1 or p16 lnk4 , documenting the specificity of p27 ⁇ ,p1 phosphorylation by hKIS. In addition, hKIS was also observed to undergo autophosphorylation.
  • hKIS was incubated in an in vitro phosphorylation assay with NH 2 -terminal or COOH- terminal GST-p27 K ⁇ p1 .
  • hKIS was found to bind the COOH-terminal domain of p27 K ⁇ p1
  • phosphorylation of GST- p27 K ⁇ p1 was detected in the NH 2 .terminal region of the protein.
  • mutational analyses were performed in this region.
  • hKIS-green fluorescent protein (GFP) fusion protein was prepared and transfected into 293 cells. In contrast to GFP alone, which showed characteristic cytoplasmic staining, hKIS-GFP localized predominantly in the nucleus. This localization was confirmed by biochemical analysis of epitope-tagged hKIS in nuclear and cytoplasmic extracts. Endogenous p27 K ⁇ p1 was detected in both compartments, though at higher levels in the nucleus. In contrast, hKIS localized primarily to the nucleus.
  • GFP green fluorescent protein
  • endogenous p27 K ⁇ p1 and hKIS were found to associate biochemically in vivo in nuclear extracts, as determined by immunoprecipitation with anti- p27 K ⁇ p1 antibody, followed by detection of hKIS by Western blot analysis.
  • cell transfection studies were performed. A human melanoma line, UM316, was transfected with hKIS, p27 K ⁇ p1 or p21 C ⁇ p1 , alone or in various combinations. Human CD2 was included as a marker for cell transfection to facilitate cell cycle analysis (Hannon and Beach, 1999; Danthinne et al, 1999).
  • NHI 293 cells were transfected with hKIS (K54R), p27 K ⁇ p1 , p27 K ⁇ p1 (S10A) mutant, alone or in various combinations.
  • Human CD2 was included as a mark for cell transfection to facilitate cell cycle analysis.
  • Transfection of hKIS and hKIS (K54R) alone did not alter cell cycle distribution, but p27 K ⁇ p1 as well as p27 K ⁇ p1 S10A arrested cells at the G1/S checkpoint as expected.
  • cotransfection of hKIS and p27 K ⁇ p1 reversed the growth arrest by p27 K ⁇ 1 .
  • Example 1 The expression vectors of Example 1 were constructed as follows. pGBT9p27 K ⁇ p1 CR, pGBT9p27 K ⁇ p1 NH 2 and pGBT9p27 K ⁇ p1 COOH were obtained by PCR amplification (p27 K ⁇ p1 CR: p27 K ⁇ p1 CR5'P CGT GAA TTC ATG TCA AAC GTG CGA GTG (SEQ ID NO:7) and p27 K,p1 CR3'P CGT GGA TCC TTA CGT CTG GCG TCG AAG GCC (SEQ ID NO:8); p27 K,p1 NH 2 : p27 K ⁇ p1 CR5'P and p27 K ⁇ p1 NH 2 3'P CTG GGA TCC TGT AGA ACT CGG GCA AGC T (SEQ ID NO: 9); p27 K ⁇ p1 COOH: p27 K ⁇ p1 COOH5'P
  • GST glutathione S-transferase
  • pGSTp27 Kip1 NH2S10M was cloned by PCR amplification (p27 Kip1 NH 3 S10M5'P GCT GGA TCC ATG TCA AAC GTG AGA GTG TCC AAC GCT CCG AG (SEQ ID NO:15)(Serine10 AGC ⁇ Alanine GCT) and p27 Kip1 NH 2 3'P) and subcloned into BamHI site of pGEX-6P.
  • pGST p16 lnk4 was obtained by EcoRI/Xhol digestion of BShpl 6 and subcloned into EcoRI/Xhol digested pGST-6P.
  • the 3'end of hKIS cDNA was mapped by rapid amplification of cDNA ends (RACE) with internal primers for hKIS and total RNA from JY-cells by using 3'RACE system (GIBCO, Gaithersburg, MID).
  • the hKIS cDNA was cloned into the Xhol site of pcDNA3.1/His (Invitrogen, Carlsbad, CA).
  • pVR1012hKIS was obtained by Xhol/Xba digestion of pcDNA3.1 hKIS and subcloned into the Sa1l/Xba site of VCL1012CMV (Vical, San Diego, CA).
  • the plasmids pVR1012p2l, pVR1012p27 Kip1 , and pVRCD2 were described elsewhere (Serrano, 1995).
  • [ 35 S]-methionine-labeled and unlabeled hKIS were produced by in vitro transcription/translation using the TNT T7-coupled reticulocyte lysate system (Promega, Madison, Wl) with pcDNA3.1 KIS as the template.
  • the crude cell lysate of the GST-fused proteins was prepared according to the manufacturer's protocol (Phannacia, Piscataway, NJ).
  • the fusion proteins were bound to gluthathione-SepharoseTM-4B beads (Pharmacia Biotech, Uppsala Sweden) in IP buffer containing 250mM KCL, 2.5mM MgCI 2 , 20 mM Hepes pH7.9, 0.1% NP40 and IxCompleteTM protease inhibitor cocktail (Boehringer, Mannheim, Germany). After 1 hour incubation at 4°C, the beads were washed three times.
  • Binding assays were performed by incubating GST-fusion protein with 20 ⁇ g [ 35 S]-methionine-labeled hKIS transcription/translation mixture at 4°C for 1 hour in IP buffer and washed 3 times. The bound proteins were analyzed by SDS-poly acrylamide gel electrophoresis (SDS-PAGE). To confirm the correct size and amount of GST-fusion proteins, the PAGE was stained with Coomassie Brilliant Blue R250 (GIBCO, Gaithersburg, MD) prior to visualizing the [ 35 S]-methionine labeled hKIS by autoradiography.
  • SDS-PAGE SDS-poly acrylamide gel electrophoresis
  • the GST-fusion proteins were digested with PreScissionTM protease (Pharmacia, Piscataway, NJ) according to the manufacturer's protocol.
  • the in vitro phosphorylation assay was carried out by incubation of CKIs with immunoprecipitated hKIS at 30°C for 30 minutes in 20 ⁇ l phosphorylation buffer (20 mM Tris/HCI pH7.5, 1 mM EGTA, 10 mm MgC1 2 , 1 mM dithiothreitol, Ix CompleteTM protease inhibitor cocktail and 10 ⁇ Ci ⁇ 32 P-ATP. The samples were analyzed by SDS-PAGE.
  • 293 cells were maintained in Dulbecco's modified Eagle's medium (GIBCO, Gaithersburg, MD) containing 10% fetal bovine serum (FBS), glutamine (2mM) plus antibiotics.
  • UM316 cells, JY-cells and a human transformed B cell line were cultured in RPMI 1640 medium (GIBCO,

