US20030082162A1 - rAAV compositions for gene therapy - Google Patents

rAAV compositions for gene therapy Download PDF

Info

Publication number
US20030082162A1
US20030082162A1 US10/267,117 US26711702A US2003082162A1 US 20030082162 A1 US20030082162 A1 US 20030082162A1 US 26711702 A US26711702 A US 26711702A US 2003082162 A1 US2003082162 A1 US 2003082162A1
Authority
US
United States
Prior art keywords
vector
promoter
haat
expression
levels
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/267,117
Inventor
Terence Flotte
Sihong Song
Barry Byrne
Michael Morgan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Florida Research Foundation Inc
Original Assignee
University of Florida Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Florida Research Foundation Inc filed Critical University of Florida Research Foundation Inc
Priority to US10/267,117 priority Critical patent/US20030082162A1/en
Priority to US10/340,112 priority patent/US20030095949A1/en
Publication of US20030082162A1 publication Critical patent/US20030082162A1/en
Priority to US11/932,912 priority patent/US20090186002A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF FLORIDA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • Alpha-1-antitrypsin (AAT) deficiency is the second most common monogenic lung disease in man, accounting for approximately 3% of all early deaths due to obstructive pulmonary disease.
  • AAT protein is normally produced in the liver, secreted into the serum and circulated to the lung where it protects the fine supporting network of elastin fibers from degradation by neutrophil elastase.
  • Current therapy for AAT deficiency includes avoidance of cigarette smoke exposure and weekly intravenous infusions of recombinant human AAT (hAAT) protein.
  • AAV adeno-associated virus
  • Recombinant AAV latency also differs from that of wtAAV in that wtAAV is rapidly converted to double-stranded DNA in the absence of helper virus (e.g., adenovirus) infection, while with rAAV leading strand synthesis is delayed in the absence of helper virus (Fisher et al., 1996; Ferrari et al., 1996).
  • helper virus e.g., adenovirus
  • Kessler et al. demonstrated that murine skeletal myofibers transduced by an rAAV vector were capable of sustained secretion of biologically active human erythropoietin (hEpo), apparently without eliciting a significant immune response against the secreted hepo. See also U.S. Pat. No. 5,858,351 issued to Podsakoffet al. Likewise, Murphy et al. (1997) have observed the expression and secretion of sustained levels of leptin in ob/ob mice after AAV muscle transduction. Brantly et al. (U.S. Pat. No.
  • 5,439,824 disclose methods for increasing expression of AAT using vectors comprising intron II of the human AAT gene.
  • the level of leptin expression observed was only in the range of 2 to 5 ng/ml.
  • Therapy for AAT deficiency requires serum levels of at least about 800 ⁇ g/ml.
  • the subject invention concerns materials and methods for gene therapy.
  • One aspect of the invention pertains to vectors which can be used to provide genetic therapy in animals or humans having a genetic disorder where relatively high levels of expression of a protein is required to treat the disorder.
  • the vectors of the invention are based on adeno-associated virus (AAV).
  • AAV adeno-associated virus
  • the vectors are designed to provide high levels of expression of heterologous DNA contained in the vector.
  • the vectors comprise AAV inverted terminal repeat sequences and constitutive or regulatable promoters for driving high levels of gene expression.
  • the subject invention also pertains to methods for treating animals or humans in need of gene therapy, e.g., to correct a genetic deficiency disorder.
  • FIG. 1 shows rAAV-AAT vector cassettes used according to the subject invention.
  • the A-AT and B-AT constructs contain the promoters from the small nuclear RNA genes, U1a and U1b, respectively.
  • the C-AT construct contains the CMV promoter, whereas the E-AT vector uses the human elongation factor 1- ⁇ (ELF in the figure) promoter.
  • ITR refers to AAV inverted terminal repeat; An refers to polyA signal; Tk refers to the HSV thymidine kinase promoter; neo refers to the Tn5 neomycin phosphotransferase gene.
  • FIG. 2 shows hAAT secretion rates in vitro from transiently transfected murine C2C12 myoblast cell line using expression vectors according to the subject invention.
  • C-AT does not differ significantly from E-AT, but both differ from A-AT and B-AT (p ⁇ 0.05)
  • AAT expression was detected using an ELISA assay specific for human AAT.
  • FIG. 3 shows hAAT secretion rates in vitro from stably transduced murine C2C12 myoblast cell line using viral particles comprising expression vectors according to the subject invention.
  • the mean rates of secretion from G418-resistant cultures 1 mo after transduction with either packaged E-AT vector or packaged C-AT vector are shown.
  • a “low” multiplicity transduction (4 ⁇ 10 5 particles/cell) and a high multiplicity transduction (4 ⁇ 10 6 particles/cell) were performed.
  • AAT expression was detected using an ELISA assay specific for human AAT.
  • FIG. 4 shows additional constructs tested for hAAT expression.
  • the murine myoblast C2C12 cells were grown in 35-mm wells with approximately 4 ⁇ 10 5 cell per well and were transfectd with 5 ⁇ g of the appropriate plasmid DNA using SUPERFECT transfection (Qiagen Inc., CA). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from three experiments (triplicate in each experiment).
  • FIGS. 5A and 5B show sustained secretion of therapeutic levels of HAAT using either the C-AT vector or the E-AT vector in either SCID or C57BL mice.
  • FIG. 5A shows the mean total serum levels of hAAT observed in groups of either SCID (squares) or C57BL (circles) mice receiving either low dose (5 ⁇ 10 11 particles) (open symbols) or high dose (1.4 ⁇ 10 13 particles) (filled symbols) single injections into muscle of the C-AT vector measured at time points ranging from 1 to 16 wk after injection.
  • FIG. 5B shows analogous data with the E-AT vector. None of these differences were significant.
  • FIG. 5C shows long term secretion of HAAT from murine muscle transduced with C-AT.
  • C57B1/6 or C57B1/6-SCID mice received 3.5 ⁇ 10 10 IU, 1.4 ⁇ 10 13 particles/mouse.
  • serum hAAT levels were still 400 ⁇ g/ml in C57B1/6-SCID and 200 ⁇ g/ml in C57B1/6. This level are comparable with the peak levels observed (800 or 400 ⁇ g/ml, respectively).
  • FIG. 6 shows an immunoblot of sera taken from several of the C-AT vector-treated mice at 11 weeks after vector administration.
  • Ten microliters of a 1:100 dilution of serum was electrophoresed by 10% SDS/PAGE, blotted, and incubated with 1:1,500 dilution of goat anti-hAAT-horseradish peroxidase conjugate (Cappel/ICN).
  • hAAT was added either to negative-control C57B1 serum (first hAAT lane) or to PBS (second hAAT) lane to final equivalent serum concentration of 100 ⁇ g/ml.
  • FIGS. 7A and 7B show that some BALB/c mice mount humoral immune responses to hAAT, which correlate with lower serum levels but no observable toxicity.
  • FIG. 7A shows serum hAAT levels
  • FIG. 7B shows serum anti-hAAT antibody levels as determined by ELISA performed on serum taken from mice injected with 1 ⁇ 10 11 particles of the C-AT vector.
  • Each set of symbols represents an individual animal ( ⁇ , no. 1 ; ⁇ , no. 2; ⁇ , no. 3). Note the inverse correlation between the presence of antibody and the presence of circulating hAAT.
  • FIG. 8 shows the persistence of rAAV-AAT vector DNA in high molecular weight form.
  • PCR products were amplified from DNA prepared by Hirt extraction from three SCID mice injected 16 wk earlier with 5 ⁇ 10 11 resistant-particles of C-AT and analyzed by Southern blot.
  • the high molecular weight Hirt pellet (genomic DNA lanes) and the low molecular weight supernatant (episomal DNA lanes) were analyzed separately.
  • Control lanes include a sample in which an hAAT cDNA plasmid was the template DNA (+) and a control in which water was the template ( ⁇ ). In this internal PCR reaction, a 500-bp product is expected regardless of whether or not the vector genome is integrated.
  • FIG. 9 shows serum hAAT in C57B1/6 mice transduced with C-AT and p43CB-AT.
  • C57B1/6 mice were injected in muscle with C-AT (3.5 ⁇ 10 10 IU/mouse, 1 ⁇ 10 12 particles/mouse) or p43CB-AT (6 ⁇ 10 9 IU, 1 ⁇ 10 12 particles/mouse).
  • the level of hAAT from p43CB-AT were projected based on an estimation of the equivalent dosage (infectious unit) of C-AT.
