CA2308575A1 - Hyperthermic inducible expression vectors for gene therapy and methods of use thereof - Google Patents

Hyperthermic inducible expression vectors for gene therapy and methods of use thereof Download PDF

Info

Publication number
CA2308575A1
CA2308575A1 CA002308575A CA2308575A CA2308575A1 CA 2308575 A1 CA2308575 A1 CA 2308575A1 CA 002308575 A CA002308575 A CA 002308575A CA 2308575 A CA2308575 A CA 2308575A CA 2308575 A1 CA2308575 A1 CA 2308575A1
Authority
CA
Canada
Prior art keywords
promoter
expression
cell
expression construct
gene
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.)
Abandoned
Application number
CA002308575A
Other languages
French (fr)
Inventor
Tom Tsang
Eugene Gerner
David T. Harris
Evan Hersh
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.)
Arizona Board of Regents of University of Arizona
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2308575A1 publication Critical patent/CA2308575A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • 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
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron

Abstract

Methods and compositions are provided for transgene expression in target cells. Expression constructs using an inducible amplification system to drive expression of a therapeutic gene or other gene of interest in mammalian host cells are provided, as well as methods therefor. Inducible expression of the transgenes at high levels under physiologic conditions results from induction by hyperthermic conditions relative to the basal temperature of the host cells.

Description

HYPERTHERMIC INDUCIBLE EXPRESSION VECTORS FOR
GENETHERAPY AND METHODS OF USE THEREOF
BACKGROUND OF THE INVENTION
1. Field of the Invention The present invention relates generally to the field of gene therapy. More particularly, it concerns methods and compositions for increasing transgene expression.
2. Description of Related Art Gene therapy now is thought to be widely applicable in the treatment of a variety of cancers and a number of other diseases. Viral vectors are one method employed as a gene delivery system. A great variety of viral expression systems have been developed and assessed for their ability to transfer genes into somatic cells. In particular, retroviral and adenovirus based vector systems have been investigated extensively over a decade. Recently, adeno-associated virus (AAV) has emerged as a potential alternative to the more commonly used retroviral and adenoviral vectors. Lipid vectors including cationic lipids and Iiposomes also are used to deliver plasmid DNA containing therapeutic genes.
The therapeutic treatment of diseases and disorders by gene therapy involves the transfer and stable or transient insertion of new genetic information into cells.
The correction of a genetic defect by re-introduction of the normal allele of a gene encoding the desired function has demonstrated that this concept is clinically feasible (Rosenberg et al., New Eng. J. Med., 323:570 (1990)). Indeed, preclinical and clinical studies covering a large range of genetic disorders currently are underway to solve basic issues dealing with gene transfer efficiency, regulation of gene expression, and potential risks of the use of viral vectors. The majority of clinical gene transfer trials that employ viral vectors perform ex vivo gene transfer WO 99/23216 PCTIiJS98123387 into target cells which are then administered in vivo. Viral vectors also may be given in vivo but repeated administration may induce neutralizing antibody.
A major issue facing potential clinical application of gene therapy is the question of how to heterologous genes expressed in clinically significant quantities in selected tissues of the subject. Gene regulatory elements provide a potential answer to that question. Gene regulatory elements such as promoters and enhancers possess cell type specific activities and can be activated by certain induction factors via responsive elements. The use of such regulatory elements as promoters to drive gene expression facilitates controlled and restricted expression of heterologous genes in vector constructs. For instance, heat shock promoters can be used to drive expression of a heterologous gene following heat shock.
US Patent Nos. 5,614,381, 5,646,410 and WO 89/00603, refer to driving transgene expression using heat shock at temperatures greater than 42°C. These temperatures are not practicable in human therapy as they can not be maintained for a sustained period of time without harm to the individual.
Gene therapy could be used in combination with a variety of conventional cancer therapy treatments including cytotoxic drugs and radiation therapies.
It has been shown that hyperthermia enhances the cell killing effect of radiation in vitro (Harisiadis et al., Cancer, 41:2131-2142 (1978)), significantly enhances tumor response in animal tumors in viva and improves the outcome in randomized clinical trials. However, the major problem with the use of hyperthermia treatment is that the hyperthermia system can not adequately heat large and deep tumors.
Thus, it would be useful to develop vectors that may be used at temperatures of 42°C and below, systemically or locally, to treat a patient such that the expression of the therapeutic genes) is activated preferentially in regions of the body that have been subjected to conditions which induce such expression.
SUMMARY OF THE INVENTION
The present invention provides methods for effecting the inducible expression of polynucleotides in cells. In particular, the use of heat shock promoters in methods for effecting the inducible expression of polynucleotides in mammalian cells is taught. The present invention overcomes deficiencies in the prior art by providing heat shock-controlled vectors that may be used at temperatures of 42°C and below. These methods may be used to treat a patient via the inducible expression of a therapeutic gene.
In one embodiment, the present invention provides a method for effecting l0 transgene expression in a mammalian cell that comprises first providing an expression construct that comprises both (i) an inducibie promoter operably linked to a gene encoding a transactivating factor and (ii) a second promoter operably linked to a selected polynucleotide. The second promoter is activated by the transactivating factor expressed by the same construct. The method then includes the step of introducing the expression construct into the cell. Finally, the cell is subjected to conditions which activate the inducible promoter and result in the expression of the selected polynucleotide.
In a preferred embodiment of the invention, the inducible promoter is a heat shock promoter and the conditions which activate the heat shock promoter are hyperthermic conditions. The hyperthermic conditions may comprise a temperature between about basal temperature and about 42°C. As used herein the basal temperature of the cell is defined as the temperature at which the cell is normally found in its natural state, for example, a cell in skin of a mammal may be at temperatures as low as 33°C whereas a cell in the liver of an organism may be as high as 39°C. In specific embodiments, the application of hyperthermia involves raising the temperature of the cell from basal temperature, most typically 37°C. to about 42°C or less. Alternatively, the hyperthermic conditions may range from about 38°C to about 41°C, or from about 39°C to about 40°C. The heat shock promoter is optionally derived from a promoter selected from the group of the heat shock protein (HSP) promoters HSP70, HSP90, HSP60, HSP27, HSP72, HSP73, HSP25 and HSP28. The ubiquitin promoter may also be used as the heat-shock inducible promoter in the expression construct. A minimal heat shock promoter derived from HSP70 and comprising the first approximately 400 by of the HSP70B
promoter may optionally be used in the invention.
In an alternative embodiment, the inducible promoter comprises a hypoxia-responsive element (HR,E). This hypoxia-responsive element may optionally contain at least one binding site for hypoxia-inducible factor-1 (HIF-1).
In one embodiment of the invention, the second promoter may be selected l0 from the group consisting of an human immunodeficiency virus-1 (HIV-1) promoter and a human immunodeficiency virus-2 (HIV-2) promoter. In preferred embodiments, the transactivating factor may be a transactivator of transcription (TAT).
The selected polynucleotide may code for a protein or a polypeptide. For instance, the selected polynucleotide may encode any one of the following proteins:
ornithine decarboxylase antizyme protein, p53, p16, neu, interleukin-1 (IL 1), interleukin-2 (IL2), interleukin -4 (IL4), interleukin-7 (IL7}, interleukin-12 (IL12), interleukin-15 (IL 15), FLT-3 ligand, granulocyte-macrophage stimulating factor (GM-CSF), granulocyte-colony stimulating factor (G-CSF}, gamma-interferon (IFNy), alpha-interferon (IFNa), tumor necrosis factor (TNF), herpes simplex virus thymidine kinase (HSV-TK), I-CAM1, human leukocyte antigen-B7 (HLA-B7), or tissue inhibitor of metalloproteinases (TIMP-3). In such an embodiment, the selected polynucleotide is positioned in a sense orientation with respect to the second promoter.
Alternatively, expression of the selected polynucleotide may involve transcription but not translation and produces a ribozyme. In this embodiment, the selected polynucleotide is also positioned in a sense orientation with respect to the second promoter.

WO 99/23216 PCTIUS98/23387~
In still another alternative embodiment, the expression of the selected polynucleotide involves transcription but not translation and results in an RNA
molecule which serves as an antisense nucleic acid. In such an embodiment, the selected polynucleotide may be the target gene, or a fragment thereof, which is 5 positioned in the expression construct in an antisense orientation with respect to said second promoter.
The expression construct may further comprise a gene encoding a selectable marker, such as hygromycin resistance, neomycin resistance, puromycin resistance, zeocin, gpt, DHFR, green fluorescent protein or histadinol. Alternatively, the expression construct may further comprise (i) a second selected polynucleotide which is operably linked to said second promoter, and (ii) an internal ribosome entry site positioned between said first and second selected polynucleotides.
The cell may be a tumor cell, a cell located within a tumor, or a cell located within a mammal. The introduction of the expression construct into the cell may occur in vitro or in vivo. In an one embodiment, the introduction of the expression construct into the cell is mediated by a delivery vehicle selected from the group consisting of liposomes, retroviruses, adenoviruses, adeno-associated viruses, lentiviruses, herpes simplex viruses, and vaccinia viruses.
In another embodiment of the invention, a method of providing a subject with a therapeutically effective amount of a product of a selected gene is provided.
This method involves providing a first expression construct which comprises an inducible promoter operably linked to a gene encoding a transactivating factor and providing a second expression construct which comprises a second promoter operably linked to a selected polynucleotide, where the second promoter is activated by the transactivating factor encoded by the first expression construct.
The first and second expression construct are introduced into the desired cell of said subject and that cell is subjected to conditions which activate the inducible promoter, so that expression of the selected polynucleotide is induced. In a preferred embodiment, the first and second expression constructs are present on the same vector. Also, the inducible promoter is preferably a heat shock promoter and the activating conditions comprise a temperature below 42°C and above about basal temperature.
The introduction of one or both of the expression constructs may be performed either ih vivo or ex vivo. The expression product of the selected polynucleotide may optionally be deleterious to a pathogen in the subject, such as a virus, bacterium, fungus, or parasite. Alternatively, the expression product of the selected polynucleotide may inhibit the growth of the cell of the subject. In still another alternative embodiment of the invention, the expression product of the selected polynucleotide replaces a deficient protein in the subject.
Alternatively, the expression product of the selected polynucleotide rnay promote nerve regeneration.
In further embodiments, there is provided a method of treating cancer in a mammal, such as a human, comprising the steps of (a) providing an expression construct that comprises (i} an inducible promoter, preferably a heat shock promoter, which is operably linked to a gene encoding a transactivating factor; and (ii) a second promoter operably linked to a selected polynucleotide, wherein the second promoter is activated by the transactivating factor; (b) introducing said expression construct into a tumor cell; and (c) subjecting the tumor cell to conditions which activate the inducible promoter so that the selected polynucleotide is expressed in high enough quantities to inhibit the growth of the tumor cell. If the inducible promoter is a heat shock promoter, the activating conditions comprise a temperature below about 42°C and above about basal temperature.
This method further may comprise treating said tumor cell with an established form of therapy for cancer which is selected from the group consisting of external beam radiation therapy, brachytherapy, chemotherapy, and surgery.
The cancer may optionally be selected from the group consisting of cancers of the brain, lung, liver, spleen, kidney, lymph node, small intestine, pancreas, blood cells, colon, stomach, breast, endometrium, prostate, testicle, ovary, vulva, cervix, skin, head and neck, esophagus, bone marrow and blood.
In one particular embodiment of the invention, the selected polynucleotide is ornithine decarboxylase antizyme protein. .After the cell is subjected to conditions which activate the inducible promoter of the expression construct in the tumor cell, the tumor cell is treated with the radioprotector WR-33278 or WR-1065. Lastly, the tumor cell is treated with radiation therapy.
Methods for provoking an immune response in a mammal, such as a human, are also provided by the present invention. The provoked immune response may constitute either a humoral immune response or a cellular immune response. In one embodiment, the method comprises (a) providing an expression construct that comprises (i) an inducible promoter, preferably a heat shock promoter, which is operably linked to a gene encoding a transactivating factor; and (ii) a second promoter operably linked to a selected polynucleotide, wherein the second promoter is activated by the transactivating factor; (b) introducing said expression construct into a cell in the mammal; and (c) subjecting the cell to conditions which activate the inducible promoter so that the selected polynucleotide is expressed highly enough to provoke an immune response in the mammal. If the inducible promoter is a heat shock promoter, the activating conditions comprise a temperature below about 42°C and above about basal temperature.
In one embodiment, the immune response which is provoked is directed against the cell in the mammal which contains the expression construct. 'The method may also optionally involve treating the cell with an established form of therapy for cancer selected from the group consisting of chemotherapy, external beam radiation therapy, brachytherapy, and surgery.
In another embodiment, there is provided an expression construct comprising (a) a gene encoding a transactivating factor; (b) an inducible promoter operably linked to the gene; (c) a selected polynucleotide; and (d) a second promoter which is operably linked to the selected polynucleotide. The second promoter of the construct is activated by the transactivating factor. In a preferred embodiment, the inducible promoter is a heat shock promoter and the expression of the selected polynucleotide can be induced by hyperthermic conditions comprising a temperature below about 42°C and above about 37°C. In an alternative embodiment, the inducible promoter of the expression construct may comprise a hypoxia-responsive element. The expression construct may also comprise a second selected polynucleotide which is also operably linked to the second promoter and separated by the first selected polynucleotide by a IRES.
A cell comprising the expression construct is also provided. The provided expression construct can also optionally be used in a method of altering the genetic material of a mammal.
Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
FIG. 1 depicts the basic vector used for quantitating heat shock promoter activity. The plasmid contains a minimal promoter derived from the HSP70B
promoter (StressGen). A reporter gene, such as Enhanced Green Fluorescence Protein (EGFP), ~i-gal, or IL-2 is easily inserted into the multiple cloning site (MCS) so that it is expressed under control of the minimal HSP70B promoter.
The plasmid also contains the neomycin and ampicillin resistance genes for selectability in mammalian cells as well as the standard elements for growth in a bacterial system. The S8 plasmid comprises the plasmid shown with EGFP inserted in the multiple cloning site.
FIG. 2 shows fluorescence activated cell sorting (FACS) histograms for DU-145 cells stably transfected with the S8 plasmid. Fluorescence increases from left to right. The top histogram is from transfected DU-145 cells which have not been IO subjected to heat shock. The bottom histogram is from transfected DU-145 cells which have been subjected to a 42°C heat shock for 1 hour.
FIG. 3 shows FACS histograms for three different populations of S8-transfected MCF7 cells. The MCF7 cells, transfected with the S8 construct, were sorted by FACS. The original population came from a polyclonal selected cell line.
That cell line's activated (i. e., cells expressing EGFP) population was separated from the non-activated population. After the sort, the positive population was grown and then re-sorted to obtain a more purely positive cell line. In this case, the polyclonal MCF7-S8-P cells were sorted twice yielding the highly positive population MCF7-S8-PS2.
FIG. 4 shows expression of EGFP in different cell lines assayed by FACS.
Cell lines were transfected with the plasmid S8. The cells were then cloned or a polygonal line was grown. In some cases the cell lines were sorted for EGFP
expression by FACS. The total mean fluorescence was quantified and graphed.
FIG. 5 shows expression of EGFP in stably transfected DU-145 cells which have been twice sorted (DU-S8-PS2) following heat shock. The DU-S8-PS2 cells were heated at either 40°C or 42°C and allowed to recover for various times. The cells were then analyzed by FACS.

