EP0932679A1 - METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-$g(a) NUCLEIC ACIDS - Google Patents

METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-$g(a) NUCLEIC ACIDS

Info

Publication number
EP0932679A1
EP0932679A1 EP97910794A EP97910794A EP0932679A1 EP 0932679 A1 EP0932679 A1 EP 0932679A1 EP 97910794 A EP97910794 A EP 97910794A EP 97910794 A EP97910794 A EP 97910794A EP 0932679 A1 EP0932679 A1 EP 0932679A1
Authority
EP
European Patent Office
Prior art keywords
promoter
tissue
expression
cells
vector
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
EP97910794A
Other languages
German (de)
English (en)
French (fr)
Inventor
Tattanahalli L. Nagabhushan
Deba P. Saha
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.)
Canji Inc
Original Assignee
Canji 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
Application filed by Canji Inc filed Critical Canji Inc
Priority to EP05013342A priority Critical patent/EP1591528A3/en
Publication of EP0932679A1 publication Critical patent/EP0932679A1/en
Ceased legal-status Critical Current

Links

Classifications

    • 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
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10351Methods of production or purification of viral material
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/85Vector systems having a special element relevant for transcription from vertebrates mammalian

Definitions

  • the human interferon alphas are a family of proteins comprising at least 24 subspecies (Zoon, K.C.,
  • interferon 9 1 (1987), Gresser, I., ed. , Academic Press, NY).
  • the interferon alphas were originally described as agents capable of inducing an antiviral state in cells but are now known as pleiotropic lymphokines affecting many functions of the immune system (Openakker et al . Experimentia 45:513 (1989)) .
  • IFN- ⁇ has been widely used for therapeutic purposes, including hairy cell leukemia, kaposi's sarcoma, renal cell carcinoma, non Hodgkin ' s lymphoma, T-cell leukemia, multiple and chronic myelogenous leukemia, malignant melanoma, bladder cell carcinoma, colon carcinoma (with 5-FU) , condyloma acuminata, rhinovirus and various forms of chronic viral hepatitis occurring as a result of hepatitis B virus (HBV) , hepatitis C virus (HCV) , non A non B virus (NANB) , or hepatitis ⁇ virus
  • HBV hepatitis B virus
  • HCV hepatitis C virus
  • NANB non A non B virus
  • IFN- ⁇ infection (Pestka AIDA Research & Human Retroviruses 8(5):776-786 (1992)). IFN- ⁇ has also been found to be highly effective against megakaryocytopoiesis and controlling thrombocytosis in patients with myeloproliferative disorders (Talpaz et al . Annals Int. Med. 99:789-792 (1983); Gisslinger et al. Lancet i:634-637 (1989); Ganser et al . Blood 70:1173-1179 (1987)).
  • Gene therapy techniques have the potential for limiting the exposure of a subject to a gene product, such as interferon, by targeting the expression of the therapeutic gene to a tissue of interest.
  • a gene product such as interferon
  • the ability to target the tissue of interest is one of the major challenges of gene therapy.
  • WIPO Patent Application Publication No. WO 93/15609 discloses the delivery of interferon genes to vascular tissue by administration of such genes to areas of vessel wall injury using a catheter system.
  • an adenoviral vector encoding a protein capable of enzymatically converting a prodrug, a "suicide gene", and a gene encoding a cytokine are administered directly into a solid tumor.
  • Transductional targeting refers to the selective entry into specific cells, achieved primarily by selection of a receptor ligand.
  • Positional targeting within the genome refers to integration into desirable loci, such as active regions of chromatin, or through homologous recombination with an endogenous nucleotide sequence such as a target gene.
  • Transcriptional targeting refers to selective expression attained by the incorporation of transcriptional promoters with highly specific regulation of gene expression tailored to the cells of interest.
  • tissue-specific promoters include the promoter for creatine kinase, which has been used to direct the expression of dystrophin cDNA expression in muscle and cardiac tissue (Cox et al . Nature 364:725-729 (1993)); and immunoglobulin heavy or light chain promoters for the expression of suicide genes in B cells (Maxwell et al . Cancer Res . 51:4299-4304 (1991)).
  • An endothelial cell-specific regulatory region has also been characterized (Roboudi et al . Mol . Cell. Biol. 14:999-1008 (1994)).
  • Amphotrophic retroviral vectors have been constructed carrying a herpes simplex virus thymidine kinase gene under the control of either the albumin or alpha- fetoprotein promoters (Huber et al . Proc . Natl. Acad. Sci. U.S.A. 88:8039-8043 (1991)) to target cells of liver lineage and hepatoma cells, respectively.
  • tissue specific promoters can be used in retroviral vectors (Hartzoglou et al . J. Biol. Chem. 265:17285-17293 (1990)) and adenovirus vectors (Friedman et al. Mol . Cell. Biol.