  • FIG. 10 shows enhancement of CMV promoter activity by a synthetic enhancer in C2C12 cells.
  • the murine myoblast C2C12 cells were grown in 35-mm wells with approximately 4 ⁇ 10 5 cell per well and were transfected with 5 ⁇ g of p43msENC-AT vector DNA using SUPERFECT transfection (Qiagen Inc, CA). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from one experiment (triplicate).
  • FIG. 11 shows secretion of hAAT from mouse liver cells (HO15) transfected with different constructs.
  • the murine liver cells (HO15) were grown in 35-mm wells with approximately 4 ⁇ 10 5 cell per well and were transfected with 5 ⁇ g of the plasmid DNA using LIPOFECTAMINE reagents (Life Technologies Inc, MD). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from two experiments (triplicate).
  • FIG. 12 shows secretion of HAAT from mouse liver cells (HO15) transfected using different methods.
  • the murine liver cells (HO15) were grown in 35-mm wells with approximately 4 ⁇ 10 5 cell per well and were transfected with 5 ⁇ g of the p43CB-AT vector using SUPERFECT (Qiagen Inc., CA), FuGENE (Boehringer Mannhem Co, IN), Lipofectin, LIPOFECTAMNE (ife Technologies Inc, MD) reagents and Calcium phosphate (CA-PO4) transfection.
  • SUPERFECT Quiagen Inc., CA
  • FuGENE FuGENE
  • Lipofectin lipofectin
  • LIPOFECTAMNE ife Technologies Inc, MD
  • CA-PO4 Calcium phosphate
  • FIG. 13 shows hAAT secretion from mouse liver transduced with rAAV.
  • C57B1/6 mice were injected with either p43CB-AT, C-AT or E-AT vector either by portal vein or tail vein injection.
  • PV portal vein injection.
  • TV tail vein injection.
  • FIG. 14 shows serum HAAT levels in C57B1/6 mice after intratracheal (IT) injection of C-AT orp43CB-AT vector. Mice received either 10 9 IU of C-AT (open circles), 10 9 IU of p43CB-AT (open triangles) or 10 10 IU of p43CB-AT (open squares).
  • FIG. 15 shows a map and nucleotide sequence for the vector of the present invention designated as C-AT.
  • FIG. 16 shows a map and nucleotide sequence for the vector of the present invention designated as E-AT.
  • FIG. 17 shows a map and nucleotide sequence for the vector of the present invention designated as DE-AT.
  • FIG. 18 shows a map and nucleotide sequence for the vector of the present invention designated as p43C-AT.
  • FIG. 19 shows a map and nucleotide sequence for the vector of the present invention designated as p43C-AT-IN.
  • This vector includes intron II from human AAT gene to enhance transcription.
  • FIG. 20 shows a map and nucleotide sequence for the vector of the present invention designated as p43CB-AT.
  • FIG. 21 shows a map and nucleotide sequence for the vector of the present invention designated as C-AT2.
  • FIG. 22 shows a map and nucleotide sequence for the vector of the present invention designated as p43 msENC-AT. This vector is similar to p43C-AT but also comprises an enhancer sequence upstream of the CMV promoter.
  • FIG. 23 shows a map and nucleotide sequence for the vector of the present invention designated as p43rmsENC-AT. This vector is the same as the p43msENC-AT vector except that the enhancer sequence is in an opposite orientation.
  • FIG. 24 shows a map and nucleotide sequence for the vector of the present invention designated as p43 msENCB-AT.
  • This vector is similar to p43CB-AT but also comprises an enhancer sequence upstream of the CMV promoter.
  • FIG. 25 shows a map and nucleotide sequence for the vector of the present invention designated as p43rmsENCB-AT. This vector is the same as p43msENCB-AT except that the enhancer sequence is in an opposite orientation.
  • the subject invention pertains to novel materials and methods for providing gene therapy to a mammal or human having a condition or disorder, such as genetic deficiency disorders, where high levels of expression of a protein are required to treat the disorder or condition.
  • a viral vector is introduced into cells of an animal wherein a therapeutic protein is produced, thereby providing genetic therapy for the animal.
  • a method of the invention comprises introducing into an animal cell or tissue an effective amount of viral particles or vector comprising a recombinant genome which includes heterologous polynucleotide encoding a protein useful in genetic therapy and that can be expressed by the cell or tissue. Expression of the heterologous polynucleotide results in production of the protein.
  • the therapeutic protein encoded by the heterologous polynucleotide is a serum protein.
  • vector material comprising the heterologous polynucleotide is integrated into a chromosome of the cell of the host animal.
  • a recombinant polynucleotide vector of the present invention is derived from adeno-associated virus (AAV) and comprises a constitutive or regulatable promoter capable of driving sufficient levels of expression of the heterologous DNA in the viral vector.
  • AAV adeno-associated virus
  • a recombinant vector of the invention comprises inverted terminal repeat sequences of AAV, such as those described in WO 93/24641.
  • a vector of the present invention comprises polynucleotide sequences of the pTR-UF5 plasmid.
  • the pTR-UF5 plasmid is a modified version of the pTRBSUF/UF1/UF2/UFB series of plasmids (Zolotukhin et al., 1996; Klein et al., 1998).
  • the pTR-UF5 plasmid contains modifications to the sequence encoding the green fluorescent protein (GFP).
  • Promoters useful with the subject invention include, for example, the cytomegalovirus immediate early promoter (CMV), the human elongation factor 1-alpha promoter (EF1), the small nuclear RNA promoters (U1a and U1b), ⁇ -myosin heavy chain promoter, Simian virus 40 promoter (SV 40 ), Rous sarcoma virus promoter (RSV), adenovirus major late promoter, ⁇ -actin promoter and hybrid regulatory element comprising a CMV enhancer/ ⁇ -actin promoter. These promoters have been shown to be active in a wide range of mammalian cells.
  • CMV cytomegalovirus immediate early promoter
  • EF1 human elongation factor 1-alpha promoter
  • U1a and U1b small nuclear RNA promoters
  • ⁇ -myosin heavy chain promoter Simian virus 40 promoter
  • SV 40 Simian virus 40 promoter
  • RSV Rous sarcoma virus promoter
  • the promoters are operably linked with heterologous DNA encoding the protein of interest.
  • operably linked it is intended that the promoter element is positioned relative to the coding sequence to be capable of effecting expression of the coding sequence.
  • Promoters particularly useful for expression of a protein in muscle cells include, for example, hybrid CMV enhancer/ ⁇ -actin promoters, CMV promoters, synthetic promoters and EF1 promoter.
  • Promoters particularly useful for expression of a protein in liver cells include, for example, hybrid CMV enhancer/ ⁇ -actin promoters and EF1 promoters.
  • inducible and cell type specific promoters are also contemplated for use with the vectors of the present invention.
  • Tet-inducible promoters (Clontech, Palo Alto, Calif.) and VP16-LexA promoters (Nettelbeck et al., 1998) can be used in the present invention.
  • the vectors can also include introns inserted into the polynucleotide sequence of the vector as a means for increasing expression of heterologous DNA encoding a protein of interest.
  • an intron can be inserted between a promoter sequence and the region coding for the protein of interest on the vector.
  • Introns can also be inserted in the coding regions.
  • Exemplified in the present invention is the use of intron H from the hAAT gene in a subject vector.
  • Transcriptional enhancer elements which can function to increase levels of transcription from a given promoter can also be included in the vectors of the invention. Enhancers can generally be placed in either orientation, 3′ or 5′, with respect to promoter sequences.
  • synthetic enhancers can be used in the present invention.
  • a synthetic enhancer randomly assembled from Spc5-12-derived elements including muscle-specific elements, serum response factor binding element (SRE), myocyte-specific enhancer factor-1 (NEF-1), myocyte-specific enhancer factor-2 (MEF-2), transcription enhancer factor-1 (TEF-1) and SP-1 can be used in vectors of the invention.
  • Heterologous polynucleotide in the recombinant vector can include, for example, polynucleotides encoding normal, functional proteins which provide therapeutic replacement for normal biological function in animals afflicted with genetic disorders which cause the animal to produce a defective protein, or abnormal or deficient levels of that protein. Proteins, and the polynucleotide sequences that encode them, which can be provided by gene therapy using the subject invention include, but are not limited to, anti-proteases, enzymes, structural proteins, coagulase factors, interleulcins, cytokines, growth factors, interferons, and lymphokines.
  • heterologous DNA in a recombinant AAV vector encodes human alpha-1-antitrypsin protein.