FIG. 6 shows expression levels of EGFP in stably transfected DU-145 cells 16 hours after exposure to heat stresses. One population of cells (DU-S8-PS2) was stably transfected with the S8 plasmid. Another population {DU-V9-PS2) was stably transfected with the V9 plasnlid, a plasmid identical to S8 except that the 5 EGFP of the V9 plasmid is operably linked to a CMV promoter, rather than HSP
70B (see FIG. 7). The cells were heated at various temperatures and allowed to recover for 15 hours. Non-transfected DU-145 cells were included as a control.
FIG. 7 shows a schematic diagram of the plasmid V9 which contains a CMV
promoter that is operably linked to the gene encoding the Enhanced Green l0 Fluorescence Protein {EGFP) FIG. 8 shows the basic vector design for a vector containing a second promoter which allows for amplification of the heat shock response. The plasmid contains a multiple cloning site {MCS) operably linked to HSP70B promoter, but also contains a therapeutic gene operably linked to a second promoter. The plasmid also contain the neomycin resistance gene, the ampicillin resistance gene, and standard elements for growth in bacteria. In the plasmid pCB, the second promoter is the HIV-1 long terminal repeat (LTR) and the therapeutic gene is IL2. In pfl2, tat is inserted in the MCS, the second promoter is the HIV-1 LTR, and the therapeutic gene is IL2. Another plasmid, p007, is the same as pfl2, except that the HIV-2 LTR is used as the second promoter.
FIG. 9 shows amplifier constructs containing the therapeutic gene IL-2 driven by either the HIV-1 or the HIV-2 promoter. The amplifier part is controlled by either the CMV or the HSP 70 promoter driving TAT expression. The plasmids also contain the neomycin resistance gene and elements for growth in bacteria.
These constructs were used in the amplifier studies of Examples 2 and 3. FIG.

shows a plasmid designated X14 containing a CMV-TAT-HIV-1-IL2 expression cassette; FIG. 9B shows a plasmid designated Y15 containing a CMV-TAT-HIV-2-IL2 expression cassette; FIG. 9C shows a plasmid designated pfl2 containing an WO 99l23Z16 PCTIUS98/Z338~

HSP-TAT-HIV-1-IL2 expression cassette; and FIG. 9D shows a plasmid designated p007 containing an HSP-TAT-HIV-2-IL2 expression cassette.
FIG 10. shows the DNA sequence of the BamH 1-HindIII fragment of p173QR from StressGen Biotechnology Corp. This fragment contains the approximately 0.4kb minimal HSP70B promoter fragment used in constructs of the specific examples, Example 1 and 3, below.
DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
1. The Present Invention Gene therapy faces two major technical problems: how to both regulate and enhance the expression of therapeutic genes in vivo. The present invention addresses both of these questions by combining hyperthermia treatment with inducible expression constructs. The inventors have demonstrated increases in the efficiency of specific, inducible gene expression.
The ability to express therapeutic genes) at very high levels and the ability to control the levels of expression are important objectives in the development of gene therapy. The inventors have created new sets of expression vectors to address these objectives. The inventors use an amplifier strategy to drive the expression of the genes) of interest. The amplifiers consist of the human HSP70B promoter driving expression of proteins that are transcriptional activators of other promoters, which, in turn, drive reporter genes. These additional promoters and their operably linked reporter genes are preferably included in the same vector with the promoter element and the gene encoding the transactivating protein.
In transfection studies of mammalian cells using human IL-2 as the reporter gene, the inventors have shown that gene expression was dramatically increased using their amplifier constructs for all temperature conditions used, compared to reporter gene expression produced by the constitutive CMV promoter or by HSP70B alone (see specific example, Example 3, below). Constructs containing both the HSF70B promoter, upstream of the human immunodeficiency virus (HIV) tat gene, and the HIV 1 or HIV2 long terminal repeats, upstream of the interleukin-2 (IL-2) gene, exhibited promoter activity at 37°C which was further amplified by heat shock. Co-transfection experiments indicated that the activities of the HSP, HSP/HIV 1 and HSP/HIV2 promoter expression constructs were 0.4, 6.9 and 83.3, respectively, times that of the CMV promoter expression construct in mammalian cells. These data indicate that, while less active than the CMV promoter by itself, this minimal heat shock promoter can be used in conjunction with a second promoter to markedly amplify gene expression while still maintaining some temperature dependence.
Earlier studies have examined the use of the heat shock promoter to drive the expression of transactivating proteins to conditionally express other promoters (Schweinfest et al., Gene, 71(1):207-2I0, 1988; EPO O1 18393; WO 89/00603, U.S. Patent No. 5,614,381; U.S. Patent No. 5,646,010; EP 0 299 127). The inventions described herein differ from these earlier approaches, for example, by use of 1) different heat shock promoters, (Schweinfest et al., use Drosophila promoters) 2) different modes of delivery (the present inventors have incorporated both promoters into a single construct - whereas others have used co-transfection) 3) different temperatures for induction (the earlier work used temperatures greater than 42°C, whereas the present invention advantageously operates at temperatures of 42°C and lower); and 4) use in gene therapy context rather industrial production.
Furthermore, the present inventors are able to use either HIV-1 or HIV-2 promoters and the present invention shows a clear distinction in the expression levels resulting from these two promoters.
In a preferred aspect of the present invention, methods of effecting transgene expression in a mammalian cell by using a heat shock inducible element are provided. The heat shock sequence is used to drive the expression of a transactivating gene. Thus, when the expression construct is subjected to hyperthermia, the expression of the transactivating element is induced. The transactivating gene acts upon a second promoter which becomes activated to drive the expression of the therapeutic gene of interest. In a particular embodiment, a promoter derived from the HSP70 promoter is employed. A particularly useful aspect of this promoter is that it has a low basal level of expression at ambient temperatures and is inducible. The present invention further provides methods of providing a subject with a therapeutically effective amount of a gene product and for inhibiting the growth of a cell or provoking an immune response.
Compositions and methods employed in order to meet the objectives of the present invention are discussed in further detail herein below.
2. Heat Shock Response The heat shock or stress response is a universal response occurring in organisms ranging from plants to primates. It is a response that can be elicited as a result of not only heat shock, but also as a result of a variety of other stresses including ischemia, anoxia, glucose deprivation, ionophores glucose and amino acid analogues, ethanol, transition series metals, drugs, hormones and bacterial and viral infections. Furthermore, there is evidence that overexpression of heat shock protein genes may be associated with enhanced proliferation and stress of tumor cells (Finch et al., Cell Growth and Differentiation 3(5):269-278, 1992). This response is characterized by the synthesis of a family of well conserved proteins of varying molecular sizes that are differently induced and localized. These proteins are among the most phylogenetically conserved and are characterized according to their weights.
The transcriptional activation of stress protein-encoding genes occurs within minutes in response to environmental and or physiological trauma. This speedy response has been attributed to the lack of introns in the vast majority of heat shock proteins. This absence of introns allows heat shock proteins to circumvent a block in intron processing that occurs at elevated temperature. Thus, the heat shock protein is translated with very high efficiency, often at the expense of other proteins.
The activation of the stress genes is mediated by the conversion of a pre-existing heat shock transcription factor (HSF) from an inactive to an active form.
There is a large difference in the molecular weight of this DNA-binding protein (e.g., 83 kDa in humans and 150 kDa in yeast). The heat shock element is a conserved upstream regulatory sequence of HSP70 to which HSF binds. Although the main function of heat shock proteins is in facilitating protein folding and preventing aggregation, it is apparent that these proteins play some role in providing 1 o an organism with a protective mechanism against environmental insult and aid recovery subsequent to trauma.
Like most eukaryotic sequence-specific transcription factors, HSF acts through a highly conserved response element found in multiple copies upstream of the heat shock gene. The heat shock response element is composed of three contiguous inverted repeats of a 5-base pair sequence whose consensus was defined as nGAAn and more recently defined as AGAAn. The regulation of HSF primarily comprises a change in activity rather than an alteration in synthesis or stability.
3. Hyperthermia Therapy Many clinical studies have shown the effectiveness of hyperthermia as an adjunctive treatment for malignancies, when used in combination with radiotherapy or chemotherapy (Hahn, G.M., Hyperthermia and Cancer, 2nd Ed., New York, Plenum, 1982; Scott, et al., Int. J. Rad. Oc. Biol. Phys. 10{ 11 ) 2219-2123, 1984;
Lindholin, et al., Rec. Res. in Cancer Res. 107:152-156, 1988. The rationale for heat application, indication and contraindications, is developed on the basis of experimental evidence that desirable physiological responses can be produced by the use of heat and on the basis of controlled clinical studies. Lehman provides a comprehensive treatise for the therapeutic use of heat in other applications (Therapeutic Heat and Cold, Rehabilitation Medicine Library, published by Williams & Wilkins, 1990, incorporated by reference) the reader is referred in particular to Chapter 9, which discusses the use of heat in the context of therapeutic interventions, both medical and surgical.
"Hyperthermia" is intended to refer to a temperature condition that is greater 5 than the ambient temperature of the subject to which the treatment is being administered. Hence, a hyperthermic temperature, as used herein, will typically range from between about 37°C to about 42°C. In preferred embodiments, the temperature will range from about 38°C to about 42°C, in other embodiments, the temperature range will be from about 39°C to about 41°C, in other embodiments, 10 the temperature will be about 40°C. With the devices currently available for the application of hyperthermia in adjuvant therapies it is possible to maintain the temperature of hyperthermia treatment to within about 0.5°C for temperatures up to 42°C. Hence, the therapeutic treatments of the present invention may be carried out at 37.0°C, 37.2°C, 37.4°C, 37.6°C, 37.8°C, 38.2°C, 38.4°C, 38.6°C, 38.8°C, 39.2°C, 15 39.4°C, 39.6°C, 39.8°C, 40.2°C, 40.4°C, 40.6°C, 40.8°C, 41.2°C, 41.4°C, 41.6°C, 41.8°C, or 42.0°C. Prior to the present invention, efficacy of hyperthermia required that temperatures within a tumors) remain above about 43°C for 30 to 60 min, while safety considerations limit temperatures in normal tissues to below 42°C.
Achieving uniform temperatures above 42°C in tumors is very difficult and often not possible.
Tissues in mammals can be heated using a number of technologies including ultrasound, electromagnetic techniques, including either propagated wave (e.g., microwaves), resistive (e.g., radiofrequency) or inductive (radiofrequency or magnetic) procedures (Hahn, G.M., Hyperthermia and Cancer, 2nd Ed., New York, Plenum, 1982; Lehman, L.B., PoStgard Med., 88(3):240-243, 1990; both herein incorporated by reference). In some simple applications, tissue temperatures can be elevated using circulated hot air or water.
U.S. Pat. No. 4,230,129 to Le Veen, herein incorporated by reference, refers to a method of heating body tissue and monitoring temperature changes in the tumor in real time with the aid of a scintillation detector. The method provides for the coupling of radiofrequency (IZF) energy to the patient's body to avoid any significant heat absorption in the fatty tissues. This is obtained by focusing the RF
energy on the tumor with an orbital movement of the applicator so that energy is not constantly being applied to the same confined area within the patient's body.
U.S. Pat. No. 3,991,770 to Le Veen, also herein incorporated by reference, teaches a method of treating a tumor in a human by placing the part of the human body containing the tumor in a radiofrequency electromagnetic field to heat the tumor tissue and cause necrosis of the tumor without damaging the adjacent normal tissue.
In preferred embodiments, of the present invention, hyperthermia is applied in combination with the gene therapy vectors disclosed herein to achieve inducible gene expression at a particular tumor site. Furthermore, the hyperthermia/gene therapy treatment regimens may be used in combination with other conventional therapies, such as the chemotherapies and radiotherapies discussed below, to effectively treat cancer. Other methods for inducing hyperthermia also are known in the art. Methods and devices for the regional and/or systemic application of hyperthernua are well know to those of skill in the art and are disclosed in for example, U.S. Patent Nos. 5,284,144; 4,230,129; 4,186,729; 4,346,716;
4,848,362;
4,815,479; 4,632,128, all incorporated herein by reference.
4. Engineering Expression Constructs In certain embodiments, the present invention involves the manipulation of genetic material to produce expression constructs that encode therapeutic genes.
Such methods involve the use of an expression construct containing, for example, a heterologous DNA encoding a gene of interest and a means for its expression, replicating the vector in an appropriate helper cell, obtaining viral particles produced therefrom, and infecting cells with the recombinant virus particles.
The gene will be a therapeutic gene, for example to treat cancer cells, to express immunomodulatory genes to fight viral infections, or to replace a gene's function as a result of a genetic defect. In the context of the gene therapy vector, the gene will be a heterologous DNA, meant to include DNA derived from a source other than the viral genorne which provides the backbone of the vector. Finally, the virus may act as a live viral vaccine and express an antigen of interest for the production of antibodies thereagainst. The gene may be derived from a prokaryotic or eukaryotic source such as a bacterium, a virus, a yeast, a parasite, a plant, or an animal. The heterologous DNA also may be derived from more than one source, i.e., a multigene construct or a fusion protein. 'The heterologous DNA also may include a regulatory sequence which may be derived from one source and the gene from a different source.
a) Therapeutic Genes The selected polynucleotide of the present invention may optionally be a therapeutic gene. Any of a wide variety of therapeutic genes are suitable for use in the vectors and methods described herein. Therapeutic genes which are suitable for application of the present invention to a particular disorder, medical condition, or disease will be discernible to one skilled in the art.
In one embodiment of the invention, the selected polynucleotide is the gene encoding for ornithine decarboxylase antizyme protein. The ornithine decarboxylase (ODC} antizyme protein is an important component of feedback regulation of intracellular polyamine pool sizes (Hayashi et al., Trends in Biochemical Sciences 21(1}:27-30, 1996, herein incorporated by reference). The levels of this protein are directly related to levels of intracellular polyarnines, which stimulate translation of antizyme message. Antizyme protein targets ornithine decarboxylase, the first and often rate-limiting enzyme in polyamine synthesis, for degradation. This protein also suppresses polyamine uptake. Thus, low levels of endogenous polyamines lead to low levels of antizyme which in turn maximizes polyamine synthesis via ODC and polyamine uptake. Conversely, high levels of endogenous polyamines cause high levels of antizyme protein, which in turn minimize polyamine synthesis via ODC and suppress polyamine uptake.