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne une nouvelle composition et des procédés pour modifier la prolifération d'une cellule, et notamment des polypeptides de hKIS de type sauvage ou mutants ainsi que des inhibiteurs de kinase dépendante des cyclines, qui contiennent des mutations empêchant leur inhibition par des kinases de sérine / de thréonine.
EP99942320A 1998-08-21 1999-08-20 COMPOSITIONS DE hKIS ET PROCEDES D'UTILISATION CORRESPONDANTS Ceased EP1104463A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US9771098P 1998-08-21 1998-08-21
US97710P 1998-08-21
PCT/US1999/018903 WO2000011165A1 (fr) 1998-08-21 1999-08-20 COMPOSITIONS DE hKIS ET PROCEDES D'UTILISATION CORRESPONDANTS

Publications (1)

Publication Number Publication Date
EP1104463A1 true EP1104463A1 (fr) 2001-06-06

Family

ID=22264764

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99942320A Ceased EP1104463A1 (fr) 1998-08-21 1999-08-20 COMPOSITIONS DE hKIS ET PROCEDES D'UTILISATION CORRESPONDANTS

Country Status (4)

Country Link
EP (1) EP1104463A1 (fr)
AU (1) AU5572599A (fr)
CA (1) CA2343099C (fr)
WO (1) WO2000011165A1 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA01010486A (es) 1999-04-15 2002-03-27 Calgene Llc Secuencias de acido nucleico para proteinas implicadas en la sintesis de tocoferol.
EP1272640A2 (fr) 2000-03-24 2003-01-08 Millennium Pharmaceuticals, Inc. 3714, 16742, 23546, et 13887 nouvelles molecules de proteine kinase et leurs utilisations
US6872815B1 (en) 2000-10-14 2005-03-29 Calgene Llc Nucleic acid sequences to proteins involved in tocopherol synthesis
WO2002012478A2 (fr) 2000-08-07 2002-02-14 Monsanto Technology, Llc Genes de la voie methyl-d-erythritol phosphate
US7161061B2 (en) 2001-05-09 2007-01-09 Monsanto Technology Llc Metabolite transporters
AU2002257237B8 (en) 2001-05-09 2008-06-05 Monsanto Technology Llc Tyra genes and uses thereof
EP1427832A4 (fr) 2001-08-17 2006-09-06 Monsanto Technology Llc Genes de methyltransferase et leurs utilisations
US7262339B2 (en) 2001-10-25 2007-08-28 Monsanto Technology Llc Tocopherol methyltransferase tMT2 and uses thereof
CA2478957C (fr) 2002-03-19 2013-07-02 Monsanto Technology, Llc Acides nucleiques et polypeptides se rapportant a l'enzyme homogentisate prenyl transferase ("hpt"), et utilisations correspondantes
EP1546334A4 (fr) 2002-08-05 2007-01-03 Monsanto Technology Llc Genes associes a la biosynthese du tocopherol et utilisation de ceux-ci