  • any of a number of different nucleotide sequences can be used, based on the degeneracy of the genetic code, to produce a protein of interest for use in the present invention. Accordingly, any nucleotide sequence which encodes a protein of interest comes within the scope of this invention.
  • Biologically active fragments and variants of a protein of interest can easily and routinely be produced by techniques well known in the art. For example, time-controlled Bal31 exonuclease digestion of the full-length DNA followed by expression of the resulting fragments and routine screening can be used to readily identify expression products having the desired activity (Wei et al., 1993).
  • polynucleotide and “polynucleotide sequence” refer to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, would encompass known analogs of natural nucleotides that can function in a similar manner as naturally-occurring nucleotides.
  • Polynucleotide sequences can include both DNA strand sequences, such as that which is transcribed into RNA, and RNA sequences.
  • the polynucleotide sequences include both full-length sequences as well as shorter sequences derived from the full-length sequences.
  • polynucleotide sequence includes sequences, such as degenerate codons of the native sequence or sequences, which may be introduced to provide codon preference in a specific host cell.
  • Polynucleotides of the invention encompass both the sense and antisense strands as either individual strands or in the duplex.
  • polynucleotides of the subject invention also encompass equivalent and variant sequences containing mutations in the exemplified sequences. These mutations can include, for example, nucleotide substitutions, insertions, and deletions as long as the variant sequence functions in a manner similar to the exemplified sequences.
  • the gene therapy methods of the invention can be performed by ex vivo or in vivo treatment of the patient's cells or tissues.
  • Cells and tissues contemplated within the scope of the invention include, for example, muscle, liver, lung, skin and other cells and tissues that are capable of producing and secreting serum proteins.
  • the vectors of the invention can be introduced into suitable cells, cell lines or tissue using methods known in the art.
  • the viral particles and vectors can be introduced into cells or tissue in vitro or in vivo. Methods contemplated include transfection, transduction, injection and inhalation.
  • vectors can be introduced into cells using liposomes containing the subject vectors, by direct transfection with vectors alone, electroporation or by particle bombardment.
  • muscle cells are infected in vivo by injection of viral particles comprising recombinant vector into muscle tissue of an animal.
  • liver cells are infected in vivo by injection of recombinant virus into either the portal vein or peripheral veins.
  • the methods and materials of the subject invention can be used to provide genetic therapy for any conditions or diseases treatable by protein or cytokine infusion such as, for example, alpha-1-antitrypsin deficiency, hemophilia, adenosine deaminase deficiency, and diabetes.
  • the methods and materials of the subject invention can also be used to provide genetic therapy for treating conditions such as, for example, cancer, autoimmune diseases, neurological disorders, immunodeficiency diseases, and bacterial and viral infections.
  • the present invention can be used to provide genetic therapy to a patient wherein cells from the patient are transformed to express and produce interleukins such as interleukin-2.
  • the skilled artisan can for the first time provide therapeutically effective levels of a serum protein through genetic therapy.
  • the therapeutically effective level of serum protein that can be obtained using the subject materials and methods is at least about 1 ⁇ g/ml of protein in serum.
  • the level of serum protein that can be obtained using the present invention is at least about 100 ⁇ g/ml of protein in the serum.
  • the level of serum protein that can be obtained by the present invention is at least about 500 ⁇ g/ml of protein in the serum.
  • Animals that can be treated with the materials and methods of the invention include mammals such as bovine, porcine, equine, ovine, feline and canine mammals.
  • mammals such as bovine, porcine, equine, ovine, feline and canine mammals.
  • the mammals are primates such as chimpanzees and humans.
  • the subject invention also concerns cells containing recombinant vectors of the present invention.
  • the cells can be, for example, animal cells such as mammalian cells.
  • the cells are human cells. More preferably, the cells are human myofibers or myoblasts, hepatocytes or lung cells.
  • a recombinant vector of the present invention is stably integrated into the host cell genome. Cell lines containing the recombinant vectors are also within the scope of the invention.
  • recombinant AAV vectors comprising the human AAT gene (hAAT) using either the CMV promoter (AAV-C-AT) or the human elongation factor 1-alpha (EF1) promoter (AAV-E-AT) to drive expression were constructed and packaged using standard techniques.
  • a murine myoblast cell line, C2C12 was transduced with each vector and expression of HAAT into the medium was measured by ELISA.
  • the EF1 promoter construct resulted in 10-fold higher hAAT expression than the CMV promoter construct.
  • In vivo transduction was performed by injecting doses of up to 1.4 ⁇ 10 13 Dnase-resistant particles of each vector into skeletal muscles of a number of different strains of mice (including C57B1/6, Balb/c, and SCID).
  • the CMV promoter construct resulted in higher levels of expression, with sustained serum levels up to 800 ⁇ g/ml in SCID mice, approximately 10,000-fold higher than those previously observed with proteins secreted from AAV vectors in muscle.
  • serum levels up to 800 ⁇ g/ml in SCID mice, approximately 10,000-fold higher than those previously observed with proteins secreted from AAV vectors in muscle.
  • At lower doses in both C57B1/6 and SCID mice expression was delayed for several weeks, but was sustained for over 10 weeks without declining.
  • increasing dosage AAV vector via transduction of skeletal muscle provides a means for replacing AAT or other serum proteins.
  • Transduction of muscle using the vectors of the subject invention presents several advantages in that it is stable, non-toxic, and relatively nonimmunogenic. Furthermore, certain transcription promoters, such as the CMV promoter, which appear to be markedly down-regulated in other contexts have been found to remain active over time as used in the subject invention.
  • microgram/ml serum levels of a therapeutic protein can be achieved. In an exemplified embodiment, the levels of in vivo protein expression achieved represent a 10,000-fold or more increase over previously published results.
  • a dose-effect relationship was demonstrable within the range of doses used, providing for further increases in expression levels as vector dose is increased.
  • recombinant AAV vectors i.e., C-AT, p43C-AT, P43CB-AT, E-AT and DE-AT comprising the human AAT gene (hAAT) using were constructed and packaged using standard techniques.
  • a murine liver cell line, HO15 was transfected with each vector and expression of hAAT into the medium was measured by ELISA.
  • transduction with the p43CB-AT vector exhibited the highest level of hAAT expression.
  • the p43CB-AT vector also gave higher levels of expression.
  • Portal vein administration appeared to be the more efficient route of administration as mice injected in this manner exhibited higher levels of expression than those receiving peripheral vein injections. Transduction of liver offers the same advantages as for muscle, but hepatocytes may be more efficient at secretion of protein.
  • the dosage of recombinant vector or the virus to be administered to an animal in need of such treatment can be determined by the ordinarily skilled clinician based on various parameters such as mode of administration, duration of treatment, the disease state or condition involved, and the like.
  • recombinant virus of the invention is administered in doses between 10 5 and 10 14 infectious units.
  • the recombinant vectors and virus of the present invention can be prepared in formulations using methods and materials known in the art. Numerous formulations can be found in Remington's Pharmaceutical Sciences, 15 th Edition (1975).
  • FIG. 1 Construction of rAAV plasmids.
  • the rAAV-AAT vector plasmids used for these experiments are depicted diagrammatically (FIG. 1). Briefly, the plasrmid pN2FAT (Garver et al., 1987) plasmid was digested with Xhol to release 1.8-kb fragment containing the human AAT cDNA along with the SV40 promoter and a polyadenylation signal.
  • This fragment was subcloned into a plasmid, pBlueScript (Stratagene) and, after the removal of the SV40 promoter by Hind III digestion and religation, the hAAT cDNA with its polyA signal was released by XbaI and XhoI digestion.
  • This 1.4-kb XbaI-XhoI fragment was then cloned in to the pTR-UF5 (an AAV-inverted terminal repeat-containing vector) plasmid (Zolotukhin et al., 1996) between the XbaI site 3′ to the CMV promoter and the XhoI site 5′ to the polyoma virus enhancer/HSVthyrnidine kinase promoter cassette, which drives neo in that construct.
  • C-AT2 is similar with C-AT except there are SV40 intron and poly (A) sequences flanking the cDNA of HAAT.
  • the p43C-AT was constructed by insertion of hAAT cDNA to an AAV-vector plasmid (p43), which has CMV promoter, intron and poly (A) sequences.
  • the p43CB-AT is derived by replacement of CMV promoter with CMV enhancer and chicken P-actin promoter sequences.
  • the p43C-AT-IN is derived from p43C-AT by insertion of intron 11 sequences of hAAT gene to HAAT cDNA (Brantly et al., 1995).
