WO1998000542A2 - Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie - Google Patents

Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie Download PDF

Info

Publication number
WO1998000542A2
WO1998000542A2 PCT/US1997/011785 US9711785W WO9800542A2 WO 1998000542 A2 WO1998000542 A2 WO 1998000542A2 US 9711785 W US9711785 W US 9711785W WO 9800542 A2 WO9800542 A2 WO 9800542A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
factor viii
human
recombinant
recombinant retrovirus
Prior art date
Application number
PCT/US1997/011785
Other languages
English (en)
Other versions
WO1998000542A9 (fr
Inventor
Douglas J. Jolly
Jack R. Barber
Stephen M. W. Chang
James G. Respess
John R. Allen
Mordechai Boder
Kimberly Chong
Dan De La Vega, Jr.
Nicholas J. Depolo
David Chi-Tang Hsu
Carlos E. Ibanez
Denice M. Mittelstaedt
Charles E. Prussak
Judith Greengard
Original Assignee
Chiron Corporation
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 Chiron Corporation filed Critical Chiron Corporation
Priority to EP97940535A priority Critical patent/EP0915975A2/fr
Publication of WO1998000542A2 publication Critical patent/WO1998000542A2/fr
Publication of WO1998000542A9 publication Critical patent/WO1998000542A9/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/7455Thrombomodulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/755Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8121Serpins
    • C07K14/8125Alpha-1-antitrypsin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8121Serpins
    • C07K14/8128Antithrombin III
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • C12N9/1211Thymidine kinase (2.7.1.21)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/644Coagulation factor IXa (3.4.21.22)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6464Protein C (3.4.21.69)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/647Blood coagulation factors not provided for in a preceding group or according to more than one of the proceeding groups
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21022Coagulation factor IXa (3.4.21.22)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21069Protein C activated (3.4.21.69)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13041Use of virus, viral particle or viral elements as a vector
    • C12N2740/13043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to methods of administration of recombinant gene transfer vehicles for the treatment of hemophilia, thrombosis, and other diseases.
  • the present invention also relates generally to recombinant retroviruses, and more specifically, to high titer recombinant retroviral particle preparations suitable for a variety of applications.
  • Hemophilia is a genetic disease characterized by a severe blood clotting deficiency.
  • hemophilia A a X- chromosome linked genetic defect disrupts the gene encoding factor VIII, a trace plasma glycoprotein which acts as a cofactor in conjunction with factor IXa in the activation of factor X.
  • factor VIII gene codes for 2,351 amino acids.
  • the protein has six domains, designated from amino to carboxy terminus as Al , A2, B, A3, C 1 , and C2, respectively (Wood et al., 1984, Nature 312:330; Vehar et al., 1984, Nature 572:337; and Toole et al., 1984, Nature 572:342) with a deduced molecular weight of about 280 kilo Daltons (kD).
  • the 980 amino acid B domain is deleted in the activated procoagulant form of the protein. Additionally, in the native protein two polypeptide chains, the heavy and light chain, flanking the B domain, are bound to a divalent calcium cation.
  • hemophilia A affects about one in every 10,000 males. Due to the resultant clotting deficiency, those afflicted with the disease suffer severe bleeding episodes due to small injuries, internal bleeding, and joint hemorrhage, which leads to arthropathy, the major cause of morbidity in hemophilia. Normal levels of factor VIII average between 50 to 200 ng/ml of blood plasma (Mannucci, P.M. in Practical
  • hemophilia A typically have plasma levels well below 2 - 60 ng/ml, while levels below about 2 ng/mL result in severe hemophilia.
  • therapy for hemophilia A involved repeated administration of human factor VIII purified from blood products pooled in lots from over 1000 donors.
  • factor VIII to total protein typically were highly impure, with an estimated purity by weight (factor VIII to total protein) of approximately 0.04%.
  • hemophilia A patients in the Unites States do not presently receive factor VIII maintenance therapy, but instead only receive the polypeptide prior to activities or events which might cause bleeding, such as surgery, or to treat spontaneous bleeding.
  • factor VIII typically 24 - 40 IU per kilogram bodyweight
  • Such therapy kept factor VIII concentrations from falling below 1% of normal (Nillson, et al., 1992, J. Internal Med. 232:23).
  • a genetic therapy affording continuous, long-term therapeutical ly effective expression levels or amounts of factor VIII i.e., to decrease the severity of or eliminate the clotting disorder associated with hemophilia A, would be of great benefit.
  • Hemophilia B A condition clinically indistinguishable from Hemophilia A is Hemophilia B, resulting from the deficiency of clotting factor IX.
  • the incidence of this condition is about 5-fold lower than that of hemophilia A, and presents many of the same therapeutic challenges and difficulties. For similar reasons, it would be of great benefit to provide a gene therapy to these patients.
  • Factor X deficiency results in a rare but serious bleeding disorder affecting 1 in 500,000 known as Stuart disease.
  • Le et al., 1997, Blood 89: 1254-9 describes therapeutic levels of functional human factor X in rats after retroviral mediated hepatic gene therapy.
  • a genetic therapy affording continuous, long term therapeutically effective expression levels or amounts of factor X, i.e., to decrease the severity of or eliminate the clotting disorder associated with hemophilia B would be of great benefit.
  • the instant invention also relates to the production and use of high titer recombinant retroviruses.
  • retroviral gene delivery methods have been most extensively utilized, in part due to: (1) the efficient entry of genetic material (the vector genome) into cells; (2) an active, efficient process of entry into the target cell nucleus; (3) relatively high levels of gene expression; (4) the potential to target particular cellular subtypes through control of the vector-target cell binding and the tissue-specific control of gene expression; (5) a general lack of pre-existing host immunity; and (6) substantial knowledge and clinical experience which has been gained with such vectors.
  • retroviruses are diploid positive-strand RNA viruses that replicate through an integrated DNA intermediate.
  • the retroviral genome upon infection by the RNA virus, the retroviral genome is reverse-transcribed into DNA by a virally encoded reverse transcriptase that is carried as a protein in each retrovirus.
  • the viral DNA is then integrated pseudo- randomly into the host cell genome of the infecting cell, forming a "provirus" which is inherited by daughter cells.
  • gag, pol and env genes which are preceded by a packaging signal ( ⁇ ), and two long terminal repeat (LTR) sequences which flank both ends.
  • packaging signal
  • LTR long terminal repeat
  • the gag gene encodes the internal structural (nucleocapsid) proteins.
  • T e pol gene codes for the RNA-dependent
  • DNA polymerase which reverse transcribes the RNA genome, and the env gene encodes the retroviral envelope glycoproteins.
  • the 5' and 3' LTRs contain c -?-acting elements necessary to promote transcription and polyadenylation of retroviral RNA.
  • Adjacent to the 5' LTR are sequences necessary for reverse transcription of the genome (the tRNA primer binding site) and for efficient encapsidation of retroviral RNA into particles (the ⁇ sequence). Removal of the packaging signal prevents encapsidation (packaging of retroviral RNA into infectious virions) of genomic RNA, although the resulting mutant can still direct synthesis of all proteins encoded in the viral genome.
  • the retrovirus When the retrovirus injects its RNA into a cell, the foreign gene is also introduced into the cell, and may then be integrated into the host's cellular DNA as if it were the retrovirus itself. Expression of this foreign gene within the host results in expression of the foreign protein by the host cell.
  • retroviruses One disadvantage, however, of recombinant retroviruses is that they principally infect only replicating cells, thereby making efficient direct gene transfer difficult. Indeed, some scientists have suggested that other, more efficient methods of gene transfer, such as direct administration of pure plasmid DNA, be utilized (Davis et al., 1993, Human Gene Therapy 4:733-740).
  • liver e.g., 70% in Rettinger et al., 1994, PNAS 97:1460-1464; 70% in Moscioni et al., 1993, Surgery 5:304- 31 1 ) in order to stimulate the rapid division of hepatocytes and thereby increase the infection of such cells.
  • retroviruses serum from primates (e.g., humans) is known to cause inactivation by an antibody independent complement lysis method.
  • retroviruses of avian, murine, feline, and simian origin are all inactivated and lysed by normal human serum.
  • the scientific literature has also reported that replication competent murine amphotropic retroviruses injected intravenously into primates are cleared within 15 minutes and that the disappearance is mediated, wholly or in part, by primate complement.
  • the present invention provides recombinant retrovirus compositions which are capable of surviving inactivation in human serum.
  • the present invention provides high titer recombinant retrovirus compositions which allow delivery of therapeutics or palliatives by routes not previously deemed possible, and without the need to induce replication of cells by chemical treatment or by excision of a target organ such as the liver. The present invention provides these, as well as other related advantages. Summary of the Invention
  • This invention provides for preparations of replication defective recombinant retrovirus expressing human factor VIII protein, wherein the recombinant retrovirus is capable of infecting human cells and is resistant to degradation by human complement.
  • the invention also provides for preparations of replication defective recombinant retrovirus expressing human factor VIII protein in which the recombinant retrovirus preparation is resistant to degradation by human complement and is capable of inducing long term systemic expression of human factor VIII when administered intravenously to a human afflicted with hemophilia A.
  • compositions and therapeutic methods of the above-described retroviral vectors expressing factor VIII protein are also provided herein as are therapeutic methods for treatment of hemophilia A by intravenous injection of these retroviral vectors.
  • the retroviral vectors of the invention can expresses a B domain-deleted form of factor VIII, which in one embodiment can be the SQN mutation of factor VIII.
  • the retroviral vectors of the invention can have a titer on HT1080 cells of greater than IO 6 , more preferably 10 7 cfu/ml and more preferably at least 10 8 cfu/ml, more preferably IO 9 cfu/ml, more preferably at least 10 10 cfu/ml, and most preferably 10" cfu/ml.
  • the present invention provides high titer compositions comprising recombinant retroviruses, as well as methods for utilizing these compositions.
  • methods are provided for obtaining measurable levels of a protein, nucleic acid molecule, or enzymatic product in a bodily fluid or cells of a human, comprising the step of administering to a human a recombinant retroviral preparation having a titer on HT1080 cells of greater than 10 ⁇ cfu/ml, wherein the recombinant retroviral preparation is capable of directing the expression of a protein, nucleic acid molecule, or enzyme which generates an enzymatic product, such that measurable levels of the protein, nucleic acid molecule, or enzymatic product may be obtained in the bodily fluid or cells of said human.
  • the titer may be greater than IO 6 cfu/ml, IO 7 cfu/ml. IO 8 cfu/ml, IO 9 cfu/ml, IO 10 cfu/ml, or I O 1 1 cfu/ml.
  • methods for obtaining measurable levels of a protein, nucleic acid molecule, or enzymatic product in a bodily fluid or cells of a human, comprising the steps of administering to a human a recombinant retroviral preparation having a titer in human serum and on HT1080 cells equivalent to its' titer in heat-inactivated serum and on HT1080 cells, wherein the recombinant retroviral preparation is capable of directing the expression of a protein, nucleic acid molecule, or enzyme which generates an enzymatic product, such that measurable levels of the protein, nucleic acid molecule, or enzymatic product may be obtained in the bodily fluid or cells of said human.
  • measurable levels of a protein, nucleic acid molecule, or enzymatic product refers to a statistically significant level of detection over background, utilizing any suitable technique (e.g., antibody-mediated detection of a protein, PCR analysis for the presence of a nucleic acid molecule, or visualization of enzymatic products).
  • any suitable technique e.g., antibody-mediated detection of a protein, PCR analysis for the presence of a nucleic acid molecule, or visualization of enzymatic products.
  • equivalent titers are deemed to be those which are substantially the same within a given assay, generally, within about three-fold of each other.
  • the recombinant retrovirus is administered to a site such as the cerebral spinal fluid, bone marrow, joints, arterial endothelial cells, rectum, buccal/sublingual, vagina, the lymph system, to an organ selected from the group consisting of lung, liver, spleen, skin, blood and brain, or to a site selected from the group consisting of tumors and interstitial spaces.
  • the recombinant retrovirus may be administered intraocularly, intranasally, sublinually, orally. topically, intravesically, intrathecally, topically, intravenously, intraperitoneally, intracranially, intramuscularly, or subcutaneously.
  • Figure 1 is a schematic illustrat on ofp31N2R5(+).
  • Figure 2 is a schematic illustrat on ofpN2R3(-).
  • Figure 3 is a schematic illustrat on ofp31N25 (+).
  • Figure 4 is a schematic illustrati on ofpN2R3(+).
  • Figure 5 is a schematic illustrat on ofpN2R5(-).
  • Figure 6 is a schematic illustrat on ofp31N25 (+).
  • Figure 7 is a schematic illustrat on ofpTK A.
  • Figure 8 is a schematic illustrat: on of pPrTK A.
  • Figure 9 is a schematic illustrat: on of pTK-1 and pTK-3.
  • Figure 10 is a bar graph which llustrates the effect of Ganciclovir on CT26, CT26 bgal and CT26TK Neo cells.
  • Figure 1 1 is a graph which illustrates the effect of tumor volume over time in a Ganciclovir dose study of mice injected with CT26TK Neo.
  • Figure 12 is a graph illustrating the effect of Ganciclovir in CT26 versus CT26TK Neo cells.
  • Figure 13 is a graph demonstrating retention of viral activity upon reconstitution of a representative recombinant retrovirus lyophilized in a formulation buffer containing mannitol.
  • Figure 14 is a graph demonstrating retention of viral activity upon reconstitution of a representative recombinant retrovirus lyophilized in a formulation buffer containing lactose.
  • Figure 15 is a graph demonstrating retention of viral activity upon reconstitution of a representative recombinant retrovirus lyophilized in a formulation buffer containing trehalose.
  • Figures 16A-16D are representative graphs comparing stability of liquid non- lyophilized recombinant retrovirus stored at -80°C versus lyophilized formulated recombinant retrovirus stored at -20°C, using various saccharides.
  • Figure 17 is a bar graph which depicts the results of a reverse transcriptase assay on samples sliced from a gel. Slice 1 is from the lowest part of the gel, and slice 8 from the highest.
  • Figure 18 depicts a 8% to 25% gradient polyacrylamide gel.
  • Lane 1 is DA/ ⁇ gal S- 500 purified;
  • Lane 2 is DA/ ⁇ gal crude supernatant;
  • Lane 3 is HIV-IT crude supernatant;
  • lane 4 is DAC 6A3 crude supernatant;
  • Lane 5 is HBc/SA2 crude supernatant; and
  • Lane 5 is molecular weight markers.
  • Figure 19 is a schematic illustration of pDHF81 1.
  • Figures 20A-D are graphs which indicate the total quantity of lactate in low seed and high seed cultures ( Figures 20A and 20B, respectively) and the level of lactate production per day in low seed and high seed cultures ( Figures 22C and 22D, respectively).
  • Figure 21 is a bar graph which depicts the titer of the cell line 2X- ⁇ -gal under different initial seeding conditions.
  • Figure 22 Factor VIII antigen was determined in citrated blood samples drawn on the indicated days. Antigen determined by a standard curve obtained using normal pooled human plasma is plotted on the left y axis; the per cent of normal human levels is indicated on the right y axis.
  • Figure 23 Factor VIII expression in rabbits. Juvenile rabbits were injected with a total of 10 9 cfu of the factor VIII vector B-del-1 in a total of 9 injections over days 0,1, and 2. Citrated plasma samples were obtained at the indicated times and human factor VIII antigen levels determined by ELISA. Rabbits 93 and 95 (controls) received formulation buffer in lieu of vector.
  • Figure 24 Serum samples were obtained from rabbits on indicated days, and human growth hormone antigen was measured using ELISA .
  • Figure 25 Serum samples were obtained from mice on indicated days and human growth hormone antigen was determined by ELISA. Since the animals were too small on Day 14 to yield sufficient sample for the ELISA measurement, sera from test animals 1-5, test animals 6-10, and control animals 11 and 12 were respectively pooled prior to measurement. Data from subsequent days were obtained from individual animals.
  • Figure 26 Sera were obtained from adult mice on the indicated days. Human growth hormone antigen was measured by ELISA. Each point represents an individual animal. Negative controls had been injected with formulation buffer; positive controls with 1E7 cfu retroviral vector; "600 ug CsA" with both vector and cyclosporin A as described in the text.
  • Figure 27 PCR localization of vector in B-del-1 treated rabbits. Rabbits #82 and 95 shown in figure 22, herein were sacrificed on Day 51 1. DNA was extracted from indicated organs, subjected to 40 cycles of PCR, and dot blotted, and hybridized to 32-P labeled probe as described in text. Several replicate PCR reactions were run. Lanes indicated '" spike” had 5 copies of amplicon added to the template.
  • Figure 28 Quantitative PCR of beta-galactosidase vector in organs of transduced mice. Juvenile mice, transduced as described in the text, were sacrificed on Day 59. DNA was obtained from indicated organs and subjected to quantitative PCR as described.
  • Figure 29 Human growth hormone antigen in organs of transduced mice. Indicated organs were lysed and analyzed by ELISA as described in the text. Results are shown normalized to the amount of total protein recovered from the organ.
  • Figure 30 Expression of human factor VIII antigen in transduced normal dogs.
  • Juvenile dogs #47 and #55 were injected with a total of 4E9 cfu of factor VIII vector B- del-1 in a total of 9 injections over days 0,1, and 2.
  • Control dog #39 received formulation buffer on the same schedule.
  • Dog #47 was boosted with the same amount of vector on days 90, 91 , and 92.
  • cuitrated plasma was obtained and analyzed by ELISA for human factor VIII antigen. Percent human level is calculated based on a level of 200 ng/ml factor VIII in pooled in normal human plasma.
  • Figure 31 Transfer of expression of human growth hormone following adoptive transfer of splenocytes from transduced donor mice to lethally irradiated syngeneic recipients. Juvenile mice were transduced with growth hormone retroviral vector as described in the text. On day 59, Donors #1-5 were sacrificed, and splenocytes from each were injected into two lethally irradiated recipients (A and B. numbered according to donor number). Every 14 days subsequent to reconstitution, serum from the recipients was monitored for growth hormone levels.
  • Figure 32 Whole blood clotting times in hemophiliac dogs treated with factor VIII vector. Two hemophiliac dogs (#28 and #33) were injected with B-del-1 as described in the text. At indicated times, citrated whole blood samples were obtained and clotting times were determined following recalcification. Clotting times of two untreated normal littermates are included for reference.
  • Figure 33 is a schematic illustration of pKTl .
  • Figure 34 is a description of all modifications carried out on retroviral vector as shown in A), resulting in the cross-less retroviral vector shown in B).
  • the cross-less retroviral backbone cloned into a prokaryotic vector is called pBA-5.
  • Figure 35 Schematic representation of pKm201-80L.
  • Figure 36 Schematic representation of pKm201-90-H.
  • Figure 37 Schematic representation of pSVF8-t ⁇ 2.
  • Figure 38 Nucleotide and predicted amino acid sequence linking the light and heavy chains of Factor VIII in pSVF8-t ⁇ 2.
  • Figure 39 Linker sequences for pSVF8-500 and pcl/1 AG ⁇ F8-5O0.
  • Figure 40 Schematic representation of pSVF8500 and pel /l AG F8-500.
  • Figure 41 Linker sequence for pSVF8-500B.
  • Vector construct refers to a nucleic acid construct which carries, and within certain embodiments, is capable of directing the expression of a nucleic acid molecule of interest.
  • the retroviral vector must include at least one transcriptional promoter/enhancer or locus defining element(s), or other elements which control gene expression by other means such as alternate splicing, nuclear RNA export, post-translational modification of messenger, or post- transcriptional modification of protein.
  • Such vector constructs must also include a packaging signal, long terminal repeats (LTRs) or portion thereof, and positive and negative strand primer binding sites appropriate to the retrovirus used (if these are not already present in the retroviral vector).
  • LTRs long terminal repeats
  • the recombinant retroviral vector may also include a signal which directs polyadenylation, selectable markers such as Neomycin resistance, TK, hygromycin resistance, phleomycin resistance histidinol resistance, or DHFR. as well as one or more restriction sites and a translation termination sequence.
  • selectable markers such as Neomycin resistance, TK, hygromycin resistance, phleomycin resistance histidinol resistance, or DHFR.
  • restriction sites and a translation termination sequence typically include a 5' LTR, a tRNA binding site, a packaging signal, an origin of second strand DNA synthesis, and a 3' LTR or a portion thereof.
  • Retroviral gene delivery vehicle refers to a retrovirus which carries at least one gene of interest.
  • the retrovirus may also contain a selectable marker.
  • the recombinant retrovirus is capable of reverse transcribing its genetic material into DNA and inco ⁇ orating this genetic material into a host cell's DNA upon infection.
  • Fractor VIII is a nonenzymatic cofactor found in blood in an inactive precursor form. Precursor factor VIII is converted to the active cofactor, factor Villa, through limited proteolysis at specific sites by plasma proteases, notably thrombin and factor IXa. The majority of factor VIII circulates as a two-chain heterodimer most likely due to intracellular or pericellular processing of the single chain gene product. The two chains are noncovalently associated in a metal ion dependent manner.
  • the "biological activity" of factor VIII refers to a function or set of functions performed by the polypeptide or fragments thereof in a biological system or in an in vitro facsimile thereof.
  • the biological activity of factor Villa is characterized by its ability to form a membrane binding site for factors IXa and X in a conformation suitable for activation of the latter by the former. This includes standard assays of factor IX or X activation, binding to phospholipids, von Willebrand factor, or specific cell surface molecules, and susceptibility to thrombin, factor IXa, activated protein C, or other specific proteases under appropriate conditions. More particularly biologically active factor VIII corrects the coagulation defect in plasma derived from individuals afflicted with hemophilia A.
  • a "factor VIII cDNA molecule” is one encoding a full-length factor VIII polypeptide, a B domain deleted factor VIII protein, or other forms of factor VIII protein with biological activity.
  • the human full-length factor VIII coding region is 7,056 nucleotides.
  • Bioly active factor IX encompasses those forms of factor IX which are capable of correcting the coagulation defect in plasma derived from individuals afflicted with hemophilia B as measured in a standard in vitro clotting assay.
  • the present invention provides for methods of administration of recombinant gene delivery vehicles for treatment of hemophilia and a variety of other disorders by gene therapy techniques.
  • the present invention provides high titer complement resistant recombinant retroviral preparations which are suitable for administration to humans. Such preparations provide the unexpected result of providing efficatious gene therapy for a variety of diseases (and by a variety of routes) that were previously not amenable for gene therapy.
  • the present invention provides recombinant retroviruses which are capable of surviving inactivation in human serum, thereby allowing more efficient gene transfer over prolonged periods of time.
  • the therapeutic proteins of the present invention may be utilized in a wide variety of gene delivery vehicles.
  • the gene delivery vehicle may be of either viral or non-viral origin (See generally, Jolly, 1994, Cancer Gene Therapy 7:51-64; Kimura, 1994, Human Gene Therapy 5:845-852; Connelly, 1995, Human Gene Therapy 6:185-193; and Kaplitt, 1994, Nature Genetics 6:148-153).
  • Gene therapy vehicles for delivery of constructs including a coding sequence of a therapeutic of the invention can be administered either locally or systemically. These constructs can utilize viral or non-viral vector approaches in in vivo or ex vivo modality. Expression of such coding sequence can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence in vivo can be either constitutive or regulated as is described in detail below.
  • Retroviral vectors As noted above, the present invention provides recombinant retroviruses which are constructed to carry or express a selected nucleic acid molecule of interest. Briefly, numerous retroviral gene delivery vehicles may be utilized within the context of the present invention, including for example those described in EP 0,415,731 ; WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; U.S. Patent No. 5,219,740; WO 9311230; WO 9310218; Vile and Hart, 1993, Cancer Res. 55:3860-3864; Vile and Hart, 1993, Cancer Res. 55:962-967; Ram et al, 1993, Cancer Res.
  • Retroviral gene delivery vehicles of the present invention may be readily constructed from a wide variety of retroviruses, including for example, B, C, and D type retroviruses as well as spumaviruses and lentiviruses (see RNA Tumor Viruses, Second Edition, Cold Spring Harbor Laboratory, 1985).
  • Preferred retroviruses for the preparation or construction of retroviral gene delivery vehicles of the present invention include retroviruses selected from the group consisting of Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus and Rous Sarcoma Virus.
  • Murine Leukemia Viruses include 4070A and 1504A (Hartley and Rowe, 1976, J. Virol. 79:19-25), Abelson (ATCC No. VR-999), Friend ATCC No. VR-245), Graffi, Gross (ATCC No. VR- 590), Kirsten, Harvey Sarcoma Virus and Rauscher (ATCC No. VR-998), and Moloney Murine Leukemia Virus (ATCC No. VR-190).
  • retroviruses may be readily obtained from depositories or collections such as the American Type Culture Collection ("ATCC"; Rockville, Maryland), or isolated from known sources using commonly available techniques.
  • retroviral gene delivery vehicles Any of the above retroviruses may be readily utilized in order to assemble or construct retroviral gene delivery vehicles given the disclosure provided herein, and standard recombinant techniques (e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, 1989; Kunkle, 1985, PNAS 52:488).
  • portions of the retroviral gene delivery vehicles may be derived from different retroviruses.
  • retrovector LTRs may be derived from a Murine Sarcoma Virus, a tRNA binding site from a Rous Sarcoma Virus, a packaging signal from a Murine Leukemia Virus, and an origin of second strand synthesis from an Avian Leukosis Virus.
  • recombinant retroviruses may be made by introducing a vector construct as discussed above, into a cell (termed a "packaging cell") which contains those elements necessary for production of infectious recombinant retrovirus which are lacking in the vector construct.
  • a wide variety of retrovector constructs may be utilized within the present invention in order to prepare recombinant retroviruses.
  • retrovector constructs comprising a 5' LTR, a tRNA binding site, a packaging signal, one or more heterologous sequences, an origin of second strand DNA synthesis and a 3' LTR, wherein the vector construct lacks gag/pol or env coding sequences.
  • LTRs Long Terminal Repeats
  • U5, R and U3 These elements contain a variety of signals which are responsible for the biological activity of a retrovirus, including for example, promoter and enhancer elements which are located within U3. LTRs may be readily identified in the provirus due to their precise duplication at either end of the genome.
  • a 5' LTR should be understood to include a 5' promoter element and sufficient LTR sequence to allow reverse transcription and integration of the DNA form of the vector.
  • the 3' LTR should be understood to include a polyadenylation signal, and sufficient LTR sequence to allow reverse transcription and integration of the DNA form of the vector.
  • the tRNA binding site and origin of second strand DNA synthesis are also important for a retrovirus to be biologically active, and may be readily identified by one of skill in the art.
  • retroviral tRNA binds to a tRNA binding site by Watson-Crick base pairing, and is carried with the retrovirus genome into a viral particle.
  • the tRNA is then utilized as a primer for DNA synthesis by reverse transcriptase.
  • the tRNA binding site may be readily identified based upon its location just downstream from the 5' LTR.
  • the origin of second strand DNA synthesis is, as its name implies, important for the second strand DNA synthesis of a retrovirus. This region, which is also referred to as the poly-purine tract, is located just upstream of the 3' LTR.
  • certain preferred retrovector constructs which are provided herein also comprise a packaging signal, as well as one or more nucleic acid molecules (e.g., heterologous sequences), each of which is discussed in more detail below.
  • retrovector constructs which lack both gag/pol and env coding sequences.
  • the phrase "lacks gag/pol or env coding sequences" should be understood to mean that the retrovector does not contain at least 20, preferably at least 15, more preferably at least 10, and most preferably at least 8 consecutive nucleotides which are found in gag/pol or env genes, and in particular, within gag/pol or env expression cassettes that are used to construct packaging cell lines for the retrovector construct.
  • Packaging cell lines suitable for use with the above-described retrovector constructs may be readily prepared (see PCT Patent Publication No. WO 95/30763; see also
  • WO 92/05266 and utilized to create producer cell lines (also termed vector cell lines or "VCLs") for the production of recombinant vector particles.
  • producer cell lines also termed vector cell lines or "VCLs”
  • packaging cell lines are made from human (e.g., HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retroviruses that are capable of surviving inactivation in human serum.
  • packaging cell lines that produce greater than recombinant retroviral particles at titer s greater than 10 ⁇ or 10 7 cfu/ml (in crude supernatant) may readily be obtained.
  • titers are generally obtained from titer assays on HT1080 cells, which produce a three-fold lower titer than titers obtained on murine 3T3 cells.
  • Alphavirus and eucaryotic layered vector sene delivery vehicles The present invention also provides a variety of alphavirus-based vectors which can function as gene delivery vehicles.
  • Such vectors can be constructed from a wide variety of alphaviruses, including for example, Sindbis viruses vectors, Semliki Forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR923; ATCC VR-1250; ATCC VR-1249; ATCC VR-532).
  • the Sindbis virus which is the prototype member of the alphavirus genus of the togavirus family is an unsegmented genomic RNA (49S RNA) of virus of approximately 11,703 nucleotides in length.
  • This virus contains a 5' cap and a 3' poly- adenylated tail, and displays positive polarity.
  • Infectious enveloped Sindbis virus is produced by assembly of the viral nucleocapsid proteins onto the viral genomic RNA in the cytoplasm and budding through the cell membrane embedded with viral encoded glycoproteins.
  • genomic 49S RNA serves as template for synthesis of the complementary negative strand.
  • This negative strand in turn serves as template for genomic RNA and an internally initiated 26S subgenomic RNA.
  • the Sindbis viral nonstructural proteins are translated from the genomic RNA while structural proteins are translated from the subgenomic 26S RNA. All viral genes are expressed as a polyprotein and processed into individual proteins by post translational proteolytic cleavage.
  • the packaging sequence resides within the nonstructural coding region, therefore only the genomic 49S RNA is packaged into virions.
  • alphavirus vector systems may be constructed and utilized within the present invention. Representative examples of such systems include those described in U.S.Patent application Nos. 08/198,450, 08/405,627 and 08/679,640, U.S. Patent Nos. 5,091,309; 5,217,879 and 5,185440, PCT Publication Nos. WO 92/10578, WO/94/21792, WO 95/27069, WO 95/27044 and WO 95/07994. Particularly preferred alphavirus vectors for use within the present invention include those which are described within U.S. Application No. 08/198,450.