  • One aspect of the invention is a method for providing a patient with an interferon alpha polypeptide comprising introducing into a tissue of interest of the patient a vector comprising a nucleic acid segment encoding an interferon alpha polypeptide, the nucleic acid segment being operatively linked to a promoter having specificity for the tissue of interest, wherein the polypeptide is expressed in the tissue of interest.
  • Another aspect of the invention is a method for increasing interferon alpha levels in a tissue of interest in a patient comprising introducing into the tissue of interest a vector comprising a nucleic acid segment encoding an interferon alpha polypeptide, the nucleic acid segment being operatively linked to a promoter having specificity for the tissue of interest, wherein the interferon alpha polypeptide is expressed in the tissue of interest in the patient.
  • Another aspect of the invention is a method for treatment of cancer responsive to interferon alpha comprising administering to a cancerous tissue a vector comprising a nucleic acid segment encoding an interferon alpha polypeptide, the nucleic acid segment encoding an interferon alpha polypeptide, the nucleic acid segment being operatively linked to a promoter having specificity for the tissue, wherein the alpha interferon polypeptide is expressed in the tissue.
  • Another aspect of the invention is a method of treatment of hepatitis comprising administering to a patient's liver a vector comprising a nucleic acid segment encoding an interferon alpha polypeptide, the nucleic acid segment being operatively linked to a promoter having specificity for liver cells, wherein the interferon alpha polypeptide is expressed in the patient's liver.
  • a further aspect of the invention is a composition
  • a composition comprising a vector comprising a nucleic acid segment encoding an interferon alpha polypeptide, the nucleic acid segment being operatively linked to a promoter having specificity for a tissue of interest.
  • Figure 1 is a graph depicting the antiproliferative effects of interferon alpha on human prostate cancer cells.
  • Figure 2 is a graph depicting luciferase activity as a measure of luciferase expression driven by promoters of liver- specific genes.
  • the instant invention provides methods for the tissue specific expression of IFN- ⁇ using tissue specific promoters.
  • IFN- ⁇ as used herein is intended to include all subclasses of interferon alpha, deletion, insertion, or substitution variants thereof, biologically active fragments, and allelic forms.
  • Biologically active refers to any anti-viral or anti-proliferative activity as measured by techniques well known in the art (see, for example, Openakker et al . , supra; Moss an J. Immunol. Methods 65:55 (1983)).
  • Recombinant interferon alphas have been cloned and expressed in E. coli by several groups (for example, Weissmann et al . Science 209:1343-1349 (1980); Sreuli et al.
  • the interferon alpha is interferon alpha 2a or 2b (see, for example, WO 91/18927), although any interferon alpha may be used.
  • Nucleic acids encoding the IFN- ⁇ polypeptide can be DNA or RNA.
  • the phrase "nucleic acid sequence encoding" refers to a nucleic acid which directs the expression of a specific protein or peptide.
  • the nucleic acid sequences include both the DNA strand sequence that is transcribed into RNA and the RNA sequence that is translated into protein.
  • the nucleic acid sequences include both the full length nucleic acid sequences as well as non-full length sequences derived from the full length protein. It is further understood that the sequence includes the degenerate codons of the native sequence or sequences which may be introduced to provide codon preference in a specific host cell.
  • vector refers to viral expression systems, autonomous self-replicating circular DNA (plasmids) , and includes both expression and nonexpression plasmids. Where a recombinant microorganism or cell culture is described as hosting an "expression vector, " this includes both extrachromosomal circular DNA and DNA that has been incorporated into the host chromosome (s) . Where a vector is being maintained by a host cell, the vector may either be stably replicated by the cells during mitosis as an autonomous structure, or is incorporated within the host's genome.
  • a vector contains multiple genetic elements positionally and sequentially oriented, i.e., operatively linked with other necessary elements such that nucleic acid in the vector encoding IFN- ⁇ can be transcribed, and when necessary, translated in transfected cells .
  • gene as used herein is intended to refer to a nucleic acid sequence which encodes a polypeptide. This definition includes various sequence polymorphisms, mutations, and/or sequence variants wherein such alterations do not affect the function of the gene product.
  • gene is intended to include not only coding sequences but also regulatory regions such as promoters, enhancers, and termination regions. The term further includes all introns and other DNA sequences spliced from the mRNA transcript, along with variants resulting from alternative splice sites.
  • plasmid refers to an autonomous circular DNA molecule capable of replication in a cell, and includes both the expression and nonexpression types.