  • rAAV vectors Packaging of rAAV vectors.
  • Vectors were packaged using a modification of the method described by Ferrari et al. (1997). Briefly, plasmids containing the AAV rep and cap genes (Li et al., 1997) and the Ad genes (E2a, E4 and VA-RNA) were co-transfected along with the appropriate AAV-AAT vector plasmid into 293 cells grown in Cell Factories (Nunc). Cells were harvested by trypsinization and disrupted by freeze-thaw lysis to release vector virions which were then purified by iodixanol gradient ultracentrifugation followed by heparin sepharose affinity column purification. Alternatively, recombinant virus can be prepared according to methods described in Zolotukhin et al. (1999).
  • Vector preparations had their physical titer assessed by quantitative competitive PCR and their biological titer assessed by infectious center assay. The presence of wild-type AAV was also assessed using these same assays with appropriate internal AAV probes.
  • the low-dose C-AT stock had a wt-like AAV particle titer (i.e., positive AAV genome PCR) equal to 0.1 times the recombinant titer but no detectable infectious wtAAV.
  • the other two preparations had wt-like AAV particle titers ⁇ 10 ⁇ 5 times the recombinant titer and no detectable infectious wtAAV.
  • cells were grown under similar conditions and were transduced with vector at multiplicities of infection ranging from 4 ⁇ 10 5 to 4 ⁇ 10 6 particles per cell. Cells were then passaged in the presence of geneticin sulfate (350 ⁇ g/ml) and geneticin-resistant clones were isolated for hAAT secretion studies.
  • AAV-C-AT and AAV-E-AT vectors were obtained from Jackson Laboratories (Bar Harbor, Me.) and were handled under specific pathogen-free conditions under a protocol approved by the University of Florida Institutional Animal Care and Use Committee. Animals were anesthetized by metaphane inhalation and aliquots of vector were injected percutaneously into the quadriceps femoris muscles of both hind limbs. The volume of vector ranged from 50 to 100 ⁇ l per injection site and the total amount of virus injected per animal ranged from 5 ⁇ 10 10 to 1.4 ⁇ 10 13 Dnase-resistant particles.
  • a second antibody (1:1000 dilution of rabbit anti-human AAT, Boehringer Mannheim) was reacted with the captured antigen at 37° C. for 1 hour.
  • Detection was performed using a third antibody incubation (1:800 dilution of goat anti-rabbit IgG-peroxidase conjugate, 37° C.) followed by ophenylenediamine (OPD, Sigma) detection and measurement of the absorbance at 490 nm.
  • ELISA assay for anti-hAAT and anti-AAV VP3 antibodies Wells were coated with antigen (1 ⁇ g of hAAT or 100 ng of VP3) at 4° C. overnight, blocked with 3% BSA and then reacted with dilutions of either test serum or with positive control antibodies at 37° C. for 1 hour. After washing, a goat-anti-mouse IgG-peroxidase conjugate was used as a secondary antibody (1:1500 dilution) to detect bound anti-AAT antibody, using a standard OPD reaction, as described above. Antibody levels were quantitated by comparison with a standard curve generated by reacting dilutions of known positive monoclonal antibodies against VP3 and hAAT.
  • Lymphocvte proliferation assays to detect cell-mediated immune responses. Lymphocyte proliferation assays were performed in order to detect T cell responses to the hAAT and VP3 antigens. Freshly isolated splenocytes were grown in primary culture in 96 well plates coated with 0, 0, 1, 1, and 10 ⁇ g of either HAAT or VP3 in RPMI-C+ medium. On day three, a pulse of 3 H-thyrmidine was added, and the cells were harvested on day 4 for lysis and scintillation counting. Phytohemagglutinin (PHA) was used as a mitogen for positive control wells. A stimulation index was calculated for each antigen dosage level by dividing the counts per minute (cpm) of 3 H-thymidine incorporated in the antigen-stimulated cells by the cpm in a control (unstimulated) well.
  • cpm counts per minute
  • AAV-AAT expression vectors containing one of the CMV, EF1, U1a or U1b promoters were constructed. Each of these constructs were transfected in to the murine C2C12 myoblast cell line. Both the EF1 and the CMV promoter were active for AAT expression, with EF1 construct (AAV-E-AT) expressing 850 ng/10 5 cells/day and the CMV construct (AAV-C-AT) expressing approximately 670 ng/10 5 cells/day, as measured by a human-specific ELISA assay for AAT (FIG. 2). This difference was not statistically significant. The levels of expression from the U1a and U1b constructs were undetectable.
  • lymphocyte proliferation assays were performed using either hAAT or AAV-VP3 for antigenic stimulation of primary splenic lymphocytes harvested at the time of animal sacrifice, 16 weeks post-vector injection. Using this method, no immune responses were detectable in any of the mice.
  • vector-specific PCR (neo primers 5 ′-TATGGGATCGGCCATTGAAC-3′, and 5′-CCTGATGCTCTTC-GTCCAGA-3′, was performed on DNA extracted from 3 SCID mice 16 weeks after injection with the C-AT vector, and PCR products were analyzed by Southern blot analysis with a 32 P-labeled vectorspecific probe (FIG. 8).
  • the state of vector DNA was analyzed using the Hirt procedure (Carter et al, 1983) to separate the low molecular weight episomal DNA from the high molecular weight fraction, which would contain integrated forms and large concatemers.
  • vector DNA was present in the high molecular weight DNA fraction, whereas in only one of the animals was there a signal in the episomal fraction. This result indicates that by 16 weeks most of the vector DNA in our animals was either integrated or in large concatemers.
  • Virus or PBS was delivered into the portal vein using a 30 g needle attached to a 100 ul capillary pipette using mouth delivery via rubber tubing and a Drummond self-locking double layer 0.8 um filter.
  • a small piece of Gel-Foam (0.5 ⁇ 0.5 cm) was applied to the injection site before the needle was removed from the portal vein. The needle was retracted from beneath the Gel-Foam and the piece was held in place with forceps while the intestines were replaced into the peritoneal cavity.
  • the muscle and skin were closed in one layer using 2 simple interrupted 3-0 nylon sutures on an FS-1 cutting needle.
  • Surgeries were performed on a thermoregulated operating board designed to maintain a temperature of 37 degrees.
  • the animals were placed under a heat lamp adjusted to maintain an ambient temperature of approximately 37 degrees and given subcutaneous fluid if there was a significant amount of blood loss during surgery.
  • Serum levels of hAAT in the mice were measured two weeks after injection. Serum levels of about 200-150 ⁇ g/ml HAAT were detected in mice receiving the p43CB-AT vector (FIG. 13). Studies using the E-AT vector show that injection of vector by portal vein led to greater levels of hAAT secretion as compared to E-AT administered by tail vein injection.
  • mice were injected intratracheally with either C-AT or p43CB-AT vector. Serum levels of HAAT in the mice were measured at day 3, 14 and 31 after injection (FIG. 14). The p43CB-AT vector mediated high levels of expression of hAAT in lung.

Abstract

The subject invention concerns materials and methods for gene therapy. One aspect of the invention pertains to vectors which can be used to effect genetic therapy in animals or humans having genetic disorders where expression of high levels of a protein of interest are required to treat or correct the disorder. The subject invention also pertains to methods for treating animals or humans in need of gene therapy to treat or correct a genetic disorder. The materials and methods of the invention can be used to provide therapeutically effective levels of a protein that is non-functional, or that is absent or deficient in the animal or human to be treated. In one embodiment, the materials and methods can be used to treat alpha-1-antitrypsin deficiency.