  • alphavirus vector constructs comprising a 5' sequence which is capable of initiating in vitro transcription of a alphavirus RNA, a nucleotide sequence encoding alphavirus non-structural proteins, a viral junction region which is active, modified to reduce viral transcription of the subgenomic fragment, or inactivated such that viral transcription of the subgenomic fragment is prevented, and a alphavirus RNA polymerase recognition sequence.
  • the vector constructs described above contain no alphavirus structural proteins in the vector constructs.
  • the selected heterologous sequence may be located downstream from the viral junction region; in the vector constructs having a second viral junction, the selected heterologous sequence may be located downstream from the second viral junction region, where the heterologous sequence is located downstream, the vector construct may comprise a polylinker located between the viral junction region and said heterologous sequence, and preferably the polylinker does not contain a wild-type Sindbis virus restriction endonuclease recognition sequence.
  • the gene delivery vehicles is a eukarytic layered expression systems (ELVS) (see PCT Patent Publication Nos. WO 95/07994 and WO 96/17072 for a detailed description of eukaryotic layered expression systems).
  • ELVS vectors may also be developed using other viral and non-viral nucleotide sequences (see PCT Patent Publication Nos. WO 95/07994 and WO 96/17072.
  • Gene delivery vehicles of the present invention also include parvovirus such as adenovirus associated virus (AAV) vectors.
  • AAV adenovirus associated virus
  • Representative examples of such vectors include the AAV vectors disclosed by Srivastava in WO 93/09239, Samulski et al, 1989, J Vir. 65:3822-3828; Mendelson et al, 1988, Virol. 766:154-165; Flotte et al, 1993, PNAS 90(22): 10613- 10617.
  • Particularly preferred AAV vectors comprise the two AAV inverted terminal repeats in which the native D-sequences are modified by substitution of nucleotides, such that at least 5 native nucleotides and up to 18 native nucleotides, preferably at least 10 native nucleotides up to 18 native nucleotides, most preferably 10 native nucleotides are retained and the remaining nucleotides of the D-sequence are deleted or replaced with non-native nucleotides.
  • the native D-sequences of the AAV inverted terminal repeats are sequences of 20 consecutive nucleotides in each AAV inverted terminal repeat (i.e., there is one sequence at each end) which are not involved in HP formation.
  • the non-native replacement nucleotide may be any nucleotide other than the nucleotide found in the native D-sequence in the same position.
  • Other employable exemplary AAV vectors are pWP-19, pWN-1, both of which are disclosed in Nahreini, 1993, Gene 724:257-262.
  • Another example of such an AAV vector is psub201. See Samulski, 1987, J. Virol. 67:3096.
  • Another exemplary AAV vector is the Double-D ITR vector. How to make the Double D ITR vector is disclosed in U.S. Patent No. 5,478,745.
  • Still other vectors are those disclosed in Carter, U.S. Patent No. 4,797,368 and Muzyczka, U.S. Patent No.
  • AAV vector employable in this invention is SSV9AFABTKneo, which contains the AFP enhancer and albumin promoter and directs expression predominantly in the liver. Its structure and how to make it are disclosed in Su, Human Gene Therapy 7:463-470, 1996. Additional AAV gene therapy vectors are described in U.S. Patent Nos. 5,354,678; 5,173,414; 5,139,941; and 5,252,479.
  • viral vectors systems may also be utilized as a gene delivery vehicle.
  • adenoviral vectors may be employed as a gene delivery vehicle.
  • Representative examples of such vectors include those described by, for example, Berkner, 1988, Biotechniques 6:616-627; Rosenfeld et al, 1991, Science 252:431-434; WO 93/9191; Kolls et ⁇ /., 1994, PN-4S 97(1):215-219; Kass-Eisler et al, 1993, PNAS
  • adenoviral gene therapy vectors employable in this invention include those described in the above referenced documents and in PCT Patent Publication No. WO 94/12649, WO 93/03769, WO 93/19191, WO 94/28938, WO 95/11984, WO 95/00655, WO 95/27071, WO 95/29993, WO 95/34671, WO 96/05320, WO 94/08026, WO 94/1 1506, WO 93/06223, WO 94/24299, WO 95/14102, WO 95/24297, WO 95/02697, WO 94/28152, WO 94/24299, WO 95/09241, WO 95/25807, WO 95/05835, WO 94/18922 and WO 95/09654.
  • administration of DNA linked to killed adenovirus as described in Curiel, 1992, Hum. Gene Ther. 5:147-154,
  • Additional exemplary he ⁇ es simplex virus vectors include HFEM/ICP6-LacZ disclosed in WO 95/04139 (Wistar Institute), pHSVlac described in Geller, 1988, Science 247:1667-1669, and in WO 90/09441 and WO 92/07945; HSV Us3::pgC-lacZ described in Fink. 1992, Human Gene Therapy 5: 11-19; and HSV 7134, 2 RH 105 and GAL4 described in EP 0453242 (Breakefield), and those deposited with the ATCC as accession numbers ATCC VR-977 and ATCC VR-260.
  • Gene delivery vehicles may also be generated from a wide variety of other viruses, including for example, poliovirus (Evans et al. , 1989, Nature 559:385-388; and Sabin,
  • viral carriers may be homologous, non-pathogenic(defective), replication competent virus (e.g. , Overbaugh et al, 1988, Science 259:906-910), and nevertheless induce cellular immune responses, including CTL.
  • Non-viral ⁇ ene delivery vehicles Other gene delivery vehicles and methods that may be employed such as, for example, nucleic acid expression vectors; polycationic condensed DNA linked or unlinked to killed adenovirus alone, for example see U.S. Serial No. 08/366,787, filed December 30, 1994, and Curiel, 1992, Hum Gene Ther 5:147-154; ligand linked DNA, for example see Wu, 1989, J Biol Chem 264:16985-16987; eucaryotic cell delivery vehicles cells, for example see U.S. Serial No. 08/240,030, filed May 9, 1994, and U.S. Serial No.
  • Particle mediated gene transfer may be employed, for example, see U.S. provisional application No. 60/023,867. Briefly, the sequence of interest can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then be incubated with synthetic gene transfer molecules such as polymeric DN A- binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, as described in Wu and Wu, 1987, J. Biol. Chem.
  • Naked DNA may also be employed.
  • Exemplary naked DNA introduction methods are described in WO 90/11092 and U.S. Patent No. 5,580,859. Uptake efficiency may be improved using biodegradable latex beads. DNA coated latex beads are efficiently transported into cells after endocytosis initiation by the beads. The method may be improved further by treatment of the beads to increase hydrophobicity and thereby facilitate disruption of the endosome and release of the DNA into the cytoplasm.
  • nucleic acid sequences can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then be incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, insulin, galactose, lactose, or transferrin.
  • synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, insulin, galactose, lactose, or transferrin.
  • Non- viral delivery systems include the use of liposomes to encapsulate DNA comprising the gene under the control of a variety of tissue-specific or ubiquitously-active promoters.
  • Further non- viral delivery suitable for use includes mechanical delivery systems such as the approach described in Woffendin et al, 1994, Proc. Natl. Acad. Sci. USA 97(24): 11581-1 1585.
  • the coding sequence and the product of expression of such can be delivered through deposition of photopolymerized hydrogel materials.
  • Other conventional methods for gene delivery that can be used for delivery of the coding sequence include, for example, use of hand-held gene transfer particle gun, as described in U.S. Patent No. 5,149,655; use of ionizing radiation for activating transferred gene, as described in U.S. Patent No. 5,206,152 and PCT Patent Publication No. WO 92/11033.
  • Exemplary liposome and polycationic gene delivery vehicles are those described in U.S. Patent Nos. 5,422,120 and 4,762,915, in PCT Patent Publication Nos. WO 95/13796, WO 94/23697, and WO 91/14445, in European Patent Publication No. 524,968 and in Starrier, Biochemistry, pages 236-240 (1975) W.H. Freeman, San Francisco; Shokai, Biochem Biophys Acct 600:1, 1980; Bayer, 1979, Biochem Biophys Acct 550:464; Rivet, 1987, Meth Enzyme 749: 119; Wang, 1987, Proc Natl Acad Sci 54:7851 ; Plant, ⁇ 989, Anal Biochem 176:420.
  • the gene delivery vehicles of the invention can be administered by intravenous delivery or by other methods described herein in order to induce long term in vivo expression of a variety of therapeutic proteins.
  • administration of a retroviral vector intravenously results in sustained, long-term systemic expression of therapeutic genes expressed by the retroviral vector.
  • methods for obtaining long-term systemic expression in vivo of a variety of proteins known to one of skill in the art are encompassed by the instant invention.
  • long-term in vivo systemic expression is obtained by intravenous delivery methods as is described in detail below (see Section H of this application).
  • the action of human complement on the retroviral vector is suppressed.
  • human packaging cell lines are used in order to inhibit the action of human complement on the retroviral vector particles (see PCT publication number US 91/06852, entitled “Packaging Cells”).
  • the terms "human complement resistant” or “resistance to human complement” when applied to a gene delivery vehicle such as a retroviral vector means that the vectors is at least 70% resistant, more preferably 80% resistant and most preferably at least 90% resistant to inactivation by human complement when tested in a standard serum inactivation assay as described in Example 1 1 herein.
  • high titer retroviral vector preparation refers to a retroviral vector preparation that has a titer greater than 10 ⁇ cfu/ml, more preferably greater than IO 7 cfu/ml, still more preferably greater than 10 8 cfu ml, more preferably greater than IO 9 cfu/ml, yet more preferably greater than 10 ⁇ cfu/ml, and most preferably greater than 10 ⁇ cfu/ml when tested on HT1080 cells in the assay for colony forming units on HT1080 cells as is described herein.
  • cfu refers to "colony forming units" when vectors contain a selectable marker.
  • the term refers to identifiable colonies when a phenotypically observable marker is used, such as blue colonies when the marker is beta - galactosidase.
  • cfu refers to units measured by a PCR-based titer.
  • HT1080 cells are transduced with the markerless vector and number of proviral DNA copies are measured by quantitative PCR; HT1080 cells transduced with a retroviral vector containing the neomycin resistance gene are used as a standard (see Section F of the Detailed Description, herein).
  • the terms "long term systemic expression” or “sustained systemic expression” as used herein in reference to in vivo expression of protein encoded by a retroviral vector mean measurable or biologically active expression into the bloodstream for 30 days, more CT/US97/11785
  • the expression would be for the life of the patient such that the retroviral vector would only need to be administered to the patient once, or that "booster" injections would have to be delivered to the patient only a few times during the lifetime of the patient.
  • measurable expression as used herein in reference to in vivo expression of a protein encoded by a retroviral vector means that the protein is produced in sufficient amounts so as to be detectable in biological fluids such as serum by an assay specific for the expressed protein.
  • systemic expression as used herein means that the proteins are expressed into the circulation and are thus useful for treatment of certain diseases. A variety of diseases discussed in detail below are amenable to treatment by this type of gene therapy.
  • measurement of human growth hormone can be determined by an ELISA assay specific for human growth hormone protein as described in Example 19 herein.
  • biologically active expression or “protein expression in biologically or therapeutically active amounts” when used herein in reference to in vivo expression of a protein encoded by a retroviral vector means that protein is produced in sufficient amounts so as to be detectable in a functional assay.
  • expression of factor VIII can be measured in a clotting assay as described in Example 18, herein.
  • other expressed proteins can be measured by specific assays for each particular protein that are known to those of skill in the art.
  • proteins Long-term in vivo expression of a variety of proteins can be effected by the methods of the invention, preferably by in vivo administration of high titer retroviral vectors as described herein.
  • a large variety of different proteins can be expressed for therapeutic applications in a number of different disease states.
  • Preferred proteins include, cytokines and immune system modulators, hormones, growth factors, vaccine antigens, and proteins for treatment of inherited diseases. 1. Gene Delivery Vehicles Expressing Human Factor VIII and Factor IX for
  • the B domain separates the second and third A domains of factor FVIII in the newly synthesized single-chain molecule.
  • the B domain extends from amino acids 712 to 1648 according to Wood et al, 1984, Nature 312:330-337.
  • Proteolytic activation of factor VIIII involves cleavage at specific Arg residues located at positions 372, 740, and 1689. Cleavages of plasma factor VIII by thrombin or Factor Xa at Arg 372 and Arg 1689 are essential for factor VIII to participate in coagulation. Therefore, activated factor VIII consists of a heterodimer comprising amino acids residues 1-372 (containing the Al domain) and residues 373-740 (containing the A2 domain), and residues 1690-2332
  • B domain deletion refers to a factor VIII protein in which some or all removal of some or all of the amino acids between residues 711 and 1694 have been deleted, and which still preserves a biologically active FVIII molecule.
  • a range of B domain deletions can exist depending on which amino acid residues are in the B domain is deleted and whereby the biological activity of the FVIII molecule is still preserved.
  • a specific B domain deletion called the SQ ⁇ exists which is created by fusing Ser 743 to Gin 1638 (Lind et al, 1995, EurJ. Biochem 323: 19-27, and PCT WO 91/09122) This deletes amino acid residues 744 to 1637 from the B domain creating a Ser- Glu-Asn (SQ ⁇ ) link between the A2 and A3 FVIII domains.
  • Factor VIII SQN deletion refers to this deletion and to other deletions which preserve the single S-Q-N tripeptide sequence and which result in the deletion of the amino acids between the two B- domain SQN sequences (See PCT WO 91/09122 for a description of this amino acid sequence).
  • B-domain deleted forms of factor VIII There are number of other B-domain deleted forms of factor VIII. cDNA's encoding all of these B-domain deleted factor VIII proteins can be inserted into retroviral vectors by using standard molecular biology techniques similar to those described in Examples 1, 2, 27 and 28 herein. For example cDNA molecules encoding the following B-domain factor VIII deletions can be constructed as described below: Eaton (1986) Biochemistry 25:8343 des 797-1562 deletion
  • a B domain deletion in which an IgG hinge region has been inserted can also be used.
  • a deletion of this type can be obtained from plasmid PSVF8-t ⁇ 2.
  • PSVF8-t ⁇ 2 is a plasmid, which was designed to link the heavy and light chains with a short hinge region from immunoglobulin A (see Figure 37).
  • the 5' untranslated leader and signal peptide are from the human Factor VIII :C cDNA, with the Kozak consensus sequence at the initiation codon as in pSVF8-302.
  • a description of this vector is included in Chapman et al, U.S. Patent No. 5,595,886.
  • the 3' untranslated region is the same fused Factor VIII and tPA sequence as found in pSVF8-80K.
  • the construction was completed in two steps: an oligomer with cohesive ends for EcoRI and Bell (1 17 bp) was cloned into a transfer vector, pF8GM7, the DNA sequence of the oligomer was checked by ml 3 subcloning and Sanger sequencing.
  • Plasmid pSVF8-500 encodes a factor VIII protein with amino acids 770 to 1656 of the full length Factor VIII deleted, according to the nomenclature used in Seq ID No. 45.
  • the threonine at position 1672 of the full-length factor VIII sequence of Seq ID No. 45 was also deleted. The following is a description of the construction of the vector.
  • the pSVF8-500 plasmid is a derivative of pSVF8-302 in which the regions coding for the 92K and 80K domains are fused with a small connecting ⁇ -region of 21 amino acids, retaining the natural proteolytic processing sites.
  • This plasmid was constructed in the following manner: ( 1 ) A Sall-Kpnl fragment of 1984 bp containing the region coding for the 92K protein (except for the carboxyl terminal end) and BstXI-Sall fragment of 2186 bp containing the region coding for the carboxyl end of the 80K protein with 3' end untranslated region were isolated by gel electrophoresis after digestion of pSVF8-302 with restriction enzymes. (2) A BclI-BstXI fragment of 1705 bp containing most of the region coding for the
  • pUCF812 is prepared from pF8-102 which is described in U.S. Patent No. 5,045,455.
  • pF8-102 is digested with Bam-Xhal and ligated into vector pUC12 by in vitro mutagenesis at a Bell site using the following primer: 5' ACT ACT CTT CAA TCT GAT CAA GAG GAA 3' (Seq ID No. 52).
  • a KpnI-EcoRI fragment containing the carboxyl end of the 92K protein and part of the ⁇ region (4 amino acids) was obtained by digestion of the Sail cassette from pSVF8-302 with Kpnl and EcoRI.
  • sequence in the linker shown in Figure 39 is different from wild type human Factor VIII.
  • the sequence was changed to inco ⁇ orate unique Nrul and Mlul restriction sites without changing the amino acid sequence. These sites were alsoused to construct other two additional B-domain deleted vectors which are described below.
  • pSV500B ⁇ Thr was constructed from pSVF8-500. The threonine deletion at position 1672 was maintained.
  • the synthetic linker shown in Figure 41 was used to construct pSV500B ⁇ Thr. The linker extends from a unique Nrul site at Ser(765) to a unique Mlul site at Ile(1659) in the pSVF8-500 vector. This linker was substituted for the corresponding region of pSVF8-500.
  • a third vector pSVF8-500B was constructed from pSV500B ⁇ Thr. This vector is identical to pSVF8-500B except that the codon for threonine 1672 was re-inserted using standard mutagenesis methods.
  • the relationship between, pSVF8-500B, pSVF8-500B, is further illustrated in the table below. Amino acid sequence numbers in the table were determined by reference to full-length factor VIII sequence depicted in Seq. ID No. 45.
  • the Bglll-Pfll 1.35 kb fragments of each modified cDNA listed above can be inserted into the retroviral vectors described herein using standard molecular biology procedures known to those of skill in the art and described herein.
  • cDNA clones containing the B domain-deleted factor VIII proteins can be excised and used to replace the Bglll to Pfll fragment in the retroviral vectors described in examples 27 and 28 herein.
  • a similar procedure can be used to construct other retroviral vectors , including those described in section A, above.
  • the full-length factor VIII cDNA can also be inserted into the retroviral vectors of the invention as described in Example 2 herein and in WO 96/21035 which is hereby inco ⁇ orated by reference in its entirety.
  • the full-length human factor VIII cDNA can be derived from the construct pSV7dF8-200 or pSV7dF8-300 described in Example 28, herein. This can be constructed similarly to the retroviral vector expressing the B-domain deleted factor VIII described in Example 28 where the cDNA fragment is cloned into the Bglll/Pfll linearized vector.
  • an AccI to Xbal 5.9 kb fragment can replace the 3.15 kb AccI to Xbal fragment of the modified cDNA in F8:4213.
  • the construction of the retroviral vector can then essentially follow the steps described in Example 28.
  • a variety of Factor VIII deletions, mutations, and polypeptide analogs of Factor VIII can also be introduced into the gene delivery vehicles of the invention including retroviral vectors by modifications of the procedures described herein. These analogs include, for instance, those described in PCT Patent Publication Nos. WO 97/03193, WO 97/03194, WO 97/03195, and WO 97/03191, all of which are hereby inco ⁇ orated by reference.
  • vectors expressing B domain deleted factor VIII or full-length factor VIII can be constructed using techniques known to those of skill in the art or described herein.
  • adenoviral vectors expressing B-domain deleted factor VIII molecules can be constructed as described in WO 94/29471.
  • Gene delivery vehicles derived from adenovirus are also provided, in which the factor VIII gene, or its various derivatives as described herein, is inserted into a variety of adenovirus-derived vectors, and subsequently recombinant vector particles are generated as described.
  • the adenovirus vectors of the invention also include the following vectors constructed to express factor VIII proteins.
  • the prototype recombinant adenovirus vectors were deleted in the early region one, [Ela/Elb, (El)] region, rendering them replication defective. Following insertion of a gene of interest into the deleted region, propagation of the recombinant El -deleted adenovirus vector is accomplished in 293 cells, a complementing human embryonic kidney cell line stably transformed with the Ad El region, which provide these Ad gene products in trans. Recombinant Ad vectors generated in this fashion can yield preparations with titers between 10" to 10 13 particles/ml (Reviewed in Berkner, 1988, Biotechniques 6:616-629).
  • second generation Ad vectors have sought to further "cripple" the capacity of the vector to replicate, and express viral-specific gene products, and to increase the capacity ofheterolgous genetic material.
  • Such vectors have been of three types: (l) El and E3 genes deleted [Bett, PNAS, 91 :8802-8806, 1994], (2) El and E4 genes deleted (Wang, 1995, Gene Ther. 2:775-783, and PCT Patent Publication Nos.
  • Ad vectors including the E1/E3, E1/E4, and "gutless" vectors, harboring the full-length factor VIII gene, or its B-domain derived derivatives, can be accomplished according to methods well-known to those skilled in the art.
  • the full-length factor VIII gene, or its B-domain derived derivatives can be inserted into plasmid pBHGl 1 (Bett, 1994, PNAS 97:8802-8806), and recombinant El/E3-deleted Ad vectors are generated after transfection of 293 cells and subsequent intracellular homologous recombination
  • the full-length factor VIII gene, or its B-domain derived derivatives can be first substituted into the El region of any of a variety of El -deleted Ad vectors and co-transfected with Cla I digested H5dll014, and recombinant El/E4-deleted Ad vectors are generated after transfection of 293-E4 cells [Wang Gene Ther., 2:775-783, (1995)], and subsequent intracellular homologous recombination
  • the full-length factor VIII gene, or its B-domain derived derivatives is first inserted into the ⁇ rAd plasmid " [Fisher, 1996,
  • Purification of recombinant "gutless" adeonvirus vector particles from helper virus can be accomplished, for example, by centrifugation over a cesium gradient, as a result of a buoyant density lower than that of helper virus.
  • a factor VIII heavy chain and light chain are inserted into separate vectors, as is demonstrated in Examples 29 and 30 herein.
  • Other adenoviral vectors including those described above and in co-owned U.S. serial number 60/025649 can be constructed to express factor VIII light chain and heavy chain using the techniques described in Examples 29 and 30.
  • Heavy and light chain constructs for inclusion into the AAV vectors of the invention can be prepared for instance as described in Chapman et al, U.S. Patent No. 5,595,886.
  • Hemophilia B can also be treated with systemically administered factor IX- expressing gene delivery vehicles including retroviral vectors.
  • Human factor IX deficiency (Christmas disease or Hemophilia B) affects primarily males because it is transmitted as sex-linked recessive trait. It affects about 2000 people in the US.
  • the human factor gene codes a 416 amino acids of mature protein.
  • the human factor IX cDNA can be obtained for instance by constructing plasmid pHflXl, as described by Kurachi and Davie, 1982, PNAS 79(21):6461-6464.
  • the cDNA sequence can be excised as a PstI fragment of about 1.5 kb, blunt ended using T4 DNA polymerase.
  • the factor cDNA fragment can be readily inserted, for example into a Srfl site introduced into a retroviral vector.
  • the cDNA fragment can be inserted into the Srfl site of a linearized pMBA retroviral vector using the procedure described in Examples 27 and 28 herein.
  • the resulting retroviral vector: pMB-F9 will be produced from HAII cells and its expression will be determined using HT1080 fibroblasts as target cells.
  • the full-length factor IX cDNA fragment can be introduced into other gene delivery vehicles described herein, such as adenoviral vectors and AAV vectors.
  • vectors expressing factor IX can be produced, for instance, as described in.
  • adenoviral vectors expressing B-domain deleted factor VIII molecules can be constructed as described in WO 94/29471.
  • Adenoviral vectors expressing factor IX protein can be produced by methods similar to those described above for factor VIII protein.
  • a factor IX cDNA can be inserted into vectors using, for instance, procedures similar to those described in Examples 29 and 30 herein for factor VIII heavy and light chains.
  • Other adenoviral vectors including those described above and in co- owned U.S. provisional application No. 60/025,649 can be constructed to express factor VIII light chain and heavy chain using the techniques similar to those of Examples 29 and 30.
  • Gene delivery vehicles expressing proteins for treatment of other hereditary disorders There are a number of proteins useful for treatment of hereditary disorders that can be expressed in vivo by the methods of invention.
  • Many genetic diseases caused by inheritance of defective genes result in the failure to produce normal gene products, for example, thalassemia, phenylketonuria, Lesch-Nyhan syndrome, severe combined immunodeficiency (SCID), hemophilia, A and B, cystic fibrosis, Duchenne's Muscular Dystrophy, inherited emphysema and familial hypercholesterole ia (Mulligan et al, 1993.
  • a number of genetic diseases have been selected for treatment with gene therapy, including adenine deaminase deficiency, cystic fibrosis, ⁇ j-antitrypsin deficiency, Gaucher's syndrome, as well as non-genetic diseases.
  • a retroviral vector can be used to treat Gaucher disease.
  • Gaucher disease is a genetic disorder that is characterized by the deficiency of the enzyme glucocerebrosidase. This enzyme deficiency leads to the accumulation of glucocerebroside in the lysosomes of all cells in the body.
  • Factor VIII or Factor IX is useful for treatment of blood clotting disorders, such as hemophilia.
  • Different forms of Factor VIII such as the B domain deleted Factor VIII construct described in Example 2 herein can be used to produce retroviral vectors expressing Factor VIII for use in the methods of the invention.
  • proteins which can be expressed in the retroviral vectors of the invention and which are useful for treatment of hereditary diseases. These include lactase for treatment of hereditary lactose intolerance, AD for treatment of ADA deficiency, and alpha- 1 antitypsin for treatment of alpha- 1 antitrypsin deficiency. See F.D.
  • familial hypercholesterolemia is a disease characterized clinically by a lifelong elevation of low density lipoprotein (LDL), the major cholesterol- transport lipoprotein in human plasma; Pathologically by the deposition of LDL-derived cholesterol in tendons, skin and arteries leading to premnature coronary heart disease; and genetically by autosomal dominant inherited trait. Hetrozygotes number about 1 in 500 persons worldwide. Their cells are able to bind cholesterol at about half the rate of normal cells. Their plasma cholesterol levels show two fold elevation starting at birth.
  • LDL low density lipoprotein
  • Homozygotes number 1 in 1 million persons. They have severe cholesterolemia with death occurring usually before age 20. The disease (Arteriosclerosis) depends on geography. It affects 15.5 per 100,000 individuals in the U.S. (20,000 total) and 3.3 per 100,000 individuals in Japan.
  • Gene delivery vehicles expressing the LDL receptor for treatment of disorders manifesting with elevated serum LDL can be constructed by techniques known to those of skill in the art.
  • An example of a retroviral vector expressing LDS receptor is shown in example 32 herein.
  • Other retroviral vectors such as those described herein can readily be constructed using PCR amplication and restriction enzyme treatment methods similar to those described in Example 31.
  • retroviral vectors expressing alpha- 1 antitypsin for treatment of alpha- 1 antitrypsin deficiency can be constructed by variations of the procedure demonstrated in Example 32, herein.
  • Sequences which encode the above-described nucleic acid molecules may be obtained from a variety of sources.
  • plasmids which contain sequences that encode altered cellular products may be obtained from a depository such as the American Type Culture Collection (ATCC, Rockville, Maryland), or from commercial sources such as Advanced Biotechnologies (Columbia, Maryland).
  • Plasmids containing cyokine genes or immunodulatory genes can be digested with appropriate restriction enzymes, and DNA fragments containing the particular gene of interest can be inserted into the gene delivery vector using standard molecular biology techniques. (See Sambrook et al, Molecular Cloning: A Laboratory Manual (2 nd Edition, Vols. 1-3, Cold Spring Harbor Laboratory (1989) or Ausbel, et al. ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York (1987).
  • cDNA sequences for use with the present invention may be obtained from cells which express or contain the sequences. Briefly, within one embodiment mRNA from a cell which expresses the gene of interest is reverse transcribed with reverse transcriptase using oligo dT or random primers. The single-stranded cDNA may then be amplified by PCR (see U.S. Patent Nos. 4,683,202, 4,683,195 and 4,800,159. See also PCR Technology: Principles and Applications for DNA Amplification, Erlich (ed.), Stockton Press, 1989) utilizing oligonucleotide primers complementary to sequences on either side of desired sequences.
  • a double-stranded DNA is denatured by heating in the presence of heat stable Taq polymerase, sequence-specific DNA primers, ATP, CTP, GTP and TTP. Double-stranded DNA is produced when synthesis is complete. This cycle may be repeated many times, resulting in a factorial amplification of the desired DNA.
  • Nucleic acid molecules which are carried and/or expressed by the recombinant retroviruses described herein may also be synthesized, for example, on an Applied Biosystems Inc. DNA synthesizer (e.g., APB DNA synthesizer model 392 (Foster City, California).
  • the recombinant retrovirus directs the expression of a substance capable of activating an otherwise inactive precursor into an active inhibitor of a pathogenic agent, or a conditional toxic palliative, which are palliatives that are toxic for the cell expressing the pathogenic condition.
  • a wide variety of inactive precursors may be converted into active inhibitors of a pathogenic agent.
  • antiviral nucleoside analogues such as AZT or ddl are metabolized by cellular mechanisms to the nucleotide triphosphate form in order to specifically inhibit retroviral reverse transcriptase, and thus viral replication (Furmam et al, 1986, Proc. Natl. Acad.
  • Retroviruses which direct the expression of a gene product (e.g., a protein) such as He ⁇ es Simplex Virus Thymidine Kinase (HSVTK) or Varicella Zoster Virus Thymidine Kinase (VZVTK) which assists in metabolizing antiviral nucleoside analogues to their active form are therefore useful in activating nucleoside analogue precursors (e.g., AZT or ddC) into their active form.
  • nucleoside analogue precursors e.g., AZT or ddC
  • nucleoside analogues whose nucleotide triphosphate forms show selectivity for retroviral reverse transcriptase but, as a result of the substrate specificity of cellular nucleoside and nucleotide kinases are not phosphorylated, will be made more efficacious.
  • the HSVTK gene may be expressed under the control of a constitutive macrophage or T-cell-specific promoter, and introduced into macrophage or T-cells.
  • Constitutive expression of HSVTK results in more effective metabolism of nucleotide analogues such as AZT or ddl to their biologically active nucleotide triphosphate form, and thereby provides greater efficacy, delivery of lower doses, less generalized toxicity, and higher potency against productive infection.
  • nucleoside analogues whose nucleotide triphosphate forms show selectivity for retroviral reverse transcriptase but, as a result of the substrate specificity of cellular nucleoside and nucleotide kinases are not phosphorylated, may also be utilized within the context of the present invention.
  • gene delivery vehicles such as recombinant retroviruses are provided which direct the expression of a substance that activates another compound with little or no cytotoxicity into a toxic product in the presence of a pathogenic agent, thereby effecting localized therapy to the pathogenic agent.
  • expression of the gene product from the recombinant retrovirus is limited to situations wherein an entity associated with the pathogenic agent, such as an intracellular signal identifying the pathogenic state, is present, thereby avoiding destruction of nonpathogenic cells.
  • This cell-type specificity may also be conferred at the level of infection, by targeting the recombinant retrovirus to cells having or being susceptible to the pathogenic condition.
  • gene delivery vehicles such as recombinant retroviruses are provided which direct the expression of a gene product(s) that activates a compound with little or no cytotoxicity into a toxic product.
  • gene products which either directly or indirectly activate a compound with little or no cytotoxicity into a toxic product may be utilized within the context of the present invention.