The radioprotector WR-33278 (N,N"-(dithiodi-2,1-ethanediyl)bis-1,3-propanediamine) is a disulfide-containing polyamine analog, which is taken up by cells using the polyamine transporter (Mitchell et al., Carcinogenesis, 16:3063-3068, 1995, herein incorporated by reference). This transporter is inhibited by antizyme. Evidence from animal models indicates that this radioprotector is taken up by at least some normal tissues to a greater extent than some tumors {Ito et al., International Journal o, f'Radiation Oncolo~r, Biology, Physics 28:899-903, 1994).
Agents like WR-33278 have been used in clinical radiotherapy in attempts to protect dose-limiting normal tissues from toxicity, without reducing the tumor I0 control effectiveness of radiotherapy (Spencer and Goa, Drugs, 50{6):1001-31, 1995, herein incorporated by reference). Rationale for the difference in uptake of WR-33278 may be that proliferating tumor cells often contain higher levels of polyamines than do non-proliferating cells in normal tissues. Thus, tumors would express higher levels of antizyme than would normal tissues.
The inventors have placed an antizyme cDNA lacking the sequences necessary for polyamine-dependent regulation under the control of the human heat shock 70B promoter. The inventors have stably transfected human prostate cancer derived DU-145 cells with this construct and have selected clones which display heat-inducible suppression of polyamine uptake (indicating heat-inducible antizyme activity). The therapeutic application of this gene therapy (HSP70B promoter regulation of antizyme expression) will be put to use in future clinical trials in men with localized prostate cancer. Patients are treated with this gene therapy, administered intratumorally, combined with systemic WR-33278 and localized radiotherapy. Expression of antizyme intratumorally is then activated by localized hyperthermia. Dose-limiting normal tissues adjacent to these prostate tumors will not express antizyme in response to hyperthermia and will take up the radioprotector WR-33278, while the tumor tissue will not take up the radioprotector because they will express antizyrne in response to hyperthermia. This strategy will allow higher doses of radiotherapy to be given to the prostate, with the intent to improve local control of prostate cancers.

In an alternative embodiment, othex metabolic products of the cytoprotective drug ethyol (also known as amifostine, WR-2721, or S-2-(3-aminopropylamino)ethylphosphororthioic acid) other than WR33278 may be used in conjunction with the expression constructs described herein. For instance, WR-1065 (2-(3-aminopropylamino)ethanethiol) may be instead used as the radioprotector.
There are many other genes that may be delivered using the vectors of the present invention. For instance, it is contemplated that the vectors of the present invention may be used to transfer tumor suppressors, antisense oncogenes and 1 o prodrug activators, such as the HSV-TK gene {Rosenfeld et al., Annals of Surgery, 225:609-618, 1997; Esandi et al., Gene Therapy, 4:280-287, 1997), for the treatment of cancer. Other genes which could optionally be used in the expression constructs of the present invention include p53, p16, p21, p27, C-CAM, HLA-B7 (Gleich, et al., Arch Otolaryngol Head Neck Surg, 124:1097-104, 1998; Heo et al., Hum. Gene Ther. 9:2031-8, 1998; Nabel et al., Proc. Nat. Acad. Sciences USA, 90: 15388-15393, 1996; Stopeck et al., Journal of Clinical Oncology, 15: 341-349, 1997), IL2 (O'Malley et al., Molecular Endocrinology, 11:667-673, 1997; Otova et al., Folia Biologica, 43:25-32, 1997), IL4 (Kling, Nature Biotechnology, 15:316-317, 1997), IL7 (Toloza et al., Annals of Surgical Oncology, 4:70-79, 1997;
Sharma et al., Cancer Gene Therapy, 3:303-313, 1996), IL12 (Hiscox and Jiang, In Vivo, 11:125-137, 1997; Chen et al., Journal of Immunology, 159:351-359, 1997), GM-CSF (Kreitman and Pastan, Blood, 90:252-259, 1997; Homick et al., Blood, 89:4437-4447, 1997; Lama et al., Haematologica, 82:239-245, 1997), IFNy (Noguchi et al., Clinical Infectious Diseases, 24:992-994, 1997; Kanemaru et al., European Archives of Oto-Rhino-Laryngology, 254:158-162, 1997; Tanaka et al., Journal of Gastroenterolo~ and Hepatology, 11:1155-1160, 1996; Imai et al., Liver, 17:88-92, 1997), I-CAM 1, and TNF {Corcione et al., Annals of the New York Academy of Sciences, 815:364-366, 1997). (All articles cited in this paragraph are herein incorporated by reference.) p53 currently is recognized as a tumor suppressor gene (Montenarh, Crit.
Rev. Oncogen, 3:233-256, 1992). High levels of mutant p53 have been found in many cells transformed by chemical carcinogenesis, ultraviolet radiation, and several viruses, including SV40. The p53 gene is a frequent target of mutational 5 inactivation in a wide variety of human tumors and already is documented to be the most frequently-mutated gene in common human cancers. It is mutated in over 50% of human NSCLC and in a wide spectrum of other tumors.
P 16"''K4 belongs to a newly described class of CDK-inhibitory proteins that also includes pl6B, p21"''~~° CIP1, SD11' ~d p27KIP1. .Lhe pl6i''tK4 gene maps to 9p21, a 1 o chromosome region frequently deleted in many tumor types. Homozygous deletions and mutations of the p16~''~K4 gene are frequent in human tumor cell lines.
This evidence suggests that the pl6a''KQ gene is a tumor suppressor gene. This interpretation has been challenged, however, by the observation that the frequency of the p 16n''K4 gene alterations is much lower in primary uncultured tumors than in 15 cultured cell lines. Restoration of wild-type p16~K4 function by transfection with a plasmid expression vector reduced colony formation by some human cancer cell lines (Okamoto, et al., Proc. Natl. Acad. Sci. USA, 91:11045-11049, 1994;
Arap, et al., Cancer Res., 55:1351-1354, 1995, both herein incorporated by reference).
C-CAM is expressed in virtually all epithelial cells. C-CAM, with an 20 apparent molecular weight of 105 kD, originally was isolated from the plasma membrane of the rat hepatocyte by its reaction with specific antibodies that neutralize cell aggregation. Recent studies indicate that, structurally, C-CAM
belongs to the immunoglobulin (Ig) superfamily and its sequence is highly homologous to carcinoembryoruc antigen (CEA). The first Ig domain of C-CAM
has been shown to be critical for cell adhesion activity.
Cell adhesion molecules, or CAMs are known to be involved in a complex network of molecular interactions that regulate organ development and cell differentiation (Edelman, Annu. Rev. Biochem., 54:135-169, 1985). Recent data indicate that aberrant expression of CAMS may be involved in the tumorigenesis of several neoplasms; for example, decreased expression of E-cadherin, which predominantly is expressed in epithelial cells, is associated with the progression of several kinds of neoplasms. Also, Giancotti and Ruoslahti, Cell, 60:849-859, 1990, incorporated herein by reference, demonstrated that increasing expression of asl~1 integrin by gene transfer can reduce tumorigenicity of Chinese hamster ovary cells in vivo. C-CAM now has been shown to suppress tumor growth in vitro and in VIVO.
Other tumor suppressors that may be employed according to the present invention. For example, the selected polynucleotide may be any one of the l0 following genes: retinoblastoma (Rb); adenomatous polyposis coli gene (APC);
deleted in colorectal carcinomas (DCC); neurofibromatosis 1 (NF-1);
neurofibromatosis 2 (NF-2); Wilin's tumor suppressor gene (WT-1); multiple endocrine neoplasia type 1 (MEN-1); multiple endocrine neoplasia type 2 (MEN-2); BRCA1; von Hippel-Lindau syndrome (VHL); mutated in colorectal cancer (MCC); p 16; p21; p57; p27; and BRCA2.
In an alternative embodiment of the invention, the methods and vectors of the present invention may be used to promote regeneration processes, such as nerve regeneration, by stimulating the production of growth factors or cytokines. In such an embodiment the selected polynucleotide may be a neurotrophic factor. For instance, the selected polynucleotide may encode ciliary neurotrophic factor (CNTF), brain-derived neurotrophic factor (BDNF), or glial cell line-derived neurotrophic factor (GDNF) (Mitsumoto et al., Science, 265:1107-1110, 1994 and Gash et al., Ann. Neurol., 44(3 Suppl 1):5121-125, 1998, both herein incorporated by reference). Alternatively, the selected polynucleotide of the expression construct may optionally encode tyrosine hydroxylase, GTP cyclohydrolase 1, or aromatic L-amino acid decarboxylase (Kang, Mov. Disord., 13 Suppl 1:59-72, 1998, herein incorporated by reference). In still another embodiment, the therapeutic expression construct may express; a growth factor such as insulin-like growth factor-I (IGF-I) (Webster, Mult. Scler., 3:113-I20, 1997, incorporated herein by reference).
Examples of other diseases for which the present vectors are useful include but are not limited to hyperproliferative diseases and disorders, such as rheumatoid arthritis or restenosis by transfer of therapeutic genes, e.g., gene encoding angiogenesis inhibitors or cell cycle inhibitors.
b) Antisense constructs Oncogenes such as ras, myc, neu, raf, erb, src, fms, jun, trk, ret, gsp, hst, bcl and abl also are suitable targets. However, for therapeutic benefit, these oncogenes would be expressed as an antisense nucleic acid, so as to inhibit the expression of the oncogene. The term "antisense nucleic acid" is intended to refer to the oligonucleotides complementary to the base sequences of oncogene-encoding DNA
and RNA. Antisense nucleic acid, when expressed in a target cell, specifically bind to their target nucleic acid and interfere with transcription, RNA processing, transport and/or translation. Targeting double-stranded (ds) DNA with polynucleotides leads to triple-helix formation; targeting RNA will lead to double-helix formation.
Antisense constructs may be designed to bind to the promoter and other control regions, exons, introns or even exon-intron boundaries of a gene.
Antisense RNA constructs, or DNA encoding such antisense RNAS, may be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject. Nucleic acid sequences comprising "complementary nucleotides" are those which are capable of base-pairing according to the standard Watson-Crick complementary rules. That is, that the larger purines will base pair with the smaller pyrimidines to form only combinations of guanine paired with cytosine ~(G:C) and adenine paired with either thyrnine (A:T), in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA.

As used herein, the terms "complementary" or "antisense sequences" mean nucleic acid sequences that are substantially complementary over their entire length and have very few base mismatches. For example, nucleic acid sequences of fifteen bases in length may be termed complementary when they have a complementary nucleotide at thirteen or fourteen positions with only single or double mismatches. Naturally, nucleic acid sequences which are "completely complementary" will be nucleic acid sequences which are entirely complementary throughout their entire length and have no base mismatches.
While all or part of the gene sequence may be employed in the context of antisense construction, statistically, any sequence 17 bases long should occur only once in the human genome and, therefore, suffice to specify a unique target sequence. Although shorter oligomers are easier to make and increase in vivo accessibility, numerous other factors are involved in determining the specificity of hybridization. Both binding affinity and sequence specificity of an oligonucleotide to its complementary target increases with increasing length. It is contemplated that oligonucleotides of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more base pairs will be used. One can readily determW a whether a given antisense nucleic acid is effective at targeting of the corresponding host cell gene simply by testing the constructs in vitro to determine whether the endogenous gene's function is affected or whether the expression of related genes having complementary sequences is affected.
In certain embodiments, one may wish to employ antisense constructs which include other elements, for example, those which include C-5 propyne pyrimidines.
Oligonucleotides which contain C-5 propyne analogues of uridine and cytidine have been shown to bind RNA with high affinity and to be potent antisense inhibitors of gene expression (Wagner et al., Science, 260:1510-1513, 1993, herein incorporated by reference).

c) Ribozyme constructs As an alternative to targeted antisense delivery, targeted ribozymes may be used. The term "ribozyme" refers to an RNA-based enzyme capable of targeting and cleaving particular base sequences in DNA or, more typically, RNA. In the present invention, ribozymes are introduced into the cell as an expression construct encoding the desired ribozymal RNA. The targets of the ribozymes are much the same as described for antisense nucleic acids.
Many ribozymes are known to catalyze the hydrolysis of phosphodiester bonds under physiological conditions. The ribozymes of the present invention catalyze the sequence specific cleavage of a second nucleic acid molecule, preferably an mRNA transcript, and optionally an mRNA transcript of an oncogene.
In general, ribozymes bind to a target RNA through the target binding portion of the ribozyme which flanks the enzymatic portion of the ribozyme. The enzymatic portion of the ribozyme cleaves the target RNA. Strategic cleavage of a target RNA
destroys its ability to directly or indirectly encode protein. After enzymatic cleavage of the target has occurred, the ribozyme is released from the target and searches for another target where the process is repeated.
In a preferred embodiment of the invention, the ribozyme is a hammerhead ribozyme, a small RNA molecule derived from plant viroids (Symons, Ann. Rev.
Biochem. 61: 641-671, 1992; Clouet-D'Orval and Uhlenbeck, RNA, 2:483-491, 1996; Haseloff and Gerlach, Nature 334:585-591, 1988; Jeffries and Syrnons, Nucleic Acids Res. 17: 1371-1377, 1989; Uhlenbeck, Nature 328:596-600, 1987;
all herein incorporated by reference).
In other embodiments, the ribozyme may be a group I intron, a hairpin ribozyme, VS RNA, a hepatitis Delta virus ribozyme or an Rnase P-RNA ribozyme (in association with an RNA guide sequence). Examples of hairpin motifs are described by Hampel et al., Nucleic Acids Res. 18:299, 1990 and Hampel and Tritz, Biochemistry 28:4929, 1989; an example of the hepatitis delta viius motif is described by Perrotta and Been, Biochemistry 31:16, 1992; an example of the RNAseP motif (associated with an external guide sequence) is described by Yuan et al., Patent No. 5,624,824; a Neurospora VS RNA ribozyme motif is described in Saville and Collins, Cell 61: 685-696, 1990, Saville and Coliins, Proc. Natl.
Acad.
Sci. USA 88: 8826-8830, 1991, Collins and Olive, Biochemistry 32: 2795-2799, 5 1993; the group I intron is described in Cech et al., U.S. Patent No.
5,354,855. The above-mentioned motifs should not be considered limiting with respect to the present invention and those skilled in the art will recognize that ribozymes that may be utilized herein comprise a specific substrate binding site which is complementary to a target mRNA. Such ribozymes also comprise an enzymatic l0 portion which imparts RNA cleaving activity to the molecule. The enzymatic portion resides within or surrounds the substrate binding site.
d) Selectable Markers In certain embodiments of the invention, the therapeutic vectors of the present invention contain nucleic acid constructs whose expression may be 15 identified in vitro or in vivo by including a marker in the expression construct.
Such markers would confer an identifiable change to the cell permitting easy identification of cells containing the expression construct. Usually the inclusion of a drug selection marker aids in cloning and in the selection of transformants.
For example, genes that confer resistance to neomycin, purornycin, hygromycin, 20 DHFR, GPT, zeocin and histidinol are useful selectable markers.
Alternatively, enzymes such as herpes simplex virus thymidine kinase (tk) may be employed.
Immunologic markers also can be employed. The selectable marker employed is not believed to be important, so long as it is capable of being expressed simultaneously with the nucleic acid encoding a gene product. Further examples of 25 selectable markers are well known to one of skill in the art and include reporters such as EGFP, ~3-gal and chloramphenicol acetyltransferase (CAT).