Description

    CROSS-REFERENCE TO A RELATED APPLICATION
  • This application claims priority from provisional application U.S. Serial No. 60/083,025, filed Apr. 24, 1998.[0001]
  • [0002] The subject invention was made with government support under a research project supported by National Institute of Health NHLBI Grant No. HL 59412. The government has certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • Alpha-1-antitrypsin (AAT) deficiency is the second most common monogenic lung disease in man, accounting for approximately 3% of all early deaths due to obstructive pulmonary disease. AAT protein is normally produced in the liver, secreted into the serum and circulated to the lung where it protects the fine supporting network of elastin fibers from degradation by neutrophil elastase. Current therapy for AAT deficiency includes avoidance of cigarette smoke exposure and weekly intravenous infusions of recombinant human AAT (hAAT) protein. Attempts to devise gene therapy strategies to replace AAT either in the lung itself or within any of a number of other tissues which are capable of AAT secretion have been limited by the short duration of expression from some vectors and by the relatively high circulating levels of AAT which is required for therapeutic effect. Methods of gene therapy have been described in U.S. Pat. No. 5,399,346. [0003]
  • It has recently been demonstrated that adeno-associated virus (AAV) vectors are capable of stable in vivo expression and may be less immunogenic than other viral vectors (Flotte et al., 1996; xiao et al., 1996; Kessler et al., 1996; Jooss et al., 1998). AAV is a nonpathogenic human parvovirus whose life cycle naturally includes a mechanism for long-term latency. In the case of wild-type AAV (wtAAV), this persistence is due to site-specific integration into a site on human chromosome 19 (the AAVSI site) in the majority of cells (Kotin et al., 1990), whereas with recombinant AAV (rAAV) vectors, persistence appears to be due to a combination of episomal persistence and integration into non-chromosome 19 locations (Afione et al., 1996; Kearns et al., 1996). Recombinant AAV latency also differs from that of wtAAV in that wtAAV is rapidly converted to double-stranded DNA in the absence of helper virus (e.g., adenovirus) infection, while with rAAV leading strand synthesis is delayed in the absence of helper virus (Fisher et al., 1996; Ferrari et al., 1996). U.S. Pat. No. 5,658,785 describes adeno-associated virus vectors and methods for gene transfer to cells. [0004]
  • Kessler et al. (1996) demonstrated that murine skeletal myofibers transduced by an rAAV vector were capable of sustained secretion of biologically active human erythropoietin (hEpo), apparently without eliciting a significant immune response against the secreted hepo. See also U.S. Pat. No. 5,858,351 issued to Podsakoffet al. Likewise, Murphy et al. (1997) have observed the expression and secretion of sustained levels of leptin in ob/ob mice after AAV muscle transduction. Brantly et al. (U.S. Pat. No. 5,439,824) disclose methods for increasing expression of AAT using vectors comprising intron II of the human AAT gene. However, the level of leptin expression observed was only in the range of 2 to 5 ng/ml. Therapy for AAT deficiency requires serum levels of at least about 800 μg/ml. Thus, there remains a need in the art for a means of providing therapeutically beneficial levels of a protein to a person in need of such treatment. [0005]
  • BRIEF SUMMARY OF THE INVENTION
  • The subject invention concerns materials and methods for gene therapy. One aspect of the invention pertains to vectors which can be used to provide genetic therapy in animals or humans having a genetic disorder where relatively high levels of expression of a protein is required to treat the disorder. The vectors of the invention are based on adeno-associated virus (AAV). The vectors are designed to provide high levels of expression of heterologous DNA contained in the vector. In one embodiment, the vectors comprise AAV inverted terminal repeat sequences and constitutive or regulatable promoters for driving high levels of gene expression. The subject invention also pertains to methods for treating animals or humans in need of gene therapy, e.g., to correct a genetic deficiency disorder.[0006]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows rAAV-AAT vector cassettes used according to the subject invention. The A-AT and B-AT constructs contain the promoters from the small nuclear RNA genes, U1a and U1b, respectively. The C-AT construct contains the CMV promoter, whereas the E-AT vector uses the human elongation factor 1-α (ELF in the figure) promoter. ITR refers to AAV inverted terminal repeat; An refers to polyA signal; Tk refers to the HSV thymidine kinase promoter; neo refers to the Tn5 neomycin phosphotransferase gene. [0007]
  • FIG. 2 shows hAAT secretion rates in vitro from transiently transfected murine C2C12 myoblast cell line using expression vectors according to the subject invention. C-AT does not differ significantly from E-AT, but both differ from A-AT and B-AT (p<0.05) AAT expression was detected using an ELISA assay specific for human AAT. [0008]
  • FIG. 3 shows hAAT secretion rates in vitro from stably transduced murine C2C12 myoblast cell line using viral particles comprising expression vectors according to the subject invention. The mean rates of secretion from G418-[0009] resistant cultures 1 mo after transduction with either packaged E-AT vector or packaged C-AT vector are shown. In each instance, a “low” multiplicity transduction (4×105 particles/cell) and a high multiplicity transduction (4×106 particles/cell) were performed. E-AT “low” and “high” are greater than “high” multiplicity C-AT (P=0.02) but are not significantly different from each other (n=3). AAT expression was detected using an ELISA assay specific for human AAT.
  • FIG. 4 shows additional constructs tested for hAAT expression. The murine myoblast C2C12 cells were grown in 35-mm wells with approximately 4×10[0010] 5 cell per well and were transfectd with 5 μg of the appropriate plasmid DNA using SUPERFECT transfection (Qiagen Inc., CA). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from three experiments (triplicate in each experiment).
  • Data from transfection experiments indicate that the expression from p43CB-AT was at least three times higher than that from C-AT in vitro. [0011]
  • FIGS. 5A and 5B show sustained secretion of therapeutic levels of HAAT using either the C-AT vector or the E-AT vector in either SCID or C57BL mice. FIG. 5A shows the mean total serum levels of hAAT observed in groups of either SCID (squares) or C57BL (circles) mice receiving either low dose (5×10[0012] 11 particles) (open symbols) or high dose (1.4×1013 particles) (filled symbols) single injections into muscle of the C-AT vector measured at time points ranging from 1 to 16 wk after injection. For each strain, the high-dose curve is significantly different from the low-dose curve (P=1.009 for SCID, P=0.02 for C57BL), but the strains do not differ from each other. FIG. 5B shows analogous data with the E-AT vector. None of these differences were significant.
  • FIG. 5C shows long term secretion of HAAT from murine muscle transduced with C-AT. C57B1/6 or C57B1/6-SCID mice received 3.5×10[0013] 10 IU, 1.4×1013 particles/mouse. One year after injection, serum hAAT levels were still 400 μg/ml in C57B1/6-SCID and 200 μg/ml in C57B1/6. This level are comparable with the peak levels observed (800 or 400 μg/ml, respectively).
  • FIG. 6 shows an immunoblot of sera taken from several of the C-AT vector-treated mice at 11 weeks after vector administration. Ten microliters of a 1:100 dilution of serum was electrophoresed by 10% SDS/PAGE, blotted, and incubated with 1:1,500 dilution of goat anti-hAAT-horseradish peroxidase conjugate (Cappel/ICN). Samples from three high-dose SCID (h1-h3), one high-dose C57B1 (h3), and three low-dose C57B1 (101-103) were included, along with one negative control (saline-injected=sal) serum to indicate the level of reactivity with endogenous mAAT. As a standard, hAAT was added either to negative-control C57B1 serum (first hAAT lane) or to PBS (second hAAT) lane to final equivalent serum concentration of 100 μg/ml. [0014]
  • FIGS. 7A and 7B show that some BALB/c mice mount humoral immune responses to hAAT, which correlate with lower serum levels but no observable toxicity. FIG. 7A shows serum hAAT levels and FIG. 7B shows serum anti-hAAT antibody levels as determined by ELISA performed on serum taken from mice injected with [0015] 1×10 11 particles of the C-AT vector. Each set of symbols represents an individual animal (□, no. 1; Δ, no. 2; ∘, no. 3). Note the inverse correlation between the presence of antibody and the presence of circulating hAAT.
  • FIG. 8 shows the persistence of rAAV-AAT vector DNA in high molecular weight form. PCR products were amplified from DNA prepared by Hirt extraction from three SCID mice injected 16 wk earlier with 5×10[0016] 11 resistant-particles of C-AT and analyzed by Southern blot. The high molecular weight Hirt pellet (genomic DNA lanes) and the low molecular weight supernatant (episomal DNA lanes) were analyzed separately. Control lanes include a sample in which an hAAT cDNA plasmid was the template DNA (+) and a control in which water was the template (−). In this internal PCR reaction, a 500-bp product is expected regardless of whether or not the vector genome is integrated.
  • FIG. 9 shows serum hAAT in C57B1/6 mice transduced with C-AT and p43CB-AT. C57B1/6 mice were injected in muscle with C-AT (3.5×10[0017] 10 IU/mouse, 1×1012 particles/mouse) or p43CB-AT (6×109 IU, 1×1012 particles/mouse). The level of hAAT from p43CB-AT were projected based on an estimation of the equivalent dosage (infectious unit) of C-AT.
  • FIG. 10 shows enhancement of CMV promoter activity by a synthetic enhancer in C2C12 cells. The murine myoblast C2C12 cells were grown in 35-mm wells with approximately 4×10[0018] 5 cell per well and were transfected with 5 μg of p43msENC-AT vector DNA using SUPERFECT transfection (Qiagen Inc, CA). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from one experiment (triplicate).