  • Representative examples of such gene products include HSVTK and VZVTK which selectively monophosphorylate certain purine arabinosides and substituted pyrimidine compounds, converting them to cytotoxic or cytostatic metabolites.
  • the gene delivery vehicles of the invention can contain a liver specific promoter to maximize the potential for liver specific expression of the exogenous DNA sequence contained in the vectors.
  • liver specific promoters include the hepatitis B X-gene promoter and the hepatitis B core protein promoter. These liver specific promoters are preferably employed with their respective enhancers.
  • the enhancer element can be linked at either the 5' or the 3' end of the nucleic acid encoding the therapeutic molecule.
  • the hepatitis B X gene promoter and its enhancer can be obtained from the viral genome as a 332 base pair EcoRV-NcoI DNA fragment employing the methods described in Twu, 1987, J.
  • the hepatitis B core protein promoter can be obtained from the viral genome as a 584 base pair BamHI-Bglll DNA fragment employing the methods described in Gerlach, 1992, Virol 759:59-66. It may be necessary to remove the negative regulatory sequence in the BamHI-Bglll fragment prior to inserting it.
  • Other liver specific promoters include the AFP (alpha fetal protein) gene promoter and the albumin gene promoter, as disclosed in EP Patent Publication 0415,731, the -1 antitrypsin gene promoter, as disclosed in Rettenger, 1994, Proc. Natl. Acad. Sci.
  • liver specific promoters in retroviral vectors.
  • a liver specific promoter as described in this section can be introduced into the vector to operably linked to gene of interest (for example factor VIII) in order to induce more liver specific expression of the protein.
  • the promoter is operably linked to the nucleic acid encoding the therapeutic molecule upstream from the latter and between the AAV vector sequences (for example between the inverted terminal repeats in psub201 or downstream of the Double D ITR sequence).
  • Example 31 Examples of the construction of retroviral vectors expressing ⁇ interferon under the control of liver-specific promoters is shown in Example 31 herein. Similar techniques can be used to construct retroviral vectors expressing other proteins under control of liver- specific promoters, for instance, by using a three-way ligation procedure analogous to the procedure described in Example 31.
  • D. Use of gene delivery vectors co-expressing therapeutic protein and a prodrug-converting enzyme The gene delivery vehicles and the therapeutic methods of delivery are useful for the long term expression of therapeutic proteins for treatment of a variety of disorders described herein. Particularly, because of the long term treatment made possible by the present invention, it can be desirable for the gene delivery vehicles of the invention to co-express a prodrug converting enzyme which converts a non-cytotoxic compound into its cytotoxic counte ⁇ art.
  • the protype of such enzymes is he ⁇ es simplex thymidine kinase (HSVTK) which converts the prodrug gancyclovir into a toxic compound.
  • HSVTK or other prodrug converting enzymes into gene delivery vehicles, particularly retroviral vectors are well known to those of skill in the art. (See eg. WO 91/02805, WO 90/07936, and WO 95/14091). Procedures for preparing gene delivery vehicles, particularly retroviral vectors which co-express a therapeutic protein and HSVTK or other prodrug converting enzymes are described herein and are also described in number of publications (See, eg. WO 93/10218.) These procedures can readily be used by those of skill in the art to introduce the HSVTK gene or a gene encoding another prodrug converting enzyme into the gene delivery vehicles of the invention. In addition, the prodrug converting enzyme can be introduced into a second vector particle which is then co- administered with the gene delivery vehicle expressing the therapeutic protein. An example of this approach is described in WO 96/21015.
  • Retroviral vectors are known to preferentially infect dividing cells. (See Miller et al, 1990, Molecular Cell Biol. 70:4239.) There are variety of techniques that may be used to increase the number of dividing cells in target tissues and thereby enhance the efficiency of target cell infection by the retroviral vectors of the invention.
  • growth factors may be used to stimulate target tissues to enter the portion of the cell cycle in which retroviral vector integration can take place.
  • growth factors and their target tissues include, but are not limited to, the following:
  • Protein S and Gas6 acting on nervous system cells and smooth muscle cells thrombin acting on smooth muscle cells, gastrointestinal epithelium, liver fat storage cells, dental pulp cells, fibroblasts, endothelial cells, mesangial cells, and astrocytes; coagulation Factor Xa acting on smooth muscle cells; nerve growth factor acting on nervous system cells; CSF-1 acting on placenta and endometrium; IGF-1 acting on kidney, bone, skin, adipose tissue, airway smooth muscle cells, gastrointestinal epithelium, neural tissue, muscle, and follicular cells; insulin acting on gastrointestinal epithelium, skin, and adipose tissue; KGF acting on urothelium, mammary epithelium, skin, liver, and gastrointestinal epithelium; TGF acting on gastrointestinal epithelium, dental pulp, neural tissue, fibroblasts.
  • connective tissue inner ear sensory epithelium, colon, bone, pneumocyte type II cells, cornea, and smooth muscle cells
  • endothelin acting on kidney, smooth muscle cells, melanocytes, cardiac muscle, and astrocytes
  • PDGF acting on kidney, airway smooth muscle cells, gastrointestinal epithelium, neural tissue, and connective tissue
  • EGF acting on kidney, skin, neural tissue, inner ear sensory epithelium, connective tissue, fibroblasts, endometrium, liver, and intestine
  • PSA acting on prostate, breast, lung, colon, ovary, liver, and kidney
  • Growth factors can also be used in combination, particularly but not limited to mixtures consisting of one or several of EGF, IGF, PDGF, FGF, or KGF.
  • multiple growth factors known to stimulate cell division of a particular target tissue can used in comination to increase the proportion of dividing cells in the tissue.
  • the actions of the above and other growth factors can also be potentiated with substances including but not limited to dextran sulfate, heparin, and other sulfated glycosaminoglycans, FBP, leukotrienes, prostaglandins, oleic acid, HGF activators, androgens, estrogens, ethanol, PF4, and TGF beta antagonists.
  • Treatment with growth factors or the other substances described above can occur by administering the substances in vivo or can also be used in other treatment modalities such as ex vivo treatment.
  • the polypeptide growth factors described herein can be administered in a variety of forms including full-length growth factors, growth factor fragments, truncated growth factors, growth factor fusion proteins and growth factor analogues.
  • Growth factor fusion proteins include fusion proteins with the full-length growth factor, truncated growth factor, growth factor analogue, and growth factor fragment.
  • growth factor fragment refers to any growth factor polypeptide that contains less than a full-length sequence, and which retains sufficient biological activity to be used in the methods of the invention.
  • growth factor analogue refers to growth factors, truncated growth factors and growth factor fragments with amino acid substitutions, deletions, additions, and modifications; and retaining the biological activity of the growth factor.
  • growth factor analogue includes splice variants, truncations, variants, alleles and derivatives of the mature protein. Analogues possess one or more of the bioactivities of the full length protein. Thus, polypeptides that are identical or contain at least 60%, preferably 70%, more preferably 80%), and most preferably 90%> amino acid sequence homology to the amino acid sequence of the mature protein wherever derived, from human or nonhuman sources, are included within this definition.
  • Growth factor analogues include variants of the growth factors described herein. Growth factor variants contain amino acid substitutions, deletions, or insertions. The amino acid substitutions can be conservative amino acid substitutions or substitutions to eliminate non-essential amino acid residues such as to alter a glycosylation site, a phosphorylation site, an acetylation site, or to minimize misfolding by substitution or deletion of one or more cysteine residues that are not necessary for function.
  • Conservative amino acid substitutions are those that preserve the general charge, hydrophobicity/hydrophilicity and/or steric bulk of the amino acid substituted, for example, substitutions between the members of the following groups are conservative substitutions: Gly/Ala, Val/Ile/Leu, Asp/Glu, Lys/ Arg, Asn/Gln, Ser/Thr/Cys and Phe/T ⁇ /Tyr.
  • the analogs herein further include peptides having one or more peptide mimics, also known as peptoids, that possess the bioactivity of the protein.
  • polypeptides containing one or more analog amino acid including, for example, unnatural amino acids, etc.
  • polypeptides with substituted linkages as well as other modifications known in the art, both naturally occurring and non-naturally occurring.
  • polypeptide also does not exclude post- expression modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations and the like.
  • glycosylations, acetylations, phosphorylations and the like are known to those of skill in the art and can be used in the methods of the invention.
  • a variety of PDGF polypeptides are known in the art.
  • nucleic acid vectors encoding naturally occurring PDGF and the production of recombinant PDGF are described in U.S. Patent No. 5,219,759, issued June 15, 1993, entitled "Recombinant DNA encoding PDGF A-chain Polypeptide Expression Vectors".
  • PDGF analogues are known in the art and can be used in the methods described herein.
  • U.S. Patent No. 5,128,321, issued July 7, 1992, entitled “PDGF Analogues and Methods of Use” and U.S. Patent No. 4,849,407, issued July 18, 1989, and entitled "Biologically Active Mosaic Proteins”.
  • IGF- 1 and IGF-2 growth factor polypeptides are also well known the art.
  • recombinant human as well as specific DNA sequences and expression vectors for the production of human IGF-1 are described in European Patent No. 0123228B1, grant published on September 19, 1993, entitled "Hybrid DNA Synthesis of Mature Insulin-like Growth Factors".
  • European Patent No. 0123228B1 grant published on September 19, 1993, entitled "Hybrid DNA Synthesis of Mature Insulin-like Growth Factors”.
  • a variety of other forms of IGF- 1 polypeptides are known in the art and can readily be produced for use in the methods of the invention. See eg. U.S. Patent No. 5,019,500, entitled "59 Valine Insulin-Like Growth Factor I and Process for Production Thereof; PCT Patent Publication No.
  • FGF growth factors encompass two families of growth factors found in a variety of different tissues and which are mitogenic factors for a number of different tissues. These two families of polypeptides have been termed acidic FGF and basic FGF.
  • acidic FGF and basic FGF A large variety of different FGF polypeptides, analogues and fragments are known in the art an can be used in the methods of the present invention. See, eg. U. S. Patent No. 5,464,774, issued Nov. 7, 1995, entitled “Bovine Basic Fibroblast Growth Factor”; U.S. Patent No. 5,155,214, entitled Basic Fibroblast Growth Factor, and U. S. Patent No. 4,994,559, entitled "Human Fibroblast Growth Factor".
  • keratinocyte growth factor that are useful in the methods of the invention are also known to those of skill in the art. See, for example, a preferred truncated form of KGF which is described in PCT Patent Publication No. 95/10434, published January 12, 1995, and entitled "A Truncated Form of KGF Having Increased Biological Activity".
  • EGF polypeptides are known in the art. See eg. PCT publication No. WO 90/08771 and U.S. Patent No. 5, 096,825, issued March 17, 1992, entitled Gene for Human Epidermal Growth Factor and Synthesis of Expression Thereof.
  • the growth factor polypeptides, fragments and analogues used in the instant invention can be produced in a variety of ways, including isolation of PDGF polypeptide from naturally occurring sources, polypeptide chain synthesis by peptide synthesis methods and production of recombinant proteins. These methods are well known to those of skill in the art and are further described herein.
  • recombinant PDGF polypeptides and analogues see U.S. Patent No. 5,045,633, issued Sept. 3, 1991 , entitled "Expression of Biologically Active PDGF Analogues in Eucaryotic Cells"; U.S. Patent No. 4,769,328, issued Sept.
  • Liver is an attractive organ for gene therapy because it is easily accessible via the circulation and is the source of a variety of proteins involved in genetic disorders, including factor VIII.
  • Gene therapy targeting the liver using the retroviral vectors of the invention can be performed with or without pretreatment to include benign hype ⁇ lasia of the liver.
  • Pretreatment to induce benign liver hype ⁇ lasia can be effected, for example, by treatment with hepatocyte growth factor (HGF) and/or transforming growth factor alpha.
  • HGF hepatocyte growth factor
  • transforming growth factor alpha See eg. Liu et al., 1994, Hepatology 79:1521.
  • a variety of different forms of HGF useful to induce liver cell proliferation are known in the art. See eg. European Patent Publication No.
  • HGF Human Leukocyte Derived Heaptacyte Growth Factor- with DNA encoding it, Recombinant Expression Vectors, and Transformant Cells Expressing it.”
  • HGF can also be produced and administered to induce liver proliferation in vivo as is described in Joplin et al, 1992, J. Clin. Invest. 90: 1284.
  • Liver cells can also be stimulated by administration of agents that mediate or potentiate the activation of endogenous HGF.
  • HGF is produced as a single chain protein that is inactive as a growth factor. Single chain HGF is subsequently cleaved into a two- chain form which is the biologically active growth factor.
  • Enzymes which are shown to convert single-chain HGF to its biologically active form are useful for inducing liver cell proliferation. Therefore, these enzymes can be administered either alone or in combination with exogenous HGF to enhance liver proliferation. Examples of such enzymes are coagulation factor Xlla (see Shiomura et al, 1995, European J.
  • HGF activator see Shiomura et al, 1992, Cytotechnology 8:219
  • HGF converting enzyme Mizuno et al, 1994, Biochem. Biophys. Res. Comm. 795:1 161
  • urokinase and tissue plasminogen activators Mars et al, 1993, Am J. Pathol. 745:949.
  • urokinase can be co-administered with HGF.
  • HGF and urokinase could be either be co-formulated or mixed immediately prior to injection.
  • HGF and urokinase were co-formulated, they could, for example, be stored at low pH in order to minimize the activity of urokinase.
  • the effects of either HGF or TGFalpha on liver stimulation have also been shown to be enhanced by prior administration of collagenase (Liu, 1994, Hepatology 79:1521).
  • Agents that stimulate release of collagenase-like materials from monocytic cell types, such as LPS or endotoxin-like substances may also be used.
  • Nonenzymatic adjuncts and costimulants of growth factors may also be used, including estrogens with HGF (Ni, 1994, Hepatology 79:183), heparin or other sulfated glycosaminoglycans with HGF (Yamakazi, 1996, Cytokine 5:178; Matsumoto, 1996, Biochem Biophys Res Commun 227:455), spermine or spermidine with HGF (Higaki, 1994, Gastroenterology 706:1024), nicotinamide with EGF (Wu (1994) Cancer Res 54:5964), or choline (Tessitore, 1997, Biochem J. 322: 151).
  • the phosphofetuin/phospho-alpha-2-HS glycoprotein pathway downregulates liver regeneration.
  • Inhibitors of this pathway may be used to upregulate liver proliferation.
  • Such inhibitors include IL6, ILIalpha, or anti-fetuin antibodies (Ohnishi, 1997, Ewr. J. Biochem. 245:753). Combinations of these agents can also be applied along with the various growth factors described above.
  • liver cell growth can also be stimulated by nutritional minipulation. A variety of different nutritional routine can be used. For instance, a period of protein deprivation followed by consumption of a high protein meal can be used to stimulate DNA synthesis and cell division. (See Mead et al, 1990, Cancer Res.
  • cyclooxigenase inhibitors such as indomethacin
  • indomethacin is known to increase DNA synthesis and cell clearance in duodenal and jejunal mucusa (see Uribe et al, 1992, Dig. Dis. Sci. 57:403-408).
  • pretreatment with indomethacin or other non-steroidal anti inflammatory drugs can be used to increase cell proliferation in the gastric mucosa.
  • prostaglandins in particular, prostraglandin ⁇ 2 can be used after introduction of the gene therapy vehicle in order to increase the retention of the transduced intestinal cells.
  • Omeprazole can be obtained and administered as described in Kakei et al, 1995, Biochem. Biophys. Res. Comm. 274:861-867.
  • An additional class of drugs that is known to induce liver proliferation is the peroxisome proliferators. These are a set of structurally diverse drugs that cause an increase in the number of peroxisomes present in hepatocytes, but which also lead to hepatocyte mitosis mimicking regeneration through unknown mechanism(s).
  • hypolipide ic drugs particularly clofibrate (Reddy and Lalwai, 1983, CRC Crit Rev Toxicol 72:1-58), and act through activation of members of the superfamily of nuclear steroid receptors known as PPAR ( peroxisome proliferator-activated receptors) and subsequent increased transcription of peroxisomal beta-oxidative enzymes (Isseman and Green, 1990, Nature 547:645-650), and possibly through down-regulation of the endoplasmic reticulum protein, BiP ( Motojima & Goto, 1992, FEBS Lett 505:207-210).
  • PPAR peroxisome proliferator-activated receptors
  • BiP Motojima & Goto, 1992, FEBS Lett 505:207-210
  • clofibrate Sigma Chemical Compan, St. Louis, MO, USA
  • WY 16463 Chem Syn, Lenexa, KS, USA
  • DEHP Aldrich, Milwaukee WI, USA
  • DHEA DHEA
  • PFDA Aldrich
  • PFOA Aldrich
  • fenofibrate Labeled by PFOA
  • gemfibrozil Lopid, Warner-Lambert , Ann Arbor MI
  • clofibric acid Sigma
  • bezafibrate Sigma
  • methyl clofenapate tibric acid
  • BR 931 4-chloro-6(2,3-xylidino)2- pyrimidinylthio-(N-beta-hydroxy-ethyl)acetamide
  • DEHA di(2ethylhexyl)adipate
  • nafenopin clinofibrate.
  • nonproteinaceous substances known to stimulate liver proliferation include 9-cis-retinoic acid (Ohmura, 1996, Life Sciences 55:PL21 1), the glycolipid, hepatopoietin B, (Michalopoulos, 1990, FASEB J 4: 176), cyclosporine A (Masuhara, 1993, Carcinogenesis 74:1579), phosphatidylethanolamine N- methyltransferase inhibitors such as 3-deazaadenosine or other methylation inhibitors (Chiang, 1979, Biochem Pharmacol 25:1897), nonsteroidal antiinflammatory drugs that activate PPAR such as flufenamic acid, fenoprofen, and ibuprofen (Lehmann, 1997, J.
  • the present invention provides high titer recombinant retroviral preparation suitable for administration to humans.
  • cell culture methods as described below are provided in order to enable the production of high titer recombinant retroviruses. Briefly, a wide variety of methods may be utilized, including for example, the use of fermenters or bioreactors, roller bottles, cell hotels or cell factories, and hollow fiber culture.
  • cells are preferably grown on microcarriers (i.e., Cytodex 1 or Cytodex 2; Pharmacia, Piscataway, N.J. at concentrations ranging from 3 to 15 g/L microcarrier.
  • suitable media, and growth conditions are described by way of a representative illustration in Example 17.
  • suitable conditions include those described above for bioreactors, with the exception that microcarrier beads are not utilized.
  • cells are grown in 850 cm 2 roller bottles ("FALCON" Corning, Corning New York) containing DMEM media, along with 15%-20% FBS.
  • the bottles are sealed to avoid contamination, although "open” bottles may also be utilized under appropriate conditions (e.g., 5% CO2).
  • the roller bottles are incubated at a temperature of 37°C, with a rotation speed of 0.5 ⁇ m/minute.
  • Cell factories may also be utilized for the large scale cell culture and production of recombinant retroviruses.
  • cell factories also termed “cell hotels” typically contain 2, 10, or 40 trays, are molded from virgin polystyrene, treated to provide a Nuclon D surface, and assembled by sonic welding one to another. Generally, these factories have two port tubes which allow access to the chambers for adding reagents or removing culture fluid.
  • a 10-layer factory provides 6000cm 2 of surface area for growing cells, roughly the equivalent of 27 T-225 flasks.
  • Cell factories are available from a variety of manufacturers, including for example Nunc. Most cell types are capable of producing high titer vector into the media for 3-6 days, allowing for multiple harvests.
  • Each cell type is tested to determine the optimal harvest time after seeding the culture and optimal number of harvest days.
  • Cells are typically initially grown in DMEM supplemented with 2-20% FBS in roller bottles until the required number of cells for seeding a cell factory is obtained. Cells are then seeded into the factories and 2 liters of culture supernatant containing vector is harvested from each day for four days. Fresh DMEM/FBS is used to replenish the cultures.
  • hollow fiber culture methods are provided for the production of recombinant retroviruses.
  • high titer retroviral production using hollow fiber cultures is based on increasing viral concentration as the cells are being cultured to a high density in a reduced volume of media.
  • Cells are fed nutrients and waste products are diluted using a larger volume of fresh media which circulates through the lumen of numerous capillary fibers.
  • the cells are cultured on the exterior spaces of the capillary fibers in a bioreactor chamber where cell waste products are exchanged for nutrients by diffusion through 30,000 Dalton pores in the capillary fibers.
  • Retroviruses which are produced from the cell lines are too large to pass through the 30,000 Dalton pore membrane, and thus concentrate in the hollow fiber bioreactor along side of the cells.
  • the volume of media being cultured on the cell side is approximately 10 to 100 fold lower then volumes required for equivalent cell densities cultured in tissue culture dishes or flasks. This decrease fold in volume inversely correlates with the fold induction of titer when hollow fiber retroviral titers are compared to tissue culture dishes or flasks.
  • This 10- 100 fold induction in titer is seen when an individual retroviral producer cell line is amiable to hollow fiber growth conditions. To achieve maximum cell density, the individual cells must be able to grow in very close proximity and on top of each other.
  • the harvesting procedure is now performed using a batch peristaltic pump drive to deliver precise volumes of fresh media to replace equal volumes of harvest material which is then delivered through thin line tubing into a collection bottle stored at 4°C.
  • the pump batch sequence is activated by a timer which can be set at specific time points and the pump can be adjusted to harvest any set volume of harvest material, twenty-four hours a day.
  • the collected supernatant can then be frozen, pooled with earlier harvests, or processed as described elsewhere.
  • This collection procedure can be used for any hollow fiber system including the Cellco (Rockville, Maryland), Unisyn (Tustin, CA), or Cellex (Coon Rapids, MN) systems including ceramic matrix high density culture systems.
  • the present invention provides methods for concentrating and purifying recombinant retroviruses, in order to increase the purity of therapeutic preparation, as well as to increase the titer of recombinant retrovirus that may be given.
  • a wide variety of methods may be utilized for increasing viral concentration and purity, including for example, precipitation of recombinant retroviruses with ammonium sulfate, polyethylene glycol ("PEG”) concentration, concentration by centrifugation (either with or without gradients such as PERCOLL, or "cushions” such as sucrose, use of concentration filters (e.g., Amicon filtration), and 2-phase separations.
  • concentration filters e.g., Amicon filtration
  • ammonium sulfate is added slowly to an appropriate concentration, followed by centrifugation and removal of the ammonium sulfate either by dialysis or by separation on a hydrophobic column.
  • concentration of recombinant retroviruses other proteinaceous debris may also be concentrated.
  • recombinant retroviruses may be concentrated from culture medium with PEG (Green et al, 1970, PNAS 67:385-393; Syrewicz et al, 1972, Appl. Micro. 24:488-494).
  • PEG Green et al, 1970, PNAS 67:385-393; Syrewicz et al, 1972, Appl. Micro. 24:488-494.
  • recombinant retroviruses may be concentrated by centrifugation, and more particularly, low speed centrifugation.
  • low speed centrifugation allows concentration of recombinant retroviruses, while avoiding the difficulties associated with pelleting that accompanies high speed centrifugation (e.g., virus destruction or inactivation).
  • high speed centrifugation e.g., virus destruction or inactivation
  • Particularly preferred methods for concentrating viruses by low-speed centrifugation are described below in more detail in Example 15.
  • recombinant retroviruses may be concentrated by an aqueous two-phase separation method.
  • polymeric aqueous two- phase systems may be prepared by dissolving two different non-compatible polymers in water.
  • Many pairs of water-soluble polymers may be utilized in the construction of such two-phase systems, including for example polyethylene glycol (“PEG”) or methylcellulose, and dextran or dextran sulfate (see Walter and Johansson, 1986, Anal. Biochem. 755:215- 242; Albertsson, "Partition of Cell Particles and Macromolecules” Wiley, New York, 1960).
  • an aqueous two-phase system may be established suitable for purifying recombinant retroviruses. Utilizing such procedures, approximately 1.4 liters of crude research grade supernatant may be reduced to a 10 mL volume, while recovering approximately 50% of the total starting retrovirus.
  • recombinant retroviruses may be prepared either from roller bottles, cell factories, or bioreactors prior to concentration.
  • daily harvests of recombinant retroviruses from producer cells is preferred, followed by addition of fresh media.
  • Removed media containing the recombinant retrovirus may be frozen at -70°C, or more preferably, stored at 2°C to 8°C in large pooled batches prior to processing.
  • the recombinant retrovirus pool is first clarified through a 0.8 um filter (1.2 um glass fiber pre-filter, 0.8 um cellulose acetate) connected in series with a 0.65 um filter (Sartorious).
  • This filter arrangement provides approximately 2 square feet of filter, and allows processing of about 15-20 liters of pooled material before clogging.
  • 0.65um cartridge (2 sq. ft.) normally suffices for volumes up to 40 liters.
  • a 5 sq. ft. filter may be required.
  • the filter is rinsed with buffer (150 mM NaCl, 25 mM
  • recombinant retroviruses are concentrated by tangential flow ultrafiltration utilizing Filtron units and Sigma Screen cassettes with a 300,000 mw cut off.
  • bioreactor material containing 12% to 16% FBS
  • 4 to 5 liters of material may be concentrated per cassette.
  • 5-6 liters of material may be concentrated per cassette.
  • cell factories containing 10% FBS 8 to 9 liters of material may be concentrated per cassette.
  • Utilizing a pressure differential of 2 psi between filtrate (8 psi) and retentate (10 psi), up to 80 liters of material may be concentrated to a volume of less than 500 ml in under two hours. This process also provides a yield of about 80%.
  • DNAse is added to a concentration of 50 U/ml. and recirculated at a lower pump speed with the filtrate line closed for 30 minutes. If retroviruses have been trapped within a gel layer formed during the ultrafiltration, this step will break down trapped retrovirus, and improve recovery.
  • Discontinuous diafiltration is then accomplished by addition of 4 liters of additional buffer, and utilizing the same cross differential pressure set forth above. Generally, recovery after this step is approximately 70%.
  • Concentrated material is then subjected to column chromatography on a Phamacia S-500 HG size exclusion gel, utilizing 50 mM NaCl and 25 mM Tris pH 7.2-7.5 as minimum salt and ionic strength concentrations. Generally, recombinant retroviruses elute off in the first peak.
  • Tangential flow filtration may once again be utilized in order to further reduce the volume to under 200 ml.
  • the concentrated material is sterilized by filtration through a 0.2 um Millipore filter (PVDF, or Sterivex).
  • methods are provided for quantitating retroviral particles utilizing non-denaturing gels (e.g., 4-15% gradient polyacrylamide gels), along with methods for estimating or quantitating the resultant products such as, for example, staining with coomassie blue or silver stain, followed by densitometry scanning.
  • non-denaturing gels e.g., 4-15% gradient polyacrylamide gels
  • methods for estimating or quantitating the resultant products such as, for example, staining with coomassie blue or silver stain, followed by densitometry scanning.
  • methods while not capable of discriminating between viable and non-viable vector particles, are advantageous because they are relatively simple and quick.
  • assays are provided for titering recombinant retrovirus in a sample. Typically, such assays may be based upon presence of a selectable marker, or formation of blue colonies.
  • recombinant retroviruses which do not include a gene coding for a selectable marker. Therefore, antibody and PCR assays, the latter of which is described below, may be employed in order to determine retrovirus titer.
  • various primers are required. Such primers can readily be designed by those skilled in the art and will depend on the retroviral vector backbone employed and the components thereof, the particular region(s) desired to be amplified, etc. Representative examples of particular primer pairs include those specific for LTR sequences, packaging signal sequences or other regions of the retroviral backbone, include primers specific for the nucleic acid molecule (i.e., non-heterologous sequence) of interest.
  • a PCR titering assay is performed by growing a known number of cells, transduced with a recombinant retrovirus on 6-well plates for at least 16 hr. before harvest. One well per plate is sacrificed for counting. Cells from the other wells are lysed and their contents isolated. DNA is prepared using a QUIAmp DNA isolation kit (QUIAgen, Inc.,Chatsworth, CA). DNAs are resuspended in 5 x IO 6 cell equivalents/ ⁇ l per sample.
  • QUIAmp DNA isolation kit QUIAgen, Inc.,Chatsworth, CA
  • a standard curve is generated using DNA isolated from 5 x IO 6 untransduced HT1080 cells (negative control) and 5 x IO 6 HT1080 cells transduced with a known vector and having one copy of that vector per cell genome (positive control), such as may be prepared from packaging cell lines transduced with a recombinant retrovirus encoding a selectable marker, e.g., neomycin resistance.
  • the standard curve is generated by combining different amounts of the positive and negative control DNA and amplifying specific sequences therefrom by PCR using primers specific to a particular region of the recombinant retrovirus.
  • a representative group of mixtures for generating a standard curve is:
  • DNA preparations are placed into their own reaction tubes in duplicate. PCR reactions (50 ⁇ l total volume) are then initiated by adding 45.0 ⁇ l of a reaction mix containing the following components per tube to be tested: 24.5 ⁇ l water, 5 ⁇ l 10X reaction PCR buffer, 4 ⁇ l of 25 mM MgCl 2 , 4 ⁇ l dNTPs (containing 2.5 mM of each of dATP, dGTP, dCTP, and dTTP), 5 ⁇ l of primer mix (100 ng or each primer), 0.25 ⁇ L TaqStart monoclonal antibody (Clontech Laboratories, Inc., Palo Alto, CA), 1.00 ⁇ L TaqStart buffer (Clontech Labs, Inc.), and 0.25 ⁇ L AmpliTaq DNA polymerase (Perkin-Elmer, Inc., Norwalk, CN).
  • thermocycler Just prior to aliquoting the reaction mix to the reaction tubes, 1 ⁇ L of ⁇ - 32 P dCTP (250 ⁇ Ci; 3000 C/mmol, 10 mCi/mL, Amersham Co ⁇ ., Arlington Heights, IL) is added into the reaction mix. After aliquoting 45.0 ⁇ L the reaction mix into each of the reaction tubes, the tubes are capped and placed into a thermocycler. The particular denaturation, annealing, elongation times and temperatures, and number of thermocycles will vary depending on size and nucleotide composition of the primer pair used. 20 - 25 amplification thermocycles are then performed.
  • the scanning results are then downloaded and plotted on a log scale as cpm (ordinate) versus percent positive control DNA (abscissa).
  • Titers infectious units/mL
  • Titers are calculated by multiplying the number of cells from which DNA was isolated by the percentage (converted to decimal form) determined from the standard curve based on the detected radioactivity, divided by the volume of recombinant retrovirus used to transduce the cells.
  • other methods of detection such as colorimetric methods, may also be employed to label the amplified products.
  • recombinant retrovirus which has been purified or concentrated as described above may be preserved by first adding a sufficient amount of a formulation buffer to the media containing the recombinant retrovirus, in order to form an aqueous suspension.
  • the formulation buffer is an aqueous solution that contains a saccharide, a high molecular weight structural additive, and a buffering component in water.
  • a “buffering compound” or “buffering component” should be understood to refer to a substance that functions to maintain the aqueous suspension at a desired pH.
  • the aqueous solution may also contain one or more amino acids.