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6682728B1 (en) * 1993-10-13 2004-01-27 The United States Of America As Represented By The Department Of Health And Human Services Efficient and selective adenoviral-mediated gene transfer into vascular neointima

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0011165A1 *

Also Published As

Publication number Publication date
CA2343099A1 (fr) 2000-03-02
CA2343099C (fr) 2006-11-14
AU5572599A (en) 2000-03-14
WO2000011165A1 (fr) 2000-03-02
WO2000011165A9 (fr) 2000-06-22

Similar Documents

Publication Publication Date Title
CA2343575C (fr) Inhibition de la migration des cellules des muscles lisses au moyen de l'heme oxygenase (1)
US7772367B2 (en) C-terminal p53 palindromic peptide that induces apoptosis of cells with aberrant p53 and uses thereof
EP0998307B1 (fr) TRAITEMENT DES MALADIES VASCULAIRES PROLIFERATIVES A L'AIDE DE p27 ET DE SES PROTEINES HYBRIDES
US5856121A (en) Growth arrest homebox gene
US6251871B1 (en) P16 expression constructs and their application in cancer therapy
US7414035B2 (en) hKIS composition and methods of use
CA2343099C (fr) Compositions de hkis et procedes d'utilisation correspondants
US6482803B1 (en) Modification of mutated P53 gene in tumors by retroviral delivery of ribozyme A
EP0799243A1 (fr) PROTEINES p53 A DOMAINES DE TETRAMERISATION MODIFIES
WO2000018426A1 (fr) Inducteurs d'apoptose
US7163925B1 (en) p16 expression constructs and their application in cancer therapy
EP0806477B1 (fr) Utilisation, pour la fabrication d'un medicament, d'un adn recombinant comprenant un adn codant pour la proteine isoforme sm1 de la chaine lourde de la myosine de type muscle lisse
WO1998019686A9 (fr) Methodes therapeutiques pour lesion vasculaire
WO1998019686A1 (fr) Methodes therapeutiques pour lesion vasculaire
US5847083A (en) Modified p53 constructs which enhance DNA binding
US20050032728A1 (en) Tumor suppression through bicistronic co-expression of p53 and p14ARF
EP1007664B1 (fr) Utilisations d'un facteur de transcription de choc thermique
CA2430745A1 (fr) Facteur maitre de transcription de formation osseuse: compositions et procedes d'utilisation
US20020152486A1 (en) Vascular specific regulatory elements contained in the desmin 5' flanking region
JP3328260B2 (ja) ミオシン重鎖sm1アイソフォームタンパク質をコードするdnaをベクターdnaに組み込んだ組換え体dna並びに該組換え体dnaを含有する微生物及び動脈硬化治療剤
US20030219421A1 (en) Calbindin-D28k protection against glucocorticoid induced cell death
McCray Jr et al. lowa Research Online
WO1997010843A1 (fr) Constructions de p53 modifiees et utilisations leur convenant

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010321

AK Designated contracting states

Kind code of ref document: A1

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

Kind code of ref document: A1

Designated state(s): DE FR GB IE NL

RIN1 Information on inventor provided before grant (corrected)

Inventor name: BOEHM, MANFRED

Inventor name: NABEL, ELISABETH G.

Inventor name: NABEL, GARY J.

17Q First examination report despatched

Effective date: 20040205

RBV Designated contracting states (corrected)

Designated state(s): DE FR GB IE NL

RIN1 Information on inventor provided before grant (corrected)

Inventor name: BOEHM, MANFRED

Inventor name: NABEL, ELISABETH G.

Inventor name: NABEL, GARY J.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20090126