  • plasmid includes both extrachromosomal circular DNA molecules and DNA that has been incorporated into the host chromosome (s) .
  • a plasmid is being maintained by a host cell, the plasmid is either being stably replicated by the cells during mitosis as an autonomous structure or is incorporated within the host's genome.
  • recombinant protein or “recombinantly produced protein” refers to a peptide or protein produced using non-native cells that do not have an endogenous copy of DNA able to express the protein.
  • the cells produce the protein because they have been genetically altered by the introduction of the appropriate nucleic acid sequence.
  • the recombinant protein will not be found in association with proteins and other subcellular components normally associated with the cells producing the protein.
  • protein and “polypeptide” are used interchangeably herein.
  • the IFN- ⁇ is provided in an expression vector comprising the following elements linked sequentially at appropriate distances for functional expression: a tissue-specific promoter, an initiation site for transcription, a 3 ' untranslated region, a 5' mRNA leader sequence, a nucleic acid sequence encoding an alpha interferon polypeptide, and a polyadenylation signal. Enhancer sequences and other sequences aiding expression and/or secretion can also be included in the expression vector. Additional genes, such as those encoding drug resistance, can be included to allow selection or screening for the presence of the recombinant vector.
  • Such additional genes can include, for example, genes encoding neomycin resistance, multi-drug resistance, thymidine kinase, beta- galactosidase, dihydrofolate reductase (DHFR) , and chloramphenicol acetyl transferase.
  • tissue-specific promoters include the promoter for creatine kinase, which has been used to direct the expression of dystrophin cDNA expression in muscle and cardiac tissue (Cox et al . Nature 364:725-729 (1993)); immunoglobulin heavy or light chain promoters for the expression of genes in B cells; albumin or alpha-fetoprotein promoters to target cells of liver lineage and hepatoma cells, respectively.
  • tissue-specific expression elements for the liver include but are not limited to HMG-CoA reductase promoter (Luskey, Mol. Cell. Biol. 7 (5) : 1881-1893 (1987)); sterol regulatory element 1 (SRE-1; Smith et al . J. Biol. Chem. 265(4) :2306-2310 (1990); phosphoenol pyruvate carboxy kinase (PEPCK) promoter (Eisenberger et al . Mol. Cell Biol. 12(3) : 1396-1403 (1992)); human C-reactive protein (CRP) promoter (Li et al . J. Biol. Chem.
  • aldolase B promoter (Bingle et al . Biochem J. 294(Pt2) :473-9 (1993)); human transferrin promoter (Mendelzon et al. Nucl. Acids Res. 18 (19) : 5717-21 (1990); collagen type I promoter (Houglum et al . J. Clin. Invest. 94(2):808-14 (1994)).
  • Exemplary tissue-specific expression elements for the prostate include but are not limited to the prostatic acid phosphatase (PAP) promoter (Banas et al . Biochim. Biophvs . Acta. 1217(2) :188-94 (1994); prostatic secretory protein of 94 (PSP 94) promoter (Nolet et al . Biochim. Biophvs. ACTA 1098(2) :247- 9 (1991) ) ; prostate specific antigen complex promoter (Casper et al. J. Steroid Biochem. Mol. Biol. 47 (l-6):127-35 (1993)); human glandular kallikrein gene promoter (hgt-1) (Lilja et al. World J.
  • PAP prostatic acid phosphatase
  • tissue-specific expression elements for gastric tissue include but are not limited to the human H + /K + -ATPase alpha subunit promoter (Tanura et al . FEBS Letters 298: (2- 3) :137-41 (1992) ) .
  • Exemplary tissue-specific expression elements for the pancreas include but are not limited to pancreatitis associated protein promoter (PAP) (Dusetti et al . J. Biol. Chem. 268 (19) :14470-5 (1993)); elastase 1 transcriptional enhancer (Kruse et al . Genes and Development 7(5):774-86 (1993)); pancreas specific amylase and elastase enhancer promoter (Wu et al. Mol. Cell. Biol. 11 (9) :4423-30 (1991); Keller et al . Genes & Dev. 4(8):1316-21 (1990)); pancreatic cholesterol esterase gene promoter (Fontaine et al . Biochemistry 30 ( 28 ): 7008-14 (1991) ) .
  • PAP pancreatitis associated protein promoter
  • tissue-specific expression elements for the endometrium include but are not limited to the uteroglobin promoter (Helftenbein et al . Annal . NY Acad. Sci. 622:69-79 (1991) ) .
  • tissue-specific expression elements for adrenal cells include but are not limited to cholasterol side- chain cleavage (SCC) promoter (Rice et al . J. Biol. Chem. 265:11713-20 (1990) .
  • SCC cholasterol side- chain cleavage
  • tissue-specific expression elements for the general nervous system include but are not limited to gamma- gamma enolase (neuron-specific enolase, NSE) promoter (Forss- Petter et al . Neuron 5(2):187-97 (1990)).
  • NSE neuron-specific enolase
  • tissue-specific expression elements for the brain include but are not limited to the neurofilament heavy chain (NF-H) promoter (Schwartz et al . J. Biol. Chem. 269(18) :13444-50 (1994)).