  • the recombinant retrovirus can also be preserved in a purified form. More specifically, prior to the addition of the formulation buffer, the crude recombinant retrovirus described above may be clarified by passing it through a filter, and then concentrated, such as by a cross flow concentrating system (Filtron Technology Co ⁇ ., Nortborough, MA). Within one embodiment, DNase is added to the concentrate to digest exogenous DNA. The digest is then diafiltrated to remove excess media components and establish the recombinant retrovirus in a more desirable buffered solution. The diafiltrate is then passed over a Sephadex S-500 gel column and a purified recombinant retrovirus is eluted.
  • a cross flow concentrating system Frtron Technology Co ⁇ ., Nortborough, MA
  • the formulation buffer is an aqueous solution that contains a saccharide, a high molecular weight structural additive, and a buffering component in water.
  • the aqueous solution may also contain one or more amino acids.
  • the crude recombinant retrovirus can also be purified by ion exchange column chromatography. This method is described in more detail in U.S. Patent Application Serial No. 08/093,436.
  • the crude recombinant retrovirus is clarified by passing it through a filter, and the filtrate loaded onto a column containing a highly sulfonated cellulose matrix.
  • the recombinant retrovirus is eluted from the column in purified form by using a high salt buffer.
  • the high salt buffer is then exchanged for a more desirable buffer by passing the eluate over a molecular exclusion column.
  • a sufficient amount of formulation buffer is then added, as discussed above, to the purified recombinant retrovirus and the aqueous suspension is either dried immediately or stored, preferably at -70°C.
  • the aqueous suspension in crude or purified form can be dried by lyophilization or evaporation at ambient temperature.
  • lyophilization involves the steps of cooling the aqueous suspension below the glass transition temperature or below the eutectic point temperature of the aqueous suspension, and removing water from the cooled suspension by sublimation to form a lyophilized retrovirus. Briefly, aliquots of the formulated recombinant retrovirus are placed into an Edwards Refrigerated Chamber (3 shelf RC3S unit) attached to a freeze dryer (Supermodulyo 12K). A multistep freeze drying procedure as described by Phillips et al.
  • recombinant retrovirus is used to lyophilize the formulated recombinant retrovirus, preferably from a temperature of -40°C to -45°C.
  • the resulting composition contains less than 10% water by weight of the lyophilized retrovirus.
  • the recombinant retrovirus is stable and may be stored at - 20°C to 25°C, as discussed in more detail below.
  • water is removed from the aqueous suspension at ambient temperature by evaporation.
  • water is removed through spray drying (EP 520,748).
  • spray drying Within the spray drying process, the aqueous suspension is delivered into a flow of preheated gas, usually air, whereupon water rapidly evaporates from droplets of the suspension.
  • Spray drying apparatus are available from a number of manufacturers (e.g., Drytec, Ltd., Tonbridge, England; Lab-Plant, Ltd., Huddersfield, England). Once dehydrated, the recombinant retrovirus is stable and may be stored at -20°C to 25°C.
  • the resulting moisture content of the dried or lyophilized retrovirus may be determined through use of a Karl-Fischer apparatus (EM Science AquastarTM VI B volumetric titrator, Cherry Hill, NJ), or through a gravimetric method.
  • the aqueous solutions used for formulation are composed of a saccharide, high molecular weight structural additive, a buffering component, and water.
  • the solution may also include one or more amino acids.
  • the combination of these components act to preserve the activity of the recombinant retrovirus upon freezing and lyophilization, or drying through evaporation.
  • a preferred saccharide is lactose
  • other saccharides may be used, such as sucrose, mannitol, glucose, trehalose, inositol, fructose, maltose or galactose.
  • combinations of saccharides can be used, for example, lactose and mannitol, or sucrose and mannitol.
  • a particularly preferred concentration of lactose is 3%-4% by weight.
  • the concentration of the saccharide ranges from 1% to 12% by weight.
  • the high molecular weight structural additive aids in preventing viral aggregation during freezing and provides structural support in the lyophilized or dried state.
  • structural additives are considered to be of "high molecular weight” if they are greater than 5000 m.w.
  • a preferred high molecular weight structural additive is human serum albumin.
  • other substances may also be used, such as hydroxyethyl-cellulose, hydroxymethyl-cellulose, dextran, cellulose, gelatin, or povidone.
  • a particularly preferred concentration of human serum albumin is 0.1 % by weight.
  • the concentration of the high molecular weight structural additive ranges from 0.1%) to 10%) by weight.
  • amino acids function to further preserve viral infectivity upon cooling and thawing of the aqueous suspension.
  • amino acids function to further preserve viral infectivity during sublimation of the cooled aqueous suspension and while in the lyophilized state.
  • a preferred amino acid is arginine, but other amino acids such as lysine, ornithine, serine, glycine, glutamine, asparagine, glutamic acid or aspartic acid can also be used.
  • a particularly preferred arginine concentration is 0.1% by weight.
  • the amino acid concentration ranges from 0.1 % to 10% by weight.
  • the buffering component acts to buffer the solution by maintaining a relatively constant pH.
  • buffers may be used, depending on the pH range desired, preferably between 7.0 and 7.8.
  • Suitable buffers include phosphate buffer and citrate buffer.
  • a particularly prefened pH of the recombinant retrovirus formulation is 7.4, and a preferred buffer is tromethamine.
  • the aqueous solution contain a neutral salt which is used to adjust the final formulated recombinant retrovirus to an appropriate iso-osmotic salt concentration.
  • Suitable neutral salts include sodium chloride, potassium chloride or magnesium chloride.
  • a preferred salt is sodium chloride.
  • Aqueous solutions containing the desired concentration of the components described above may be prepared as concentrated stock solutions.
  • a particularly preferred method of preserving recombinant retroviruses in a lyophilized state for subsequent reconstitution comprises the steps of (a) combining an infectious recombinant retrovirus with an aqueous solution to form an aqueous suspension, the aqueous suspension including 4% by weight of lactose, 0.1% by weight of human serum albumin, 0.03%) or less by weight of NaCl, 0.1 %> by weight of arginine, and an amount of tromethamine buffer effective to provide a pH of the aqueous suspension of approximately 7.4, thereby stabilizing the infectious recombinant retrovirus; (b) cooling the suspension to a temperature of from -40°C to -45°C to form a frozen suspension; and (c) removing water from the frozen suspension by sublimation to form a lyophilized composition having less than 2% water by weight of the lyophilized composition, the composition being capable of infecting mammalian cells upon reconstitution. It is preferred that the recombinant retrovirus
  • mannitol and lactose lyophilized recombinant retrovirus formulations were assayed for preservation of viral activity under various storage temperatures as a function of time.
  • Figure 3 illustrates the results of assays of trehalose recombinant retrovirus formulations for preservation of viral activity under various storage temperatures as a function of time.
  • Figure 4 depicts a comparison of the viral infectivity of frozen formulated recombinant retrovirus (-80°C) as a liquid and the viral infectivity of lyophilized recombinant retrovirus stored at -20°C.
  • Mannitol formulations may lose considerable activity upon lyophilization (5-6 fold), but appear to remain stable subsequent to the lyophilization event. Although not preferable, such a loss is acceptable assuming sufficient amounts of retrovirus are present in the aqueous solution.
  • the lyophilized or dehydrated retroviruses of the subject invention may be reconstituted using a variety of substances, but are preferably reconstituted using water. In certain instances, dilute salt solutions which bring the final formulation to isotonicity may also be used. In addition, it may be advantageous to use aqueous solutions containing components known to enhance the activity of the reconstituted retrovirus. Such components include cytokines, such as IL-2, polycations, such as protamine sulfate, or other components which enhance the transduction efficiency of the reconstituted retrovirus. Lyophilized or dehydrated recombinant retrovirus may be reconstituted with any convenient volume of water or the reconstituting agents noted above that allow substantial, and preferably total solubilization of the lyophilized or dehydrated sample.
  • high titer recombinant retroviral particles of the present invention may be administered to a wide variety of locations including, for example, into sites such as the cerebral spinal fluid, bone marrow, joints, arterial endothelial cells, rectum, buccal/sublingual, vagina, the lymph system, to an organ selected from the group consisting of lung, liver, spleen, skin, blood and brain, or to a site selected from the group consisting of tumors and interstitial spaces.
  • the recombinant retrovirus may be administered intraocularly, intranasally, sublinually, orally, topically, intravesically, intrathecally, topically, intravenously, intraperitoneally, intracranially, intramuscularly, or subcutaneously.
  • Other representative routes of administration include gastroscopy, ECRP and colonoscopy, which do not require full operating procedures and hospital ization, but may require the presence of medical personnel.
  • compositions of the present invention include the following:
  • Intravenous (IV) administration can occur under a variety of protocols known to those of skill in the art.
  • retroviral vector particles can be formulated for IV administration as described above and administered as a single injection.
  • the retroviral vector particles can be delivered in a multiple injection protocol.
  • An example of a multiple injection protocol is administration for three times a day for several consecutive days or on alternate days.
  • the multiple injection schedule can be carried out over a number of days for example a week or 10 days or two weeks.
  • the injection schedule can also be repeated.
  • the total number of vector particles delivered can dispersed in varying amounts of formulation buffer.
  • the retroviral vectors can be delivered as an injection or as an IV formulation such as an IV drip which can be delivered over a longer period of time.
  • IV administration is a preferred route of administration for retroviral vectors expressing secretory proteins such as Factor VIII and human growth hormone (see e.g. examples 18-21 herein). Oral administration is easy and convenient, economical (no sterility required), safe
  • the recombinant retrovirus over dosage can be treated in most cases, and permits controlled release of the active ingredient of the composition (the recombinant retrovirus).
  • the active ingredient of the composition the recombinant retrovirus
  • there may be local irritation such as nausea, vomiting or diarrhea, erratic abso ⁇ tion for poorly soluble drugs, and the recombinant retrovirus will be subject to "first pass effect" by hepatic metabolism and gastric acid and enzymatic degradation.
  • there can be slow onset of action efficient plasma levels may not be reached, a patient's cooperation is required, and food can affect abso ⁇ tion.
  • Preferred embodiments of the present invention include the oral administration of recombinant retroviruses that express genes encoding erythropoietin, insulin, GM-CSF cytokines, various polypeptides or peptide hormones, their agonists or antagonists, where these hormones can be derived from tissues such as the pituitary, hypothalamus, kidney, endothelial cells, liver, pancreas, bone, hemopoetic marrow, and adrenal.
  • Such polypeptides can be used for induction of growth, regression of tissue, suppression of immune responses, apoptosis, gene expression, blocking receptor-ligand interaction, immune responses and can be treatment for certain anemias, diabetes, infections, high blood pressure, abnormal blood chemistry or chemistries (e.g., elevated blood cholesterol, deficiency of blood clotting factors, elevated LDL with lowered HDL), levels of Alzheimer associated amaloid protein, bone erosion/calcium deposition, and controlling levels of various metabolites such as steroid hormones, purines, and pyrimidines.
  • the recombinant retroviruses are first lyophilized, then filled into capsules and administered.
  • Buccal/sublingual administration is a convenient method of administration that provides rapid onset of action of the active component(s) of the composition, and avoids first pass metabolism. Thus, there is no gastric acid or enzymatic degradation, and the abso ⁇ tion of recombinant retroviruses is feasible. There is high bioavailability, and virtually immediate cessation of treatment is possible. Conversely, such administration is limited to relatively low dosages (typically about 10-15 mg), and there can be no simultaneous eating, drinking or swallowing.
  • Preferred embodiments of the present invention include the buccal/sublingual administration of recombinant retroviruses that contain genes encoding self and/or foreign MHC, or immune modulators, for the treatment of oral cancer; the treatment of Sjogren's syndrome via the buccal/sublingual administration of such recombinant retroviruses that contain IgA or IgE antisense genes; and, the treatment of gingivitis and periodontitis via the buccal/sublingual administration of IgG or cytokine antisense genes.
  • Rectal administration provides a negligible first pass metabolism effect (there is a good blood/lymph vessel supply, and absorbed materials drain directly into the inferior vena cava), and the method is suitable of children, patients with emesis, and the unconscious.
  • the method avoids gastric acid and enzymatic degradation, and the ionization of a composition will not change because the rectal fluid has no buffer capacity (pH 6.8; charged compositions absorb best).
  • lipidophilic and water soluble compounds are preferred for abso ⁇ tion by the rectal mucosa, and abso ⁇ tion enhancers (e.g., salts, EDTA, NSAID) may be necessary.
  • abso ⁇ tion enhancers e.g., salts, EDTA, NSAID
  • Preferred embodiments of the present invention include the rectal administration of recombinant retroviruses that contain genes encoding colon cancer antigens, self and/or foreign MHC, or immune modulators.
  • Nasal administration avoids first pass metabolism, and gastric acid and enzymatic degradation, and is convenient.
  • nasal administration is useful for recombinant retrovirus administration wherein the recombinant retrovirus is capable of expressing a polypeptide with properties as described herein. Conversely, such administration can cause local irritation, and abso ⁇ tion can be dependent upon the state of the nasal mucosa.
  • Pulmonary administration also avoids first pass metabolism, and gastric acid and enzymatic degradation, and is convenient. Further, pulmonary administration permits localized actions that minimize systemic side effects and the dosage required for effectiveness, and there can be rapid onset of action and self-medication. Conversely, at times only a small portion of the administered composition reaches the brochioli/alveoli, there can be local irritation, and overdosing is possible. Further, patient cooperation and understanding is preferred, and the propellant for dosing may have toxic effects.
  • Preferred embodiments of the present invention include the pulmonary administration of recombinant retroviruses that express genes encoding IgA or IgE for the treatment of conditions such as asthma, hay fever, allergic alveolitis or fibrosing alveolitis, the CFTR gene for the treatment of cystic fibrosis, and protease and collagenase inhibitors such as ⁇ -1 -antitrypsin for the treatment of emphysema.
  • Ophthalmic administration provides local action, and permit prolonged action where the administration is via inserts.
  • Preferred embodiments of the present invention include the ophthalmic administration of recombinant retroviruses that express genes encoding IgA or IgE for the treatment of hay fever conjunctivitis or vernal and atomic conjunctivitis; and ophthalmic administration of recombinant retroviruses that contain genes encoding melanoma specific antigens (such as high molecular weight-melanoma associated antigen), self and/or foreign MHC, or immune modulators.
  • Transdermal administration permits rapid cessation of treatment and prolonged action leading to good compliance. Further, local treatment is possible, and avoids first pass metabolism, and gastric acid and enzymatic degradation. Conversely, such administration may cause local irritation, is particularly susceptible to tolerance development, and is typically not preferred for highly potent compositions.
  • Preferred embodiments of the present invention include the transdermal administration of recombinant retroviruses that express genes encoding IgA or IgE for the treatment of conditions such as atopic dermatitis and other skin allergies; and transdermal administration of recombinant retroviruses encoding genes encoding melanoma specific antigens (such as high molecular weight- melanoma associated antigen), self and/or foreign MHC, or immune modulators.
  • Vaginal administration provides local treatment and one preferred route for hormonal administration. Further, such administration avoids first pass metabolism, and gastric acid and enzymatic degradation, and is preferred for administration of compositions wherein the recombinant retroviruses express peptides.
  • Preferred embodiments of the present invention include the vaginal administration of recombinant retroviruses that express genes encoding self and/or foreign MHC, or immune modulators.
  • Other preferred embodiments include the vaginal administration of genes encoding the components of sperm such as histone, flagellin, etc., to promote the production of sperm-specific antibodies and thereby prevent pregnancy. This effect may be reversed, and/or pregnancy in some women may be enhanced, by delivering recombinant retroviruses vectors encoding immunoglobulin antisense genes, which genes interfere with the production of sperm- specific antibodies.
  • Intravesical administration permits local treatment for urogenital problems, avoiding systemic side effects and avoiding first pass metabolism, and gastric acid and enzymatic degradation. Conversely, the method requires urethral catheterization and requires a highly skilled staff.
  • Preferred embodiments of the present invention include intravesical administration of recombinant retrovirus encoding antitumor genes such as a prodrug activation gene such thymidine kinase or various immunomodulatory molecules such as cytokines.
  • Endoscopic retrograde cystopancreatography (ERCP) (goes through the mouth; does not require piercing of the skin) takes advantage of extended gastroscopy, and permits selective access to the biliary tract and the pancreatic duct.
  • the method requires a highly skilled staff, and is unpleasant for the patient.
  • routes of administration described herein may be accomplished simply by direct administration using a needle, catheter or related device.
  • one or more dosages may be administered directly in the indicated manner: into the cerebral spinal fluid at dosages greater than or equal to 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 ' ° or 10 ' ' cfu; into bone marrow at dosages greater than or equal to IO 5 , IO 6 , IO 7 , IO 8 , IO 9 , 10 I O or I O 1 1 cfu; into joint(s) at dosages greater than or equal to IO 5 , IO 6 , IO 7 , IO 8 , IO 9 , IO 10 or IO 1 ' cfu; intravenously at dosages greater than or equal to 10 , 10 or IO 1 ' cfu; intra-arterially at dosages greater than or equal to IO 5 , IO 6 , IO 7 , IO 8 , IO 9 , IO 10 or IO 1 1 cfu;
  • Recombinant retrovirus may be delivered to the target from outside of the body (as an outpatient procedure) or as a surgical procedure, where the vector is administered as part of a procedure with other pu ⁇ oses, or as a procedure designed expressly to administer the vector.
  • Other routes and methods for administration include the non-parenteral routes as well as administration via multiple sites.
  • viral vectors and particularly retroviral vectors preferentially infect liver cells and spleen cells in order to effect sustained expression of proteins such as factor VIII and human growth hormone. Furthermore, as described herein expression of proteins in the liver is useful therapeutically, for example, for production of proteins secreted by the liver, and for localized production of therapeutic proteins in the liver which can be used in the treatment of liver diseases.
  • the gene delivery vehicles of the invention including recombinant retroviruses are preferentially administered intravenously, generally in a peripheral vein which is readily accessible.
  • the gene delivery vehicles of the invention may also be delivered into the hepatic artery or in the portal vein in order to have more effective delivery to the liver.
  • cannulation of the portal vein can be accomplished by a simple dissection of the umbilical vein, thereby eliminating a laparotomy (Storer et al, 1966, Am J. Surg 777:56-68, and Weigand et al, 1983, Rec Res Cancer Res 56:90-92).
  • the ligamentum teres is cut 4 to 5 cm above the umbilicus and a polyethlyene catheter is threaded into the umbilical vein until reaching the junction with the left branch of the portal vein.
  • a brisk return of blood through the catheter indicates the proper entry into the portal system.
  • gene delivery vehicles of the invention can be delivered by direct injection into the liver using standard medical procedures.
  • the recombinant viruses of the invention can be administered after induction of cell proliferation by a growth factor, or may be co-administered with a growth factor.
  • the growth factors of the invention are administered by parenteral, topical, oral or by local administration.
  • the growth factors are administered parenterally, eg. intravenously, subcutaneously, intradermally, or intramuscularly.
  • the growth factors are administered intravenously.
  • Administration of the therapeutic agent of the invention can be accomplished by, for example, injection, catheterization, laser-created perfusion channels, cannulization, a particle gun, and a pump.
  • the growth factors of the invention are typically administered with a pharmaceutical carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Such carriers are well known to those of ordinary skill in the art.
  • Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • compositions may contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • the therapeutic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. Liposomes are included within the definition of a pharmaceutically acceptable carrier.
  • liposomes refers to, for example, the liposome compositions described in U.S. Patent No. 5,422,120, WO 95/13796, WO 94/23697, WO 91/14445 and EP 524,968 Bl . Liposomes may be pharmaceutical carriers for the polypeptides of the invention.
  • the growth factors of the invention are administered in therapeutically effective amounts.
  • therapeutically effective amount refers to an amount of a growth factor that is capable of stimulating cell division in a target tissue in vivo. Stimulation of cell proliferation in a target tissue means that the number of dividing cells in the target tissue is greater than in the absence of treatment.
  • the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and particular growth factor that is used. The effective amount for a given situation can be determined by routine experimentation and will vary from growth factor to growth factor.
  • a dose of 1 ug/kg to 2 mg/kg body weight, and more preferably from 10 ug/kg to 200 ug/kg body weight is used.
  • a dose of 100 ug/kg to 5 mg/kg body weight, or more preferably a dose of 1 mg/kg to 50 mg/kg body weight is used.
  • Dose amounts for the other growth factors used in the claimed methods are known to those of skill in the art or can readily be determined experimentally.
  • Clofibrate or the other proxisome proliferators, can be administered by IP injection (5-500 mg/kg), or orally (5-500 mg/kg). More preferably the dosages are 10-100 mg/kg.
  • a typical dosing schedule is daily administration for 3-10 days.
  • a tapered dosing can alternatively be employed.
  • retroviral vectors can be administered, preferably intravenously, at doses ranging from 1E5 to 1E11 cfu per injection. Injection schedules of one to three times daily, for one to ten days, will be employed. Repeat administrations of retroviral vector with or without repeat clofibrate or growth factor dosing can be performed.
  • Collagenase can be administered at a dosage of from .05-5 U/kg, more preferably 0.5-2 U/kg) (Liu (994) Hepatology 19:1521 ).
  • Estrogens with HGF or other growth factors can be administered at a dosage of 0.01-20 ug/g, and more preferably 0.5-2 ug/g.
  • Heparin or other sulfated glycosaminoglycans administered with HGF or other growth factors can be administered at a dose of 100-10,000 U/kg, more preferably 2000-6000 U/kg.
  • 9-cis-retinoic acid is administered at dosages of 10-200 mg/kg, more preferably 60- 100 mg/kg.
  • Cyclosporine A is delivered at a dose of 0.1 -100 mg/kg, more preferably 10-50 mg/kg.
  • Prostaglandins El and/or II ; isoproterenol can be administered at dosages of 0.5-100 ug/kg; more preferably 1-10 ug/kg with or without glucagon at a dose of 0.1.1-10 mg/kg, more preferably 0.5-2 mg/kg.
  • Phosphatidylethanolamine N-methyltransferase inhibitors such as 3-deazaadenosine or other methylation inhibitors are administered at a dose of 1-50 mg/kg, more preferably 5-15 mg/kg.
  • Liver mitogen nafenopin can be administered at a dose of 0.1- 1000 mg/kg, more preferably 50-300 mg/kg.
  • Cyproterone acetate can be administered at a dose of (0.1 - 600 mg/kg, more preferably 10-100 mg/kg.
  • Triiodothyrone can be administered at at dose of 0.1-10 mg/kg, more preferably 0.5-5.0 mg/kg.
  • the N2R5 construct is mutated by site-directed in vitro mutagenesis to change the ATG start codon to ATT preventing gag expression.
  • This mutagenized fragment is 200 base pairs (bp) in length and flanked by Pst I restriction sites.
  • the Pst I-Pst I mutated fragment is purified from the SK+ plasmid and inserted into the Pst I site of N2 MoMLV 5' LTR in plasmid pUC31 to replace the non-mutated 200 bp fragment.
  • the plasmid pUC31 is derived from pUC19 (Stratagene, La Jolla, CA) in which additional restriction sites Xho I, Bgl II, BssH II and Neo I are inserted between the EcoR I and Sac I sites of the polylinker. This construct is designated pUC3 l/N2R5gM.
  • a 1.0 kilobase (Kb) MoMLV 3' LTR EcoR I-EcoR I fragment from N2 is cloned into plasmid SK + resulting in a construct designated N2R3 " .
  • a 1.0 Kb Cla I-Hind III fragment is purified from this construct.
  • the Cla I-Cla I dominant selectable marker gene fragment from pAFVXM retroviral vector (Kriegler et al, 1984, Ce77 55:483; St. Louis et al, 1988, PNAS 55:3150-3154), comprising a SV40 early promoter driving expression of the neomycin (neo) phosphotransferase gene, is cloned into the SK + plasmid.
  • This construct is designated SK SV2- «eo A 1.3 Kb Cla I-BstB I gene fragment is purified from the SK + SV2- «eo plasmid.
  • KT-3B or KT-1 vectors are constructed by a three part ligation in which the Xho I- Cla I fragment containing the gene of interest and the 1.0 Kb MoMLV 3' LTR Cla I-Hind III fragment are inserted into the Xho I-Hind III site of pUC31/N2R5gM plasmid. This gives a vector designated as having the KT-1 backbone.
  • the 1.3 Kb Cla I-BstB I neo gene fragment from the pAFVXM retroviral vector is then inserted into the Cla I site of this plasmid in the sense orientation to yield a vector designated as having the KT-3B backbone.
  • retroviral vectors encoding a full-length factor VIII cDNA. Further discussion is also provided in PCT Patent Publication No. WO 96/21035. Due to the packaging constraints of retroviral vectors and because selection for transduced cells is not a requirement for therapy, a retroviral backbone, e.g. , KT- 1 , lacking a selectable marker gene is employed.
  • a gene encoding full-length factor VIII can be obtained from a variety of sources.
  • One such source is the plasmid pCIS-F8 (see EP 0 260 148), which contains a full-length factor VIII cDNA whose expression is under the control of a CMV major immediate-early (CMV MIE) promoter and enhancer.
  • the factor VIII cDNA contains approximately 80 bp of 5' untranslated sequence from the factor VIII gene and a 3' untranslated region of about 500 bp.
  • a CMV intron sequence, or "cis" element between the CMV promoter and the factor VIII sequence lies a CMV intron sequence, or "cis" element.
  • the cis element spanning about 280 bp, comprises a splice donor site from the CMV major immediate-early promoter about 140 bp upstream of a splice acceptor from an immunoglobulin gene.
  • a plasmid, designated pJW-2 encoding a retroviral vector for expressing full length factor VIII is constructed using the KT-1 backbone from pKT-1. Briefly, in order to facilitate directional cloning of the factor VIII cDNA insert into pKT-1, the unique Xho I site is converted to a Not I site by site directed mutagenesis. The resultant plasmid vector is then opened with Not I and Cla I. pCIS-F8 is digested to completion with Cla I and Eag I, for which there are two sites, to release the fragment encoding full-length factor VIII. This fragment is then ligated into the Not I/Cla I restricted vector to generate a plasmid designated pJW-2.
  • a plasmid vector encoding a truncation of about 80% (approximately 370 bp) of the 3' untranslated region of the factor VIII cDNA, designated pND-5, is constructed in a pKT-1 vector as follows: As described for pJW-2, the pKT-1 vector employed has its Xho I restriction site replaced by that for Not I.
  • the factor VIII insert is generated by digesting pCIS-F8 with Cla I and Xba I, the latter enzyme cutting 5' of the factor VIII stop codon. The approximately 7 kb fragment containing all but the 3' coding region of the factor VIII gene is then purified.
  • pCIS-F8 is also digested with Xba I and Pst I to release a 121 bp fragment containing the gene's termination codon. This fragment is also purified and then ligated in a three way ligation with the larger fragment encoding the rest of the factor VIII gene and Cla I/Pst I restricted BLUESCRIPT® KS + plasmid (Stratagene, supra) to produce a plasmid designated pND-2.
  • the unique Sma I site in pND-2 is then changed to a Cla I site by ligating Cla I linkers (New England Biolabs, Beverly, MA) under dilute conditions to the blunt ends created by a Sma I digest. After recircularization and ligation, plasmids containing two Cla I sites are identified and designated pND-3.
  • the factor VIII sequence in pND-3 is cloned as follows into a plasmid backbone derived from a Not I/Cla I digest of pKT-1 (a pKT-1 derivative by cutting at the Xho I site, blunting with Klenow, and inserting a Not I linker (New England Biolabs)), which yields a 5.2 kb Not I/Cla I fragment.
  • pCIS-F8 is cleaved with Eag I and Eco RV and the resulting fragment of about 4.2 kb, encoding the 5' portion of the full-length factor VIII gene, is isolated.
  • pND-3 is digested with Eco RV and Cla I and a 3.1 kb 71
  • This expression vector is designated p25D and has the exact backbone as pCISF8 above.
  • the Hpa I site at the 3' of the FVIII8 cDNA in p25D is modified to Cla-I by oligolinkers.
  • An Ace I to Cla I fragment is clipped out from the modified p25D plasmid. This fragment spans the B-domain deletion and includes the entire 3' two-thirds of the cDNA.
  • An Ace I to Cla I fragment is removed from the RetrovectorTM JW-2 above, and replaced with the modified B-domain deleted fragment just described. This is designated B-del-1.
  • BC10ME (Patek et al, 1982. Cell Immuno 72:113, ATCC# TIB85) L33env, and BCenv (L33e > and BCewv express HIV-1 IIIBeHv, Warner et al, 1991 , AIDS Res. and Human Retrovirus 7:645)
  • L33env L33e > and BCewv express HIV-1 IIIBeHv, Warner et al, 1991 , AIDS Res. and Human Retrovirus 7:645
  • Non-transduced cells are also analyzed for factor VIII expression and compared with KT-ND5 transduced cells to determine the effect of transduction on protein expression.
  • Murine cell lines L33-KT-ND5, L33e «v-KT-ND5, L33e «v, L33, BC10ME, BC10ME-KT-ND5, BCeov, and BCe «v-KT-ND5, are tested for expression of the KT-ND5 molecule.
  • Cells are grown to subconfluent density and the supernatant is removed following centrifugation at 200 xg. The samples are diluted and assayed by the Coamatic® Factor VIII assay (KabiVitrum Diagnostica, Molndal, Sweden).
  • Coamatic® Factor VIII assay Karlin, Molndal, Sweden
  • the assay is performed as follows: 100 ⁇ l of culture media are mixed with 200 ⁇ l of lx working buffer diluted according to the maufacturer's instructions. Fifty ⁇ l of the mixture are prewarmed at 37°C for 3-4 minutes in wells of a microtiter plate. The 50 ⁇ l of factor reagent from the kit, prewared to 37°C are added, and the mixture incubated at 37°C for 4 min., after which 50 ⁇ l of chromogenic substrate (7.7mg S2765 plus 0.2 mg of thrombin inhibitor 12581 in a mannitol carrier, diluted to a total of 6.0 ml with sterile water) are added.
  • Transduction of Human Cells Cell lines transduced with KT-ND5 are examined for expression of factor VIII. Non-transduced cells are analyzed to compare with KT-ND5 transduced cells and determine the effect that transduction has on expression.
  • the packaging cell line, HX (WO92/05266), are seeded at 5.0 x 10 s cells on a 10 cm tissue culture dish on day 1 with Dulbecco's Modified Eagle Medium (DMEM) and 10% fetal bovine serum (FBS). On day 2, the media is replaced with 5.0 ml fresh media 4 hours prior to transfection.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • a standard calcium phosphate-DNA co-precipitation is performed by mixing 40.0 ⁇ l 2.5 M CaC , 10 ⁇ g plasmid DNA, and deionized H 2 O to a total volume of 400 ⁇ l.