  • FIG. 11 shows secretion of hAAT from mouse liver cells (HO15) transfected with different constructs. The murine liver cells (HO15) were grown in 35-mm wells with approximately 4×10[0019] 5 cell per well and were transfected with 5 μg of the plasmid DNA using LIPOFECTAMINE reagents (Life Technologies Inc, MD). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from two experiments (triplicate).
  • FIG. 12 shows secretion of HAAT from mouse liver cells (HO15) transfected using different methods. The murine liver cells (HO15) were grown in 35-mm wells with approximately 4×10[0020] 5 cell per well and were transfected with 5 μg of the p43CB-AT vector using SUPERFECT (Qiagen Inc., CA), FuGENE (Boehringer Mannhem Co, IN), Lipofectin, LIPOFECTAMNE (ife Technologies Inc, MD) reagents and Calcium phosphate (CA-PO4) transfection. Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA. Each bar represents the mean of results from one experiment (triplicate).
  • FIG. 13 shows hAAT secretion from mouse liver transduced with rAAV. C57B1/6 mice were injected with either p43CB-AT, C-AT or E-AT vector either by portal vein or tail vein injection. PV=portal vein injection. TV=tail vein injection. [0021]
  • FIG. 14 shows serum HAAT levels in C57B1/6 mice after intratracheal (IT) injection of C-AT orp43CB-AT vector. Mice received either 10[0022] 9 IU of C-AT (open circles), 109 IU of p43CB-AT (open triangles) or 1010 IU of p43CB-AT (open squares).
  • FIG. 15 shows a map and nucleotide sequence for the vector of the present invention designated as C-AT. [0023]
  • FIG. 16 shows a map and nucleotide sequence for the vector of the present invention designated as E-AT. [0024]
  • FIG. 17 shows a map and nucleotide sequence for the vector of the present invention designated as DE-AT. [0025]
  • FIG. 18 shows a map and nucleotide sequence for the vector of the present invention designated as p43C-AT. [0026]
  • FIG. 19 shows a map and nucleotide sequence for the vector of the present invention designated as p43C-AT-IN. This vector includes intron II from human AAT gene to enhance transcription. [0027]
  • FIG. 20 shows a map and nucleotide sequence for the vector of the present invention designated as p43CB-AT. [0028]
  • FIG. 21 shows a map and nucleotide sequence for the vector of the present invention designated as C-AT2. [0029]
  • FIG. 22 shows a map and nucleotide sequence for the vector of the present invention designated as p43 msENC-AT. This vector is similar to p43C-AT but also comprises an enhancer sequence upstream of the CMV promoter. [0030]
  • FIG. 23 shows a map and nucleotide sequence for the vector of the present invention designated as p43rmsENC-AT. This vector is the same as the p43msENC-AT vector except that the enhancer sequence is in an opposite orientation. [0031]
  • FIG. 24 shows a map and nucleotide sequence for the vector of the present invention designated as p43 msENCB-AT. This vector is similar to p43CB-AT but also comprises an enhancer sequence upstream of the CMV promoter. [0032]
  • FIG. 25 shows a map and nucleotide sequence for the vector of the present invention designated as p43rmsENCB-AT. This vector is the same as p43msENCB-AT except that the enhancer sequence is in an opposite orientation.[0033]
  • DETAILED DISCLOSURE OF THE INVENTION
  • The subject invention pertains to novel materials and methods for providing gene therapy to a mammal or human having a condition or disorder, such as genetic deficiency disorders, where high levels of expression of a protein are required to treat the disorder or condition. In one method of the subject invention, a viral vector is introduced into cells of an animal wherein a therapeutic protein is produced, thereby providing genetic therapy for the animal. In one embodiment, a method of the invention comprises introducing into an animal cell or tissue an effective amount of viral particles or vector comprising a recombinant genome which includes heterologous polynucleotide encoding a protein useful in genetic therapy and that can be expressed by the cell or tissue. Expression of the heterologous polynucleotide results in production of the protein. Preferably, the therapeutic protein encoded by the heterologous polynucleotide is a serum protein. In a preferred embodiment, vector material comprising the heterologous polynucleotide is integrated into a chromosome of the cell of the host animal. [0034]
  • In one embodiment, a recombinant polynucleotide vector of the present invention is derived from adeno-associated virus (AAV) and comprises a constitutive or regulatable promoter capable of driving sufficient levels of expression of the heterologous DNA in the viral vector. Preferably, a recombinant vector of the invention comprises inverted terminal repeat sequences of AAV, such as those described in WO 93/24641. In a preferred embodiment, a vector of the present invention comprises polynucleotide sequences of the pTR-UF5 plasmid. The pTR-UF5 plasmid is a modified version of the pTRBSUF/UF1/UF2/UFB series of plasmids (Zolotukhin et al., 1996; Klein et al., 1998). The pTR-UF5 plasmid contains modifications to the sequence encoding the green fluorescent protein (GFP). [0035]
  • Promoters useful with the subject invention include, for example, the cytomegalovirus immediate early promoter (CMV), the human elongation factor 1-alpha promoter (EF1), the small nuclear RNA promoters (U1a and U1b), α-myosin heavy chain promoter, [0036] Simian virus 40 promoter (SV40), Rous sarcoma virus promoter (RSV), adenovirus major late promoter, β-actin promoter and hybrid regulatory element comprising a CMV enhancer/β-actin promoter. These promoters have been shown to be active in a wide range of mammalian cells.
  • The promoters are operably linked with heterologous DNA encoding the protein of interest. By “operably linked,” it is intended that the promoter element is positioned relative to the coding sequence to be capable of effecting expression of the coding sequence. [0037]
  • Promoters particularly useful for expression of a protein in muscle cells include, for example, hybrid CMV enhancer/β-actin promoters, CMV promoters, synthetic promoters and EF1 promoter. Promoters particularly useful for expression of a protein in liver cells include, for example, hybrid CMV enhancer/β-actin promoters and EF1 promoters. [0038]
  • Also contemplated for use with the vectors of the present invention are inducible and cell type specific promoters. For example, Tet-inducible promoters (Clontech, Palo Alto, Calif.) and VP16-LexA promoters (Nettelbeck et al., 1998) can be used in the present invention. [0039]
  • The vectors can also include introns inserted into the polynucleotide sequence of the vector as a means for increasing expression of heterologous DNA encoding a protein of interest. For example, an intron can be inserted between a promoter sequence and the region coding for the protein of interest on the vector. Introns can also be inserted in the coding regions. Exemplified in the present invention is the use of intron H from the hAAT gene in a subject vector. Transcriptional enhancer elements which can function to increase levels of transcription from a given promoter can also be included in the vectors of the invention. Enhancers can generally be placed in either orientation, 3′ or 5′, with respect to promoter sequences. In addition to the natural enhancers, synthetic enhancers can be used in the present invention. For example, a synthetic enhancer randomly assembled from Spc5-12-derived elements including muscle-specific elements, serum response factor binding element (SRE), myocyte-specific enhancer factor-1 (NEF-1), myocyte-specific enhancer factor-2 (MEF-2), transcription enhancer factor-1 (TEF-1) and SP-1 (Li et al., 1999; Deshpande et al., 1997; Stewart et al., 1996; Mitchell et al., 1989; Briggs et al., 1986; Pitluk et al., 1991) can be used in vectors of the invention. [0040]
  • Heterologous polynucleotide in the recombinant vector can include, for example, polynucleotides encoding normal, functional proteins which provide therapeutic replacement for normal biological function in animals afflicted with genetic disorders which cause the animal to produce a defective protein, or abnormal or deficient levels of that protein. Proteins, and the polynucleotide sequences that encode them, which can be provided by gene therapy using the subject invention include, but are not limited to, anti-proteases, enzymes, structural proteins, coagulase factors, interleulcins, cytokines, growth factors, interferons, and lymphokines. In an exemplified embodiment, heterologous DNA in a recombinant AAV vector encodes human alpha-1-antitrypsin protein. [0041]
  • As those of ordinary skill in the art will appreciate, any of a number of different nucleotide sequences can be used, based on the degeneracy of the genetic code, to produce a protein of interest for use in the present invention. Accordingly, any nucleotide sequence which encodes a protein of interest comes within the scope of this invention. Biologically active fragments and variants of a protein of interest can easily and routinely be produced by techniques well known in the art. For example, time-controlled Bal31 exonuclease digestion of the full-length DNA followed by expression of the resulting fragments and routine screening can be used to readily identify expression products having the desired activity (Wei et al., 1993). [0042]