  • tissue-specific expression elements for lymphocytes include but are not limited to the human CGL- 1/granzyme B promoter (Hanson et al . J. Biol. Chem. 266 (36):24433-8 (1991)); the terminal deoxy transferase (TdT) , lambda 5, VpreB, and lck (lymphocyte specific tyrosine protein kinase p561ck) promoter (Lo et al . Mol. Cell. Biol. 11(10) :5229-43 (1991)); the humans CD2 promoter and its
  • tissue-specific expression elements for the colon include but are not limited to .pp60c-src tyrosine kinase promoter (Talamonti et al . J. Clin. Invest 91(l):53-60 (1993)); organ-specific neoantigens (OSNs) , mw 40kDa (p40) promoter (Ilantzis et al . Microbiol . Immunol . 37(2): 119-28 (1993)); colon specific antigen-P promoter (Sharkey et al . Cancer 73(3 supp.) 864-77 (1994)).
  • tissue-specific expression elements for breast cells include but are not limited to the human alpha- lactalbumin promoter (Thean et al . British J. Cancer. 61(5) :773-5 (1990) ) .
  • Other elements aiding specificity of expression in a tissue of interest can include secretion leader sequences, enhancers, nuclear localization signals, endosmolytic peptides, etc.
  • these elements are derived from the tissue of interest to aid specificity.
  • Techniques for nucleic acid manipulation of the nucleic acid sequences of the invention such as subcloning nucleic acid sequences encoding polypeptides into expression vectors, labelling probes, DNA hybridization, and the like are described generally in Sambrook et al .
  • DNA encoding a sequence of interest is isolated and cloned, one can express the encoded proteins in a variety of recombinantly engineered cells. It is expected that those of skill in the art are knowledgeable in the numerous expression systems available for expression of DNA encoding. No attempt to describe in detail the various methods known for the expression of proteins in prokaryotes or eukaryotes is made here.
  • the expression of natural or synthetic nucleic acids encoding a sequence of interest will typically be achieved by operably linking the DNA or cDNA to a promoter (which is either constitutive or inducible) , followed by incorporation into an expression vector.
  • the vectors can be suitable for replication and integration in either prokaryotes or eukaryotes .
  • Typical expression vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of polynucleotide sequence of interest.
  • expression plasmids which contain, at the minimum, a strong promoter to direct transcription, a ribosome binding site for translational initiation, and a transcription/translation terminator.
  • the expression vectors may also comprise ' generic expression cassettes containing at least one independent terminator sequence, sequences permitting replication of the plasmid in both eukaryotes and prokaryotes, i.e., shuttle vectors, and selection markers for both prokaryotic and eukaryotic systems . See Sambrook et al .
  • the constructs of the invention can be introduced into the tissue of interest in vivo or ex vivo by a variety of methods.
  • the vector is introduced to cells by such methods as microinjection, calcium phosphate precipitation, liposome fusion, or biolistics.
  • the DNA is taken up directly by the tissue of interest.
  • the constructs are packaged into a viral vector system to facilitate introduction into cells .
  • Viral vector systems useful in the practice of the instant invention include adenovirus, herpesvirus, adeno- associated virus, minute virus of mice (MVM) , HIV, Sindbis virus, and retroviruses such as Rous sarcoma virus, and MoMLV.
  • the constructs of the instant invention are inserted into such vectors to allow packaging of the interferon expression construct, typically with accompanying viral DNA, infection of a sensitive host cell, and expression of the interferon- ⁇ gene .
  • a particularly advantageous vector is the adenovirus vector disclosed by Wills et al . Hum. Gene Therapy 5:1079-1088 (1994) .
  • the recombinant IFN- ⁇ constructs of the invention are conjugated to a cell receptor ligand for facilitated uptake (e.g., invagination of coated pits and internalizatio of the endosome) through a DNA linking moiety (Wu et al . J. Biol. Chem. 263:14621-14624 (1988); WO 92/06180).
  • the DNA constructs of the invention can be linked through a polylysine moiety to asialo-oromucocid, which is a ligand for the asialoglycoprotein receptor of hepatocytes .
  • viral envelopes used for packaging the constructs of the invention can be modified by the addition of receptor ligands or antibodies specific for a receptor to permit receptor-mediated endocytosis into specific cells (e.g., WO
  • the DNA constructs of the ' invention are linked to viral proteins, such as adenovirus particles, to facilitate endocytosis (Curiel et al . Proc . Natl . Acad. Sci. U.S.A. 88:8850-8854 (1991)).
  • molecular conjugates of the instant invention can include microtubule inhibitors (WO/9406922); synthetic peptides mimicking influenza virus hemagglutinin (Plank et al . J. Biol. Chem. 269:12918- 12924 (1994)); and nuclear localization signals such as SV40 T antigen (W093/19768) .
  • treatment is intended to refer to the introduction of nucleic acid encoding an alpha interferon to a patient for the purpose of exposing a tissue of interest, especially a tissue having one or more cells demonstrating some pathology, to alpha interferon.