  • 29 2 3 cells (WO 92/05266) ( these are 293 cells expressing gag and pol) are transfected with the vector DNA and the plasmid pMLP-VSVG ( or other VSVG encoding plasmids) to yield VSVG psuedotyped vector particles that are harvested and stored as described above.
  • DA amphotropic cell line derived from a D17 cell line ATCC No. 183, WO 92/05266
  • cells are seeded at 5.0 x 10 5 cells/10 cm tissue culture dish in 10 ml DMEM and 10%) FBS, 4 ⁇ g/ml polybrene (Sigma, St. Louis, MO) on day 1.
  • FBS 4 ⁇ g/ml polybrene (Sigma, St. Louis, MO)
  • 3.0 ml, 1.0 ml and 0.2 ml of the freshly collected virus-containing HX media is added to the cells.
  • the cells are incubated with the virus overnight at 37°C.
  • the media is removed and 1.0 ml DMEM, 10% FBS with 800 ⁇ g/ml G418 is added to the plate.
  • a G418 resistant pool is generated over a period of a week. The pool of cells is dilution cloned by removing the cells from the plate and counting the cell suspension, diluting the 74
  • the titer of the individual clones is determined by infection of HT 1080 cells, (ATCC No. CCL 121). On day 1, 5.0 x 10 5 HT1080 cells are plated on each well of a 6 well microtiter plate in 3.0 ml DMEM, 10% FBS and 4 ⁇ g/ml polybrene. On day 2, the supernatant from each clone is serially diluted 10 fold and used to infect the HT1080 cells in 1.0 ml aliquots. The media is replaced with fresh DMEM, 10% FBS media, and the cells incubated with the vector overnight at 37°C, 10% CO 2 .
  • transduced cells On day 3, selection of transduced cells is performed by replacing the media with fresh DMEM, 10% FBS media containing 800 ⁇ g/ml G418. Cells are incubated at 37°C, 10%) CO2 for 14 days at which time G418 resistant colonies are scored at each dilution to determine the viral titer of each clone as colony forming units(cfu)/ml.
  • cell lines are derived that produce greater than or equal to IO 6 cfu/ml in culture.
  • the packaging cell line HX is transduced with vector generated from the DA vector producing cell line in the same manner as described for transduction of the DA cells from HX supernatant.
  • Transduction of the DA or HX cells with vectors lacking a neo selectable marker was performed as described above. However, instead of adding G418 to the cells on day 3, the cells are cloned by limiting dilution. Titer is analyzed as described above.
  • DA cells are seeded at 5.0 x 10 5 cells on a 10 cm tissue culture dish on day 1 with DMEM and 10%> irradiated (2.5 megarads minimum) FBS. On day 2, the media is replaced with 5.0 ml fresh media 4 hours prior to 7 11785
  • a standard calcium phosphate-DNA coprecipitation is performed by mixing 60 ⁇ l 2.0 M CaC , 10 ⁇ g MLP-G plasmid, 10 ⁇ g KT-ND5 retroviral vector plasmid, and deionized water to a volume of 400 ⁇ l.
  • Four hundred microliters of the DNA-CaCl2 solution is added dropwise with constant agitation to 400 ⁇ l 2X precipitation buffer (50 mM HEPES-NaOH, pH 7.1 , 0.25 M NaCl and 1.5 mM Na2HPO4-NaH2PO4). This mixture is incubated at room temperature for 10 minutes. The resultant fine precipitate is added to a culture dish of DA cells plated the previous day.
  • the cells are incubated with the DNA precipitate overnight at 37°C. On day 3, the medium is removed and fresh medium is added. The supernatant containing G-pseudotyped virus is removed on day 4, passed through a 0.45 ⁇ l filter and used to infect the DA packaging cell.
  • DA cells are seeded at 5.0 x 10 5 cells on a 10 cm tissue culture dish in 10 ml DMEM and 10% FBS, 4 mg/ml polybrene (Sigma, St. Louis, MO) on day 1.
  • 10 ml DMEM 10% FBS, 4 mg/ml polybrene (Sigma, St. Louis, MO)
  • FBS 4 mg/ml polybrene
  • 2.0 ml 2.0 ml, 1.0 ml or 0.5 ml of the freshly collected and filtered G-pseudotyped virus containing supernatant is added to the cells.
  • the cells are incubated with the virus overnight at 37°C.
  • the medium is removed and 10 ml DMEM, 10% irradiated FBS with 800 ⁇ g/ml G418 is added to the plate. Only cells that have been transduced with the vector and contain the neo selectable marker will survive.
  • a G418 resistant pool is generated over the period of 1-2 weeks.
  • the pool is tested for expression and then dilution cloned by removing the cells from the plate, counting the cell suspension, diluting the cell suspension down to 10 cells/ml and adding 0.1 ml to each well (1 cell/well) of a 96-well plate. Cells are incubated for 2 weeks at 37°C, 10% CO2 Twenty-four clones are selected and expanded up to 24-well plates, then 6-well plates, and finally 10 cm plates, at which time the clones are assayed for expression and the supernatants are collected and assayed for viral titer as described above.
  • the extended S + L assay determines whether replication competent, infectious virus is present in the supernatant of the cell line of interest.
  • the assay is based on the empirical observation that infectious retroviruses generate foci on the indicator cell line MiCl ] (ATCC No. CCL 64.1).
  • the MiCl] cell line is derived from the MvlLu mink cell line (ATCC No. 97/11785
  • CCL 64 by transduction with Murine Sarcoma Virus (MSV). It is a non-producer, non- transformed, revertant clone containing a replication defective murine sarcoma provirus, S + , but not a replication competent murine leukemia provirus, L". Infection of MiClj cells with replication competent retrovirus "activates" the MSV genome to trigger "transformation" which results in foci formation.
  • MSV Murine Sarcoma Virus
  • Supernatant is removed from the cell line to be tested for presence of replication competent retrovirus and passed through a 0.45 ⁇ filter to remove any cells.
  • MvlLu cells are seeded at 1.0 x 10 5 cells per well (one well per sample to be tested) of a 6 well plate in 2 ml DMEM, 10%> FBS and 8 ⁇ g/ml polybrene.
  • MvlLu cells are plated in the same manner for positive and negative controls on separate 6 well plates. The cells are incubated overnight at 37°C, 10%> CO2.
  • 1.0 ml of test supernatant is added to the Mvl Lu cells.
  • the negative control plates are incubated with 1.0 ml of media.
  • the positive control consists of three dilutions (200 focus forming units (ffu), 20 ffu and 2 ffu each in 1.0 ml media) of MA virus (referred to as pAM in Miller et al, Molec. and Cell Biol. 5:431 , 1985) which is added to the cells in the positive control wells. The cells are incubated overnight. On day 3, the media is aspirated and 3.0 ml of fresh DMEM and 10% FBS is added to the cells. The cells are allowed to grow to confluency and are split 1 : 10 on day 6 and day 10, amplifying any replication competent retrovirus.
  • MA virus referred to as pAM in Miller et al, Molec. and Cell Biol. 5:431 , 1985
  • the media on the MvlLu cells is aspirated and 2.0 ml DMEM and 10%) FBS is added to the cells.
  • the MiCli cells are seeded at 1.0 x 10 5 cells per well in 2.0 ml DMEM, 10%> FBS and 8 ⁇ g/ml polybrene.
  • the supernatant from the MvlLu cells is transferred to the corresponding well of the MiClj cells and incubated overnight at 37°C, 10% CO2.
  • the media is aspirated and 3.0 ml of fresh DMEM and 10%> FBS is added to the cells.
  • the cells are examined for focus formation (appearing as clustered, refractile cells that overgrow the monolayer and remain attached) on the monolayer of cells.
  • the test article is determined to be contaminated with replication competent retrovirus if foci appear on the MiCli cells. Using these procedures, it can be shown that the HBV core producer cell lines are not contaminated with replication competent retroviruses. 77
  • production cells are cocultivated with an equivalent number of Mus dunni (NIH NIAID Bethesda, MD) cells.
  • Small scale cocultivations are performed by mixing of 5.0 x 10 5 Mus dunni cells with 5.0 x 10 5 producer cells and seeding the mixture into 10 cm plates (10 ml standard culture media/plate, 4 ⁇ g/ml polybrene) at day 0. Every 3-4 days the cultures are split at a 1 :10 ratio and 5.0 x 10 5 Mus dunni cells are added to each culture plate to effectively dilute out the producer cell line and provide maximum amplifcation of RCR.
  • culture supernatants are harvested, passed through a 0.45 ⁇ cellulose-acetate filter, and tested in the MdH marker rescue assay.
  • Large scale cocultivations are performed by seeding a mixture of 1.0 x 10 8 Mus dunni cells and 1.0 x 10 8 producer cells into a total of twenty T-150 flasks (30 ml standard culture media/flask, 4 ⁇ g/ml polybrene). Cultures are split at a ratio of 1 : 10 on days 3, 6, and 13 and at a ratio of 1 :20 on day 9.
  • the final supernatants are harvested, filtered and a portion of each is tested in the MdH marker rescue assay.
  • the MdH marker rescue cell line is cloned from a pool of Mus dunni cells transduced with LHL, a retroviral vector encoding the hygromycin B resistance gene (Palmer et al, 1987, PNAS 84: 1055-1059).
  • the retroviral vector can be rescued from MdH cells upon infection of the cells with RCR.
  • One ml of test sample is added to a well of a 6- well plate containing IO 5 MdH cells in 2 ml standard culture medium (DMEM with 10% FBS, 1%) 200 mM L-glutamine, 1%> non-essential amino acids) containing 4 ⁇ g/ml polybrene. Media is replaced after 24 hours with standard culture medium without polybrene.
  • HT1080 cells are set up at 2 x IO 4 cells per well in six well tissue culture plates containing 2 mis standard growth media (DME + 10% FBS ).
  • ND-5 FVIII retroviral vector particles from a confluent vector producing cell line are harvested as a HX-ND-5 clone. They are filtered through .45 ⁇ m syringe filters prior to testing the supernatants. (Alternatively the filtered media supernatants may be frozen at 80 in aliquots for later use.) Polybrene is added to each well such that the final concentration is 8 ⁇ g per ml. Thirty minutes later, either diluted or undiluted retroviral vector supernatant is added to duplicate wells.
  • Typical volumes and dilutions are 0.5 ml per well and four or more 1 :3 serial dilutions in growth media.
  • two wells are transduced with the same volume of growth media only.
  • the wells are refeed with 2mls of fresh media and the cells allowed to reach confluence, which may typically be about day four or five.
  • the cells are again refeed with one ml per well fresh growth media. Twenty four hours later the media is harvested and filtered as above.
  • the expression of vector transduced human cells for FVIII is detected by the Coamatic R assay as described above in Example 2B 1. Activity is assayed relative to supernatant from the control wells by counting the cells per well from the two control wells and normalizing FVIII expression data per lx IO 6 cells per 24 hours.
  • the intestinal epithelium is an attractive site for gene delivery due to its rapidly proliferating tissue mass and the known location of stem cells in the crypts of Lieberkuhn.
  • the accessibility of the retroviral vector to these stem cells can be improved in animal models 79
  • Blood is collected from the tail vein and assayed for factor VIII production by a sandwich ELISA specific for human factor VIII (according to the modified procedure of Zatloukal, K., et al, PNAS 97:5148-5152, 1994).
  • the ELISA is based on two Diagnostica). ESH 4 (25 ⁇ g/ml in 1.0 M NaHCO 3 /0.5 M NaCl, pH 9.0) is coupled to the ELISA plates overnight at 4°C, washed with 0.1%o Tween 20 in PBS, and blocked with 1%> BSA in PBS.
  • the samples are applied in 0.05 M Tris-HCl/1 M NaCl/2% BSA, pH 7.5, over 4 hr at room temperature, the plates are washed, and ESH 8 (2.5 ⁇ g/ml in 0.05 M Tris-HCl/1 M NaCl/2% BSA, pH 7.5,) which has been biotinylated with N- hydroxysuccinimidobiotin (Pierce, Rockford, IL.) is added for 2 hr at room temperature.
  • the color reaction is performed with peroxidase-conjugated streptavidin (Boehringer Mannheim, Indianapolis, IN.) and o- phenylenediamine dihydrochloride as substrate.
  • the human factor VII c standard from the National Institute for Biological Standards and Control, Hertfordshire, U.K.
  • normal rat plasma are used as references.
  • Lyophilized recombinant retrovirus containing the gene for Factor VIII expression is formulated into an enteric coated tablet or gel capsule according to known methods in the art. These are described in the following patents: US 4,853,230, EP 225,189, AU 9,224,296, AU 9,230,801, and WO 92144,52. /US97/11785
  • the capsule is administered orally to be targeted to the jejunum.
  • expression of Factor VIII is measured in the plasma and blood by the Coamatic R Factor VIII assay as described in Example 2B1.
  • retroviral vectors are based on the N2 vector (Keller et al., Nature 575:149-154, 1985). Briefly, 5' and 3' Eco RI LTR fragments (2.8 and 1.0 Kb, respectively) (Armentano, J. Vir. 67:1647, 1987; Eglitis, Science 250:1395, 1985) are initially subcloned into the Eco RI site of plasmids SK (Stratagene, San Diego, CA) and pUC31.
  • pUC31 is a modification of pUC 19 (Stratagene, San Diego, CA) carrying additional restriction sites (Xho I, Bgl II, BssH II, and Neo I) between the Eco RI and Sac I sites of the polylinker.
  • Plasmid N2R3+/- is thereby created from ligation of the SK + plasmid with the 1.0 Kb 3' LTR fragment.
  • the plasmids p31N2R5+/- and p31N2R3+/- are constructed from the ligation of pUC31 with the 2.8 Kb 5' LTR and packaging signal (Y) or the 1.0 Kb 3' LTR fragment, respectively.
  • N2 refers to the vector source
  • R refers to the fact that the fragment is an Eco RI fragment
  • 5 and 3 refer to 5' or 3' LTRs
  • + or - refers to the orientation of the insert (see Figures 1-6 for examples of LTR subclones).
  • a 1.2 Kb Cla I/Eco RI 5' LTR and W fragment from N2 is subcloned into the same sites of an SK vector. This vector is designated pN2CR5.
  • the 5' LTR containing a 6 bp deletion of the splice donor sequence (Yee et al, Cold Spring Harbor, Quantitative Biology, 51:1021, 1986) is subcloned as a 1.8 Kb Eco RI fragment into pUC31.
  • This vector is designated p31N25D[+], Figure 6.
  • HSV- 1 thymidine kinase gene The coding region and transcriptional termination signals of HSV- 1 thymidine kinase gene (HSVTK) are isolated as a 1.8 Kb Bgl II/Pvu II fragment from plasmid 322TK (3.5 kb Bam HI fragment of HSV-1 (McKnight et al.) cloned into Bam HI of pBR322 (ATCC No. 31344)) and cloned into Bgl II/Sma I-digested pUC31. This construct is designated pUCTK. For constructs which require deletion of the terminator signals, pUCTK is digested with Sma I and Bam HI and the 0.3 Kb fragment containing the (A) n signal is removed.
  • the remaining coding sequences and sticky-end Bam HI overhang are reconstituted with a double- stranded oligonucleotide made from the following oligomers: 5' GAG AGA TGG GGG AGG CTA ACT GAG 3' (SEQUENCE ID. NO. 1) 5' GAT CCT CAG TTA GCC TCC CCC ATC TCT C 3' (SEQUENCE ID. NO. 2)
  • pTKD A Figure 7.
  • the oligonucleotides are designed to destroy the Sma I site while maintaining the Ava I site without changing the translated protein.
  • the plasmid pPrTKDA ( Figure 8), which contains the HSVTK promoter and coding sequence (lacking an (A) signal), is constructed as follows.
  • pTKD A is linearized with Bgl II treated with alkaline phosphatase, and gel purified.
  • a 0.8 Kg fragment contained the HSVTK transcriptional promoter is isolated as a Bam HI/Bgl II fragment from p322TK.
  • retroviral provectors expressing HSVTK from a constitutive promoter The retroviral provectors pTK-1 and pTK-3 are constructed essentially as described below.
  • HSVTK coding sequences lacking transcriptional termination sequences are isolated as a 1.2 Kb Xho I/Bam HI fragment from pTKDA ( Figure 2).
  • TK- 1 Figure 9
  • pTK-3 is constructed by linearizing TK-1 with Bam HI, filling in the 5' overhang and blunt-end ligating a 5'-f ⁇ lled Cla I/Cla I fragment containing the bacterial lac UV5 promoter, SV40 early promoter, plus Tn5 neo r gene obtained from pAFVXM retroviral vector (Krieger et al, 1984, Cell 59:483; St. Louis et al, 1988, PNAS 55:3150). Kanamycin-resistant clones are isolated and individual clones are screened for the proper orientation by restriction enzyme analysis (Figure 9).
  • constructs were used to generate infectious recombinant vector particles in conjunction with a packaging cell line, such as DA as described above.
  • CT26 cells colon tumor 26, Brattain, Baylor College of Medicine, Houston, TX
  • CT26 cells are transduced with G-pseudotyped TK-3 vector. Twenty-four hours after adding the viral supernatant, the CT26 cells are placed under G-418 selection (450 lg/ml). After 10 days incubation, a G-418 selected pool is obtained and designated CT26TK Neo.
  • CT26 TK Neo cells were seeded into six 10 cm plates at a density of 2.5 X 10 per plate.
  • each of two other cell types CT26 and CT26 beta-gal (this cell line was transduced with a retroviral vector carrying the reporter gene ⁇ -galactosidase from E. coli.), were also seeded into six 10 cm plates as controls.
  • Five plates of each cell type were treated twice per day for four consecutive days with medium containing ganciclovir concentrations of 100 ⁇ g/ml, 50 ⁇ g/ml, 25 ⁇ g/ml, 12.5 ⁇ g/ml and 6.25 ⁇ g/ml.
  • One plate of each cell type was left untreated. Afterwards, the cells were removed from each dish using trypsin-EDTA, resuspended in DMEM with 10% FBS and counted.
  • the data in Figure 10 shows that even the lowest dose of ganciclovir had a 83
  • mice In order to test whether in vivo transduction of a murine tumor could be used to treat the disease, an experiment was performed to determine the optimal concentration of ganciclovir necessary to eliminate a tumor that was transduced and selected in vitro to assure 100%) transduction. Twelve groups of 3 mice each are injected with 2 X 10 CT26TK Neo cells. Six groups of mice are injected with these cells intraperitoneally (I.P.) and six groups of mice are injected subcutaneously (S.C). Two other groups of 3 mice each are injected with 2 X 10 5 unmodified CT26 cells (as a control) either I.P. or S.C.
  • mice in the 125 mg/Kg, 250 mg/Kg and 500 mg/Kg treated groups were dead due to the toxic effects of ganciclovir.
  • Mice in the 15.63 mg/Kg, 31.25 mg/Kg and 62.5 mg/Kg treated groups were treated for an additional 7 days and were able to tolerate the treatment. Tumor measurements were made for 23 days Figure 11).
  • CT26TK Neo grew only slightly slower than unmodified CT26 in the absence of ganciclovir.
  • Complete tumor regression was seen in the groups of mice treated with the 62.5 mg/Kg regimen. Partial tumor regression was seen in the 31.25 mg/Kg treated groups. Little or no effect was seen in the 15.63 mg/Kg treated groups as compared to the 2 untreated control groups. Even though there was some toxicity observed in the 62.5 mg/Kg 85
  • mice In order to determine whether ganciclovir has an effect on the growth of unmodified CT26 tumor cells in vivo, 2 groups of 7 mice are injected S.C. with 2 X 10 s unmodified CT26 cells and 2 groups of 7 mice are injected S.C. with 2 X 10 5 CT26TK Neo cells. Seven days after tumor implantation, one group of CT26 injected mice and one group of CT26TK Neo injected mice are placed on a twice daily (AM and PM) regimen of I.P. ganciclovir at 62.5 mg/Kg. These mice are treated for 12 days or until the CT26TK Neo injected animals have no detectable tumor burden. Tumor growth is monitored over a three week period.
  • AM and PM twice daily
  • mice injected with CT26 and treated with ganciclovir had tumors that were somewhat smaller than untreated mice injected with CT26, indicating a small HSVTK - independent inhibition of tumor growth (Figure 12). However, a dramatic decrease in tumor burden was observed if, and only if, CT26 TKneo containing mice were treated with ganciclovir ( Figure 12).
  • AY-27 cells that are transduced with DA/TK-3.
  • AY-27 cells are rat carcinoma cells which have been induced by N-[4-(5-nitro-2-furyl)-2-thiazolyl] formamide ( Selman et al, 1986, J. Urol 756:141, 1986).
  • the AY-27 cells are transduced with G-pseudotyped TK-3 vector. Twenty-four hours after adding the viral supernatant, the AY-27 cells are placed under G- 418 selection (450 ⁇ g/ml). After 10 days incubation, a G-418 selected pool is obtained and designated AY-27TK Neo.
  • AY-27TK Neo cells are seeded into six 10 cm plates at a density of 2.5 X 10 per plate.
  • 2 galactosidase from E. coli. are also seeded into six 10 cm plates as controls. Five plates of each cell type are treated twice per day for four consecutive days with medium containing ganciclovir concentrations of 100 ⁇ g/ml, 50 ⁇ g/ml, 25 ⁇ g/ml, 12.5 ⁇ g/ml and 6.25 ⁇ g/ml. One plate of each cell type is left untreated. Afterwards, the cells are removed from each dish using trypsin- ⁇ DTA, resuspended in DM ⁇ M with 10%) FBS and counted. The data generated can be used to determine the concentration which has the most cytotoxic effect o the AY-27, AY-27 beta-gal, or AY-27 TK Neo cells.
  • Each dose regimen consists of 2 daily AM and PM I.P. injections of ganciclovir. The experiment is summarized in Table B below.
  • Rats are treated for 12 days, eliminating those that die due to the higher concentrations of ganciclovir.
  • Tumor measurements are made for 20 days, assessing tumor regression in order to determine an optimal ganciclovir concentration.
  • AY-27 rat bladder carcinoma cells are transplanted into the bladders of 20 male Fischer 344 rats (Charles River Breeding Laboratories, Portage, MD).
  • tumor-bearing rats weighing between 200 to 300 grams are anesthetized, their bladders are surgically exteriorized and evacuated of urine with a 27- gauge needle.
  • Viral vector particles are instilled into the bladders of 5 rats at 10 6 to 10 ' ° in cfu 200 to 2,000 ⁇ l of formulation buffer.
  • the addition of 4 to 8 ⁇ g/ml of polybrene increases the efficiency of transduction.
  • the cystotomy is repaired with 7-zero nonabsorbable suture.
  • the virus is allowed to incubate in the presence of the tumor cells for 0.5 to 1.0 hour by keeping the animal anesthetized and thereby preventing voiding.
  • Ten control rats receive 500 ⁇ l of formulation buffer only.
  • 200 to 2,000 ⁇ l of vector can be instilled directly into the bladder by catheterization through the urethra following urine evacuation and rinsing with saline.
  • the rats are placed on twice daily (AM and PM) injections of I.P. ganciclovir at the previously determined optimum dose (e.g. 62.5 mg/Kg body weight) for 4 to 12 days. Finally, the rats receive a single daily dose of ganciclovir until the end of the experiment (1 to 10 weeks). Whole bladders are removed and tumor growth is measured.
  • a urinary (Foley) catheter is inserted through the urethra into the bladder and secured in place.
  • the bladder is evacuated of urine and washed with 100 to 500 mis of sterile saline.
  • Recombinant retroviruses containing the gene for thymidine kinase expression are instilled through the catheter into the bladder at 10 5 to 10 1 ' cfu in 10 to 500 ml of formulation buffer preferably containing 4 to 8 ⁇ g/ml of polybrene, or other enhancing excipients.
  • the viral particles are allowed to incubate for 0.25 to 12 hours prior to removal of the catheter. After 1 to 7 days, ganciclovir is administered at 1 to 5mg/Kg l.V.
  • the vector can be readministered multiple times (2 to 20), followed by ganciclovir administration. Due to the frequency of granulocytopenia and thrombocytopenia in patients receiving ganciclovir, it is recommended that neutrophil and platelet counts be performed every two days during the dosing of the drug. Tumor regression is monitored by x-ray and/or biopsy and the treatment repeated if required.
  • the KT-ND5 viral supernatant in formulation buffer with 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipient, is nebulized using a DeVilbiss #15 Atomizer (DeVilbiss Health Care Division, Somerset, PA) designed to produce 0.3 to 0.5 ⁇ m particles (Rousculp, 1992, Human Gene Therapy 5:471-477).
  • the aerosol produced by this nebulizer uses compressed air in a laminar flow hood. The mist is directed into a polypropylene tube, and the condensed vapor, as well as the control viral supernatant, is resterilized by 0.22 ⁇ m filtration. Retroviral particles pass through this filter without any significant loss of functional activity.
  • Rats are anesthetized and the trachea is exposed by anterior midline incision.
  • Recombinant retroviral particles expressing the factor VIII gene product are diluted in 300 ⁇ l of formulation buffer, with or without 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipient, at 10 5 to 10 10 cfu ml and instilled into the trachea though a small gauge needle.
  • Control animals are administered 300 ⁇ l of formulation buffer only. The incision is sutured and the rats are allowed to recover. Two to fourteen days following viral instillation, blood is drawn from the tail vein and examined for factor VIII production as described in Example 2Hi.
  • the recombinant retrovirus is administered using a DeVilbiss #15 Atomizer (as described above) for 2 to 60 minutes. Two to fourteen days following administration of the recombinant retoviruses, expression of Factor VIII is measured in the blood and plasma by the Coamatic R Factor VIII assay (as described in Example 2B1) . 90
  • Cottontail rabbit papillomavirus provides an animal model for the highly oncogenic human papillomavirus (HPV).
  • PapiUomas can be induced in the cottontail rabbit and virus infection leads to three different outcomes in the rabbit (Lin, Y. et el., J Virol 67:382, 1993).
  • twelve cottontail rabbits E. Johnson, Rago, KA
  • B strain of CRPV Stevens, J.
  • papillomas of 6 animals are injected with 25 to 100 ⁇ l of formulation buffer, with or without 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipient, at 10 5 to 10 10 cfu/ml though a small gauge needle.
  • control animals are administered 25 to 100 ⁇ l of formulation buffer only.
  • the rabbits are placed on twice daily (AM and PM) injections of I.P.
  • ganciclovir at the previously determined optimum dose (e.g. 62.5 mg/Kg body weight) for 4 to 12 days. Finally, the rabbits receive a single daily dose of ganciclovir until the end of the experiment ( 1 to 10 weeks). Papilloma regression is visually monitored for 2 to 14 days. 1 85
  • the clinical cutaneous lesions that result from the human papillomavirus include common warts, filiform warts, plantar warts, and anogenital warts (reviewed in Cobb et al, 1990, J. Am. Acad. Derm. 22:547).
  • patients are divided into two groups.
  • TDS transdermal delivery system
  • TDS Transdermal delivery systems
  • the major components of TDS are a controlled release device composed of polymers, the drug to be administered, excipients, and enhancers, and a fastening system to fix the device to the skin.
  • a number of polymers have been described which include gelatin, gum arabic, paraffin waxes, and cellulose acetate phthalate (Sogibayasi, K., et ⁇ /., J. Controlled Release, 29:177-185, 1994).
  • the second group receives 100 to 500 ⁇ l of vector particle in formulation buffer only. The areas are covered and the viral particles are allowed to incubate for 0.25 to 12 hours prior to removal of the TDS. After 1 to 7 days, ganciclovir is administered at 1 to
  • adenovirus isolation and purification of adenovirus is described by Green et al. (Methods in Enzymology 58: 425, 1979). Specifically, five liters of Hela cells (3-6.0 x 10 5 cells/ml) are infected with 100-500 plaque forming units (pfu) per ml of adenovirus type 2 (Ad2) virions (ATCC No. VR-846). After incubation at 37°C for 30-40 hours, the cells are placed on ice, harvested by centrifugation at 230 xg for 20 minutes at 4°C, and resuspended in Tris-HCl buffer (pH 8.1).
  • the pellets are mechanically disrupted by sonication and homogenized in trichlorotrifluoroethane prior to centrifugation at 1,000 xg for 10 min.
  • the upper aqueous layer is removed and layered over 10 mis of CsCl (1.43 g/cm 3 ) and centrifuged in a SW27 rotor for 1 hour at 20,000 ⁇ m.
  • the opalescent viral band is removed and adjusted to 1.34 g/cm 3 with CsCl and further centrifuged in a Ti 50 rotor for 16-20 hours at 30,000 ⁇ m.
  • the visible viral band in the middle of the gradient is removed and stored at 4°C until purification of adenoviral DNA.
  • the adenovirus band is incubated with protease for 1 hour at 37°C to digest proteins.
  • the particles After centrifugation at 7,800 xg for 10 minutes at 4°C, the particles are solubilized in 5% SDS at room temperature for 30 minutes before being extracted with equal volumes of phenol. The upper aqueous phase is removed, re-extracted with phenol, extracted three times with ether, and dialyzed in Tris buffer for 24 hours. The viral Ad2 DNA is precipitated in ethanol, washed in ethanol, and resuspended in Tris-EDTA buffer (pH 8.1). Approximately 0.5 mg of viral Ad2 DNA is isolated from virus produced in 1.0 L of cells. 93
  • the viral Ad2 DNA is digested with EcoR I and separated by electrophoresis on a 1% agarose gel.
  • the resulting 2.7 Kb Ad2 EcoR I D fragments, located in the Ad2 coordinate region 75.9 to 83.4, containing the E3/19K gene are eluted by electrophoresis, phenol extracted, ethanol precipitated, and dissolved in Tris-EDTA (pH 8.1).
  • E3/19K Gene into KT-3B The E3/ 19K gene is cloned into the EcoR I site of PUC 1813.
  • PUC 1813 is prepared as essentially described by Kay et al. (Nucleic Acids Research 75:2778, 1987) and Gray et al. (PNAS 50:5842, 1983).
  • the E3/19K is retrieved by EcoR 1 digestion and the isolated fragment is cloned into the EcoR I site of phosphatase-treated pSP73 plasmid. This construct is designated SP-E3/19K.
  • the orientation of the SP-E3/19K cDNA is verified by using appropriate restriction enzyme digestion and DNA sequencing.
  • the 5' end of the cDNA is adjacent to the Xho I site of the pSP73 polylinker and the 3' end adjacent to the Cla I site.
  • the Xho I-Cla I fragment containing the E3/19K cDNA in either sense or antisense orientation is retrieved from the SP-E3/19K construct and cloned into the Xho I-Cla I site of the KT-3BB retroviral. This construct is designated KT- 3B/E3/19K.
  • Ad2 DNA E3/19K gene including the amino terminal signal sequence, followed by the intraluminal domain and carboxy terminal cytoplasmic tail which allow the
  • E3/19K protein to embed itself in the endoplasmic reticulum (ER) is located between viral nucleotides 28,812 and 29,288. Isolation of the Ad2 E3/19K gene from the viral genomic
  • DNA is accomplished by PCR amplification, with the primer pair shown below: 94
  • the forward primer corresponds to the Ad2 nucleotide sequences 28,812 to 28,835. (Seq ID No. 3) 5'-TATATCTCCAGATGAGGTACATGATTTTAGGCTTG-3'
  • the reverse primer corresponds to the Ad2 nucleotide sequences 29,241 to 29,213. (Seq ID No. 4) 5'-TATATATCGATTCAAGGCATTTTCTTTTCATCAATAAAAC-3'
  • both primers contain a five nucleotide "buffer sequence" at their 5' ends for efficient enzyme digestion of the PCT amplicon products.
  • This sequence in the forward primer is followed by the Xho I recognition site and by the Cla I recognition site in the reverse primer.
  • the E3/19K gene is flanked by Xho I and Cla I recognition sites.
  • Amplification of the E3/19K gene from Ad2 DNA is accomplished with the following PCR cycle protocol: Temperature°C Time (min) No. Cycles
  • the transformed bacterial cells are plated on LB plates containing ampicillin. The plates are incubated overnight at 37°C, bacterial colonies are selected and DNA prepared from them. The DNA is digested with Xho I and Cla I. The expected endonuclease restriction cleavage fragment sizes for plasmids containing the E3/19K gene are 780 and 1,300 bp.
  • 293 2-3 cells (a cell line derived from 293 cells ATCC No. CRL 1573, (WO 92/05266) 5.0 x 10 5 cells are seeded at approximately 50%> confluence on a 6 cm tissue culture dish. The following day, the media is replaced with 4 ml fresh media 4 hours prior to transfection.
  • a standard calcium phosphate-DNA coprecipitation is performed by mixing 10.0 ⁇ g of KT-3B/E3/19K plasmid and 10.0 ⁇ g MLP G plasmid with a 2M CaCl 2 solution, adding a lx HEPES buffered saline solution, pH 6.9, and incubating for 15 minutes at room temperature.
  • the calcium phosphate-DNA coprecipitate is transferred to the 293 2-3 cells, which are then incubated overnight at 37°C, 5% CO2. The following morning, the cells are rinsed three times in lx PBS, pH 7.0. Fresh media is added to the cells, followed by overnight incubation at 37°C, 10%) CO2. The following day, the media is collected off the cells and passed through a 0.45 ⁇ filter. This supernatant is used to transduce packaging and tumor cell lines. Transient vector supernatant for other vectors are generated in a similar fashion.
  • Packaging cell line transduction is performed as described in Example 2C.
  • EBV transformed cell lines BLCL
  • other suspension cell lines are transduced by co-cultivation with irradiated producer cell line, such as DA-E3/19K.
  • irradiated (10,000 rads) producer line cells are plated at 5.0 x 10 5 cells/6 cm dish in growth media containing 4 ⁇ g/ml polybrene. After the cells have been allowed to attach for 2-24 hours, 10 6 suspension cells are added. After 2-3 days, the suspension cells are removed, pelleted by centrifugation, resuspended in growth media containing 1 mg/ml G418, and seeded in 10 wells of a round bottom 96 well plate. The cultures were expanded to 24 well plates, then to T-25 flasks.
  • Radio-immuno precipitation assay (RIP A) lysates are made from selected cultures for analysis of E3/19K expression.
  • RIPA lysates are prepared from confluent plates of cells. Specifically, the media is first aspirated off the cells. Depending upon the size of the culture plate containing the cells, a volume of 100 to 500 ml ice cold RIPA lysis buffer (10 mM Tris, pH 7.4; 1% Nonidet P40; 0.1% SDS; 150 mM NaCl) is added to the cells. Cells are removed from plates using a micropipet and the mixture is transferred to a microfuge tube. The tube is centrifuged for 5 minutes to precipitate cellular debris and the supernatant is transferred to another tube.
  • the supernatants are electrophoresed on a 10% SDS- polyacrylamide gel and the protein bands are transferred to an Immobilon membrane in CAPS buffer ( 10 mM CAPS (Aldrich, Milwaukee, WI) pH 11.0; 10% methanol) at 10 to 60 CT/US97/11785
  • CAPS buffer 10 mM CAPS (Aldrich, Milwaukee, WI) pH 11.0; 10% methanol
  • the membrane is transferred from the CAPS buffer to 5% Blotto (5% nonfat dry milk; 50 mM Tris, pH 7.4; 150 mM NaCl; 0.02% Na azide, and 0.05% Tween 20) and probed with a mouse monoclonal antibody to E3/19K (Severinsson et al, 1985, J. Cell. Biol. 707:540-547). Antibody binding to the membrane is detected by the use of ' ⁇ I-Protein A.
  • Non-Transduced Cells Decreased Levels of Class I Expression Compared to Non-Transduced Cells.