e) Multigene Constructs and IRES
In certain embodiments of the invention, the use of internal ribosome binding sites (IRES) elements are used to create multigene, or polycistronic, messages. IRES elements are able to bypass the ribosome scanning model of 5' methylated Cap dependent translation and begin translation at internal sites.
IRES
elements from two members of the picanovirus family (polio and encephalomyocarditis) have been described (Pelletier and Sonenberg, Nature, 334:320-325, 1988), as well as an IRES from a mammalian message (Macejak and Sarnow, Nature, 353:90-94, 1991). IRES elements can be linked to heterologous l0 open reading frames. Multiple open reading frames can be transcribed together, each separated by an IRES, creating polycistronic messages. By virtue of the IR.ES
element, each open reading frame is accessible to ribosomes for e~cient translation. Multiple genes can thus be efficiently expressed using a single promoter/enhancer to transcribe a single message.
Any heterologous open reading frame can be linked to IRES elements. This includes genes for secreted proteins, multi-subunit proteins encoded by independent genes, intracellular or membrane-bound proteins and selectable markers.
Control Regions In order for the expression construct to affect expression of a transcript encoding a therapeutic gene, the polynucieotide encoding the therapeutic gene will be under the transcriptional control of a promoter and a polyadenylation signal. A
"promoter" refers to a DNA sequence recognized by the synthetic machinery of the host cell, or introduced synthetic machinery, that is required to initiate the specific transcription of a gene. A polyadenylation signal refers to a DNA sequence recognized by the synthetic machinery of the host cell, or introduced synthetic machinery, that is required to direct the addition of a series of nucleotides on the end of the MRNA transcript for proper processing and trafficking of the transcript out of the nucleus into the cytoplasm for translation. The phrase "under transcriptional control" means that the promoter is in the correct location in relation to the polynucleotide to control RNA polymerase initiation and expression of the polynucleotide.
The term promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA
polymerase II.
Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 by of DNA, and containing one or more recognition sites for transcriptional activator or repressor proteins.
At least one module in each promoter functions to position the start site for RNA synthesis. The best known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 by upstream of the start site, although a number of promoters have recently been .shown to contain 2o functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 by apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.
Where a human cell is targeted, it is preferable to position the polynucleotide coding region adjacent to and under the control of a promoter that is capable of WO 99/23216 PCTNS98/2338?

being expressed in a human cell. Generally speaking, such a promoter might include either a human or viral promoter. A list of promoters is provided in Table 1.

PROMOTER
Immunoglobulin Heavy Chain Immunoglobulin Light Chain T-Cell Receptor HLA DQ a and DQ (3 ', ~i-Interferon Interleukin-2 Interleukin-2 Receptor MHC Class II 5 MHC Class II HLA-DRa (3-Actin Muscle Creatine Kinase Prealbumin (Transthyretin) Elastase~ I
Metallothionein Collagenase Albumin Gene a-Fetaprotein i-Globin (3-Globin c-fos c-HA-ras Insulin Neural Cell Adhesion Molecule (NCAM) aL.Antitrypsin H2B (TH2B) Histone Mouse or Type I Collagen Glucose-Regulated Proteins (GRP94 and GRP78) Rat Growth Hormone Human Serum Amyloid A (SAA) Troponin I (TN I) Platelet-Derived Growth Factor Duchenne Muscular Dystrophy Polyoma Retroviruses Papilloma Virus Hepatitis B Virus Human Immunodeficiency Virus Cytomegalovirus Gibbon Ape Leukemia Virus The particular promoter that is employed to control the expression of the therapeutic gene is not believed to be critical, so long as it is capable of being activated by the gene product linked to the inducible promoter. In a preferred embodiment of the invention, the transactivating protein is tat, and the promoter 5 which is operably linked to the therapeutic gene is the HIV-1 or HIV-2 LTRs.
For example, a promoter element containing an AP-1 site would respond to the inducible expression of the c-jun or c-fos proteins. Other suitable transactivating factor/promoter combination would be known by one skilled in the art.
The promoter which controls expression of the gene encoding the 1 o transactivating factor must be an inducible promoter. An inducible promoter is a promoter which is inactive or exhibits relatively low activity except in the presence of an inducer substance. Some examples of promoters that may be included as a part of the present invention include, but are not limited to, MT II, MMTV, collagenase, stromelysin, SV40, marine MX gene, a-2-macroglobulin, MHC class I
15 gene h-2kb, proliferin, tumor necrosis factor, or thyroid stimulating hormone a gene. The associated inducers of these promoter elements are shown in Table 2.
The Egr-1 promoter and the multidrug resistance gene (MDR 1 ) promoter are also options for inducible promoters. In preferred embodiments the inducible promoter is heat shock inducible and is derived from one of the following promoters:
20 HSP70, HSP90, HSP60, HSP27, HSP72, HSP73, HSP25, ubiquitin, and HSP28. In another preferred embodiment, the inducible promoter comprises a hypoxia-responsive element, such as those responsive to HIF-1. It is understood that any inducible promoter may be used in the practice of the present invention and that all such promoters would fall within the spirit and scope of the claimed invention.

Element ~ Inducer MT II Phorbol Ester (TPA) Heavy metals MMTV (mouse Glucocorticoids mammary tumor virus) (3-Interferon poly(rI)X
poly(rc) Adenovirus 5 E2 Ela c-jun Phorbol Ester (TPA), H202 Collagenase Phorbol Ester (TPA) Stromelysin Phorbol Ester (TPA), IL-1 SV40 Phorbol Ester (TPA) Murine MX Gene Interferon, Newcastle Disease Virus GRP78 Gene A23187 a-2 Macroglobulin IL-6 Vimentin Serum MHC Class I Gene H- Interferon 2kB

HSP70 Ela, SV40 Large T Antigen Proliferin Phorbol Ester-TPa Tumor Necrosis FactorFMA

Thyroid Stimulating Thyroid Hormone Hormone a Gene In particularly preferred embodiments, the tat protein is used as the transactivating factor. The genome of HIV-1 and HIV-2 share a great deal of similarities with the Simian immunodeficiency viruses (SIVS) and they have been extensively studied. It was discovered that in addition to the gag, env, pal genes that are common to all retroviruses, there are a number of regulatory genes that are important in HIV transcription. The viral tat protein is one such regulatory factor and it is characterized by its ability to greatly increase the activity of the HIV-1 and HIV-2 promoter (Sodroski et al., J. Virol, 55(3):831-835,1985a; Sodroski, et al., Science, 229(4708):74-77, 1985b; Sodroski, et al., Science, 228(4706):1430-1434, Sodroski, et al., Science, 228(4706):1430-1434, 1985c; Sodroski et al., Science, 227(4683):171-173, 1985d; which are all incorporated by reference herein). Tat is thought to bind with the transactivation response element (TAR) in the HIV LTR
and increase the steady state levels of the HIV specific RNA. There is also evidence suggesting that tat can act more like a traditional transcription factor in that it can interact with several transactivator proteins. Tat and adenovirus transactivator EIA can act synergistically in increasing the levels of steady state RNA (Laspia et al., Genes Dev., 4(12B):2397-2408, 1990, herein incorporated by reference). Thus, a way to increase further the activity of the HIV-LTR/TAT
constructs is to incorporate ElA into the same construct.
Where a cDNA insert is employed, one will typically desire to include a polyadenylation signal to effect proper polyadenylation of the gene 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. Such polyadenylation signals as that from SV40, bovine growth hormone, and the herpes simplex virus thymidine kinase gene have been found to function well in a number of target cells.
5. Methods of Gene Transfer In order to effect transgene expression in a cell, it is necessary to first introduce or transfer the therapeutic expression constructs of the present invention into a cell. Such transfer may employ viral or non-viral methods of gene.transfer.
This section provides a discussion of methods and compositions of gene transfer.

A. Non-viral Transfer In a preferred embodiment, the therapeutic constructs of the present invention e.g., various genetic (i.e., DNA) constructs must be delivered into a cell.
In certain preferred situations, the introduction of the expression construct into a cell is mediated by non-viral means.
Several non-viral methods for the transfer of expression constructs into cultured mammalian cells are contemplated by the present invention. These include calcium phosphate precipitation {Graham and Van Der Eb, virology, 52:456-467, 1973; Chen and Okayama, Mol. Cell. Biol., 7:2745-2752, 1987; lZippe et al., Mol.
Cell Biol., 10:689-695, 1990) DEAE-dextran (Gopal, Mol. Cell Biol., 5:1188-1190, 1985), electroporation (Tur-Kaspa et al., Mol. Cell Biol., 6:716-718, 1986;
Potter et al., Proc. Nat. Acad. Sci. USA, 81:7161:7165, 1984), direct microinjection (Harland and Weintraub, J. Cell Biol., 1 D 1:1094-1099, 1985), DNA-loaded liposomes (Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190, 1982; Fraley et al., Proc. Natl. Acad. Sci. USA, 76:3348-3352, 1979), cell sonication (Fechheimer et al., Proc. Natl. Acad. Sci. USA, 84:8463-8467, 1987), gene bombardment using high velocity microprojectiles (Yang et al., Proc. Natl.
Acad.
Sci. USA, 87:9568-9572, 1990}, and receptor-mediated transfection (Wu and Wu, J.
Biol. Chem., 262:4429-4432, 1987; Wu and Wu, Biochemistry, 27:887-892, 1988).
(Articles cited in this paragraph are herein incorporated by reference.}
Once the construct has been delivered into the cell the nucleic acid encoding the therapeutic gene may be positioned and expressed at different sites. In certain embodiments, the nucleic acid encoding the therapeutic gene may be stably integrated into the genome of the cell. This integration may be in the cognate location and orientation via homologous recombination {gene replacement) or it may be integrated in a random, non-specific location (gene augmentation). In yet further embodiments, the nucleic acid may be stably maintained in the cell as a separate, episomal segment of DNA. Such nucleic acid segments or "episomes"
encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. How the expression construct is delivered to a cell and where in the cell the nucleic acid remains is dependent on the type of expression construct employed.
In a particular embodiment of the invention, the expression construct may be entrapped in a liposome. Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution.
The lipid components undergo self rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers.
The addition of DNA to cationic liposomes causes a topological transition from liposomes to optically birefringent liquid-crystalline condensed globules.
These DNA-lipid complexes are potential non-viral vectors for use in gene therapy.
Liposome-mediated nucleic acid delivery and expression of foreign DNA in vitro has been very successful. Using the (3-lactamase gene, Wong et al., Gene, 10:87-94, 1980, demonstrated the feasibility of liposome-mediated delivery and expression of foreign DNA in cultured chick embryo, HeLa, and hepatoma cells.
Nicolau et al., (Methods Enzymol., 149:157-176, 1987, herein incorporated by reference) accomplished successful liposome-mediated gene transfer in rats after intravenous injection. Also included are various commercial approaches involving "lipofection" technology.
In certain embodiments of the invention, the liposome may be complexed with a hemagglutinating virus (HVJ}. This has been shown to facilitate fusion with the cell membrane and promote cell entry of liposome-encapsulated DNA. In other embodiments, the liposome may be complexed or employed in conjunction with nuclear nonhistone chromosomal proteins (HMG-1). In yet further embodiments, the liposome may be complexed or employed in conjunction with both HVJ and HMG-1. In that such expression constructs have been successfully employed in WO 99/23216 PC'T/US98/23387 transfer and expression of nucleic acid in vitro and in vivo, then they are applicable for the present invention.
Other vector delivery systems which can be employed to deliver a nucleic acid encoding a therapeutic gene into cells are receptor-mediated delivery vehicles.
5 These take advantage of the selective uptake of macromolecules by receptor-mediated endocytosis in almost all eukaryotic cells. Because of the cell type-specific distribution of various receptors, the delivery can be highly specific (Wu and Wu, Adv. DrugDelivety Rev., 12:159-167, 1993).
Receptor-mediated gene targeting vehicles generally consist of two 10 components: a cell receptor-specific ligand and a DNA-binding agent.
Several ligands have been used for receptor-mediated gene transfer. The most extensively characterized ligands are asialoorosomucoid (ASOR) (Wu and Wu, J. Biol. Chem., 262:4429-4432, 1987) and transferrin (Wagner et al., Proc. Natl. Acad. Sci.
87(9):3410-3414, 1990). Recently, a synthetic neoglycoprotein, which recognizes 15 the same receptor as ASOR, has been used as a gene delivery vehicle (Ferkol et al., FASEB J., 7:1081-1091, 1993; Perales et al., Proc. Natl. Acad. Sci USA, 91:4086-4090, 1994) and epidermal growth factor (EGF) has also been used to deliver genes to squamous carcinoma cells (Myers, EPO 0273085).
In other embodiments, the delivery vehicle may comprise a ligand and a 20 liposome. For example, Nicolau et al. Methods Enzymol., 149:157-176, 1987, employed lactosyl-ceramide, a galactose-terminal asialganglioside, incorporated into Iiposomes and observed an increase in the uptake of the insulin gene by hepatocytes. Thus, it is feasible that a nucleic acid encoding a therapeutic gene also may be specifically delivered into a cell type such as prostate, epithelial or tumor 25 cells, by any number of receptor-ligand systems with or without liposomes.
For example, the human prostate-specific antigen (Watt et al., Proc. Natl. Acad.
Sci., 83(2):3166-3170, 1986) may be used as the receptor for mediated delivery of a nucleic acid in prostate tissue.