  • a "cancerous" tissue is intended to refer to a tissue in which one or more cells is classified as cancerous, malignant, tumorous, precancerous, transformed, or as an adenoma or carcinoma, or any other synonym commonly used in the art for these conditions.
  • noncancerous cell as used herein is understood in the art as excluded from the definition of cancerous or cancer cell, and can include normal cells and cells displaying some pathological feature such as infection by a virus, bacterium, parasite, or other organism, cells affected by a hereditable condition that renders them less optimal than normal or wild type counterparts, cells affected by some presumed non- infectious disease state such as diabetes, etc., and cells which have survived any of these stresses, etc.
  • Treatment or therapy of any condition which would benefit from administration of IFN- ⁇ can begin prior to the diagnosis of the condition or at any time after diagnosis of a condition.
  • a patient suspected of having a precancerous lesion or an increased probability of developing some type of cancer can be treated with the compositions of the invention.
  • a person exposed to a pathogen, such as hepatitis B virus can be treated with the compositions of the invention before hepatitis is diagnosed.
  • suspected carriers of HBV or patients likely to become carriers can be treated after gross symptoms of the disease have improved.
  • the constructs of the invention are useful in the therapy of various cancers, hepatitis and other conditions in which the administration of IFN- ⁇ to raise IFN- ⁇ levels in tissues is advantageous, including but not limited to ulcerative colitis, rhinovirus infections, condyloma acuminata, laryngeal papillomitis; HIV infection, fibrosis, allergic diseases due to excess IL-4 and IgE production, and granulomatous disorders, such as Crohn ' s disease.
  • any tissue can be targeted for which some tissue-specific expression element, such as a promoter, can be identified, of particular interest is the tissue specific administration of IFN- ⁇ to raise IFN- ⁇ levels in cancerous tissues, such as human prostate carcinoma and hepatoma tissues.
  • the constructs of the invention can be used to raise IFN- ⁇ levels in tissues in pathological conditions in which non-cancerous cells are deficient in interferon production, i.e,, produce less interferon than the healthy cells, such as in chronic hepatitis B virus carriers (Nouri-Aria et al . Hepatolo ⁇ v 14 (6) : 1308-1311 (1991)).
  • the recombinant constructs are targeted to neighboring tissues or cells to raise the local concentration of interferon alpha in a cell population of interest.
  • compositions of the invention will be formulated for administration by manners known in the art acceptable for administration to a mammalian subject, preferably a human.
  • the compositions of the invention can be administered directly into a tissue by injection or into a blood vessel supplying the tissue of interest.
  • the compositions of the invention are administered "locoregionally" , i.e., intravesically, intralesionally, and/or topically.
  • the compositions of the invention are administered systemically by injection, inhalation, suppository, transdermal delivery, etc.
  • the compositions are administered through catheters or other devices to allow access to a remote tissue of interest, such as an internal organ.
  • the compositions of the invention can also be administered in depot type devices, implants, or encapsulated formulations to allow slow or sustained release of the compositions.
  • compositions for administration which comprise a solution of the compositions of the invention dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
  • an acceptable carrier preferably an aqueous carrier.
  • aqueous carriers may be used, e.g., water, buffered water, 0.8% saline, 0.3% glycine, hyaluronic acid and the like.
  • These compositions may be sterilized by conventional, well known sterilization techniques, or may be sterile filtered.
  • the resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • concentration of the compositions of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • compositions of the invention may also be administered via liposomes.
  • Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like.
  • the composition of the invention to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to a desired target, such as antibody, or with other therapeutic or immunogenic compositions.
  • liposomes either filled or decorated with a desired composition of the invention of the invention can delivered systemically, or can be directed to a tissue of interest, where the liposomes then deliver the selected therapeutic/immunogenic peptide compositions .
  • Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol.
  • the selection of lipids is generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream.
  • a variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al . Ann. Rev. Biophvs. Bioeng. 9:467 (1980), U.S. Patent Nos. 4,235,871, 4,501,728, 4,837,028, and 5 , 019 , 369 , incorporated herein by reference .