  • As used herein, the terms “polynucleotide” and “polynucleotide sequence” refer to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, would encompass known analogs of natural nucleotides that can function in a similar manner as naturally-occurring nucleotides. Polynucleotide sequences can include both DNA strand sequences, such as that which is transcribed into RNA, and RNA sequences. The polynucleotide sequences include both full-length sequences as well as shorter sequences derived from the full-length sequences. It is understood that a particular polynucleotide sequence includes sequences, such as degenerate codons of the native sequence or sequences, which may be introduced to provide codon preference in a specific host cell. Polynucleotides of the invention encompass both the sense and antisense strands as either individual strands or in the duplex. [0043]
  • The polynucleotides of the subject invention also encompass equivalent and variant sequences containing mutations in the exemplified sequences. These mutations can include, for example, nucleotide substitutions, insertions, and deletions as long as the variant sequence functions in a manner similar to the exemplified sequences. [0044]
  • The gene therapy methods of the invention can be performed by ex vivo or in vivo treatment of the patient's cells or tissues. Cells and tissues contemplated within the scope of the invention include, for example, muscle, liver, lung, skin and other cells and tissues that are capable of producing and secreting serum proteins. The vectors of the invention can be introduced into suitable cells, cell lines or tissue using methods known in the art. The viral particles and vectors can be introduced into cells or tissue in vitro or in vivo. Methods contemplated include transfection, transduction, injection and inhalation. For example, vectors can be introduced into cells using liposomes containing the subject vectors, by direct transfection with vectors alone, electroporation or by particle bombardment. In an exemplified embodiment, muscle cells are infected in vivo by injection of viral particles comprising recombinant vector into muscle tissue of an animal. In another embodiment, liver cells are infected in vivo by injection of recombinant virus into either the portal vein or peripheral veins. [0045]
  • The methods and materials of the subject invention can be used to provide genetic therapy for any conditions or diseases treatable by protein or cytokine infusion such as, for example, alpha-1-antitrypsin deficiency, hemophilia, adenosine deaminase deficiency, and diabetes. The methods and materials of the subject invention can also be used to provide genetic therapy for treating conditions such as, for example, cancer, autoimmune diseases, neurological disorders, immunodeficiency diseases, and bacterial and viral infections. For example, the present invention can be used to provide genetic therapy to a patient wherein cells from the patient are transformed to express and produce interleukins such as interleukin-2. [0046]
  • Using the materials and methods of the subject invention, the skilled artisan can for the first time provide therapeutically effective levels of a serum protein through genetic therapy. In a preferred embodiment, the therapeutically effective level of serum protein that can be obtained using the subject materials and methods is at least about 1 μg/ml of protein in serum. Preferably, the level of serum protein that can be obtained using the present invention is at least about 100 μg/ml of protein in the serum. Most preferably, the level of serum protein that can be obtained by the present invention is at least about 500 μg/ml of protein in the serum. [0047]
  • Animals that can be treated with the materials and methods of the invention include mammals such as bovine, porcine, equine, ovine, feline and canine mammals. Preferably, the mammals are primates such as chimpanzees and humans. [0048]
  • The subject invention also concerns cells containing recombinant vectors of the present invention. The cells can be, for example, animal cells such as mammalian cells. Preferably, the cells are human cells. More preferably, the cells are human myofibers or myoblasts, hepatocytes or lung cells. In a preferred embodiment, a recombinant vector of the present invention is stably integrated into the host cell genome. Cell lines containing the recombinant vectors are also within the scope of the invention. [0049]
  • In an exemplified embodiment, recombinant AAV vectors comprising the human AAT gene (hAAT) using either the CMV promoter (AAV-C-AT) or the human elongation factor 1-alpha (EF1) promoter (AAV-E-AT) to drive expression were constructed and packaged using standard techniques. A murine myoblast cell line, C2C12, was transduced with each vector and expression of HAAT into the medium was measured by ELISA. In vitro, the EF1 promoter construct resulted in 10-fold higher hAAT expression than the CMV promoter construct. In vivo transduction was performed by injecting doses of up to 1.4×10[0050] 13 Dnase-resistant particles of each vector into skeletal muscles of a number of different strains of mice (including C57B1/6, Balb/c, and SCID). In vivo, the CMV promoter construct resulted in higher levels of expression, with sustained serum levels up to 800 μg/ml in SCID mice, approximately 10,000-fold higher than those previously observed with proteins secreted from AAV vectors in muscle. At lower doses in both C57B1/6 and SCID mice, expression was delayed for several weeks, but was sustained for over 10 weeks without declining. Thus, increasing dosage AAV vector via transduction of skeletal muscle provides a means for replacing AAT or other serum proteins.
  • Transduction of muscle using the vectors of the subject invention presents several advantages in that it is stable, non-toxic, and relatively nonimmunogenic. Furthermore, certain transcription promoters, such as the CMV promoter, which appear to be markedly down-regulated in other contexts have been found to remain active over time as used in the subject invention. Using the materials and methods of the subject invention, microgram/ml serum levels of a therapeutic protein can be achieved. In an exemplified embodiment, the levels of in vivo protein expression achieved represent a 10,000-fold or more increase over previously published results. In addition, a dose-effect relationship was demonstrable within the range of doses used, providing for further increases in expression levels as vector dose is increased. [0051]
  • In another embodiment of the invention, recombinant AAV vectors i.e., C-AT, p43C-AT, P43CB-AT, E-AT and DE-AT comprising the human AAT gene (hAAT) using were constructed and packaged using standard techniques. A murine liver cell line, HO15, was transfected with each vector and expression of hAAT into the medium was measured by ELISA. In vitro, transduction with the p43CB-AT vector exhibited the highest level of hAAT expression. In vivo, the p43CB-AT vector also gave higher levels of expression. Portal vein administration appeared to be the more efficient route of administration as mice injected in this manner exhibited higher levels of expression than those receiving peripheral vein injections. Transduction of liver offers the same advantages as for muscle, but hepatocytes may be more efficient at secretion of protein. [0052]
  • The dosage of recombinant vector or the virus to be administered to an animal in need of such treatment can be determined by the ordinarily skilled clinician based on various parameters such as mode of administration, duration of treatment, the disease state or condition involved, and the like. Typically, recombinant virus of the invention is administered in doses between 10[0053] 5 and 1014 infectious units. The recombinant vectors and virus of the present invention can be prepared in formulations using methods and materials known in the art. Numerous formulations can be found in Remington's Pharmaceutical Sciences, 15th Edition (1975).
  • All publications and patents cited herein are expressly incorporated by reference. [0054]
  • Materials and Methods
  • Construction of rAAV plasmids. The rAAV-AAT vector plasmids used for these experiments are depicted diagrammatically (FIG. 1). Briefly, the plasrmid pN2FAT (Garver et al., 1987) plasmid was digested with Xhol to release 1.8-kb fragment containing the human AAT cDNA along with the SV40 promoter and a polyadenylation signal. This fragment was subcloned into a plasmid, pBlueScript (Stratagene) and, after the removal of the SV40 promoter by Hind III digestion and religation, the hAAT cDNA with its polyA signal was released by XbaI and XhoI digestion. This 1.4-kb XbaI-XhoI fragment was then cloned in to the pTR-UF5 (an AAV-inverted terminal repeat-containing vector) plasmid (Zolotukhin et al., 1996) between the [0055] XbaI site 3′ to the CMV promoter and the XhoI site 5′ to the polyoma virus enhancer/HSVthyrnidine kinase promoter cassette, which drives neo in that construct. This yielded the pAAV-CMV-AAT construct (C-AT). Analogous constructs using the promoter from the small nuclear RNA proteins, U1a and U1b, (to give the A-AT and B-AT constructs, respectively) and human elongation factor 1-alpha (EF1) promoter (to give the E-AT construct) were constructed by substituting each of these promoter cassettes in place of the CMV promoter, between the KpnI and XbaI sites.
  • The construct, DE-AT derived from E-AT by deletion of the silencer (352 bp) by SAC II-cut (Wakabayashi-Ito et al., 1994). C-AT2 is similar with C-AT except there are SV40 intron and poly (A) sequences flanking the cDNA of HAAT. The p43C-AT was constructed by insertion of hAAT cDNA to an AAV-vector plasmid (p43), which has CMV promoter, intron and poly (A) sequences. The p43CB-AT is derived by replacement of CMV promoter with CMV enhancer and chicken P-actin promoter sequences. The p43C-AT-IN is derived from p43C-AT by insertion of [0056] intron 11 sequences of hAAT gene to HAAT cDNA (Brantly et al., 1995).