3fi In another embodiment of the invention, the expression construct may simply consist of naked recombinant DNA or plasmids. Transfer of the construct may be performed by any of the methods mentioned above which physically or chemically permeabilize the cell membrane. This is applicable particularly for transfer in vitro, however, it may be applied for in vivo use as well.
Dubensky et al. {Proc. Nat. Acad. Sci. USA, 81:7529-7533, 1984), successfully. injected polyomavirus DNA in the form of CaP04 precipitates into liver and spleen of adult and newborn mice demonstrating active viral replication and acute infection.
Benvenisty and Neshif, Proc. Nat. Acad. Sci. USA, 83:9551-9555, 1986, also demonstrated that direct intraperitoneal injection of CaP04 precipitated plasmids results in expression of the transfected genes. It is envisioned that DNA
encoding a CAM may also be transferred in a similar manner in vivo and express CAM.
Another embodiment of the invention for transferring a naked DNA
expression construct into cells may involve particle bombardment. This method depends on the ability to accelerate DNA coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them (Klein et al., Nature, 327:70-73, 1987, herein incorporated by reference). Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical current, which in turn provides the motive force (Yang et al., Proc. Natl. Acad. Sci. USA, 87:9568-9572, 1990).
The microprojecriles used have consisted of biologically inert substances such as tungsten or gold beads.
B. Viral Vector-Mediated Transfer Another method of achieving gene transfer is via viral transduction using infectious viral particles as a delivery vehicle, for example, by transformation with an adenovirus vector of the present invention as described herein below.
Alternatively, retroviral or bovine papilloma virus may be employed, both of which permit permanent transformation of a host cell with a genes) of interest.
Thus, in one example, viral infection of cells is used in order to deliver therapeutically WO 99/23216 PCT/US98l23387 significant genes to a cell. Typically, the virus simply will be exposed to the appropriate host cell under physiologic conditions, permitting uptake of the virus.
Though adenovirus is exemplified, the present methods may be advantageously employed with other viral vectors, as discussed below. Such methods will be familiar to those of ordinary skill in the art.
a) Adenovirus Adenovirus is particularly suitable for use as a gene transfer vector because of its mid-sized DNA genome, ease of manipulation, high titer, wide, target-cell range, and high infectivity. The roughly 36 kB viral genome is bounded by 100-200 base pair (bp) inverted terminal repeats (ITR), in which are contained cis-acting elements necessary for viral DNA replication and packaging. The early (E) and late (L) regions of the genome that contain different transcription units are divided by the onset of viral DNA replication.
The E1 region (ElA and E1B) encodes proteins responsible for the regulation of transcription of the viral genome and a few cellular genes. The expression of the E2 region (E2A and E2B) results in the synthesis of the proteins for viral DNA replication. These proteins are involved in DNA replication, late gene expression, and host cell shut off (Renan, 1990). The products of the late genes (L1, L2, L3, L4 and L5), including the majority of the viral capsid proteins, are expressed only after significant processing of a single primary transcript issued by the major late promoter (MLP). The MLP (located at 16.8 map units) is particularly efficient during the late phase of infection, and all the mRNAs issued from this promoter possess a 5' tripartite leader (TL) sequence which makes them preferred mRNAs for translation.
In order for adenovirus to be optimized for gene therapy, it is necessary to maximize the carrying capacity so that large segments of DNA can be included.
It also is very desirable to reduce the toxicity and immunologic reaction associated with certain adenoviral products. The two goals are, to an extent, coterminous in that elimination of adenoviral genes serves both ends. By practice of the present invention, it is possible to achieve both these goals while retaining the ability to manipulate the therapeutic constructs with relative ease.
The large displacement of DNA is possible because the cis elements required for viral DNA replication all are localized in the inverted terminal repeats (ITR) ( 100-200 bp) at either end of the linear viral genome. Plasmids containing ITR's can replicate in the presence of a non-defective adenovirus. Therefore, inclusion of these elements in an adenoviral vector should permit replication.
In addition, the packaging signal for viral encapsulation is localized between 194-385 by (0.5-1.1 map units) at the left end of the viral genome. This signal mimics the protein recognition site in bacteriophage ~, DNA where a specific sequence close to the left end, but outside the cohesive end sequence, mediates the binding to proteins that are required for insertion of the DNA into the head structure. E1 substitution vectors of Ad have demonstrated that a 450 by (0-1.25 map units) fragment at the left end of the viral genome could direct packaging in 293 cells.
Previously, it has been shown that certain regions of the adenoviral genome can be incorporated into the genome of mammalian cells and the genes encoded thereby expressed. These cell lines are capable of supporting the replication of an adenoviral vector that is deficient in the adenoviral function encoded by the cell line. There also have been reports of complementation of replication deficient adenoviral vectors by "helping" vectors, e.g., wild-type virus or conditionally defective mutants.
Replication-deficient adenoviral vectors can be complemented, in traps, by helper virus. This observation alone does not permit isolation of the replication-deficient vectors, however, since the presence of helper virus, needed to provide replicative functions, would contaminate any preparation. Thus, an additional element was needed that would add specificity to the replication and/or packaging of the replication-deficient vector. That element, as provided for in the present invention, derives from the packaging function of adenovirus.
It has been shown that a packaging signal for adenovirus exists in the left end of the conventional adenovirus map. Later studies showed that a mutant with a deletion in the ElA (194-358 bp) region of the genome grew poorly even in a cell line that complemented the early (ElA) function. When a compensating adenoviral DNA (0-353 bp) was recombined into the right end of the mutant, the virus was packaged normally. Further mutational analysis identified a short, repeated, position-dependent element in the left end of the Ad5 genome. One copy of the l0 repeat was found to be su~cient for efficient packaging if present at either end of the genome, but not when moved towards the interior of the Ad5 DNA molecule.
By using mutated versions of the packaging signal, it is possible to create helper viruses that are packaged with varying efficiencies. Typically, the mutations are point mutations or deletions. When helper viruses with low efficiency packaging are grown in helper cells, the virus is packaged, albeit at reduced rates compared to wild-type virus, thereby permitting propagation of the helper.
When these helper viruses are grown in cells along with virus that contains wild-type packaging signals, however, the wild-type packaging signals are recognized preferentially over the mutated versions. Given a limiting amount of packaging factor, the virus containing the wild-type signals are packaged selectively when compared to the helpers. If the preference is great enough, stocks approaching homogeneity should be achieved.
b) Retrovirus The retroviruses are a group of single-stranded RNA viruses characterized by an ability to convert their RNA to double-stranded DNA in infected cells by a process of reverse-transcription. The resulting DNA then stably integrates into cellular chromosomes as a provirus and directs synthesis of viral proteins.
The integration results in the retention of the viral gene sequences in the recipient cell and its descendants. The retroviral genome contains three genes - gag, pol and env - that code for capsid proteins, polymerase enzyme, and envelope components, respectively. A sequence found upstream from the gag gene, termed ~f, functions as a signal for packaging of the genome into virions. Two long terminal repeat 5 (LTR) sequences are present at the 5' and 3' ends of the viral genome. These contain strong promoter and enhancer sequences and also are required for integration in the host cell genome.
In order to construct a retroviral vector, a nucleic acid encoding a promoter is inserted into the viral genome in the place of certain viral sequences to produce a 10 virus that is replication-defective. In order to produce virions, a packaging cell line containing the gag, pol and env genes but without the LTR and 'IJ components is t constructed. When a recombinant plasmid containing a human cDNA, together with the retroviral LTR and ~l' sequences is introduced into this cell, line (by calcium phosphate precipitation for example), the ~ sequence allows the RNA
15 transcript of the recombinant plasmid to be packaged into viral particles, which are then secreted into the culture media. The media containing the recombinant retroviruses is collected, optionally concentrated, and used for gene transfer.
Retroviral vectors are able to infect a broad variety of cell types. However, integration and stable expression of many types of retroviruses require the division 20 of host cells.
An approach designed to allow specific targeting of retrovirus vectors recently was developed based on the chemical modification of a retrovirus by the chemical addition of galactose residues to the viral envelope. This modification could permit the specific infection of cells such as hepatocytes via 25 asialoglycoprotein receptors, should this be desired.
A different approach to targeting of recombinant retroviruses was designed in which biotinylated antibodies against a retroviral envelope protein and against a specific cell receptor were used. The antibodies were coupled via the biotin components by using streptavidin (Roux et al., Proc. Natl Acad. Sci. USA, 86:9079-90$3, 19$9). Using antibodies against major histocompatibility complex class I
and class II antigens, the infection of a variety of human cells that bore those surface antigens was demonstrated with an ecotropic virus in vitro.
c) Adeno-associated Irrus AAV utilizes a linear, single-stranded DNA of about 4700 base pairs.
Inverted terminal repeats flank the genome. Two genes are present within the genome, giving rise to a number of distinct gene products. The first, the cap gene, produces three different virion proteins (VP), designated VP-1, VP-2 and VP-3.
The second, the rep gene, encodes four non-structural proteins (NS). One or more of these rep gene products is responsible for transactivating AAV
transcription.
The three promoters in AAV are designated by their location, in map units, in the genome. These are, from left to right, p5, p19 and p40. Transcription gives rise to six transcripts, two initiated at each of three promoters, with one of each pair being spliced. The splice site, derived from map units 42-46, is the same for each transcript. The four non-structural proteins apparently are derived from the longer of the transcripts, and three virion proteins all arise from the smallest transcript.
AAV is not associated with any pathologic state in humans. Interestingly, for efficient replication, AAV requires "helping" functions from viruses such as herpes simplex virus I and II, cytomegalovirus, pseudorabies virus and, of course, adenovirus. The best characterized of the helpers is adenovirus, and many "early"
functions for this virus have been shown to assist with AAV replication. Low level expression of AAV rep proteins is believed to hold AAV structural expression in check, and helper virus infection is thought to remove this block.
The terminal repeats of the AAV vector can be obtained by restriction endonuclease digestion of AAV or a plasmid such as p201, which contains a modified AAV genome {Sarnulski et al. J. hirol., 61(10):3096-3101, 19$7, herein incorporated by reference), or by other methods known to the skilled artisan, including but not limited to chemical or enzymatic synthesis of the terminal repeats based upon the published sequence of AAV. The ordinarily skilled artisan can determine, by well-known methods such as deletion analysis, the minimum sequence or part of the AAV ITRs which is required to allow function, i.e., stable and site-specific integration. The ordinarily skilled artisan also can determine which minor modifications of the sequence can be tolerated while maintaining the ability of the terminal repeats to direct stable, site-specific integration.
AAV-based vectors have proven to be safe and effective vehicles for gene delivery in vitro, and these vectors are being developed and tested in pre-clinical and clinical stages for a wide range of applications in potential gene therapy, both ex vivo and in vivo.
AAV-mediated efficient gene transfer and expression in the lung has led to clinical trials for the treatment of cystic fibrosis (Carter and Flotte, Ann.
N. Y. Acad.
Sci., 770:79-90, 1995; Flotte et al., Proc.. Natl. Acad. Sci. USA, 90:10613-10617, 1993). Similarly, the prospects for treatment of muscular dystrophy by A.AV-mediated gene delivery of the dystrophin gene to skeletal muscle, of Parkinson's disease by tyrosine hydroxylase gene delivery to the brain, of hemophilia B by Factor IX gene delivery to the liver, and potentially of myocardial infarction by vascular endothelial growth factor gene to the heart, appear promising since AAV-mediated transgene expression in these organs has recently been shown to be highly efficient (Fisher et al., J. Yirol, 70:520-532, 1996; Flotte et al., Proc.
Natl. Acad.
Sci. USA, 90:10613-10617, 1993; Kaplitt et al., Nat. Genet., 8:148-153, 1994;
Kaplitt et al., Arm Thord. Surg., 62:1669-1676, 1996; Koeberl et al., Proc.
Natl.
Acad. Sci. USA, 94:1426-1431, 1997; McCown et al., Brain Res., 713:99-107, 1996; Ping et al., Microcirculation, 3:225-228, 1996; Xiao et al., J. virol., 70:8098-8108, 1996).
d) Oth er oral Vectors Other viral vectors may be employed as expression constructs in the present invention. Vectors derived from viruses such as vaccinia viruses (Ridgeway, In:

Vectors: A survey of molecular cloning vectors and their uses, Rodriguez RL, Denhardt DT. ed., Stoneham: Butterworth, pp. 467-492, 1988; Baichwal and Sugden, In: Gene Transfer, Kucherlapati R, ed., New York, Plenum Press, pp.

148, 1986; Coupar et al., Gene, 68:1-10, 1988) canary pox viruses, lentiviruses and herpes viruses may be employed.
6. Cell Targets The methods and vectors of the present invention may be used to target a wide variety of cells, organs, and tissues within a mammal.
In some embodiments, the expression constructs described herein are used to l0 treat cancer. The cell which is targeted may be either a tumor cell, a cell within a tumor, or a cell near a tumor. The tumor may optionally be in the brain, lung, liver, spleen, kidney, bladder, lymph node, small intestine, pancreas, colon, stomach, breast, endometrium, prostate, testicle, vulva, cervix, ovary, skin, head and neck, esophagus, bone marrow, or blood. One of ordinary skill in the art will be able to readily discern an appropriate therapeutic gene to be expressed in a given tumor type.
In alternative embodiments, a medical condition other than cancer is being treated. For instance, the present invention provides for highly effective protein replacement therapy. In such a case, a specific type of cell, tissue, or organ may be targeted for expression of a protein which is underexpressed in the subject, especially if the activity of the protein is limited to that specific cell type, tissue, or organ. Again, one of ordinary skill in the art will be able to discern which cells are most appropriately targeted.
The expression construct may be introduced into the cell of interest through an in vitro, ex vivo, or in vivo method. Much gene therapy is currently performed ex vivo, since the transfection or transduction of an isolated cell is often more efficient. The choice of method of introduction will be dependent upon the cell type, tissue, or organ being targeted, as well as the particular delivery vehicle chosen. One of ordinary skill in the art can readily navigate such a choice.
Since the expression constructs of the present invention require induction to be active, in many cases the expression construct may be delivered to a larger part of the subject's body than just the cell, tissue, or organ in which expression is desired. Exposure of the subject to the activating conditions which induce expression of the transferred expression constructs can then be limited to achieve specificity of expression. In many cases, this is preferred. For instance, exposure of a subject to radiation therapy is preferably limited to only those areas necessary.
Application of hyperthermia will generally also be limited in its range. In other embodiments, the activating conditions may be conditions inherent to the targeted cell itself. For instance, the hypoxic environment of a tumor will trigger expression when the expression construct has an inducible promoter containing a hypoxia-responsive element. In such cases, the resulting expression, will be by its very nature, very localized, even if delivery of the expression construct was not 7. Combination Therapy The expression constructs of the present invention may advantageously be combined with one or more traditional clinical therapies. One goal of current cancer research is to fmd ways to improve the efficacy of chemo- and radiotherapy.
One way is by combining such traditional therapies with gene therapy. For example, the herpes simplex-thymidine kinase (HS-tk) gene, when delivered to brain tumors by a retroviral vector system, successfully induces susceptibility to the antiviral agent ganciclovir. However, the effective use of gene therapy in combination with traditional cancer therapies has been hindered by the need for clinically significant expression of the genes once they have been transferred to the target cell.
To kill cells, inhibit cell growth, inhibit metastasis, decrease tumor size and otherwise reverse or reduce the malignant phenotype of tumor cells, using the methods and compositions of the present invention, one would generally introduce an expression construct of the present invention into the "target" cell and induce expression by the application of hyperthermia or other conditions which activate the inducible promoter. This gene therapy may be combined with compositions comprising other agents effective in the treatment of cancer. These compositions 5 would be provided in a combined amount effective to kill or inhibit proliferation of the cell. This process may involve introducing the expression construct and the agents} or factors) into the cell at the same time. This may be achieved by contacting the cell with a single composition or pharmacological formulation that includes both agents, or by exposing the cell to two distinct compositions or 10 formulations, at the same time, wherein one composition includes the expression construct and the other includes the agent.
Alternatively, the gene therapy/hyperthermia treatment may precede or follow the other agent treatment by intervals ranging from minutes to weeks.
In embodiments where the other agent and expression construct are applied separately 15 to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and expression construct would still be able to exert an advantageously combined effect on the cell.
In such instances, it is contemplated that one would contact the cell with both modalities within about 12-24 hours of each other and, more preferably, within 20 about 6-12 hours of each other, with a delay time of only about 12 hours being most preferred. In some situations, it may be desirable to extend the time period for treatment significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (1, 2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations.
It also is conceivable that more than one administration of either expression 25 construct or the other agent will be desired. Various combinations may be employed, where the expression construct is "A" and the other agent is "B", as exemplified below:
A/B/A B/A/B B/B/A A/A/B B/A/A A/B/B B/B/B/A B/B/A/B