  • a liposome suspension containing a composition of the invention may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the composition of the invention being delivered, and the stage of the disease being treated.
  • nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more compositions of the invention of the invention, and more preferably at a concentration of 25%- 75%.
  • compositions of the invention are preferably supplied in finely divided form along with a surfactant and propellant.
  • Typical percentages of compositions of the invention are 0.01%-20% by weight, preferably 1%-10%.
  • the surfactant must, of course, be nontoxic, and preferably soluble in the propellant.
  • Representative of such agents are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride.
  • Mixed esters such as mixed or natural glycerides may be employed.
  • the surfactant may constitute 0.1%-20% by weight of the composition, preferably 0.25-5%.
  • the balance of the composition is ordinarily propellant.
  • a carrier can also be included, as desired, as with, e.g., lecithin for intranasal delivery.
  • the constructs of the invention can additionally be delivered in a depot-type system, an encapsulated form, or an implant by techniques well-known in the art. Similarly, the constructs can be delivered via a pump to a tissue of interest.
  • the compositions of the invention are administered ex vivo to cells or tissues explanted from a patient, then returned to the patient. Examples of ex vivo administration of gene therapy constructs include Arteaga et al . Cancer Research 56(5):1098- 1103 (1996); Nolta et al . Proc Natl . Acad. Sci . USA 93(6):2414-9 (1996); Koc et al .
  • LNCaP androgen dependent, ATCC #CRL 1740
  • PC-3 cells androgen independent, ATCC #CRL 1435
  • DU-145 androgen independent, ATCC #HTB 81
  • the cells were grown in 5 different concentrations (10, 10 2 , 10 3 , 10 ⁇ 10 5 IU/ml) of interferon- ⁇ 2b
  • PC-3 was cultured in Ham's F12 K medium (GIBCO BRL) supplemented with 7% fetal bovine serum
  • DU-145 was cultured in DMEM (GIBCO BRL) supplemented with 10% fetal calf serum
  • LnCaP was cultured in RPMI 1640 (GIBCO BRL ) supplemented with 5% fetal bovine serum.
  • Antiproliferative effects of interferon were measured by MTT assay (Mossman J. Immunol. Methods 65:55 (1983)).
  • PC-3 and DU-145 cells showed a consistent sensitivity to increasing concentrations of interferon plateauing at 10 4 IU/ml.
  • An expression vector was constructed having the complete cDNA sequence of interferon alpha 2b (IFN- ⁇ 2b) and the complete signal sequence for IFN- ⁇ 2b (Sreuli et al . Science 209:1343-1347 (1980); Goeddel et al . Nature 290:20-26 (1981); Henco et al . J. Mol. Biol. 185:227-260 (1985)) under control of an approximately 600 bp basal promoter for the prostate specific antigen gene (PSA) for tissue specific expression of IFN- ⁇ in prostate carcinoma cells.
  • PSA prostate specific antigen gene
  • full-length IFN- ⁇ 2b cDNA having its putative signal leader at the 5 ' end was cloned into the polycloning site at Malawi and Eco RI downstream of the CMV promoter in the mammalian expression vector PCDNA3 (Invitrogen) to create plasmid DIFN.
  • the 5 ' flanking sequence of the PSA gene containing the PSA promoter was inserted into the vector, replacing the CMV promoter, to create plasmid PSADIFN.
  • flanking sequences including basal promoters, from four human liver specific genes, albumin (HAL), ⁇ l- antitrypsin (HAT) , alpha feto protein (AFP) , and hydroxy-methyl- glutaryl CoA reductase (HMG) , were subcloned from ATCC 65731, ATCC 61597, ATCC 65735, and ATCC 59567, respectively, into pCRScript vector for use in interferon gene delivery and its tissue specific expression in hepatic cells (Luskey Mol. Cell. Biol. 7(5) :1881-1893 (1987); Minghetti et al . J. Biol. Chem.
  • pGLC contains the luciferase gene under the control of the SV40 promoter and the SV40 enhancer.
  • Luciferase expression by the four liver specific promoter sequences in the transfected cells was compared with the control plasmid pGLC and the vector pGL3.
  • the data are summarized in Figure 2 (HAL result not shown) .
  • Three cell types were transfected by DNA constructs having the salient features indicated. Solid bars represent human hepatoma HepG2 cells; cross-hatched bars represent Chinese hamster ovary cells (CHO) ; diagonal bars represent human hepatoma Hep3B cells (Hep3B) .
  • pGLB is a negative control plasmid
  • pGLC is a positive control plasmid
  • HAT denotes the human ⁇ l-antitrypsin promoter
  • HMG denotes the human hydroxy-methyl-glutaryl CoA reductase promoter
  • AFP denotes the human ⁇ -feto protein promoter.
  • HAT human ⁇ l-antitrypsin
  • HAT promoter Expression driven by the HAT promoter can be further optimized by constructing an expression vector with a liver specific enhancer sequence such as the human ⁇ -fetoprotein enhancer (Watanabe, et al . J. Biol. Chem. 262 ( 10 ): 4812-4818
  • a liver specific enhancer sequence such as the human ⁇ -fetoprotein enhancer (Watanabe, et al . J. Biol. Chem. 262 ( 10 ): 4812-4818