  • Cell lines transduced with the KT3b-E3/19K-vector are examined for MHC class I molecule expression by FACS analysis.
  • Non-transduced cells are also analyzed for MHC class I molecule expression and compared with E3/19K transduced cells to determine the effect of transduction on MHC class I molecule expression.
  • Murine cell lines BC10ME, BC10ME-E3/19K, P815 (ATCC No. TIB 64), and P815-E3/19K, are tested for expression of the H-2D d molecule on the cell surface.
  • Cells grown to subconfluent density are removed from culture dishes by treatment with Versene and washed two times with cold (4°C) PBS plus 1% BSA and 0.02% Na-azide (wash buffer) by centrifugation at 200g. Two million cells are placed in microfuge tubes and pelleted in a microfuge at 200g before removing the supernatant.
  • Cell pellets are resuspended with the H-2D d -specific Mab 34-2- 12s (50 ml of a 1:100 dilution of purified antibody, ATCC No. HB 87) and incubated for 30 min at 4°C with occasional mixing.
  • Antibody labeled cells are washed two times with 1 ml of wash buffer (4°C) prior to removing the supernatant.
  • Cells are resuspended with a biotinylated goat anti-mouse kappa light chain Mab (50 ml, of a 1:100 dilution of purified antibody) (Amersham, Arlington Height, IL) and incubated for 30 min at 4°C.
  • Rats are anesthetized and one eye is instilled with 5 to 100 ⁇ l of recombinant retroviral particles at a concentration of 10 5 to 10 10 cfu/ml in formulation buffer, with or without 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipient,.
  • Five to one hundred ⁇ l of solution containing formulation buffer only is added to the other eye to be used as a control.
  • the solution is allowed to incubate for 1 hour before each eye is rinsed 3 times with 100 ⁇ l of saline.
  • Two to seven days following the treatment the rat is sacrificed, the cornea is removed, and homogenized in 2 ml ice cold RIPA lysis buffer. Expression of E3/19K is detected by Western blot analysis as described in Example 6E1.
  • the cornea may be incubated for 1 hour in 1 ml of retroviral vector particles at a concentration of 10 5 to 10 10 cfu/ml in formulation buffer, with or without 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipient, just prior to surgical attachment.
  • the progress of the transplant is monitored by visually observing tissue viability.
  • the nasal route has been shown to be effective for the administration of a number of molecules due to the extensive network of capillaries located under the nasal mucosa. This facilitates effective systemic abso ⁇ tion and when the drug is administered with abso ⁇ tion promoters, abso ⁇ tion occurs rapidly with high bioavailability (review in Gizurarson et al. , 1990, Acta Pharm 2:105).
  • One group of six Fischer-344 rats are used for nasal administration of the retroviral vector particles for Factor VIII.
  • One to fifty ⁇ l of retroviral vector particles at IO 9 cfu/ml in formulation buffer, with or without 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipients are applied with a pipette inserted about 3 to 5 mm into each nostril.
  • Another group is administered formulation buffer without vector in the same manner. Blood samples are collected from the jugular or tail vein 1 to 14 days later and assayed for factor VIII production as described in Example 2Hi.
  • Two groups of patients are used in this study.
  • One group of patients receives 100 to 500 ⁇ l of retroviral vector particles at IO 9 cfu/ml in formulation buffer, with or without 4 to 8 ⁇ g/ml of polybrene or other transduction enhancing excipients, applied to each nostril via nasal spray or nasal drops.
  • Another group receives formulation buffer only applied in the same manner. Blood samples are collected 1 to 14 days later and assayed for factor VIII production as described in Example 2B1.
  • Vector pDHF828 containing the full-length human growth hormone gene is constructed essentially as follows. Briefly, plasmid pDHF81 1, was constructed by removing the Xhol- Clal fragment of the KT- 1 retroviral vector described above, and inserting the following oligonucleotide linkers by ligation of the cohesive ends:
  • the linkers were annealed at 65°C for 20 minutes, 42°C for 20 minutes, 37°C for 20 minutes, and room temperature for 2 hours.
  • the concentrations of both oligonucleotides was 18mM and the salt concentration was 100 mM NaCl.
  • 50ml of 1.8 mM annealed linker was digested with Clal overnight to generate Clal ends.
  • 3nM of KT-1 Xhol - Clal fragment was mixed with 90nM of linker, and the resultant mixture incubated at 15°C for 3 hours.
  • the ligated DNA sample was transformed into DH-5 ⁇ competent cells, followed by screening of transformants. Plasmid chGH 800 containing the full length cDNA of the hGH gene (Martial et al,
  • the pDHF828 plasmid was then introduced into the HX packaging cell, using standard procedures and assayed using the HGH Chemi luminescence Kit (HGH 100T) ( Nichols Institute, San Juan Capistrano, CA.), according to a preferred modification of the kit protocol.
  • HGH 100T HGH Chemi luminescence Kit
  • Test samples were centrifuged for 5' at top speed in a microfuge before using them in order to remove fibrin and other debris. All samples were measured in quadruplicate, including the standards.
  • the incubations are performed in 12 xl7 polypropylene tubes that have been stored in the dark.
  • HX/HGH retrovector producing cell lines were generated with titers of 4.8 x 10 cfu/ml. Introduction of the plasmid into DX packaging cells resulted in
  • Crude recombinant retrovirus is obtained from a Celligan bioreactor (New Brunswick, New Brunswick, NJ) containing DA cells transformed with the recombinant retrovirus bound to the beads of the bioreactor matrix. The cells release the recombinant retrovirus into the growth media that is passed over the cells in a continuous flow process.
  • the media exiting the bioreactor is collected and passed initially through a 0.8 micron filter then through a 0.65 micron filter to clarify the crude recombinant retrovirus.
  • the filtrate is concentrated utilizing a cross flow concentrating system (Filtron, Boston, MA).
  • DNase Intergen, New York, NY
  • concentrate Approximately 50 Units of DNase (Intergen, New York, NY) per ml of concentrate is added to digest exogenous DNA.
  • the digest is diafiltrated using the same cross flow system to 150 mM NaCl, 25 mM tromethamine, pH 7.2.
  • the diaf ⁇ ltrate is loaded onto a
  • the formulation buffer containing lactose was prepared at a 2X concentrated stock solution.
  • the formulation buffer contains 25 mM tromethamine, 70 mM NaCl, 2 mg/ml arginine, 10 mg/ml HSA, and 100 mg/ml lactose in a final volume of 100 mis at a pH 7.4.
  • the purified recombinant retrovirus is formulated by adding one part 2X lactose formulation buffer to one part S-500 purified recombinant retrovirus.
  • the formulated recombinant retrovirus can be stored at -70°C to -80°C or dried.
  • the formulated retrovirus is lyophilized in an Edwards Refrigerated Chamber
  • the lyophilized recombinant retrovirus is reconstituted with 1.0 ml water.
  • the infectivity of the reconstituted recombinant retrovirus is determined by a titer activity assay.
  • the assay is conducted on HT 1080 fibroblasts or 3T3 mouse fibroblast cell line (ATCC No. CCL 163). Specifically, 1 x 10 5 cells are plated onto 6 cm plates and incubated overnight at 37°C, 10%> CO2. Ten microliters of a dilution series of reconstituted recombinant retroviruses are added to the cells in the presence of 4 mg/mL polybrene (Sigma, St. Louis, MO) and incubated overnight at 37°C, 10%. CO2. Following incubation, cells are selected for neomycin resistance in G418 containing media and incubated for
  • the recombinant retrovirus utilized in this example was purified as described in Example 9A.
  • the formulation buffer containing mannitol was prepared as a 2X concentrated stock solution.
  • the formulation buffer contains 25 mM tromethamine, 35 mM NaCl, 2 mg/ml arginine, 10 mg/ml HSA and 80 mg/ml mannitol at a final volume of 100 mis at a pH 7.4.
  • the purified recombinant retrovirus is formulated by adding one part mannitol formulation buffer to one part S-500 purified recombinant retrovirus.
  • the formulated recombinant retrovirus can be stored at this stage at -70°C to -80°C or dried.
  • the formulated retrovirus is dried in an Edwards Refrigerated Chamber (3 Shelf RC3S unit) attached to a Supermodulyo 12K freeze dryer.
  • the vials are stoppered under a vacuum following nitrogen gas bleeding to 700 mbar. Upon removal, vials are crimped with aluminum seals.
  • formulated liquid product was stored at both -80°C and at -20°C in cycling freezers.
  • the viral infectivity of these samples were compared to the viral infectivity of lyophilized samples, Figure 14.
  • the lyophilized samples were stored at - 20°C, refrigerator temperature and room temperature. Activity of the samples upon reconstitution are determined using the titer assay described in Example 9A.
  • Figure 14 demonstrates that storage in lyophilized form at -20°C to refrigerator temperature retains significant viral activity as compared to recombinant retrovirus stored in liquid at -80°C or -20°C, permitting less stringent temperature control during storage.
  • the recombinant retrovirus utilized in this example was purified as described in Example 9A.
  • the formulation buffer containing trehalose was prepared as a 2X concentrated stock solution.
  • the formulation buffer contains 25 mM tromethamine, 70 mM NaCl, 2.0 mg/ml arginine, 10.0 mg/ml HSA and 100 mg/ml trehalose at a final volume of 100 mis at a pH 7.2.
  • the purified recombinant retrovirus is formulated by adding one part trehalose formulation buffer to one part S-500 purified recombinant retrovirus.
  • the formulated recombinant retrovirus can be stored at this stage at -70°C to -80°C or dried.
  • the formulated retrovirus is dried in an Edwards Refrigerated Chamber (3 Shelf RC3S unit) attached to a Supermodulyo 12K freeze dryer.
  • the vials are stoppered under a vacuum following nitrogen gas bleeding to 700 mbar. Upon removal, vials are crimped with aluminum seals.
  • formulated liquid product was stored at both -80°C and at -20°C in cycling freezers.
  • the viral infectivity of these samples was compared to the viral infectivity of lyophilized samples, Figure 15.
  • the lyophilized samples were stored at - 20°C, refrigerator temperature and room temperature. Activity of the samples upon reconstitution are determined using the titer assay as described in Example 9A.
  • Figure 15 demonstrates that storage in lyophilized form at -20°C to refrigerator temperature retains similar viral activity as compared to recombinant retrovirus stored in liquid at -80°C to -20°C permitting less stringent temperature control during storage.
  • Viral infectivity of liquid formulated recombinant retrovirus samples stored at -80°C was compared to viral infectivity of lyophilized formulated recombinant retrovirus stored at -20°C. Initially, a bulk of recombinant retrovirus was received and formulated in four different ways as shown below. The formulated recombinant retrovirus was then frozen in bulk for 1.5 months subsequent to being quick thawed and freeze dried. Positive controls were stored at -80°C for comparison with lyophilized samples which were stored at -20°C after freeze-drying. The formulations are listed below:
  • the y-axis on each of the 4 graphs (A, B, C, D) represent the normalized titer.
  • t 0, a titer value was established for both the -80°C liquid sample and the -20°C lyophilized sample.
  • the titer obtained was divided by the zero time point titer value and the % of original entered onto the graph.
  • Virus bands may be identified by their relative molecular weight and by reverse transcriptase activity (RT).
  • RT reverse transcriptase activity
  • the pu ⁇ ose of this assay is to quantify the activity of reverse transcriptase (RT), an enzyme exclusively associated with all retroviruses. The relative amount of retrovirus in a sample can be determined by measuring the activity of this enzyme in a given preparation.
  • moloney murine leukemia virus reverse transcriptase (Pharmacia, Newark,
  • Thymidine 5' - Triphosphate (30-50 Ci/mmol). This mixture is incubated for 1 hour at 37°C in a water bath. Following incubation the sample is placed on ice. Approximately 1.0 ml of 2N HCl is added to the cooled sample. The precipitated radiolabled DNA fragments are vacuum filtered onto glass fiber filters using a Millipore sampling manifold (Millipore, Philadelphia, PA). The filters are washed, dried, placed in scintillation cocktail, and counted in a Beckman LS5000TD scintillation counter (Beckman, Dallas, TX). EXAMPLE 11
  • D Dl 7 dog cells
  • 2 human embryonic kidney cell line 293
  • all packaging cells were grown in DMEM media (Irvine Scientific, Santa Ana, CA) supplemented with 10% heat inactivated fetal bovine serum (Hyclone, Salt Lake City, UT).
  • Each of the four packaging cell lines was transduced with a G-pseudotyped pCB ⁇ - gal (see Burns et al., 1993, Proc. Nat'l. Acad. Sci. USA 90:8033-8037, for a pseudotyping procedure).
  • the selected cell pools were dilution cloned (except for the amphotropic vector producing human cell line which was maintained as a non-clonal pool).
  • Vector producing cell lines producing the highest titer on HT1080 cells (ATCC No. CCL 121) were selected. Titers were determined approximately 2 days after transduction by G418 colony forming units or by X-gal staining of the monolayers.
  • Retrovirus containing supernatants were collected from each of the confluent monolayers of the individual amphotropic envelope (DA and 2A) or xenotropic envelope (DX or 2X) vector producing cell lines and filtered through a 0.45 ⁇ m filter prior to aliquoting and storage at -70°C.
  • DA and 2A amphotropic envelope
  • DX or 2X xenotropic envelope
  • Serum inactivation titer assays were performed as follows: Serum was drawn from at least two different human volunteers, chimpanzees, baboons, and macaque monkeys. Approximately 20-70 mL of blood was collected from each donor into serum separating tubes (Becton Dickinson, Los Angeles, CA). Blood was allowed to clot for 20-30 minutes at room temperature, after which time the samples were centrifuged at 2000 x g for 10 minutes at 4°C. Serum was frozen in approximately 1.1 mL aliquots and stored at -80°C.
  • HFB hollow fiber bioreactor
  • the growth media should be what ever the cell line has been adapted to. All liquids in the HFB when originally shipped should be aspirated and replaced with the complete growth media.
  • the cells When seeding the bioreactor, the cells should not have been split more than 48 hours earlier and should be in log growth phase at the time of harvest for the seeding of the HFB. The cells are harvested by trypsinazation and pelleted by centrifugation.
  • the cell pellet is then resuspended in 4 mL of 25% pre-conditioned media and delivered to the extra-capillary space by syringe using the side syringe ports found on the HFB. After seeding the HFB, allow the cells to adhere for 20 to 30 minutes before starting the circulation pump. During this time replace the media used to condition the HFB with 100-200 ml using 25% pre-conditioned media.
  • the circulation feed pump is initiated with the starting flow rate set at 25 mL/min. (setting 5 with 2 long pump pins). After 1 hour from the time of switching the pump on, a one mL sample of media is collected in order to record the initial levels of lactate and ammonia.
  • the original Cellco supplied reservoir feeding cap is exchanged for the larger reservoir cap (Unisyn-vender part #240820) adapted for the Cellco system with the addition of tubing and male luer lock fittings.
  • This reservoir cap will accommodate the large 2 Liter Corning bottles. (To avoid the exchange of the reservoir caps during a culture run, initiate the culture run with the larger reservoir cap which can also support smaller bottle sizes.)
  • daily lactate readings are assayed and recorded, one can calculate the daily levels of lactate production of the culture in order to determine when the culture reaches maximum cell density when the rate of lactate decreases and levels off.
  • Figure 20 is a representative graph of data generated over a two week period of the vector producer cell line 2X-B-GAL 17-14.
  • one HFB was seeded with 1.3 x IO 7 cells (to represent the low seed culture) the other seeded with 1.6 x IO 8 cells.
  • the cell line (2x- ⁇ - GAL 1 7.14) was able to initiate a good hollow fiber run under low and high seeding conditions. Being able to incubate the HFB with fewer cells, is only convenient for reducing the effort required for generating the number of cells required to start a culture. However a low seed start also extends the time it takes to reach optimal cell densities which usually yield the highest titers.
  • the cell line used adapted very well to hollow fiber cultures which eventually required daily media changes of 500 ml per day.
  • Figure 20-B demonstrates lactate concentrations of a culture which required daily 500 mL exchanges of fresh media after reaching day 7 of culture.
  • Figure 20-D is one indication of the health of the culture by tracking the amount of lactate being produced on a daily basis.
  • the transduced cells were stained for ⁇ GAL activity and individual cells counted on a hemocytometer giving a titer based on the number of blue cells /mL (BCT/mL).
  • BCT/mL blue cells /mL
  • optimum titers were obtained on day 7 of the high seed culture at 1.8 xlO 8 BCT/mL from a 72 hour blue cell titer on 293 cells.
  • the bottom layer (approximately 20 mL) is carefully eluted, and the inte ⁇ hase (approximately 1 mL) is collected in a 15 mL conical FALCON tube.
  • the inte ⁇ hase containing vector is diluted to 10 mL by addition of PBS, and incubated at 37°C in order to bring the solution to room temperature and destabilize the micelles.
  • KC1 is added to a final concentration 0.4 M, and mixed well. The tube is then placed on ice for ten minutes, and spun for 2 minutes at 2,000 ⁇ m in a bench-top centrifuge. The supernatant is removed and filtered through a 0.45 um syringe filter.
  • the other half of the inte ⁇ hase containing vector is separated by S-500 Sephadex chromatography in IX PBS.
  • the vector- containing solution may be further subjected to concentration utilizing an MY-membrane Amicon filter, thereby reducing the volume from 10 to 14 mL, down to less than 1 mL.
  • DX/ND7 ⁇ gal Clone 87 Utilizing a Cell Factory DX/ND7 ⁇ gal clone 87, an expression vector, was grown in cell factories. Cells were grown in DMEM supplemented with Fetal Bovine Serum in roller bottles until enough cells to seed 20 10-layer cell factories (NUNC) at a 1 :3 dilution were obtained. Each 10- layer cell factory is seeded with approximately 0.8 liters of cell medium.
  • NUNC 10-layer cell factories
  • Cells were seeded into the cell factory by pouring media containing cells into the factory so that the suspensions evenly fill the 10 layers. The factory is then carefully tilted away from the port side to prevent the suspension from redistribution in the common tube. Finally, the cell factory is rotated into its final upright position. A hepavent filter is attached to each port. The factory was then placed in a CO2 incubator.
  • Verification of the vector was performed by transduction of HT1080 cells. These cells were harvested 2 days later and stained for ⁇ -gal protein. The titer of the supernatant was determined to be 2 x 10 7 /ml.
  • Producer cell lines DA ⁇ gal and HX/DN-7 were cultured in a culture flask and a roller bottle, respectively, containing Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% fetal bovine serum plus 1 mM L-Glutamine, Sodium pyruvate, non-essential amino acids and antibiotics. Viral supernatant was harvested from the flask and roller bottle, and were filtered through a 0.45 um syringe filter. The filtered supernatants were stored either at 4°C (HX/ND7), or frozen at -70°C (DA ⁇ -gal).
  • DMEM Dulbecco's Modified Eagle's Medium
  • Viral supernatant was aliquoted into 50 ml sterile OAKRIDGE screw cap tubes, and placed into an SS34 rotor for use in a Sorvall centrifuge. The tubes were spun for 1 hour at 16,000 ⁇ m (25,000g-force) at 4°C. Upon completion of the spin, the tubes were removed, the supernatant decanted and a small opaque pellet resuspended in the DMEM media described above.
  • Virus Concentrated virus was titered on HT1080 cells plated 24 hours earlier at a cell density of 2x10 ⁇ cells per well in a six well plate + 4 ug/ml polybrene. Briefly, virus preps were diluted from 1/10 to 1/10,000 and 50 ul of each dilution was used to infect one well from the six well plate. Plates were incubated overnight at 37°C. Forty-eight hours later, cells were fixed and stained with X-gal. The results are set forth below in Table 1. Table 1. Virus Concentration through Low Speed Centrifugation
  • virus recovery ranged from 30% to 99%o, with the best recovery being obtained from human producer cells (HX/ND7; recovery ranged from 91% to 99%).
  • EXAMPLE 16 Concentration of Recombinant Retroviruses by Ultrafiltration S-500 purified supernatant containing the ⁇ -gal expressing recombinant retrovirus DX CB- ⁇ gal and partially concentrated supernatant containing the same virus were each filtered through a 0.45 um filter, and loaded into a CENTRIPREP- 100 filter (product #4308, Amicon, MA). The supernatants were kept at a temperature of 4 °C throughout this procedure, including during centrifugation. The CENTRIPREP filters were spun three times each for 45 to 60 minutes at 500 x G. Between each spin the filtrate was decanted. The retentate was thus sequentially reduced, such that the initial 15 ml (or 10 ml) volume was reduced to approximately 0.6 mL per unit.
  • the resultant titer was determined by assaying HT1080 target cells set up at a concentration of 1 x 10 5 cells per well 24 hours prior to transduction of the viral sample. Cells were transduced in the presence of 8 ug/ml polybrene and 2 mL growth media (DMEM plus 10% FBS) per well. As shown in Table 1 below, approximately one hundred percent of the virus was recovered utilizing this procedure (note that titers are in BCFU/ml).
  • Producer cells are frozen in DMEM media containing 10% to 20% FBS, and 5 to 15% DMSO, at a concentration of 1 x 10 7 cells/ml/vial. Cells are frozen in a controlled rate freezer (Series PC, Controlled Rate Freezing System, Custom Biogenic Systems, Warren MI) at a rate of from 1 to 10 °C per minute. Frozen cells are stored in liquid nitrogen.
  • DMEM media containing 10% to 20% FBS, and 5 to 15% DMSO, at a concentration of 1 x 10 7 cells/ml/vial.
  • Cells are frozen in a controlled rate freezer (Series PC, Controlled Rate Freezing System, Custom Biogenic Systems, Warren MI) at a rate of from 1 to 10 °C per minute. Frozen cells are stored in liquid nitrogen.
  • Expanded cell culture is used to inoculate a "CELLIGEN PLUS" bioreactor (5 liter working volume; New Brunswick, Edison, N.J.).
  • the cells are grown on microcarriers (i.e., Cytodex 1 or Cytodex 2; Pharmacia, Piscataway, N.J.) at a concentration of 3 to 15 g/L microcarrier.
  • Initial inoculation densities are from 4 to 9 cells/bead at half to full volume for 2 to 24 hours.
  • the media constituents for vims production are DMEM-high glucose (Irvine
  • Glucose ranges from 6 to 0.25 g/L, Lactate from 1 to 25 mM, and Ammonia ranges from 0.5 to 30 mM. Cells are incubated in the bioreactor for 5 to 25 days.
  • abso ⁇ tion plate 2 was placed on abso ⁇ tion plate 2, covered with parafilm, and incubated at room temperature shaking at 80 ⁇ m.
  • 300 ul donkey anti-sheep IgG conjugated to alkaline phosphatase was absorbed on abso ⁇ tion plate 3 at 1 :30,000 in DB2, covered and incubated at room temperature shaking at 80 ⁇ m.
  • plate 1 was washed and 200 ul of absorbed sheep anti-VIII:C was transferred from plate 2 to plate 1.
  • Plate 1 was covered with parafilm and incubated at room temperature for 1-2 hrs, shaking at 80 ⁇ m.
  • plate 1 was washed and 200 ul absorbed donkey anti-sheep conjugate was transferred from plate 3 to plate 1. Plate 1 was covered and incubated 1 hr at room temperature shaking at 80 ⁇ m.
  • Readings were compared to a standard curve using duplicate dilutions of NHP (pooled normal plasma, George King, Overland Park, Kansas, USA) as a factor VIII source diluted in 1 :3 rabbit plasma at 3-fold dilutions starting at 1 : 10 NHP.
  • NHP pooled normal plasma, George King, Overland Park, Kansas, USA
  • Figure 23 shows the data in this experiment extended through Day 429.
  • the two high-expressing rabbits described above maintain their expression levels through day 429.
  • Medium-level expressing rabbits do as well, while the control rabbits remain negative.
  • expression is observed at least from Day 56-429, indicating >373 days of expression from a single course of vector administration.
  • Retroviral Vector Human growth hormone-expressing retroviral vectors were produced as described in Example 8 herein. Large scale production in Cell Factories was performed as described for DX/ND7 in Example 14 herein.
  • the retroviral vector was lactose formulated as described in Example 9 herein.
  • the titer was 8E8.
  • Vector was diluted 5-fold in formulation buffer, and 1 ml injections were given to 500 g juvenile rabbits 3 time per day for three days as described in Example 18 for factor VIII vector. Serum was collected on days 0 (prebleed), 7, 14, and every 7 days thereafter. Serum was analyzed for human growth hormone content using a commercial ELISA kit ( Boehringer Mannheim Biochemicals, Indianapolis, Indiana, USA) according to the directions of the manufacturer.
  • Retroviral vectors Human growth hormone-expressing retroviral vectors were produced as described in
  • Example 8 herein.
  • Large scale production in Cell Factories was performed as described for DX/ND7 in Example 14, herein.
  • the retroviral vector was then lactose formulated as described in Example 9, herein.
  • the titer was 8E8.
  • Three week old mice were given a single injection of 200 ul vector via the tail vein.
  • Human growth hormone was analyzed in mouse serum using a commercial ELISA kit ( Boehringer Mannheim Biochemicals) according to the instructions of the manufacturer. The results are shown in Figure 25. Due to the low serum volumes that could be obtained from young mice relative to the volumes required for ELISA analysis, pooled test mice were tested on Day 14, and therefore the results shown on this date represent the average value for the indicated animals.
  • mice All three pools (two test, on control) yielded values of ⁇ 50 pg/ml on Day 14. By Day 28, animals had grown sufficiently that individual animals could be tested. Animals 2 and 3 displayed hGH levels of approximately 300 pg/ml on Day 28; animal 9 showed a level; of 200 pg/ml. All other mice were ⁇ 100 pg/ml. The two controls, Animals #11 and 12, were 0 throughout the test period. By day 48, all 10 test animals displayed increasing levels of hGH, and all were clearly above 100 pg/ml, with animals #1, 2, 3, 4, 5, and 10 showing levels of 300-600 pg/ml. Therefore, mice injected a single time with high-titer formulated retroviral vector expressed human growth hormone at high levels for at least 20 days following a lag phase of 14-48 days.
  • Human growth hormone-expressing retroviral vectors were produced as described in Example 8 herein. Large scale production was carried out in Cell Factories as described for DX/ND7 in Example 14 herein. The retrovial vectors were then lactose formulated as described in Example 9, herein. The titer was 8E8 Adult mice were given a single injection of 100 ⁇ l vector via the tail vein. One group of mice also received 600 ⁇ g cyclosporin A once daily i.m. starting 1 day prior to injection of vector and continuing through Day 13. Human growth hormone was analyzed in mouse serum using a commercial ELISA kit (Boehringer Mannheim Biochemicals) according to the instructions of the manufacturer. The results are shown in Figure 26.
  • “Positive control” refers to mice that received no cyclosporin. High level (400-1200 pg/ml) human growth hormone was observed on Day 3, declining to zero by Day 14. Negative controls (formulation buffer-injected) expressed no human growth hormone. The injection of cyclosporin A using the above schedule neither augmented nor diminished the growth hormone response. Therefore, intravenous injection of high titer formulated retroviral vectors induces high level expression of growth hormone in adult animals with no lag phase.
  • Example 22 Use of Peroxisome Proliferators to Induce Liver Mitosis and Retroviral Transduction
  • Balb/C mice were treated IP (500 ⁇ l) or via gavage (200 ⁇ l) with the peroxisome proliferator WY 14643 (Chem Syn, Lenexa, Kansas, USA) (15 mg/ml) once per day for 4 days.
  • WY 14643 Chem Syn, Lenexa, Kansas, USA
  • 200 ul beta galactosidase retrovector (1E9 cfu/ml, DA-Bgal) was injected via tail vein.
  • the mice were sacrificed, tissues were fixed in 2%> formaldehyde/PBS for 24 hr, and blocked into slices of 2-5 mm.
  • Sections were rinsed in PBS and stained in fresh XGAL solution (5 mM potassium ferricyanide/5 mM potassium ferrocyanide/ 2 mM MgC12/ 0.5mg/ml XGAL in DMF (Gold Scientific)/ IX PBS). Tissue was embedded in paraffin (-50 degrees), sectioned at 5 microns (PML, San Diego), and counterstained with Hematoxylin/Eosin.
  • Sections were compared from both periportal and peripheral regions from normal liver ( XGAL stain only), control liver (no WY 16463, with vector), WY control liver ( WY 16463 only, no vector), liver from group I (IP WY 16463 plus vector), liver from group II ( gavage WY 16463 plus vector), and spleen from Group I. Numerous blue cells were observed in both treatment groups, both periportally and on the liver periphery. Very rare blue cells were observed in the control liver (without mitogen). No blue cells were seen in the normal liver (no mitogen or vector) or the WY liver (mitogen, no vector).
  • PCR was performed on each sample (0.5 uM primer LTR 492S (CCC TGT GCC TTA TTT GAA CTA ACC) (Seq ID No. 12), 0.55 uM primer LTR 1072AS (CCC ACC ACA ACC ACA TAT CCC TCC) (Seq ID No.13), IX PCR Buffer, 2.5 mM MgC12, 200 uM dNTPs, 2.5 U Taq in a total volume of 50 ul (Perkin-Elmer)). The reactions are incubated 10 min at 94o, then subjected to 32 cycles of 94ox 30 sec, 68o x 30 sec, 72o x 30 sec.
  • the amplicon consisted of a 580 bp region spanning the LTR/Psi region of the vector backbone (see Example 1). Each sample was subjected to four replicate reactions. Two other replicate reactions were spiked with 5 copies of positive control DNA (DA6A3 HBV-IT VCL genomic DNA; see Townsend et al, 1997, J. Virol. 77:3365, to control for interference with PCR amplification. Ten microliters of each amplification reaction were dot-blotted onto a nylon membrane (Zeta Probe).
  • the membrane was prehybridized in buffer (6X SSPE, 30% formamide, 5X Denhardt's Solution, 0.5% SDS, 1.5 mg/ml herring sperm DNA) at 42o for 60 min, the hybridized at 42o for 4 hr in the same buffer to a 32P-end labeled probe (LTR-P - CCA GTC CTC CGA TTG ACT G [Seq ID No. 14]) corresponding to a sequence internal to the LTR-Psi amplicon.
  • the blot was washed (at room temperature in 2x SSPE/1% SDS, then 3x 10 min at 42o in 2x SSPE/1% SDS) and exposed to film.
  • the film was inspected for number of positive blots per total number of reactions.
  • Tissue samples from Rabbit #82, which expressed high levels of human factor VIII protein, and from Rabbit #95, a control animal that had received no vector, are shown in Figure 27.
  • the spiked reactions from all tissues were positive, indicating that negatives are not due to PCR inhibiting substances from the tested tissues.
  • Liver samples from 10 locations, a spleen sample, and samples from two brains locations were all negative in the control rabbit.
  • the brain sample was negative in the expressing rabbit, while spleen and all 10 liver samples were positive in the expressing rabbit.
  • the results suggest that liver and spleen have been transduced in the vector-treated rabbit, that marked cells are still present after 511 days, and that these are candidate organs for the source of the circulating factor VIII.
  • Juvenile mice were treated with DA/beta-gal vector as in Example 20 herein. Expressing mice were sacrificed at Day 59 or 105. Brain, liver, kidney, lung, and spleen were analyzed for vector sequences by quantitative PCR. Quantitative PCR is performed as described above, except that 1 ⁇ l of alpha-32P-dCTP is included in the PCR buffer, and a standard curve is prepared from the same positive control DNA in 3 -fold dilutions from 1000 copies/sample down to 1.4 copies/sample, maintaining a constant concentration of 500 ng DNA/reaction by adding human PBMC DNA to each dilution.
  • liver and spleen are the major sites of transduction and synthesis of genes of interest when retroviral vectors are introduced intravenously.
  • Recombinant retroviral vector expressing B domain deleted factor VIII was constructed, packaged, and expressed as described in Example 2 herein. Large scale production was carried out in Cell Factories as described for DX/ND7 in Example 14 herein. The retroviral vector was then lactose formulated as described in Example 9 herein. High titer formulated retroviral vector was injected into 8 week old normal juvenile dogs according to the following schedule: 4E8 vector particles were injected into the cephalic vein at 2.5 hr intervals, three times per day for three days (Day 0, 1 , and 2).
  • citrated plasma was obtained by venipuncture of the cephalic vein and analyzed for human factor VIII antigen as in Example 18, except that the diluent was 1 :3 citrated dog plasma (Harlan).
  • nonresponder displayed 0-8 ng/ml human factor VIII antigen in their plasmas.
  • Experimental Dog 55 (“responder”) displayed levels of 40-110 ng/ml factor VIII.
  • Levels of factor VIII in Dog 55 peaked around Day 160, gradually declining until Day 279, the last date tested.
  • the nonresponder dog #47 was boosted with dosage schedule recapitulating that used on day 0,1 , and 2.
  • dog 47 displayed 95 ng/ml, comparable to the 77 ng/ml found in the unboosted responder dog 55 on that date.
  • boosted Dog 47 did not maintain these levels, which declined to barely detectable levels by Day 125.
  • Example 25 Transduction of Splenocytes and Possible Stem Cell Transduction bv Intravenous