AIA/B/B A/B/A/B ABIBIA BB/A/A BIAIBIA BIA/A/B BIBIBIA
A/A/AIB B/A/A/A A/B/A/A AIAIBIA AIBBB B/A/B/B B/B/AIB
Other combinations are contemplated. Again, to achieve cell killing, both agents are delivered to a cell in a combined amount effective to kill the cell.
Agents or factors suitable for use in a combined therapy are any chemical compound or treatment method that induces DNA damage when applied to a cell.
Such agents and factors include radiation and waves that induce DNA damage such as, y-irradiation, X-rays, UV-irradiation, microwaves, electronic emissions, and the like.
In one embodiment of the invention, the radiation therapy which is combined with the gene therapy constitutes external beam radiation. The external beam radiation treatment typically delivers high-energy radiation, such as high-energy x-ray beams.
Alternatively, internal radiation, or brachythcrapy, may be used in combination with the gene therapy. Methods of delivering brachytherapy include intracavitary or interstitial placement of radiation sources, instillation of colloidal solutions, and parenteral or oral administration. Sealed sources are encapsulated in a metal, wire, tube, needle, or the like. Unsealed radioactive sources are prepared in a suspension or solution.
Encapsulated radioactive elements are placed in body cavities or inserted directly into tissues with suitable applicators. The applicator is usually placed into the body cavity or tissue surgically or using fluoroscopy. The applicators, usually plastic or metal tubes, may be sutured into or near the tumor to hold them in place.
The radioactive isotope is later placed into the applicator ("afterloading").
Radiative implants are used in the treatment of cancers of the tongue, lip, breast, vagina, cervix, endometrium, rectum, bladder, and brain. Encapsulated sources may also be left within a patient as permanent implants. "Seeding" with small beads of radioactive material is an approach used for the treatment of localized prostate cancers, and localized, but inoperable, lung cancers.
A variety of chemical compounds, also described as "chemotherapeutic agents," function to induce DNA damage, all of which are intended to be of use in the combined treatment methods disclosed herein. Chemotherapeutic agents contemplated to be of use, include, e.g., adriamycin, 5-fluorouracil (SFU), etoposide (VP-16), camptothecin, actinomycin-D, mitomycin C, cisplatin (CDDP) and even hydrogen peroxide. The invention also encompasses the use of a combination of one or more DNA damaging agents, whether radiation-based or l0 actual compounds, such as the use of X-rays with cisplatin or the use of cisplatin with etoposide.
For example, in treating cancer according to the invention, one would contact the tumor cells with an agent in addition to the expression construct and induce the expression of the gene by application of hyperthermia. This may be achieved by applying hyperthermia locally at the tumor site or systemically to the individual. This treatment may be in combination with irradiation of the tumor with radiation such as X-rays, UV-light, gamma-rays or even microwaves.
Alternatively, the tumor cells may be contacted with the agent by administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a compound such as, adriamycin, 5-fluorouracil, etoposide, camptothecin, actinomycin-D, mitomycin C, or more preferably, cisplatin. The agent may be prepared and used as a combined therapeutic composition, or kit, by combining it with a therapeutic expression construct, as described above.
Agents that directly cross-link nucleic acids, specifically DNA, are envisaged to facilitate DNA damage leading to a synergistic, antineoplastic combination with the expression constructs of the present invention. Agents such as cisplatin, and other DNA alkylating agents may be used. Cisplatin is not absorbed orally and must therefore be delivered via injection intravenously, subcutaneously, intratumorally or intraperitoneally.

Agents that damage DNA also include compounds that interfere with DNA
replication, mitosis and chromosomal segregation. Such chemotherapeutic compounds include adriamycin, also known as doxorubicin, etoposide, veraparnil, podophyllotoxin, and the like. Widely used in a clinical setting for the treatment of neoplasms, these compounds are administered through bolus injections intravenously at doses ranging from 25-75 mg/m2 at 21 day intervals for adriamycin, to 100 mg/m2 for etoposide intravenously or double the intravenous dose orally.
Agents that disrupt the synthesis and fidelity of nucleic acid precursors and subunits also lead to DNA damage. As such a number of nucleic acid precursors have been developed. Particularly useful are agents that have undergone extensive testing and are readily available. As such, agents such as 5-fluorouracil (5-FU), are preferentially used by neoplastic tissue, making this agent particularly useful for targeting to neoplastic cells. Although quite toxic, 5-FU, is applicable in a wide range of carriers, including topical, however intravenous administration with doses ranging from 450-1000 mg/m2/day being commonly used.
Other factors that cause DNA damage and have been used extensively include what are commonly known as y-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other forms of DNA damaging factors are also contemplated such as microwaves and UV-irradiation. It is most likely that all of these factors effect a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes. Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 weeks), to single doses of 2000 to 6000 roentgens. Dosage ranges for radioisotopes vary widely, and depend on the half life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
The skilled artisan is directed to "Remington's Pharmaceutical Sciences"
15th Edition, chapter 33, in particular pages 624-652. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologics standards.
The regional delivery of the therapeutic expression constructs of the present invention to patients with cancers is a preferred method for delivering a therapeutically effective gene to counteract the clinical disease being treated.
Similarly, the chemo- or radiotherapy may be directed to a particular, affected region of the subject's body. Alternatively, systemic delivery of expression construct and/or the agent may be appropriate in certain circumstances, for example, where extensive metastasis has occurred.
In addition to combinations of gene therapies with chemo- and radiotherapies, it also is contemplated that combination of multiple gene therapies will be advantageous. For example, targeting of p53 and p16 mutations at the same time may produce an improved anti-cancer treatment. Any other tumor-related gene conceivably can be targeted in this manner, for example, p21, Rb, APC, DCC, NF-1, NF-2, BRCA2, p16, FHIT, WT-1, MEN-I, MEN-II, BRCA1, VHL, FCC, MCC, ras, myc, neu, raf, erb, src, jyrts, jun, trk, ret, gsp, hst, bcl and abl.
8. Pharmaceutical Compositions and Routes of Administration It is contemplated that the therapeutic compositions of the present invention may be administered, in vitro, ex vivo or in vivo. Thus, it will be desirable to prepare the complex as a pharmaceutical composition appropriate for the intended application. Generally this will entail preparing a pharmaceutical composition that is essentially free of pyrogens, as well as any other impurities that could be harmful to humans or animals. One also will generally desire to employ appropriate salts and buffers to render the complex stable and allow for complex uptake by target cells.

The compositions of the present invention comprise an effective amount of the expression construct or a viral vector or .liposome carrying the expression construct, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. Such compositions also can be referred to as inocula. The 5 phrases "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. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and 1 o absorption delaying agents and the like. The use of such media and agents for pharmaceutically 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 also can be incorporated into the compositions.
15 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 also can 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 20 of microorganisms.
The therapeutic compositions of the present invention may include classic pharmaceutical preparations for use in therapeutic regimens, including their administration to humans. Administration of therapeutic compositions according to the present invention will be via any common route so long as the target tissue or 25 cell is available via that route. This includes oral, nasal, buccal, rectal, vaginal or topical. Alternatively, administration will be by orthotopic, intradermal subcutaneous, intramuscular, intraperitoneal, or intravenous injection. Such compositions would normally be administered as pharmaceutically acceptable compositions that include physiologically acceptable carners, buffers or other excipients. For application against tumors, direct intratumoral injection, injection of a resected tumor bed, regional (e.g., lymphatic) or systemic administration is contemplated. It also may be desired to perform continuous perfusion over hours or days via a catheter to a disease site, e.g., a tumor or tumor site.
The therapeutic compositions of the present invention are administered advantageously in the form of injectable compositions either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared. These preparations also may be emulsified. A
typical composition for such purpose comprises a pharmaceutically acceptable carrier. For instance, the composition may contain about 100 mg of human serum albumin per milliliter of phosphate buffered saline. Other pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like may be used. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil and injectable organic .
esters such as ethyloleate. Aqueous carriers include water, alcoholiclaqueous solutions, saline solutions, parenteral vehicles such as sodium chloride, R.inger's dextrose, etc. Intravenous vehicles include fluid and nutrient replenishers.
Preservatives include antimicrobial agents, anti-oxidants, chelating agents and inert gases. The pH and exact concentration of the various components of the pharmaceutical composition are adjusted according to well known parameters.
Additional formulations which are suitable for oral administration also are contemplated. Oral formulations include such typical excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. The compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders. When the route is topical, the form may be a cream, ointment, salve or spray.
An effective amount of the therapeutic agent is determined based on the intended goal, for example (i) inhibition of tumor cell proliferation, (ii) elimination or killing of tumor cells, or (iii) gene transfer for short- or long-term expression of a therapeutic gene. The term "unit dose" refers to physically discrete units suitable for use in a subject, each unit containing a predetermined quantity of the therapeutic composition calculated to produce the desired responses, discussed above, in association with its administration, i. e., the appropriate route and treatment regimen. The quantity to be administered, both according to number of treatments and unit dose, depends on the subject to be treated, the state of the subject and the result desired. Multiple gene therapeutic regimens are contemplated by the present inventors In one embodiment, a vector encoding a therapeutic gene is used to treat cancer patients. Typical amounts of a viral vector used in gene therapy of cancer is 106-lOls PFU/dose e. 106 107 10g 109 10~° 1011 1012 1013 1014 and 101s) ( &> > > > > > > > >
wherein the dose is divided into several injections at different sites within a solid tumor. The treatment regimen also involves several cycles of administration of the gene transfer vector over a period of 3-10 wk. Administration of the vector for longer periods of time from months to years may be necessary for continual therapeutic benefit.
In another embodiment of the present invention, a viral vector encoding a therapeutic gene may be used to vaccinate humans or other mammals. Typically, an amount of virus effective to produce the desired effect, in this case vaccination, would be administered to a human or other mammal so that long term expression of the transgene is achieved and a host immune response develops. It is contemplated that a series of injections, for example, a primary injection followed by two booster injections, would be sufficient to induce an long term immune response. A
typical dose would be from 106 to 1015 PFU/injection depending on the desired result.
Low doses of antigen generally induce a strong cell-mediated response, whereas high doses of antigen generally induce an antibody-mediated immune response.
Precise amounts of the therapeutic composition also depend on the judgment of the practitioner and are peculiar to each individual.

9. Examples The following specific examples are intended to illustrate the invention and should not be construed as limiting the scope of the claims.

The Heat Shock Promoter Can Induce Expression of a Reporter Gene Vector Constructs. To study the ability of the HSP70 promoter to induce gene expression, either a minimal heat shock (HS) promoter or a minimal CMV
promoter was inserted upstream of a reporter gene in a plasmid containing l0 neomycin and ampicillin selectable markers. The basic design of a plasmid (MS) containing one multiple cloning site in operable position to a promoter derived from HSP70 is shown in Figure 1. MS was constructed by replacing the CMV promoter in pcDNA3.0 (Invitrogen, Inc.) with a minimal HSP70B promoter (SEQ ID NO:1, Figure 10), a 0.4 kb fragment (HindIII-BarnHl) of the human heat shock protein 70B {HSP70B) promoter, obtained from StressGen, Inc.
Activity of the minimal HS and CMV promoters were determined by transfecting human cancer cells, MCF7 human breast carcinoma cells and DU 145 human prostate carcinoma cells, with the plasmid S8. The S8 plasmid, derived from the MS vector of Figure 1, contains the minimal HSP70B promoter operably linked to the gene encoding Enhanced Green Fluorescence Protein (EGFP). S8 was constructed by inserting the EGFP gene from pEGFP-1 (Clonetech, Inc.) into the multiple cloning site (MCS) of M5.
Cell Culture and Transfection. Human DU-145 prostate cancer derived cells and MCF-7 human breast cancer derived cells were transfected with the S8 vector described above. To isolate cells stably transfected with S8, cultures were transfected using standard calcium phosphate precipitation methods. Cells containing the integrated plasmids were selected for their ability to proliferate in the presence of neomycin. Heat shock was administered by completely submersing culture flasks in a temperature controlled (t0.1 °C) waterbath.
One positive clone, clone 4, and a polyclonal line were selected with geneticin from the MCF7 cells' transfection. One polyclonal line was selected with geneticin from the DU-145 cell's transfection. (In each case, the cells were selected with geneticin for 2 weeks.) The selected lines were then analyzed and sorted by FACS.
Isolation of Positive Cell Lines. Cells expressing high levels of EGFP in l0 response to heat shock were selected both using conventional serial dilution methods and by fluorescence activated cell sorting {FACS) methods. Expression of EGFP was quantitated using flow cytometry. The Enhanced Green Fluorescence Protein (EGFP) excites at 490 run allowing it to be viewed under a fluorescence microscope or analyzed by FACS. Cells expressing EGFP were sorted from cells not expressing EGFP by using the FACS method. This was done with Geneticin-selected cell lines. The reason that this is required is that in a polyclonal cell line there are populations that have the S8 plasmid integrated in a way that interferes with the expression of the reporter gene. By sorting the cells these populations can be removed leaving the purely positive population for further analysis.
As seen in Figure 2, mean fluorescence from EGFP in DU-145 cells stably transfected with the minimal heat shock promoter driving EGFP (S8) and growing at 37°C was approximately 10 relative fluorescence units. Four hours after exposure to 42°C heat shock for one hour, the mean relative fluorescence was 7-9 times greater. Relative gene expression was subsequently quantitated by measuring changes in relative fluorescence in stably transfected cells. The sorting by FACS of MCF7 cells transfected with the S8 plasmid is illustrated in Figure 3.
Kinetic Studies. Heat exposure survival studies were conducted to evaluate the optimal times/temperatures at which MCF7 cells could be heated without causing massive cell death. For 40°C and 42°C up to 1 hour cell death was found to be negligible with less than a 3% cell death. At 44°C for a time of only 30 minutes almost 50% of the cells had died.
Using the optimal survival times above, initial kinetic studies were 5 performed. Heating transfected MCF7 cells for 1 hour at 40°C and 42°C produced more EGFP than heating for only 30 minutes when assayed by FACS. The optimal recovery time for the cells after heating was 4 hour. Any additional recovery time did not increase the levels of EGFP. For heat treatments done at 44°C
for 30 min, the recovery time took longer with 8 hours being maximal.
10 EGFP Expression at 37°C-44°C in Various Cell Lines. The following heating/recovering times were used as identified in the kinetic studies for testing the inducibility of EGFP driven by the HSP70-derived promoter in all of the inventors' transfected cell lines:

40°C - 1 h of heat treatment, 4 h of recovery 42°C - 1 h of heat treatment, 4 h of recovery 44°C - 30 min of heat treatment, 8 h of recovery Using these temperatures/times the following cell lines were tested for EGFP
expression:
MCF7: breast carcinoma parental cell line.
Du145: prostate carcinoma parental cell line.
MCF7-S8-P: MCF7 cells transfected with the S8 plasmid, polyclonal line.
MCF7-S8-PS1: MCF7-S8-P cells that were sorted for EGFP
expression by FACS once.
MCF7-S8-PS2: MCF7-S8-PS 1 cells that were resorted for EGFP
expression by FACS.
MCF7-S8-4: Clone 4 of the MCF7 S8 transfection.
MCF7-S8-451: MCF7-S8-4 cells sorted once for EGFP expression.
Du145-S8-P: Du145 cells transfected with the S8 plasmid, polyclonal line.
The expression seen from the transfected lines of EGFP driven by the HSP70-derived promoter is shown in Figure 4. As the temperature increases the relative amount of EGFP also increases. These data show that the inventors' heat shock promoter does indeed respond to heat. However, at 37°C there was still expression of EGFP.
EGFP Expression in Stably Transfected DU-145 Cells After Heat Shock.
The induction of endogenous heat shock promoters is transient and temperature dependent. When DU-145 cells, stably transfected with the minimal HS promoter driving EGFP expression (S8) and selected twice by FACS {DU-S8-PS2 cells), were heat shocked for various times and at various temperatures, reporter gene expression was temperature-dependent and expression was transient with maximal values at 15-24 hours after the inducing stress (Figure 5). These results indicate that the promoter is transiently activated under the conditions used here and that EGFP is unstable, since fluorescence decreases after 15-24 hours in these cells.
The minimal heat shock promoter activity increases transiently by approximately 3 fold after a 40°C heat shock for either 1 or 2 hours. Promoter activity increases 13 l0 and 25 fold after 42°C heat shock for either 1 or 2 hours, respectively.
Comparison of Expression of EGFP Under Control of Heat Shock and CMV Promoters. The data presented in Figure 6 show that minimal heat shock promoter activity in DU-S8-PS2 cells is temperature-dependent over the range from 37-43°C. In contrast, DU-145 cells stably transfected with V9, a vector in which the CMV promoter drives EGFP expression (Figure 7), express nearly 50% higher levels of promoter activity than do these same cells transfected with the minimal heat shock promoter and induced with 43°C heat shock. The CMV promoter activity is essentially unaffected by temperature in these cells. The temperature-dependence of the minimal HS promoter is not specific to the DU-145 cells.