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biochemistry (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Saccharide Compounds (AREA)
EP97910794A 1996-10-18 1997-10-16 METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-$g(a) NUCLEIC ACIDS Ceased EP0932679A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP05013342A EP1591528A3 (en) 1996-10-18 1997-10-16 Method and compositions for delivery and expression of interferon-alpha nucleic acids

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US73381596A 1996-10-18 1996-10-18
US733815 1996-10-18
PCT/US1997/017928 WO1998017801A1 (en) 1996-10-18 1997-10-16 METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-α NUCLEIC ACIDS

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP05013342A Division EP1591528A3 (en) 1996-10-18 1997-10-16 Method and compositions for delivery and expression of interferon-alpha nucleic acids

Publications (1)

Publication Number Publication Date
EP0932679A1 true EP0932679A1 (en) 1999-08-04

Family

ID=24949214

Family Applications (2)

Application Number Title Priority Date Filing Date
EP97910794A Ceased EP0932679A1 (en) 1996-10-18 1997-10-16 METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-$g(a) NUCLEIC ACIDS
EP05013342A Withdrawn EP1591528A3 (en) 1996-10-18 1997-10-16 Method and compositions for delivery and expression of interferon-alpha nucleic acids

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP05013342A Withdrawn EP1591528A3 (en) 1996-10-18 1997-10-16 Method and compositions for delivery and expression of interferon-alpha nucleic acids

Country Status (20)

Country Link
EP (2) EP0932679A1 (es)
JP (2) JP2001502540A (es)
KR (1) KR20000049243A (es)
AR (1) AR013618A1 (es)
AU (1) AU4808097A (es)
BR (1) BR9712362A (es)
CA (1) CA2269100A1 (es)
CZ (1) CZ131699A3 (es)
DE (1) DE932679T1 (es)
ES (1) ES2136583T1 (es)
HU (1) HUP9904219A2 (es)
ID (1) ID24500A (es)
IL (1) IL129387A0 (es)
NO (1) NO991839L (es)
NZ (1) NZ335134A (es)
PL (1) PL332856A1 (es)
SK (1) SK50699A3 (es)
TR (1) TR199901190T2 (es)
WO (1) WO1998017801A1 (es)
ZA (1) ZA979295B (es)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9912925D0 (en) * 1999-06-03 1999-08-04 Angeletti P Ist Richerche Bio Adenoviral vectors encoding interferon and their use in gene therapy
US8058248B2 (en) * 2001-04-26 2011-11-15 The United States Of America As Represented By The Secretary Of Agriculture Foot and mouth disease virus vaccine comprising interferons
US7217796B2 (en) 2002-05-24 2007-05-15 Schering Corporation Neutralizing human anti-IGFR antibody
EP1694850B1 (en) 2003-11-12 2011-06-29 Schering Corporation Plasmid system for multigene expression
PE20050928A1 (es) 2003-11-21 2005-11-08 Schering Corp Combinaciones terapeuticas de anticuerpo anti-igfr1
BRPI0417451A (pt) * 2003-12-10 2007-04-10 Canji Inc método de tratar um tumor resistente a interferon
ES2356830T3 (es) 2004-12-03 2011-04-13 Schering Corporation Biomarcadores para preselección de pacientes para terapia de anti-igf-1r.
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
KR101449587B1 (ko) * 2006-09-14 2014-10-10 메드제닉스 메디칼 이스라엘 리미티드 장기 지속형 약물 제형
ES2439954T3 (es) 2007-02-23 2014-01-27 Her Majesty The Queen, In Right Of Canada, As Represented By The Minister Of National Defence Vectores víricos recombinantes para la prevención y la protección contra la infección por alfavirus
BRPI1012951A2 (pt) 2009-06-09 2016-07-26 Defyrus Inc "administração de interferon para profilaxia ou tratamento de infecção por patôgeno"
JP2013532152A (ja) 2010-06-15 2013-08-15 メドジェニクス・メディカル・イスラエル・リミテッド 長期持続性の医薬製剤
CN110636854A (zh) * 2017-03-31 2019-12-31 阿卡尼斯生物技术F&E有限责任两合公司 非黑素瘤皮肤癌(nmsc)的预防和治疗