  • Retroviral Vector Data in Example 24 herein show transduction of spleen and liver in long-term expressing animals treated with retroviral vector intravenously.
  • an adoptive transfer experiment was carried out. Fragments of spleens (1/4 to 1/3 spleen) were collected from BALB/c mice which had received 1E7 cfu DA-827 51 days earlier via tail vein injection as described in Example 20 herein. Spleens were processed individually. Single-cell suspensions were prepared in sterile Hank's balanced salt solution (HBSS; Irvine Scientific) by passing tissue fragments through a 70 mm nylon mesh strainer (Becton Dickenson #2350).
  • HBSS Hank's balanced salt solution
  • mice After pelleting, cells from each spleen were resuspended in 0.8 ml HBSS; 0.3 ml of this suspension was injected intravenously into lethally irradiated (900 R) recipient BALB/c mice. An estimated 6 - 7 x 10 6 cells were transferred to each recipient. Serum levels of hGH were monitored every two weeks in recipients as in Example 20. levels of hGH expression in recipients roughly corresponded to the levels that had been present in the original donors (See Figure 31), suggesting that these levels correlated with transduced cell number. Peak expression was observed 56 days following spleen cell transfer, with a decrease by day 70. The increase of expression with time as the animals reconstituted indicates that stem cells have been transduced.
  • Example 26 Expression of Human Factor VIII in Hemophiliac Dogs Following Intravenous Expression of Vector
  • Two factor Vlll-deficient hemophiliac dogs (8 weeks old) were obtained from the closed colony at U. North Carolina, Chapel Hill. Two normal littermates were also obtained and used without further treatment to provide baseline values for assays.
  • the two test dogs were injected with retroviral vector as in Example 24, herein, except that the injection dates were Days 1,2, and 3. Blood samples were obtained on Days 4,7,10, 14, and every 7 days subsequently. Citrated samples were used for measurements of whole blood clotting time (See Figure 32). On day -4, samples from the hemophiliac dogs were incoagulable (WBCT > 60 min), whereas the normal littermates had WBCT of 6-8 minutes.
  • the WBCT of the normal littermates were maintained throughout the experiment.
  • the WBCT of the injected hemophiliac dogs dropped to 12-16 minutes. This represents expression of pg levels of factor VIII ( Dr. Tim Nichols, UNC-Chapel Hill, personal communication).
  • the hemophiliac dog WBCT was again incoagulable (>50 min). Starting at day 35, the clotting time was again reduced, reaching 17 minutes for one dog and 16 minutes for the second, indicating a second peak of factor VIII clotting activity.
  • This data demonstrates the successful expression of biologically active factor VIII in dogs by intravenous administration of a retroviral vector expressing a B-domain deleted factor VIII protein.
  • pKT-1 is described in Example 1 herein and is shown schematically in Figure 33.
  • the construction of the pKT-1 starting material used in the present example is further described in detail in PCT WO 95/30763 and in co-owned U.S. Serial No. 08/721,327, both of which are hereby inco ⁇ orated by reference in their entirity.
  • two stop codons were introduced in the DNA sequence of the packaging signal sequence in order to increase the safety of these vectors. All modifications are summarized in Fig.
  • pBA-5a 36 and the resulting retroviral backbones are called pBA-5a, pBA-5b, and pBA-5c. Further details on the contruction of pBA-5a, pBA-5b, and pBA-5c are provided in co-owned U.S. Serial No. 08/721,327 and co-owned application, attorney docket 1147.004, filed May 5, 1997, entitled "Crossless Retroviral Vectors" and which is also hereby inco ⁇ orated by reference.
  • This example describes modification of the extended packaging signal ( ⁇ +) by PCR on the template KT-1 using primers that introduce two stop codons in the extended packaging signal sequence.
  • the template pKT-1 contains the modification ATT at the normal ATG start site of gag (position 621-623 of SEQ ID NO: 15).
  • the start site was further modified to the stop codon, TAA, and an additional stop codon TGA was added to replace the codon TTA at position 645-647 of SEQ ID NO: 15.
  • two sets of PCR reactions were carried out on pKTl , each introducing one stop codon.
  • the primers for the PCR were designed such that the two PCR products had overlapping regions and a splice-overlap extension PCR (SOE-PCR) was carried out with the two PCR products in order to combine the two introduced stop codons on one strand.
  • SOE-PCR splice-overlap extension PCR
  • the first set of oligonucleotides introducing the change from ATT to TAA were 5'-GGG- AGT-GGT-AAC-AGT-CTG-GCC-TTA-ATT-CTC-AG (SEQ ID NO: 16) and 5'-CGG- TCG-ACC-TCG-AGA-ATT-AAT-TC (SEQ ID NO: 17) and the second set of oligonucleotides introducing the change from TTA to TGA were 5'CTG-GGA-GAC-GTC- CCA-GGG-ACT-TC (SEQ ID NO: 18) and 5 * GGC-CAG-ACT-GTT-ACC-ACT-CCC- TGA-AGT-TTG-AC (SEQ ID NO: 19).
  • the flanking primers of the final 708 base pair PCR product introduced the Aat l and the Xhol sites, at the 5' and 3', respectively.
  • the ends of the 708 base pair product were blunted and phosphorylated and the product introduced into the S ⁇ l and EcoRV digested vector pBluescript SK- (Stratagene, San Diego, Calif).
  • the resulting plasmid was designated pBA-2, and is shown diagramatically in Figure 34.
  • Retroviral envelope sequence was removed upstream of the 3' LTR between the Clal site and the TAG stop codon of the envelope coding sequence.
  • the DNA sequence was modified by PCR such that the TAG stop codon was replaced by a Clal site and the 97 nucleotides upstream from this new Clal site to the original Clal site were deleted, as well as the 212 base pairs of retroviral sequence downstream of the 3' LTR.
  • PCR product was cloned into pPCRII (Invitrogen, San Diego, Calif.) using the TA cloning kit (Invitrogen, San Diego, Calif.) and called pBA-1.
  • pBA-2 (described in section A above) was digested with-Y ⁇ l and Aatll which deleted a part of the multiple cloning site and into this linearized vector the 780 base pair fragment from N ⁇ el to Aatll from pKTl was cloned, resulting in the plasmid pBA- 3.
  • This plasmid pBA-3 combined the shortened 5' LTR with the above described packaging region including the two introduced stop codons.
  • ⁇ BA-1 was digested with Clal and Apal resulting in a 640 base pair fragment that was cloned into the Clal and Apal digested pB A-3 resulting in the plasmid pBA-4.
  • This plasmid combines the above described 5 * LTR and the packaging signal with the 3' LTR.
  • pBA-4 was digested with p ⁇ l and EcoRI, ends blunted and religated in order to remove extraneous 3' polylinker sites, resulting in plasmid pBA-5a.
  • pBA-5a was cut with ⁇ otI (blunted) and EcoRI and introduced into Smal and EcoRI digested pUC 18 (GIBCO/BRL, Gaithersburg, MD) resulting in pB A-5b.
  • This construct moved the retroviral vector from a pBluescript into an alternate pUC18 vector.
  • pBA-5c is constructed as identical to pUC-18 derived pBA-5b except for the Xhol/Clal multicloning site was introduced into pUC-19.
  • This example further describes several modifications of the retroviral vector pBA-5b which result in a vector with multiple unique restriction enzyme sites for convenient cloning.
  • the he ⁇ es simplex virus thymidine kinase (HSVTK) gene was retrieved by digesting pBH-1 with Xhol and ⁇ coRI resluting in a 1.2kb fragment.
  • pBH-1 was prepared as described in WO 91/02805 entitled "Recombinant Retrovimses Delivering Vector Constmcts to Target Cells" which is hereby inco ⁇ orated by reference.
  • the neomycin gene driven by the SV40 promoter was retrieved by digesting pKT-1 with ⁇ coRI and BstBI resulting in a 1.3 kb fragment. Both fragments were cloned into a Xhol and Cla digested pBA-5b resulting in the retroviral vector pMO-TK.
  • the TK gene from the retroviral vector pMO-TK was isolated as a Xhol-Clal fragment and inserted into the Xhol-Clal digested pBA-5b, resulting in the plasmid pBA- 5bTK.
  • pBA-5bTK was digested with Hindlll and Hindi, overhanging ends removed using T4 polymerase and blunt ends ligated unsing the T4 DNA ligase, resulting in plasmid pTJBA-5bTK with 16 bases (TGC ATG CCT GCA GGT C) removed from upstream of the 5 'LTR.
  • the plasmid pTJBA-5bTK has a BamHI upstream of the
  • 5'LTR 5'LTR. It is desirable to remove this BamHI site since it is a common site used for cloning.
  • the BamHI-containing TK gene in pTJBA-5bTK was replaced by the IL-2 gene via Xhol-Clal digest, resulting in plasmid pTJBA-5bIL-2.
  • the plasmid pTJBA-5bIL-2 was digested with BamHI, the ends filled in with the Klenow fragment and the ends religated, resulting in pTJBA-5bIL-2 (BamHI del.).
  • the IL-2 gene from pTJBA-5bIL-2 (BamHI del.) is deleted via Xhol-Clal digest and replaced with an linker that introduces a multiple cloning site (MCS) and codes for the restriction enzyme sites 5' -Xhol-Apal-Bglll-Notl- NruI-Sall-Hindlll-BamHI-Clal- 3'.
  • MCS multiple cloning site
  • the sequence of the two primers that is used to produce the linker are as follows: 5 ' - TCG AGG GGC CCA GAT CTG CGG CCG CTC GCG AGT CGA CAA GCT TGG ATC CAT - 3' (Seq ID No.
  • This example describes several modifications of the retroviral vector pBA-5b which result in a vector coding for the human placental alkaline phosphatase gene (PLAP), driven by the SV40 promoter.
  • PLAP human placental alkaline phosphatase gene
  • the plasmid pBA-8bLl was constructed in a three-way ligation using the following three fragments: The Ndel-Clal fragment from pBA-5b (described above) containing the 3'LTR and the pUC18 backbone, the Clal-Hindlll fragment from pCI-PLAP coding for PLAP and the Hindlll-Ndel fragment from pBA-6bLl containing the 5'LTR and the SV40 promoter.
  • Plasmid pBA-6bLl is based on pBA-6b (described above) where the HIVenv/rev coding region was deleted via Xhol-Clal digest and replaced with the Ll linker coding for several restriction enzyme sites including Xhol at the 5'end and Clal at the 3'end.
  • a B-domain deleted factor VIII cDNA fragment thus constmcted was obtained by XhoI/NotI digestion, as described below.
  • a retroviral vector (pMBF8) expressing B- domain-deleted factor VIII is constmcted from the expression plasmid pSVF8-200 which is prepared as described in Truett, 1985, DNA 4:333) and in U.S. Patent No. 5,045,455, and which was deposited with the ATCC on July 17, 1985, and which has been assigned ATCC number 40190.
  • Plasmid SVF8-302 was constmcted in a similar manner as pSVF8-200, which is described in detail in Truett and in U.S. Patent No. 5,045,455. Construction of pSVF8- 302 is also described in U.S. Patent No. 5,595,886.
  • the full-length cDNA sequence of the factor III is shown in Seq ID No. 44 and the full-length amino acid sequence is shown in Seq ID No. 45.
  • the cDNA sequence of the B- domain deleted SQN deletion is shown in Seq ID No. 46 and the SQN deletion amino acid sequence is shown in Seq. ID No. 47.
  • Fragment 1 encompassing nt 5500-6248 of pSVF8-200 (see Figure 8 of Truett), was obtained by VENT-PCR using factor VIII primers encoding a PF1M1 site at the 3' end and the 5' SQN sequence plus a Hindlll site at the 5' end.
  • the 5' primer encompasses the region 2446-2460 of the 5' SQN and the region 5144-5167 just downstream of the 3' SQN sequence. Thus, this fragment spans the sequence between the two SQN sites within the B domain (positions 2461 and 5142).
  • the particular primer sequences used were:
  • Fragment 1 was blunt-end cloned into vector SK- that had been cut with Smal and dephosphorylated, forming pSK-Pfl.
  • Fragment 2 encompassing nt 1190 and 2448 was isolated following Hindlll digestion and cloned into the Hindlll site of SK-Pfl to form SK- Pfl-Hind. The orientation of the insert was determined using AccI and Pstl digests.
  • pSVF8- 200 was digested with Hpal and religated to remove two small Hpal fragments 3' to the factor VIII cDNA insert, forming pF8-30O-del-Hpa.
  • Fragment 3 encompassing nt 5885-7604, was isolated following PflMI and NotI digestion and cloned into SK-Pfl-Hind following PflMI and NotI digestion of the latter to form pF8:213.
  • Fragment 4 (encompassing nt .104- 133 to 1204) was obtained following VENT-PCR of PSV7dF8-300 with primers containing 5' Xhol and 3' AccI sites respectively. The 5' primer encompasses nt 104-133 and the 3' primer encompasses nt 1200-1224.
  • pF8:213 was digested sequentially with Tjol followed by AccI and ligated to Fragment 4 which was digested with Xhol and ⁇ ccI, pF8:4213.
  • the primer sequences for the 5' UT and Xhol primers were:
  • the crossless backbones pBA-9b, pBA-5a, pBA-5b and pBA-5c were modified by linearizing with Clal, blunt ending and religating in the presence of NotI phosphorylated linkers.
  • the modified cDNA fragment was cloned into the XhoI/NotI linearized vectors.
  • the modified cDNA described above is cloned into the backbone pMBA backbone described above which has been digested with Xhol and NotI
  • the plasmid used as a template in the PCR reaction was pCMVKmHSTB, a B deleted factor VIII expression construct.
  • the amplified fragment was digested with Sal I, and cloned into a CMV expression vector, pCMVKmLINK digested with Sal I and Xho I.
  • This plasmid was called pCMVKm90H.
  • pCMVkMLINK is an expression vector containing the CMV promoter/intron, a polylinker for cloning genes of interest, and a bovine growth homone polyadenylation signal.
  • pCMVKm90H was digested with Sal I and Bam HI, the Bam HI site was filled in with T4 DNA polymerase, and this fragment was cloned into the rAAV vector pKm201CMV-CI digested with Sal I and Eco RV.
  • pKm201CMV-CI contains the inverted terminal repeats of AAV, the CMV promoter, the chimeric intron from pCI (Promega, Madison, WI), and the bovine growth hormone polyadenylation signal.
  • the final AAV vector was called pKm201- 90H.
  • This plasmid was called pCMVKmF8L (for factor VIII leader).
  • pCMVKmF8L for factor VIII leader
  • the following primers were used: forward, 5'-TCGGCTCGAGGCATCAACGGGAAATAACTCGT-3' (Seq ID No. 32), and reverse, 5'-CCGACTCGAGTCAGTAGAGGTCCTGTGCCTC-3' (Seq ID No. 33).
  • pCMVkMHSTB served as the template for the PCR.
  • the amplified fragment included sequences from amino acid 1645 of factor VIII to the STOP codon after amino acid 2332. This included the last four amino acids of the B domain and the complete light chain.
  • rAAV The heavy and light chain constructs, pKm201 -80L and pKm201 -90H, were packaged following standard procedures for the production of rAAV (Zhou et al, 1994, J. Exp. Med. 179: 1867-1875). rAAV was purified as described (Wang et al, 1995, Proc. Natl. Acad. Sci. USA 92:12416-12420) and used to infect 293 cells plated in 6-well plates. Supernatants of infected cells were collected at 48 h after infection and assayed for biologically acitve factor VIII by Coamatic Factor VIII (KabiVitrum, Sweden) following manufacturer's instructions.
  • Retroviral Vectors with Liver-Specific Promoters (a) Preparation of Interferon -alpha Sequences Utilizing PCR Construction of the Retroviral Vector Encoding Interferon- ⁇ 2a. ⁇ 2b. ⁇ 2c. ⁇ 54. and ⁇ 76 DNA clones of interferon- ⁇ 2a, ⁇ 2b, ⁇ 2c ⁇ 54, and ⁇ 76 are prepared utilizing PCR.
  • the interferon- ⁇ 2a DNA is obtained from a cDNA library as described by Goeddel et al, 1980, Nature 257:411-416.
  • interferon- ⁇ 2b DNA is obtained from a cDNA library described by Streuli et al, 1980, Science 209:1343-1347, and the interferon- ⁇ 2c DNA is obtained as described by Saveliev et al, 1986, Antibiot. Med. Biotekhnol. 5:592-596.
  • DNA clones of interferon- ⁇ 54 and ⁇ 76 are prepared utilizing PCR. Both the interferon- ⁇ 54 DNA and the interferon- ⁇ 76 DNA are obtained as described in U.S. Patent No. 4,975,276 and U.S. Patent No. 5,098,703.
  • a reaction mixture is then prepared according to procedures specified by New England Biolabs (Beverly, MA). More specifically, a reaction mixture is prepared containing 1 ug purified plasmid, 10 ul of 10X ThermoPol reaction buffer, 2 ul 2.5 mM of each dATP, dCTP, dGTP and dTTP, 0.5 ul of 2 units/100 ul Vent polymerase, 1 - 3 ul of lOOmM MgSO 4 , and 0.5-1.0 ug of the primers specified below (Mattila et al, 1991, Nucleic Acids Research 79:4967-4973, Eckert, K.A. and Kunkel, T. A., PCR Methods and Applications 1, 17-24, 1991).
  • the coding region is identical except for AAA codon at position 23 for interferon- ⁇ 2a and AGA codon at position 23 for interferon- ⁇ 2b.
  • the sense primer for both interferon- ⁇ 2a and ⁇ 2b is from the 5' region of the coding sequence, 6 bp upstream from the ATG start codon until 25 bp downstream from the start codon.
  • the 5' end of the primer contains the Xho I restriction site. (Seq ID No. 36) 5'-3': CGCG CCG CTC GAG TCT ACA ATG GCC TTG ACC TTT GCT TTA CTG G
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3' end of the interferon gene.
  • the 5' end of the primer contains the Cla I restriction site. (Seq ID No. 37) 5'-3': GCG CCC ATC GAT TCA TTC CTT ACT TCT TAA ACT TTC TTG CAA G
  • the sense primer is from the 5' region of the coding sequence, 7 bp upstream from the ATG start codon until 28 bp downstream from the start codon.
  • the 5' end of the primer contains the Xho I restriction site.
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3 'end of the interferon gene.
  • the 5' end of the primer contains the Cla I restriction site. (Seq ID No. 39) 5'-3": CC ATC GAT TCA ATC CTT CCT CCT TAA TCT TTT TTG CAA G
  • the sense primer is from the 5' region of the coding sequence, 6 bp upstream from the ATG start codon until 24 bp downstream from the start codon.
  • the 5' end of the primer contains the Xho I restriction site.
  • 5'-3' CGCG CCG CTC GAG TCT ACA ATG GCT TTG CCT TTT GCT TTA CTG
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3' end of the interferon gene.
  • the 5' end of the primer contains the Cla I restriction site.
  • 5'-3' GCG CCC ATC GAT TTA TTC CTT CCT CCT TAA CCT TTC TTG CAA GCG CCC ATC GAT TTA TTC CTT CCT CCT TAA CCT TTC TTG CAA G
  • the sense primer is from the 5' region of the coding sequence, 7 bp upstream from the ATG start codon until 28 bp downstream from the start codon.
  • the 5' end of the primer contains the Xho I restriction site.
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3 'end of the interferon gene.
  • the 5' end of the primer contains the Cla I restriction site. (Seq ID No. 56) 5'-3': CC ATC GAT TCA ATC CTT CCT CCT TAA TCT TTT TTG CAA G
  • the reaction mixture is brought up to 100 ⁇ l with DI H 2 O, and each tube is placed into a thermocycler (Gene Amp PCR System 9600, Perkin-Elmer, Cetus, Calif.).
  • the PCR program regulates the temperature of the reaction vessel first at 94°C for 10 minutes to hot start reactions. Vent polymerase is added next and the cycle begins with 94°C for 2 minutes, next at 56°C for 30 - 60 seconds, and 72°C for 30 - 60 seconds. This cycle is repeated 35 times. After the 35th cycle, product ends are sealed by the reactions at 72 °C for 3 - 5 minutes. Reactions are held at 4°C. If necessary, PCR amplification can be optimized using touchdown or stepdown PCR protocol (White, B. A., PCR Cloning Protocols 67, 39- 45, 1997).
  • the PCR product from the reaction yields the interferon- ⁇ 2a, ⁇ 2b and ⁇ 2c gene.
  • the solution is transferred to a fresh 1.5 ml microfuge tube. Fifty microliters of 3 M sodium acetate is added to this solution followed by 500 ⁇ l of chloroform :isoamy alcohol (24:1). The mixture is vortexed and then centrifuged at 14,000 ⁇ m for 5 minutes. The aqueous phase is transferred to a fresh mirofuge tube and 1.0 ml 100% EtOH is added. This solution is incubated at -20°C for 4.5 hours, and then centrifuged at 10,000 ⁇ m for 20 minutes.
  • the supernatant is decanted, and the pellet rinsed with 500 ul of 70% EtOH.
  • the pellet is dried by centrifugation at 10,000 ⁇ m under vacuum and then resuspended in 10 ul deionized H2O.
  • One microliter of the PCR product is analyzed by electrophoresis in a 1.0% agarose gel.
  • PCR products approximately 570 bp in length, are digested with Xho I and Cla I restriction endonucleases, electrophoresed through a 1.0% agarose gel and the DNA is purified from the gel slice by Geneclean II (Bio 101, Vista, California).
  • Xho I-Cla I PCR products are subcloned into the respective sites of pBluescript KSII+ (Stratagene, La Jolla, California). These constmcts are designated KSII+ Xho-Cla IFN- ⁇ 2a, ⁇ 2b, ⁇ .2c, ⁇ 54 and ⁇ 76 respectively, and are verified by DNA sequencing.
  • Retroviral Vector Backbone - pBA ⁇ bLl
  • the pBA ⁇ bLl retroviral vector backbone is described above. Two fragments are purified for the construction of the interferon ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76 pBA ⁇ bLl retroviral vector. First, pBA ⁇ bLl is digested with Xho I and Cla I and the 6.2 kb fragment containing the retroviral vector backbone is isolated.
  • the 570 Xho I-Cla I bp fragment containing the interferon- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76 gene is isolated and inserted into the Xho I-Cla I sites of pBA ⁇ bll .
  • the vector constmcts are designated pBA6b-IFN ⁇ 2a, pBA6b-IFN ⁇ 2b, pBA6b-IFN ⁇ 2c, pBA6b- IFN ⁇ 54 and pBA6b-IFN ⁇ 76, respectively.
  • the retroviral vector backbone, pLXSN is available by Clontech (Palo, Alto, Calif). Two fragments are purified for the constmction of the LXSN retroviral vector encoding interferon- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76. First, pLXSN is digested with Xho I and Bam HI and the 5.9 kb fragment containing the retroviral vector backbone is isolated.
  • the 570 bp Xho-Bam HI fragment containing either the interferon- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 or ⁇ 76 gene is isolated and inserted into the respective sites of pLXSN.
  • This vector construct is designated pLXSN- IFN ⁇ 2a, pLXSN-IFN ⁇ 2b, P LXSN-IFN ⁇ 2c, pLXSN- IFN ⁇ 54 and P LXSN-IFN ⁇ 76.
  • HIV retroviral vector backbone is made, similar to V653RSN as described by Parolin et al. (J. Virol. 65:3888-3895, 1994).
  • KSII+ Xho-Cla IFN- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76 the 570 bp Xho I-Xba I fragment containing the interferon- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76 is isolated and inserted into the Xho I-Xba I sites of a CMV expression vector, pCI (Promega, Madison, WI) and is designated pCI -IFN ⁇ 2a, IFN ⁇ 2b, IFN ⁇ 2c, IFN ⁇ 54 and IFN 76, respectively.
  • pCI Promega, Madison, WI
  • vector constmcts are sequenced. These vector constmcts are designated V653- IFN ⁇ 2a, V653-IFN ⁇ 2b, V653-IFN ⁇ 2c, V653-IFN ⁇ 54 and V653-IFN ⁇ 76, respectively.
  • the sense primer for both interferon- ⁇ 2a and ⁇ 2b is from the 5' region of the coding sequence, 6 bp upstream from the ATG start codon until 25 bp downstream from the start codon.
  • the 5' end of the primer contains the Bam HI restriction site. (Seq ID No. 40) 5'-3': CC GGA TCC TCT ACA ATG GCC TTG ACC TTT GCT TTA CTG G
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3' end of the interferon gene.
  • the 5' end of the primer contains the Not I restriction site.
  • 5'-3' CGCG CCG GCG GCC GC TCA TTC CTT ACT TCT TAA ACT TTC TTG CAA G b.
  • Interferon- ⁇ 2c For interferon- ⁇ 2c, the sense primer is from the 5' region of the coding sequence, 7 bp upstream from the ATG start codon until 28 bp downstream from the start codon.
  • the 5' end of the primer contains the Bam HI restriction site.
  • 5'-3' CC GGA TCC CAT CCA ATG GCC CTG TCC TTT TCT TTA CTT ATG G
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3 'end of the interferon gene.
  • the 5' end of the primer contains the Not I restriction site. (Seq ID No. 43) 5'-3': CGCG CCG GCG GCC GC TCA ATC CTT CCT CCT TAA TCT TTT TTG CAA G
  • the sense primer for both interferon- ⁇ 54 is from the 5' region of the coding sequence, 6 bp upstream from the ATG start codon until 25 bp downstream from the start codon.
  • the 5' end of the primer contains the Bam HI restriction site. (Seq ID No. 57) 5'-3':CC GGA TCC TCT ACA ATG GCT TTG CCT TTT GCT TTA CTG
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3' end of the interferon gene.
  • the 5' end of the primer contains the Not I restriction site. (Seq ID No. 58) 5'-3':CGCG CCG GCG GCC GC TTA TTC CTT CCT CCT TAA CCT TTC TTG CAA G
  • the sense primer is from the 5' region of the coding sequence, 7 bp upstream from the ATG start codon until 28 bp downstream from the start codon.
  • the 5' end of the primer contains the Bam HI restriction site.
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3 'end of the interferon gene.
  • the 5' end of the primer contains the Not I restriction site.
  • 5'-3' CGCG CCG GCG GCC GC TCA ATC CTT CCT CCT TAA TCT TTT TTG CAA G
  • PCR products approximately 570 bp in length, are digested with Bam HI and Not I restriction endonucleases, electrophoresed through a 1.0% agarose gel and the DNA is purified from the gel slice by Geneclean II (Bio 101, Vista, California).
  • This Bam HI-Not I PCR product is subcloned into the respective sites of pBluescript KSII+ (Stratagene, La Jolla, California). These constmcts, designated KSII+ Bam-Not IFN- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76 are verified by DNA sequencing.
  • Two fragments are purified for the construction of the interferon ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 and ⁇ 76 pBA8bLl retroviral vector.
  • pBA ⁇ bLl is digested with Not I and Srf l and the 6.1 kb fragment containing the retroviral vector backbone is isolated.
  • the 570 bp Sma I-Not I fragments containing the interferon-cc2a, ⁇ 2b, ⁇ 2c, ⁇ 54 or ⁇ 76 are isolated and inserted into the Srf I- Not I sites of pBA8bLl .
  • These vector constmcts are designated pBA8b-IFN- ⁇ 2a, pBA8b- IFN- ⁇ 2b, pBA8b- ⁇ 2c, pBA8b-IFN- ⁇ 54 and pBA8b-IFN ⁇ 76.
  • Retroviral Vector Backbone - pBA6bLl/TK
  • HSV he ⁇ es simplex vims
  • TK thymidine kinase
  • Example 27B and inserted into the Sal I/Cla I sites of pSP72 plasmid (Promega, Madison, WI) and designated pSP72-TK.
  • the human interleukin-2 (IL-2) cDNA is excised from KT3-IL-2 (described in PCT Patent Publication Nos. WO 94/21792 and WO 96/21015) and inserted into the Xho I/Cla I sites of pBluescript SK- and designated pBSSK-IL2.
  • the Hind Ill/Sma I double digest of pSP72-TK excises TK cDNA and this fragment is inserted into the Hind Ill/Sma I sites of pBSSK-IL2.
  • This construct has the following: Xho I-IL2 cDNA-Cla I-Hind III-TK cDNA-Sma I sequence and is designated pBSSK-IL2/TK. From KSII+ Xho-Cla IFN- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 or ⁇ 76, the 570 Xho I-Cla I bp fragment containing the interferon- ⁇ 2a, oc2b, ⁇ 2c, ⁇ 54 or ⁇ 76 gene is isolated.
  • the IL2 fragment is excised by Xho I/Cla I double digestion, and replaced with Xho/Cla I fragments containing the interferon- ⁇ 2a, ⁇ 2b, ⁇ 2c, ⁇ 54 or ⁇ 76 genes.
  • the vector constmcts are designated pBSSK-IFN ⁇ 2a /TK, pBSSK-IFN ⁇ 2b /TK, pBSSK-
  • IFN ⁇ 2c /TK IFN ⁇ 2c /TK
  • pBSSK-IFN ⁇ 54/TK IFN ⁇ 2c /TK
  • pBSSK-IFN ⁇ 76/TK The Xho I/Not I fragments from pBSSK-IFN ⁇ 2a TK, pBSSK-IFN ⁇ 2b /TK, pBSSK-IFN ⁇ 2c /TK, pBSSK- IFN ⁇ 54 /TK and pBSSK-IFN ⁇ 76 /TK are excised and inserted into the Xho I-Not I sites of pBA ⁇ bll.
  • di-cistronic retroviral vectors encoding IFN ⁇ 2a, ⁇ 2b, ⁇ 2c ⁇ 54 and ⁇ 76 are designated pBA ⁇ bLl IFN ⁇ 2a/TK, pBA ⁇ bLl IFN ⁇ 2b/TK, pBA6bLl IFN ⁇ 2c/TK, pBA ⁇ bLl IFN ⁇ 54/TK and pBA ⁇ bLl IFN ⁇ 76/TK.
  • Retroviral Vector Backbone with a Liver-specific promoter The ApoE enhancer-alpha 1 -antitrypsin promoter yields high levels of expression from liver cells (Okuyama et al, 1996, Hum Gene Ther 7:637-645).
  • the retroviral vector encoding the liver-specific promoter is constmcted as follows.
  • ApoE enhancer cassette The apolipoprotein E (Apo E) enhancer sequence has been identified as a 154 bp fragment by Shachter et al, 1993, J. LipidRes 5 ⁇ :1699-1707.
  • the ApoE enhancer cassette was generated by sets of four oligonucleotides that span the entire sequence. For the first reaction, four oligonucleotides were synthesized that were phosphorylated at the 5' end. The first oligonucleotide is the sense strand and contains Hind III restriction site at the 5' end.
  • the second oligonucleotide is the sense strand, spanning the second half of the Apo E enhancer and contains the Hind III restriction site at the 3' end.
  • the third oligonucleotide is the antisense strand of the second half of the Apo E enhancer and contains the Hind III site at the 5' end.
  • the fourth oligonucleotide is the antisense strand of the first half of the Apo E enhancer and contains the Hind III site at the 3 ' end.
  • This set of four oligonucleotides of approximately 80 bases in length are annealed, and ligated into the dephosphory lated Hind III sites of pBluescript KSII+ (Stratagene, La Jolla, California) following standard molecular cloning protocols. Recombinants are selected and sequenced. Correct monomer Apo E enhancer elements are excised by Hind III, electrophoresed through a polyacrylamide gel and the DNA is purified following procedures described by Asubel et al., Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York (1987).
  • the monomer enhancer element is ligated into the dephosphorylated Hind III sites of pBluescript KSII+ at an insert: vector ratio of 10:1 in order to generate recombinants containing dimers.
  • Recombinant plasmids were assessed by DNA sequencing to select clones which carried both enhancer fragments in the same orientation with the 5' end closer to the Eco RV site and the 3' end closer to the Cla I site within the multi-cloning site of pBluescript. This constmct is designated KSII+ Hind III ApoE(x2)
  • oligonucleotide For the second reaction, another set of four oligonucleotides were synthesized that were phosphorylated at the 5' end.
  • the first oligonucleotide is the sense strand and contains Eco RI restriction site at the 5' end.
  • the second oligonucleotide is the sense strand, spanning the second half of the APO E enhancer and contains the Eco RI restriction site at the 3' end.
  • the third oligonucleotide is the antisense strand of the second half of the Apo E enhancer and contains the Eco RI site at the 5' end.
  • the fourth oligonucleotide is the antisense strand of the first half of the Apo E enhancer and contains the Eco RI site at the 3' end.
  • this set of four oligonucleotides of approximately 80 bases in length are annealed, and ligated into the dephosphorylated Eco RI sites of pBluescript KSII+ (Stratagene, La Jolla, California). Recombinants are selected and sequenced.
  • the 300 base pair Pst I/Eco RV fragment from KSII+ Eco RI ApoE(x2) is inserted into the Pst I and Eco RV sites within the multi-cloning sites of KSII+ Hind III ApoE(x2). This constmct is designated KSII+ ApoE(x4), and contains 4 copies of the 154 base pair Apo E enhancer.
  • the human alpha 1 -antitrypsin (hAAT) promoter has been identified as a 400 base pair region by Shen et al, 1989, DNA 5:101-108.
  • the hAAT promoter DNA is obtained from a genomic library as was done by Long et al, 1984, Biochem 25:4828-4837, or can be obtained from ATCC (ATCC No. 61596).
  • a PCR reaction is conducted in order to isolate the hAAT promoter.
  • the 5' end of the primer contains the Sma I restriction site.
  • the second primer corresponding to the anti-sense nucleotide sequence contains the Not I restriction site.
  • This PCR product approximately 400 bp in length, is digested with Sma I and Not I and is cloned into the Sma I and Not I sites within the multi-cloning sites of KSII+ ApoE(x4). This constmct is confirmed by sequencing and is designated KSII+ ApoE/hAAT.
  • Interferon- ⁇ 2a and ⁇ 2b The sense primer for both interferon- ⁇ 2a and ⁇ 2b is from the 5' region of the coding sequence, 6 bp upstream from the ATG start codon until 25 bp downstream from the start codon. The 5' end of the primer contains the Not I restriction site.
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3' end of the interferon gene.
  • the 5' end of the primer contains the Cla I restriction site. (Seq ID No. 72) 5'-3': GCG CCC ATC GAT TCA TTC CTT ACT TCT TAA ACT TTC TTG CAA G
  • the product of this PCR reaction is digested with Not I and Cla I and purified.
  • KSII+ ApoE/hAAT is digested with Not I and Cla I and the 1 kb fragment containing the Apo E enhancer and the hAAT promoter is isolated. From pBA ⁇ bLl , the 6.1 kb Xho/Cla I retroviral vector backbone is isolated.
  • the second primer corresponds to the anti-sense nucleotide sequence, extending 31 bp into the coding sequence from the 3' end of the interferon gene.
  • the 5' end of the primer contains the Cla I restriction site.
  • KSII+ ApoE/hAAT is digested with Not I and Cla I and the 1 kb fragment containing the Apo E enhancer and the hAAT promoter is isolated. From pB A9b, the 4.8 kb Xho/Cla I retroviral vector backbone is isolated.
  • TK ApoE/hAAT-IFN ⁇ 2b The thymidine kinase (TK) gene is excised form pTJ5b-TK with Xho/Cla I and inserted into the Xho I/Cla I sites of KSII+ ApoE hAAT. This constmct is designated KSII+ TK/ApoE/hAAT.
  • PCR products containing either the IFN ⁇ 2a or IFN ⁇ 2b genes described in the example above is digested with Not I and Cla I and purified.
  • the Xho/Not fragment containing the TK/ ApoE/hAAT sequence is excised from KSII+ TK ApoE/hAAT.
  • From pBA ⁇ bLl, the 6.1 kb Xho/Cla I retroviral vector backbone is isolated.
  • a the Xho I/Not I fragment containing the TK gene and the
  • Apo E enhancer with the hAAT promoter (b) the Not 1/ Cla I fragment encoding the IFN ⁇ 2a or ⁇ 2b gene and (c) the Xho I/Cla I fragment encoding the retroviral vector backbone are joined to generate pBA6 TK/ApoE/hAAT-IFN ⁇ 2a or pBA6 TK/ApoE/hAAT-IFN ⁇ 2b.
  • TK thymidine kinase
  • PCR products containing either the IFN ⁇ 2a or IFN ⁇ 2b genes described in the example above is digested with Not I and Cla I and purified.
  • the Xho/Not fragment containing the TK ApoE/hAAT sequence is excised from KSII+ TK ApoE/hAAT.
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO:l: GAGAGATGGG GGAGGCTAAC TGAG 24
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO:3: TATATCTCCA GATGAGGTAC ATGATTTTAG GCTTG 35
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • SEQUENCE DESCRIPTION SEQ ID NO : 6 : CGCGTCGACA TCGATGCGGC CGCCCGGGCG GATCC 35
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • SEQUENCE DESCRIPTION SEQ ID NO: 9: CCCGAGAGAT GGGGGAGGCT AACTGAG 27
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • xi SEQUENCE DESCRIPTION: SEQ ID NO: 13: CCCACCACAA CCACATATCC CTCC 24
  • MOLECULE TYPE DNA (genomic)
  • SEQUENCE DESCRIPTION SEQ ID NO: 14: CCAGTCCTCC GATTGACTG 19