Expression of IL-2 Can Be Amplifeed by the Use of a HIV Promoter and tat in a Construct.
Initial Amplifier Studies. Studies involving new constructs capable of amplifying a therapeutic gene's expression were performed. To demonstrate the principle of the amplifier idea, several constructs were produced. The constructs contain a constitutive promoter, the CMV promoter, rather than a heat-shock induible promoter. These constructs are the plasmids L-27, X 14; RR I 3, Y 15, and SS 10. Table 3, below, shows the promoters/genes present in each plasmid and the ~8 amount of iL-2 produced. Four of the plasmids were obtained from a plasmid containing two multiple cloning sites. In these four plasmids, the CMV
promoter was inserted upstream of either the tat gene or a multiple cloning site (MCS) and either the HIV 1 or HIV2 long terminal repeats (LTRs) was inserted upstream of the mouse interleukin-2 (IL-2) gene. The plasmids X 14 and Y 15 are shown schematically in Figure 9A and 9B. The L-27 plasmid served as a reference. IL-was measured from tissue culture supernatants by ELISA using the IL-2 EASIA
kit (Medgenix Diagnostics, Fleurus, Belgium). The sensitivity of the kit is estimated at O.lIU IL-2/ml. In this study, SW480 cells were transfected with the lipid Dosper l0 (see the transfection protocol of Example 3, below.).

Plasmid Name Promoter/gene Amount of IL-2 in LU.
Lipid alone Dosper .48 L-27 CMV/IL-2 15.63 RR13 HIV 1/IL-2 17.56 CMV/multiple cloning site X 14 HI V 1 /IL-2 173 . 7 CMV/TAT
SS 10 HIV2/IL-2 12.83 CMV/multiple cloning site Y 15 HIV2/IL-2 440.55 CMV/TAT

WO 99/2321b PCT/US98I23387 It can be seen from this study that the complete amplifier constructs are capable of increased expression over the CMV promoter. Also, the production of the transactivating factor, TAT, is required for this increased production.

Heat Inducible Amplifiers.
Vector Construction. To determine if a second promoter could be used to increase the activity of the minimal HS promoter, MCF-7 cells were transiently transfected with a series of vectors, including pCB, pfl2, and p007 (Figures 8 and 9). Using a plasmid containing two multiple cloning sites, the minimal heat shock promoter was inserted upstream of either the tat gene or a multiple cloning site (MCS) and either the HIV 1 or HIV2 long terminal repeats (LTRs) was inserted upstream of the mouse interleukin-2 (IL-2) gene. The plasmids also each carried neomycin and ampicillin selectable markers.
The plasmid fll was first created by inserting a 0.5 kb EcoRI fragment, containing the interleukin-2 (IL-2) coding region (see GenBank accession no.
577834), from plasmid CS into the EcoRI site of the vector MS (see specific example, Example 1, above). The plasmid C8 was constructed by inserting a 1 kb BarnHI fragment, containing the 0.4 kb HSP70B fragment upstream of a MCS from plasmid B4527 (see Tsang, et al., Biotechniques 20:51-52, 1996 and Tsang et al., 2o Biotechniques 22:68, 1997, both herein incorporated by reference}, into the BamHI
site of plasmid DNP-1 (Tsang et al., 1996, and Tsang et al., 1997), which contains the HIV1 LTR upstream of the IL-2 coding region. The vector f12 (Figure 9) was then constructed by inserting a 0.4 kb NotI fragment, containing the coding region for the HIV tat gene, into the NotI site of C8. An intermediate vector D10 was constructed by inserting the 1 kb BamH I fragment containing the minimal promoter into plasmid MNP-7 (Tsang et al., 1996, and Tsang et al., 1997), which contains the HIV2 LTR upstream of the IL-2 coding region. Plasmid 007 (Figure 9) was created by inserting the 0.4 kb Not I fragment, encoding the tat gene, into the Not I site of D 10.
Transfection Protocol. Transfection were performed according to the published procedure (Stopeck, et al., Cancer Gene Therapy, 5:119-126, 1998.) 5 MCF-7 cells were plated in either a 6 well or 12 well plates. The next day, cells were washed with Hanks Buffered Saline Solution and replaced with a 1 rnl transfection solution. The transfection solution was a 4:1 lipid to DNA mass ratio of either Dosper (1,3-Di-Oleoyloxy-2-(6-Carboxy-spermyl)-propylamid, from Boehringer Mannheim) or Dmrie C ( 1,2-dimyristyloxypropyl-3dimethyl-hydroxy 10 ethyl ammonium bromide, from Gibco BRL) with either 1.25 ~.g or 2.5 p,g of plasmid DNA in serum-free OptiMEM (from Gibco BRL). Fetal bovine serum (FBS) was immediately added to each well to a final concentration of 10%
(vol/vol). Dmrie C was determined to be a better lipid then Dosper. Cells were incubated for 24 hours before heating and 24 hours after heating prior to IL-2 15 quantitation.
Heat-Induced Amplification Studies. In one set of experiments, cells transfected with the pCB, pfl2, or p007 plasmids were assayed for IL-2 expression activity. IL-2 was measured from tissue culture supernatants by ELISA using the IL-2 EASIA kit (Medgenix Diagnostics, Fleurus, Belgium). The sensitivity of the 20 kit is estimated at 0. lIU IL-2/ml. The data from this set of experiments are shown in Table 4, below. This table shows IL-2 expression levels in MCF7 cells which were transfected with Dosper, heated 24 hours later, and were then assayed by ELISA 49 hours after the transfection. The plasmids L-27 (a plasmid used for reference that expresses IL-2 driven by the CMV promoter), 007, f12, and C8 were 25 all tested.

__ _. -LU. of II,-2 . .

temperature:37C 39C 41C 42C 44C

heat shock continuous continuous1 hr lhr 0.5 hr .

duration:

Lipid alone2.03 0.50 0.41 0.53 0.53 L-27 14.2 9.88 5.95 9.88 7.80 007 336.76 318.49 334.02 373.74 389.27 F12 8.40 6.88 49.93 60.02 88.13 C8 9.19 8.03 11.74 8.73 16.37 From this study it can be seen that pfl2 is responsive to heating and produces larger quantities of the heat shock amplifier construct IL-2 than either pC8 or pL-27 does. At 37°C, pfl2 produced 5-fold more IL-2 than its CMV
driven control, L-27. When cells were heat shocked at 39°C overnight, pfl2 produced 7-fold more IL-2 than the CMV driven controls at 37°C. A 1 hour heat shock treatment at 41°C or 42°C increased expression from the amplifier constructs by as much as 26-fold, compared to the CMV-driven control vector at 37°C.
(However, the p007 plasmid at 37°C is already near its maximal activity and does not increase expression levels greatly with heat.) The activity of pfl2 is also at a high level at 37°C. These results showed that the amplifier strategy can augment the levels of gene expression at temperatures between about 37°C and about 42°C.
In a different set of experiments, variations in transfection efficiencies were accounted for by co-transfection with a control plasmid in which the CMV
promoter was upstream of ~i-galactosidase. The general protocol for these experiments was to transfect cells 24 hours after subculture, heat shock cultures an additional 24 hours later, change culture medium and then collect medium for measurement of IL-2 levels 24 hours later. As seen in Table 5, below, the activity of the CMV promoter was only minimally affected by heat shock. The minimal heat shock promoter activity was very low in cells maintained at 37°C
and was induced over 20 fold by heat shock at 42°C. As seen in the stably transfected cells, the minimal heat shock promoter activity was only about one half that of the CMV
promoter.

Interleukin-2 (IL2) Expression*
Vector Promoter 37°C 42°C** Fold (42/3?) Relative***
L27 CMV-IL2 82.6 93.4 1.1 1.0 C8 HSP-MCS 84.7 70.6 0.8 1.9 fll HSP-IL2 2.3 54.0 23.7 0.4 (1) f12 HSP-TAT 107.6 347.4 3.2 , 6.9 (17) 007 HSP-TAT 747.5 1642.9 2.2 83.3 (208) * values in IU IL2 produced per mg cell protein in 24 hours ** heat shock was for 1 hour *** based on 42°C values and co-transfection with CMV-~-gal The HIV1 promoter, in the absence of tat expression, was similar to that of the CMV promoter and was nearly independent of heat shock. However, when the minimal heat shock promoter was used to express tat, reporter gene expression was dramatically increased after 42°C heat shock. In cells transiently transfected with heat shock promoter/tat and HIV1/IL-2, IL-2 production was similar to that for heat shock promoter/MCS and HIV 1/IL-2 in cells maintained at 37°C. This activity was increased over 3 fold, and to levels nearly 7 fold greater than CMV promoter activity by itself, after 42°C HS.
The HS promoter/tat and HIV2/IL-2 transfected cells showed substantial reporter gene expression in cells maintained at both 37 and after 42°C
heat shock.
Relative promoter activity, measured by IL-2 production was over 80 fold higher than that for the CMV promoter. alone. Temperature regulation was reduced, with reporter gene expression approximately 2 times higher after 42°C heat shock 1 o compared to the same activity in cells maintained at 37°C.
The temperature dependence of reporter gene expression was not influenced by the presence of a second promoter. As shown in Table b, below, reporter gene expression, in cells transiently transfected with the minimal heat shock promoter/tat and HIV2/IL-2 containing plasmid, increased in a temperature-dependent manner between 37 and 44°C. These results are qualitatively similar to those seen in Figures 4 and 6 for cells stably transfected with only the minimal heat shock promoter.

Ii.-2 Expression ~IU/ml)*
Vector Promoter 37°C 39°C 40°C 41°C
42°C 44°C
C8 HSP-MCS 7.2 9.3 6.0 4.8 5.3 7.0 fl2 HSP-TAT 40.6 -- -- -- 133.1 -MCF7 breast cancer cells were transiently transfected with vectors as shown;
heat shocked for 1 hr 24 hrs later; media were collected and IL2 measured 24 hrs after heat shock Animal Studies Mouse models of human cancer, with the histologic features and metastatic 5 potential resembling tumors seen in humans, can be treated with the therapeutic compositions of the present invention. In one embodiment of the present invention, SCID mice are injected with human tumor cells stably transfected with reporter constructs in which the HSP70B promoter is driving the expression of TAT and in which the HIV-1 or HIV-2 promoter is driving either EGFP or IL-2 expression.
10 After growing the tumors to an appropriate measurable size of for example, 1 cm in diameter, the tumors are heated using ultrasound to temperatures up to about 42°C.
Gene expression is quantitated at various times after heating by either removing the tumor, making tissue slices and measuring fluorescence from EGFP or measuring tumor tissue levels of IL2 using ELISA. Using another embodiment of the present 15 invention, human tumor cells are injected into SCID mice. The tumors grown to an appropriate measurable size and injected with DNA-lipid complexes. Tumors are heated using ultrasound and gene expression measured at times after heating.
The efficacy of these treatments is indicated by a decrease in the size of the tumor, a decrease in metastatic activity, a decrease in cell proliferation or a halt in the tumor 20 growth as a result of the administration of the therapeutic compositions of the present invention.
Various modifications and variations of the present invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific 25 preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in the art are intended to be within the scope of the following claims.