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4238778C2 (de) * 1992-11-12 1995-11-02 Max Delbrueck Centrum Vektor zur Expression von therapeutisch relevanten Genen
US5631236A (en) * 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
DE69434594T2 (de) * 1993-10-25 2006-09-21 Canji, Inc., San Diego Rekombinante adenoviren-vektor und verfahren zur verwendung
US5830686A (en) * 1994-01-13 1998-11-03 Calydon Tissue-specific enhancer active in prostate
DE4407859C1 (de) * 1994-03-04 1995-03-02 Max Planck Gesellschaft Vektor für die leberspezifische Gentherapie
JP4216350B2 (ja) * 1994-09-19 2009-01-28 大日本住友製薬株式会社 動物細胞感染用の組換えdnaウイルスベクター
US5789244A (en) * 1996-01-08 1998-08-04 Canji, Inc. Compositions and methods for the treatment of cancer using recombinant viral vector delivery systems

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9817801A1 *

Also Published As

Publication number Publication date
ES2136583T1 (es) 1999-12-01
JP2008301832A (ja) 2008-12-18
AR013618A1 (es) 2001-01-10
CA2269100A1 (en) 1998-04-30
NO991839L (no) 1999-06-15
TR199901190T2 (xx) 1999-07-21
ZA979295B (en) 1998-04-20
EP1591528A2 (en) 2005-11-02
NZ335134A (en) 2001-05-25
SK50699A3 (en) 2000-05-16
JP2001502540A (ja) 2001-02-27
PL332856A1 (en) 1999-10-25
CZ131699A3 (cs) 1999-07-14
NO991839D0 (no) 1999-04-16
BR9712362A (pt) 1999-08-31
WO1998017801A1 (en) 1998-04-30
AU4808097A (en) 1998-05-15
HUP9904219A2 (hu) 2000-12-28
EP1591528A3 (en) 2005-11-16
ID24500A (id) 2000-07-20
IL129387A0 (en) 2000-02-17
KR20000049243A (ko) 2000-07-25
DE932679T1 (de) 2000-03-09

Similar Documents

Publication Publication Date Title
JP2008301832A (ja) インターフェロン−α核酸の送達および発現のための方法および組成物
US20080166373A1 (en) METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-alpha NUCLEIC ACIDS
Miller Human gene therapy comes of age
US20070244064A1 (en) Methods and compositions to induce antitumor response
JP4219335B2 (ja) インターフェロンβのような分泌タンパク質をコードする遺伝子を使用する治療のための方法および組成物
AU723660B2 (en) Tissue specific expression of retinoblastoma protein
WO1998021228A9 (en) Tissue specific expression of retinoblastoma protein
Narumi et al. Adenovirus vector-mediated perforin expression driven by a glucocorticoid-inducible promoter inhibits tumor growth in vivo
AU2007242949A1 (en) Methods and compositions for delivery and expression of interferon-alpha nucleic acids
AU3521301A (en) Methods and compositions for delivery and expressions of interferon-alpha nucleic acids
WO2000022124A2 (en) Methods and compositions to induce antitumor response
MXPA99003563A (es) Metodos y composiciones para la distribucion y expresion de acidos nucleicos de interferon
WO1997035993A2 (en) Psa positive regulating (psar) sequences and uses thereof
Xian‐Jun et al. Tissue‐specific activity of heterologous viral promoters in primary rat hepatocytes and Hep G2 cells
AU3521101A (en) Methods and compositions for delivery and expression of interferon-alpha nucleic acids
IL129922A (en) A unique form of expression for retinoblastoma protein tissue
US7087582B1 (en) Combination for site-specifically transforming cells in vivo comprising a double-balloon catheter and nucleic acid comprising a gene encoding P21
CN1240480A (zh) 运送和表达干扰素-α核酸的方法和组合物
MXPA99004499A (es) Expresion de proteina de retinoblastoma especifica para tejido

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: 19990511

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: LT PAYMENT 19990511;LV PAYMENT 19990511;RO PAYMENT 19990511

ITCL It: translation for ep claims filed

Representative=s name: RITORATO CON ISTANZA DEL 2.9.99;ING. A. GIAMBROCON

ITCL It: translation for ep claims filed

Representative=s name: ING. A. GIAMBROCONO & C. S.R.L.

EL Fr: translation of claims filed
REG Reference to a national code

Ref country code: ES

Ref legal event code: BA2A

Ref document number: 2136583

Country of ref document: ES

Kind code of ref document: T1

TCAT At: translation of patent claims filed
RTI1 Title (correction)

Free format text: METHODS AND COMPOSITIONS FOR DELIVERY AND EXPRESSION OF INTERFERON-ALPHA NUCLEIC ACIDS

DET De: translation of patent claims
17Q First examination report despatched

Effective date: 20030602

APBN Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2E

APBR Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3E

APAF Appeal reference modified

Free format text: ORIGINAL CODE: EPIDOSCREFNE

APBT Appeal procedure closed

Free format text: ORIGINAL CODE: EPIDOSNNOA9E

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: 20070329

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1021992

Country of ref document: HK