Abstract

L'invention concerne des procédés permettant d'obtenir des niveaus mesurables d'une protéine, d'une molécule d'acide nucléique ou d'un produit enzymatique dans un liquide biologique ou des cellules d'un sujet humain; ces procédés comprennent l'étape d'administration à un sujet humain d'une préparation rétrovirale recombiné avec un titre sur des cellules HT1080 supérieur à 105 cfu/ml, dans laquelle la préparation rétrovirale recombiné est capable de diriger l'expression d'une protéine, d'une molécule d'acide nucléique, ou d'un enyzme générant un produit enzymatique, de telle sorte que des niveaux mesurables de la protéine, la molécule d'acide nucléique, ou le produit enzymatique puissent être obtenus dans le liquide biologique ou les cellules du sujet humain.
PCT/US1997/011785 1996-07-03 1997-07-02 Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie WO1998000542A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP97940535A EP0915975A2 (fr) 1996-07-03 1997-07-02 Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US64560196A 1996-07-03 1996-07-03
US08/645,601 1996-07-03
US69638196A 1996-08-13 1996-08-13
US08/696,381 1996-08-13
US86930997A 1997-06-04 1997-06-04
US08/869,309 1997-06-04

Publications (2)

Publication Number Publication Date
WO1998000542A2 true WO1998000542A2 (fr) 1998-01-08
WO1998000542A9 WO1998000542A9 (fr) 1998-06-04

Family

ID=27417749

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1997/011784 WO1998000541A2 (fr) 1996-07-03 1997-07-02 Procedes d'administration de porteurs fournissant des genes recombinants pour le traitement d'une maladie chez l'homme
PCT/US1997/011785 WO1998000542A2 (fr) 1996-07-03 1997-07-02 Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US1997/011784 WO1998000541A2 (fr) 1996-07-03 1997-07-02 Procedes d'administration de porteurs fournissant des genes recombinants pour le traitement d'une maladie chez l'homme

Country Status (2)

Country Link
EP (2) EP0915975A2 (fr)
WO (2) WO1998000541A2 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999029848A1 (fr) * 1997-12-05 1999-06-17 The Immune Response Corporation Vecteurs et genes a expression accrue
WO2000017375A2 (fr) * 1998-09-23 2000-03-30 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Procede de traitement de l'hemophilie par therapie genique in vivo a l'aide de vecteurs retroviraux
WO2001070968A2 (fr) * 2000-03-22 2001-09-27 Octagene Gmbh Production de facteurs de coagulation sanguine recombines dans des lignees de cellules humaines
WO2010126370A2 (fr) 2009-04-29 2010-11-04 Academisch Medisch Centrum Bij De Universiteit Van Amsterdam Moyens et procédés pour contrebalancer, prévenir et/ou déterminer une insuffisance cardiaque, ou un risque d'insuffisance cardiaque
WO2011120474A1 (fr) 2010-04-01 2011-10-06 Centro De Ingeniería Genética Y Biotecnología Méthode pour inhiber la réplication du vih dans des cellules de mammifères et chez l'homme
EP2390346A1 (fr) 2010-05-28 2011-11-30 Universiteit Twente Marqueurs d'ostéoarthrose
WO2012105826A1 (fr) 2011-02-01 2012-08-09 Erasmus University Medical Center Rotterdam Utilisation de microarn dans le diagnostic et la thérapie du vieillissement

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6404736B1 (en) 1997-06-20 2002-06-11 Telefonaktiebolaget L M Ericsson (Publ) Call-routing efficiency with a network access server
WO1999011292A2 (fr) * 1997-09-02 1999-03-11 Chiron Corporation Compositions et methodes de traitement de l'arthrite par therapie genique
AU1107199A (en) * 1997-10-21 1999-05-10 Chiron Corporation Methods for cancer immunotherapy using retroviral vectors expressing gamma interferon
WO1999060110A2 (fr) * 1998-05-20 1999-11-25 The University Of Tennessee Research Corporation Proteines a enveloppe stable pour des vecteurs retroviraux
FR2817559B1 (fr) * 2000-12-06 2003-12-12 Genodyssee Procede de determination d'un ou plusieurs polymorphisme(s) fontionnel(s) dans la sequence nucleique d'un gene "candidat" fonctionnel preselectionne et ses applications
US7667089B2 (en) * 2004-04-09 2010-02-23 National Chung Hsing University Transgenic mammal secreting B-domain deleted human FVII in its milk
KR102011532B1 (ko) 2011-09-30 2019-08-16 블루버드 바이오, 인코포레이티드. 개선된 바이러스 형질도입을 위한 화합물
WO2014041500A2 (fr) * 2012-09-12 2014-03-20 Centre For Bioseparation Technology-Vit Facteur viii de coagulation à double mutation et procédés associés
EP3413896B1 (fr) 2016-02-12 2021-03-17 Bluebird Bio, Inc. Compositions d'augmentation de vcn et ses procédés d'utilisation

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU648261B2 (en) * 1989-08-18 1994-04-21 Novartis Vaccines And Diagnostics, Inc. Recombinant retroviruses delivering vector constructs to target cells
EP0644946A4 (fr) * 1992-06-10 1997-03-12 Us Health Particules vecteurs resistantes a l'inactivation par le serum humain.
AU7516594A (en) * 1993-07-28 1995-02-28 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Pre-binding of retroviral vector particles with complement components to enable the performance of human gene therapy (in vivo)
FR2713657B1 (fr) * 1993-12-13 1996-03-01 Transgene Sa Nouveaux vecteurs pour le traitement du sida.
US5952225A (en) * 1994-08-17 1999-09-14 Genetic Therapy, Inc. Retroviral vectors produced by producer cell lines resistant to lysis by human serum
AU4608096A (en) * 1994-12-30 1996-07-24 Chiron Corporation Production and administration of high titer recombinant retroviruses
US5681746A (en) * 1994-12-30 1997-10-28 Chiron Viagene, Inc. Retroviral delivery of full length factor VIII
GB9517263D0 (en) * 1995-08-23 1995-10-25 Cancer Res Campaign Tech Expression systems

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6642028B1 (en) 1997-12-05 2003-11-04 The Immune Response Corporation Vectors and genes exhibiting increased expression
WO1999029848A1 (fr) * 1997-12-05 1999-06-17 The Immune Response Corporation Vecteurs et genes a expression accrue
WO2000017375A2 (fr) * 1998-09-23 2000-03-30 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Procede de traitement de l'hemophilie par therapie genique in vivo a l'aide de vecteurs retroviraux
WO2000017375A3 (fr) * 1998-09-23 2000-07-27 Vlaams Interuniv Inst Biotech Procede de traitement de l'hemophilie par therapie genique in vivo a l'aide de vecteurs retroviraux
HRP20020767B1 (en) * 2000-03-22 2011-09-30 Octapharma Biopharmaceuticals Gmbh Production of recombinant blood clotting factors in human cell lines
WO2001070968A2 (fr) * 2000-03-22 2001-09-27 Octagene Gmbh Production de facteurs de coagulation sanguine recombines dans des lignees de cellules humaines
WO2001070968A3 (fr) * 2000-03-22 2001-12-13 Octagene Gmbh Production de facteurs de coagulation sanguine recombines dans des lignees de cellules humaines
WO2010126370A2 (fr) 2009-04-29 2010-11-04 Academisch Medisch Centrum Bij De Universiteit Van Amsterdam Moyens et procédés pour contrebalancer, prévenir et/ou déterminer une insuffisance cardiaque, ou un risque d'insuffisance cardiaque
EP2907879A2 (fr) 2009-04-29 2015-08-19 Academisch Medisch Centrum bij de Universiteit van Amsterdam Moyens et procédés pour combattre, prévenir et/ou déterminer une insuffisance cardiaque, ou un risque d'insuffisance cardiaque
WO2011120474A1 (fr) 2010-04-01 2011-10-06 Centro De Ingeniería Genética Y Biotecnología Méthode pour inhiber la réplication du vih dans des cellules de mammifères et chez l'homme
US9205128B2 (en) 2010-04-01 2015-12-08 Centro De Ingenieria Genetica Y Biotechnologia Method for inhibiting HIV replication in mammal and human cells
US10434137B2 (en) 2010-04-01 2019-10-08 Centro De Ingenieria Genetica Y Biotecnologia Method for inhibiting HIV replication in mammal and human cells
EP2390346A1 (fr) 2010-05-28 2011-11-30 Universiteit Twente Marqueurs d'ostéoarthrose
WO2011149352A1 (fr) 2010-05-28 2011-12-01 Universiteit Twente Marqueurs d'ostéo-arthrite
WO2012105826A1 (fr) 2011-02-01 2012-08-09 Erasmus University Medical Center Rotterdam Utilisation de microarn dans le diagnostic et la thérapie du vieillissement

Also Published As

Publication number Publication date
EP0915975A2 (fr) 1999-05-19
EP0951544A2 (fr) 1999-10-27
WO1998000541A3 (fr) 1998-03-19
WO1998000541A2 (fr) 1998-01-08

Similar Documents

Publication Publication Date Title
US6818439B1 (en) Methods for administration of recombinant gene delivery vehicles for treatment of hemophilia and other disorders
US5681746A (en) Retroviral delivery of full length factor VIII
WO1998000542A2 (fr) Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie
WO1998000542A9 (fr) Procedes d'administration d'excipients d'apport de genes recombines dans le traitement de l'hemophilie
EP0710288B1 (fr) Vecteurs adenoviraux pour le traitement de l'hemophilie
EP1005376B1 (fr) Compositions utilisees en therapie genique dans le traitement de l'hemophilie
WO1996021035A9 (fr) Production retrovirale du facteur viii entier
CA2278616C (fr) Adenovirus comprenant des proteines d'hexon modifiees
CA2343575C (fr) Inhibition de la migration des cellules des muscles lisses au moyen de l'heme oxygenase (1)
WO1998000541A9 (fr) Procedes d'administration de porteurs fournissant des genes recombinants pour le traitement d'une maladie chez l'homme
Chuah et al. Gene therapy for hemophilia
WO1996037626A1 (fr) Integration de vecteurs de recombinaison dans une position specifique du genome d'un eucaryote, assuree par une proteine chimere du type integrase
US7052904B2 (en) Hybrid adeno-retroviral vector for the transfection of cells
US20020082224A1 (en) Non-immunogenic prodrugs and selectable markers for use in gene therapy
US7615537B2 (en) Methods for treating blood coagulation disorders
WO1996021014A2 (fr) Production et administration de retrovirus recombines a titre eleve
WO1995007360A1 (fr) Compositions specifiques des cellules endotheliales, et procedes de preparation et d'utilisation
AU2002224771A1 (en) Induction of blood vessel formation by polynucleotides encoding sphingosine kinases
US20030212030A1 (en) Novel adenoviral vector for transferring human genes in vivo
WO1996033282A2 (fr) Transduction ex vivo a fort rendement de cellules a l'aide de preparations de retrovirus de recombinaison a titre eleve
WO1996036365A1 (fr) Therapie genique de carcinomes hepatocellulaires par expression de genes specifiques du cancer
US20230101788A1 (en) Gene therapy
JPH10511951A (ja) 高力価組換えレトロウイルスの産生および投与
Hortelano et al. Therapeutic approaches for haemophilia

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: PAT. BUL. 01/98, UNDER INID (30) "PRIORITY DATA", REPLACE "NOT FURNISHED" BY "08/869309"

121 Ep: the epo has been informed by wipo that ep was designated in this application
COP Corrected version of pamphlet

Free format text: PAGES 1/43-43/43, DRAWINGS, REPLACED BY NEW PAGES 1/41-41/41; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

WWE Wipo information: entry into national phase

Ref document number: 1997940535

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: JP

Ref document number: 98504518

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1997940535

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: CA

WWW Wipo information: withdrawn in national office

Ref document number: 1997940535

Country of ref document: EP