WO 99/23216 PCT/US9$/23387 SEQUENCE LINING
<110> Tsang, Tom Gerner, Eugene vl.
Harris, David T.
Hersh, Evan <120> Hyperthermic Inducible Erpression Vectors for Gene Therapy and Methods of Use Thereof <130> 15907-0016 <140>
<141>
<150> US 60/064,088 <151> 1997-11-03 <160> 1 <170> PatentIn Ver. 2.0 <210> 1 <211> 469 <212> DNA
<213> Homo sapiens <400> 1 ggatcctcca cagccccggg gagaccttg~ ~tctaaagtt gctgcttt~g cagctctgcc 60 acaaccgcgc gtcctcagag ccagccggc~ ~gagctaaaa ccttccccgc gtttctttca 120 gcagccctga gtcagaggcg ggctggcct= gcaagtagcc ccccagcc~t cttcggtctc 180 acggaccgat ccgcccgaac cttctcccg: ggtcagcgcc gcgctgcgcc gcccggctga 240 ctcagcccgg gcgggcgggc gggaggctct cgactgggcg ggaaggtgcg ggaaggttcg 300 cggcggcggg gtcggggagg tgcaaaagg~ tgaaaagccc gtggacggag ctgagcagat 360 ccggccgggc tggcggcaga gaaaccgcag ggagagcctc actgctgagc gcccctcgac 420 gcgggcggca gcagcctccg tggcctccac catccgacaa gaagcttac 469 SUBSTITUTE SHEET (RULE 28)

Claims (46)

WHAT IS CLAIMED IS:
1. A method of effecting expression of a selected polynucleotide in a mammalian cell comprising:
(a) providing an expression construct, said expression construct comprising (i) an inducible promoter operably linked to a gene encoding a transactivating factor; and (ii) a second promoter operably linked to said selected polynucleotide, wherein said second promoter is activated by said transactivating factor;
(b) introducing said expression construct into said cell; and (c) subjecting said cell to conditions which activate said inducible promoter, wherein said conditions result in the expression of said selected polynucleotide.
2. The method of claim 1, wherein said inducible promoter is a heat shock promoter and the conditions which activate said inducible promoter are hyperthermic conditions.
3. The method of claim 2, wherein said hyperthermic conditions comprise a temperature between about basal temperature for said cell and about 42°C.
4. The method of claim 3, wherein said hyperthermic conditions comprises a temperature between about 37°C and about 42°C.
5. The method of claim 4, wherein said hyperthermic conditions comprise a temperature between about 38°C and about 41°C.
6. The method of claim 5, wherein said hyperthermic conditions comprise a temperature between about 39°C and about 40°C.
7. The method of claim 2, wherein said heat shock promoter is derived from a promoter selected from the group consisting of the HSP70, HSP90, HSP60, HSP27, HSP72, HSP73, HSP25, ubiquitin, and HSP28 promoters.
8. The method of claim 1, wherein said inducible promoter comprises a hypoxia-responsive element.
9. The method of claim 1, wherein said second promoter is selected from the group consisting of an HIV-1 promoter and an HIV-2 promoter, and said transactivating factor is tat.
10. The method of claim 1, wherein the expression of said selected polynucleotide results in the production of a polypeptide, a protein, a ribozyme, or an antisense nucleic acid.
11. The method of claim 1, wherein said selected polynucleotide encodes a protein selected from the group consisting of ornithine decarboxylase antizyme protein, p53, p 16, neu, IL1, IL2, IL4, IL7, IL 12, IL 15, FLT-3 ligand, GM-CSF; G-CSF, IFN.gamma., IFN.alpha., TNF, HSV-TK, I-CAM1, HLA-B7, and TIMP-3.
12. The method of claim 1, wherein said expression construct further comprises a gene encoding a selectable marker.
13. The method of claim 1, wherein said expression construct further comprises (i) a second selected polynucleotide operably linked to said second promoter; and (ii) an internal ribosome entry site positioned between said first and second selected polynucleotides.
14. The method of claim 1, wherein said cell is a tumor cell.
15. The method of claim 1, wherein the introduction of said expression construct into said cell is mediated by a delivery vehicle selected from the group consisting of liposomes, retroviruses, adenoviruses, adeno-associated viruses, lentiviruses, herpes simplex viruses, and vaccinia viruses.
16. The method of claim 1, wherein the introduction of said expression construct into said cell occurs in vitro.
17. The method of claim 1, wherein the introduction of said expression construct into said cell occurs in vivo.
18. A method of providing a subject with a therapeutically effective amount of an expression product of a selected polynucleotide comprising:
(a) providing a first expression construct, said expression construct comprising an inducible promoter operably linked to a gene encoding a transactivating factor;
(b) providing a second expression construct, said second expression construct comprising a second promoter operably linked to said selected polynucleotide, wherein said second promoter is activated by said transactivating factor;
(c) introducing said first and second expression constructs into a cell of said subject; and (d) subjecting said cell to conditions which activate said inducible promoter, wherein expression of said selected polynucleotide is induced by said conditions.
19. The method of claim 18, wherein said inducible promoter is a heat shock promoter and the conditions which activate said inducible promoter comprise a temperature between about basal temperature and about 42°C.
20. The method of claim 19, wherein said first and second expression constructs are on the same vector.
21. The method of claim 20, wherein the introduction of said expression constructs into said cell occurs ex vivo.
22. The method of claim 18, wherein the introduction of said expression constructs into said cell occurs in vivo.
23. The method of claim 18, wherein the expression product of said selected polynucleotide is harmful to a pathogen in said subject, wherein said pathogen is selected from the group consisting of viruses, bacteria, fungi, and parasites.
24. The method of claim 18, wherein the expression product of said selected polynucleotide inhibits the growth of said cell.
25. The method of claim 18, wherein the expression product of said selected polynucleotide replaces a deficient protein in said subject.
26. The method of claim 18, wherein the expression product of said selected polynucleotide promotes nerve regeneration.
27. A method of treating cancer in a mammal comprising the steps of:
(a) providing an expression construct, said expression construct comprising (i) an inducible promoter operably linked to a gene encoding a transactivating factor; and (ii) a second promoter operably linked to a selected polynucleotide, wherein said second promoter is activated by said transactivating factor;
(b) introducing said expression construct into a tumor cell; and (c) subjecting said tumor cell to conditions which activate said inducible promoter, wherein said conditions result in the expression of said selected polynucleotide and the expression product of the selected polynucleotide is expressed in an amount effective to inhibit the growth of said tumor cell.
28. The method of claim 27, wherein said inducible promoter is a heat shock promoter and the conditions which activate said inducible promoter are hyperthermic conditions comprising a temperature between about basal temperature and about 42°C.
29. The method of claim 27, further comprising treating said tumor cell with at least one established form of therapy for cancer which is selected from the group consisting of external beam radiation therapy, brachytherapy, chemotherapy, and surgery.
30. The method of claim 27, further comprising:
(d) treating said tumor cell with the radioprotector WR-33278 or WR-1065 after subjecting it to hyperthermic conditions; and (e) in a final step, treating said tumor cell with radiation therapy, wherein said selected polynucleotide encodes ornithine decarboxylase antizyme protein.
31. The method of claim 27, wherein said mammal is a human.
32. The method of claim 27, wherein said cancer is selected from the group consisting of cancers of the brain, lung, liver, bladder, spleen, kidney, lymph node, small intestine, pancreas, blood cells, colon, stomach, breast, endometrium, prostate, testicle, ovary, skin, vulva, cervix, head and neck, esophagus, bone marrow, and blood.
33. A method for provoking an immune response in a mammal comprising:
(a) providing an expression construct, said expression construct comprising (i) an inducible promoter operably linked to a gene encoding a transactivating factor; and (ii) a second promoter operably linked to a selected polynucleotide, wherein said second promoter is activated by said transactivating factor;
(b) introducing said expression construct into a cell in the mammal; and (c) subjecting said cell to conditions which activate said inducible promoter, wherein said conditions result in the expression of said selected polynucleotide and the expression product of the selected polynucleotide is expressed in an amount effective to provoke an immune response in said mammal, said immune response being selected from the group consisting of a humoral immune response and a cellular immune response.
34. The method of claim 33, wherein the inducible promoter is a heat shock promoter and the conditions which activate said inducible promoter are hyperthermic conditions comprising a temperature between about basal temperature and about 42°C.
35. The method of claim 33, wherein the immune response is directed against said cell.
36. The method of claim 35, further comprising treating said cell with an established form of therapy for cancer selected from the group consisting of chemotherapy, external beam radiation therapy, brachytherapy, and surgery.
37. The method of claim 33, wherein said mammal is a human.
38. A method of altering the genetic material of a mammal, comprising:
(a) providing an expression construct, said expression construct comprising (i) an inducible promoter operably linked to a gene encoding a transactivating factor; and (ii) a second promoter operably linked to said selected polynucleotide, wherein said second promoter is activated by said transactivating factor; and (b) introducing said expression construct into a cell of said mammal.
39. An expression construct comprising:
(a) a gene encoding a transactivating factor;
(b) an inducible promoter operably linked to said gene;
(c) a selected polynucleotide; and (d) a second promoter operably linked to said selected polynucleotide, said second promoter being activated by said transactivating factor.
40. The expression construct of claim 39, wherein said inducible promoter is a heat shock promoter and expression of said selected polynucleotide is induced by hyperthermic conditions, said hyperthermic conditions comprising a temperature between about 37°C and about 42°C.
41. The expression construct of claim 40, wherein said heat shock promoter is derived from a promoter selected from the group consisting of the HSP70, HSP90, HSP60, HSP27, HSP72, HSP73, HSP25, ubiquitin, and HSP28 promoters.
42. The expression construct of claim 39, wherein said inducible promoter comprises a hypoxia-responsive element.
43. The expression construct of claim 39, wherein said second promoter is selected from the group consisting of HIV-1 promoter and HIV-2 promoter and said transactivating factor is selected from the group consisting of tat.
44. The expression construct of claim 39, wherein expression of said selected polynucleotide results in the production of a polypeptide, protein, ribozyme or antisense molecule.
45. The expression construct of claim 39, wherein said expression construct further comprises (i) a second selected polynucleotide operably linked to said second promoter; and (ii) an internal ribosome entry site positioned between said first and second selected polynucleotides.
46. A cell comprising the expression construct of claim 39.
CA002308575A 1997-11-03 1998-11-03 Hyperthermic inducible expression vectors for gene therapy and methods of use thereof Abandoned CA2308575A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US6408897P 1997-11-03 1997-11-03
US60/064,088 1997-11-03
PCT/US1998/023387 WO1999023216A2 (en) 1997-11-03 1998-11-03 Hyperthermic inducible expression vectors for gene therapy and methods of use thereof

Publications (1)

Publication Number Publication Date
CA2308575A1 true CA2308575A1 (en) 1999-05-14

Family

ID=22053477

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002308575A Abandoned CA2308575A1 (en) 1997-11-03 1998-11-03 Hyperthermic inducible expression vectors for gene therapy and methods of use thereof

Country Status (9)

Country Link
EP (1) EP1029041A2 (en)
JP (1) JP2001521740A (en)
KR (1) KR20010031762A (en)
CN (1) CN1299411A (en)
AU (1) AU1377299A (en)
BR (1) BR9813917A (en)
CA (1) CA2308575A1 (en)
IL (1) IL135943A0 (en)
WO (1) WO1999023216A2 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19951765A1 (en) * 1999-10-27 2001-05-03 Thomas Schweder Host-vector systems for the overproduction of thermolabile enzymes in psychrophilic organisms
US6579522B1 (en) 2000-06-27 2003-06-17 Genvec, Inc. Replication deficient adenoviral TNF vector
US7285414B2 (en) 2000-09-26 2007-10-23 Emory University Viruses targeted to hypoxic cells and tissues
ATE418342T1 (en) * 2000-09-26 2009-01-15 Univ Emory VIRUSES AGAINST HYPOXIC CELLS AND TISSUES
US20030086903A1 (en) 2001-11-02 2003-05-08 Genvec, Inc. Therapeutic regimen for treating cancer
DK1541674T3 (en) 2002-06-18 2011-03-14 Eisai R&D Man Co Ltd Primary cultured adipocytes for gene therapy
US20060015951A1 (en) * 2002-10-14 2006-01-19 Karsten Eulenberg Proteins involved in the regulation of energy homeostasis
JP2008048601A (en) * 2004-06-15 2008-03-06 Nippon Kosei Kagaku Kenkyusho:Kk Method for producing therapeutically useful product and system
JP2008540347A (en) * 2005-05-03 2008-11-20 フェテリネールメディツィニシュ ウニベルジテート ウィーン Osmotic capsule
US20090136465A1 (en) * 2007-09-28 2009-05-28 Intrexon Corporation Therapeutic Gene-Switch Constructs and Bioreactors for the Expression of Biotherapeutic Molecules, and Uses Thereof
CN103667345B (en) * 2013-12-02 2015-10-28 东南大学 A kind of Antioncogene magnetic composite nano particle and preparation method
WO2017133175A1 (en) * 2016-02-04 2017-08-10 Nanjing Legend Biotech Co., Ltd. Engineered mammalian cells for cancer therapy
CN108913661A (en) * 2018-06-22 2018-11-30 安徽 A kind of preparation and extracting method of human research's candidate stem cell
CN114317588B (en) * 2020-09-29 2023-06-27 中国农业科学院生物技术研究所 Accurate methylation regulation and control return circuit of crop high temperature response formula genome
IT202200000788A1 (en) * 2022-01-19 2023-07-19 Bruno Cell S R L Temperature-sensitive induction of the myogenic and/or adipogenic program in cells producing muscle and/or adipose tissue
CN116536349B (en) * 2023-05-22 2024-02-20 安徽农业大学 Application of soybean GmMLP34 gene in regulation and control of high temperature resistance of plants

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3991770A (en) * 1974-01-24 1976-11-16 Leveen Harry H Method for treating benign and malignant tumors utilizing radio frequency, electromagnetic radiation
EP0299127A1 (en) * 1987-07-16 1989-01-18 IntraCel Corporation A method for the expression of recombinant genes under stimulation from an inducively expressed activator protein
EP0455424A3 (en) * 1990-05-02 1992-04-29 Merck & Co. Inc. Mammalian inducible promoter cascade system
FR2717187B1 (en) * 1994-03-10 1996-05-31 Transgene Sa Combined use of two expression cassettes for the production of a protein of interest.

Also Published As

Publication number Publication date
JP2001521740A (en) 2001-11-13
IL135943A0 (en) 2001-05-20
BR9813917A (en) 2001-10-30
KR20010031762A (en) 2001-04-16
CN1299411A (en) 2001-06-13
AU1377299A (en) 1999-05-24
WO1999023216A3 (en) 1999-08-05
WO1999023216A2 (en) 1999-05-14
EP1029041A2 (en) 2000-08-23

Similar Documents

Publication Publication Date Title
CA2308575A1 (en) Hyperthermic inducible expression vectors for gene therapy and methods of use thereof
EP0784693B1 (en) Viral vectors for gene therapy
AU1755299A (en) Vector for tissue-specific replication and gene expression
WO1994012629A9 (en) Episomal vectors for gene therapy
US20070244064A1 (en) Methods and compositions to induce antitumor response
JPH10509880A (en) Vectors for tissue-specific replication
JP2004501650A (en) Replication deficient adenovirus TNF vector
JP2002506651A (en) Multi-gene vector
US6709858B1 (en) Hyperthermic inducible expression vectors for gene therapy and methods of use thereof
ES2268695T3 (en) ADENOVIRUS DEFECTIVE RECOMBINANTS WITH AN INACTIVATED VAT2 GEN.
US7056897B2 (en) Inducible expression vectors and methods of use thereof
US6451579B1 (en) Regulated expression of recombinant proteins using RNA viruses
Tomita et al. Gene therapy with transcription factor decoy oligonucleotides as a potential treatment for cardiovascular diseases
Engelhard Gene therapy for brain tumors: the fundamentals
Kouraklis Gene therapy for cancer
US7098191B2 (en) Hyperthermic inducible expression vectors for gene therapy and methods of use thereof
WO1997016547A1 (en) ADENOVIRUS-ANTISENSE K-ras EXPRESSION VECTORS AND THEIR APPLICATION IN CANCER THERAPY
MXPA00004293A (en) Hyperthermic inducible expression vectors for gene therapy and methods of use thereof
WO2000022124A2 (en) Methods and compositions to induce antitumor response
Clayman Gene therapy for head and neck cancer
JP7406263B2 (en) Modified adenovirus and medicines containing it
JPH10127287A (en) New adenovirus vector for transducing human gene-in organism
US5863794A (en) SV40 viral vectors for targeted integration into cells
US20040146856A1 (en) Anti-neoplastic viral agents
FR2737221A1 (en) Viral vector for gene therapy includes heterologous regulatory sequence - so that viral genes are functional only in complementing cells, also derived infectious particles

Legal Events

Date Code Title Description
FZDE Discontinued