  • Packaging of rAAV vectors. Vectors were packaged using a modification of the method described by Ferrari et al. (1997). Briefly, plasmids containing the AAV rep and cap genes (Li et al., 1997) and the Ad genes (E2a, E4 and VA-RNA) were co-transfected along with the appropriate AAV-AAT vector plasmid into 293 cells grown in Cell Factories (Nunc). Cells were harvested by trypsinization and disrupted by freeze-thaw lysis to release vector virions which were then purified by iodixanol gradient ultracentrifugation followed by heparin sepharose affinity column purification. Alternatively, recombinant virus can be prepared according to methods described in Zolotukhin et al. (1999). [0057]
  • Vector preparations had their physical titer assessed by quantitative competitive PCR and their biological titer assessed by infectious center assay. The presence of wild-type AAV was also assessed using these same assays with appropriate internal AAV probes. The highdose C-AT stock had a particle-titer of 2.0×10[0058] 14 particles/ml and an infectious titer of 5.0×1011 infectious units (i.u.)/ml (particle to i.u. ratio=400:1). The low-dose C-AT measured 8×1012 particles/ml and 1.2×1010 i.u./ml (particle to i.u.=667:1). For the E-AT experiments, the titers were 1×1013 particles/ml and 2.5×1010 i.u./ml (particle to i.u.=400:1). The low-dose C-AT stock had a wt-like AAV particle titer (i.e., positive AAV genome PCR) equal to 0.1 times the recombinant titer but no detectable infectious wtAAV. The other two preparations had wt-like AAV particle titers <10−5 times the recombinant titer and no detectable infectious wtAAV.
  • In vitro transfection and transduction experiments. The C2C12 murine myoblast line was used for in vitro transfection and transduction experiments. Cells were grown in 35-mm wells with approximately 4×10[0059] 5 cells per well and transfected with 5 μg of each plasmid DNA using SUPERFECT (Qiagen Corp.). Secretion of hAAT into the medium was assessed at 2 days after transfection using an antigen-capture ELISA assay with standards (Brantly et al., 1991). An SV40 promoter luciferase-expression plasmid, pGL2 (Promega), was used as an internal control. For transduction experiments, cells were grown under similar conditions and were transduced with vector at multiplicities of infection ranging from 4×105 to 4×106 particles per cell. Cells were then passaged in the presence of geneticin sulfate (350 μg/ml) and geneticin-resistant clones were isolated for hAAT secretion studies.
  • In vivo injection of AAV-C-AT and AAV-E-AT vectors into murine muscle. Mice strains (C57B1/6, SCID, and Balb/c) were obtained from Jackson Laboratories (Bar Harbor, Me.) and were handled under specific pathogen-free conditions under a protocol approved by the University of Florida Institutional Animal Care and Use Committee. Animals were anesthetized by metaphane inhalation and aliquots of vector were injected percutaneously into the quadriceps femoris muscles of both hind limbs. The volume of vector ranged from 50 to 100 μl per injection site and the total amount of virus injected per animal ranged from 5×10[0060] 10 to 1.4×1013 Dnase-resistant particles.
  • Antigen capture ELISA assay for hAAT expression. Microtiter plates ([0061] Immulon 4, Dynex Technologies, Chantilly, Va.) were coated with 100 μl of a 1:200 dilution of goat anti-human AAT (CAPPEL/ICN) in Vollers buffer (Na2CO3=2.76 g, NaHCO3=1.916 g, NaN3=0.2 g, d.H2O=1 liter, Adjust PH=9.6) overnight at 4° C. After washing, standards and unknown samples containing HAAT were incubated in the plates at 37° C. for 1 hour. After blocking in 3% BSA in PBS-Tween 20 at 37° C. for 1 hour, a second antibody (1:1000 dilution of rabbit anti-human AAT, Boehringer Mannheim) was reacted with the captured antigen at 37° C. for 1 hour. Detection was performed using a third antibody incubation (1:800 dilution of goat anti-rabbit IgG-peroxidase conjugate, 37° C.) followed by ophenylenediamine (OPD, Sigma) detection and measurement of the absorbance at 490 nm.
  • ELISA assay for anti-hAAT and anti-AAV VP3 antibodies. Wells were coated with antigen (1 μg of hAAT or 100 ng of VP3) at 4° C. overnight, blocked with 3% BSA and then reacted with dilutions of either test serum or with positive control antibodies at 37° C. for 1 hour. After washing, a goat-anti-mouse IgG-peroxidase conjugate was used as a secondary antibody (1:1500 dilution) to detect bound anti-AAT antibody, using a standard OPD reaction, as described above. Antibody levels were quantitated by comparison with a standard curve generated by reacting dilutions of known positive monoclonal antibodies against VP3 and hAAT. [0062]
  • Lymphocvte proliferation assays to detect cell-mediated immune responses. Lymphocyte proliferation assays were performed in order to detect T cell responses to the hAAT and VP3 antigens. Freshly isolated splenocytes were grown in primary culture in 96 well plates coated with 0, 0, 1, 1, and 10 μg of either HAAT or VP3 in RPMI-C+ medium. On day three, a pulse of [0063] 3H-thyrmidine was added, and the cells were harvested on day 4 for lysis and scintillation counting. Phytohemagglutinin (PHA) was used as a mitogen for positive control wells. A stimulation index was calculated for each antigen dosage level by dividing the counts per minute (cpm) of 3H-thymidine incorporated in the antigen-stimulated cells by the cpm in a control (unstimulated) well.
  • Following are examples which illustrate procedures for practicing the invention. These examples should not be construed as limiting. All percentages are by weight and all solvent mixture proportions are by volume unless otherwise noted. [0064]
  • EXAMPLE 1 In vitro Studies in Murine C2C12 Myoblasts
  • In order to determine the relative strength of a number of constitutively active promoters in the context of AAV-AAT vectors, packageable AAV-AAT expression vectors containing one of the CMV, EF1, U1a or U1b promoters (FIG. 1) were constructed. Each of these constructs were transfected in to the murine C2C12 myoblast cell line. Both the EF1 and the CMV promoter were active for AAT expression, with EF1 construct (AAV-E-AT) expressing 850 ng/10[0065] 5 cells/day and the CMV construct (AAV-C-AT) expressing approximately 670 ng/105 cells/day, as measured by a human-specific ELISA assay for AAT (FIG. 2). This difference was not statistically significant. The levels of expression from the U1a and U1b constructs were undetectable.
  • In order to better characterize the level and duration of expression in the setting of vector transduction, cultures of C2C12 cells were transduced with either AAV-E-AT or AAV-C-AT at multiplicities of infection ranging from 4×10[0066] 5 to 4×106 Dnase-resistant particles per cell. Cells were then selected for expression of the neo gene (present in each of the AAV constructs) by growth in G418-containing medium. Several cell clones and pooled cell populations were independently analyzed for AAT expression at four weeks post-transduction (FIG. 3). There was a clear trend toward higher levels of expression at higher multiplicities of infection, and the E-AT construct expressed at least 10-fold greater quantities under all conditions in these long-term cultures. The most active E-AT clone expressed hAAT at a rate of over 1400 ng/105 cells/day.
  • EXAMPLE 2 In vivo Expression of hAAT from Murine Skeletal Muscle
  • In order to determine whether the AAV-AAT constructs would be active in vivo in skeletal muscle, doses of vector were injected into the quadriceps femoris muscle of mice. Circulating serum levels of HAAT were then measured for 11 to 15 weeks after the initial injection. Four saline-injected animals from each mouse strain served as controls. In the case of the C-AT vector (FIG. 5A), levels of expression were sufficient to achieve serum levels in excess of 800 μg/ml in SCID mice after a single injection of 1.4×10[0067] 13 particles. A dose-effect relationship was observed, with expression levels in SCID being at least 20-fold lower at the 5×1011 particle dose. The levels of expression increased over the first several weeks after injection and were stable thereafter until the time of sacrifice. Since HAAT has a half-life of less than 1 week, this indicated continuous expression. Levels from C57B1/6 mice were comparable, and also achieved values close to the therapeutic range. In similar studies, two of three Balb/c mice injected with 1×1011 particles of the C-AT vector did not express hAAT at detectable levels. Both of these were found to have developed high levels of anti-hAAT antibodies.
  • Surprisingly, expression levels from the AAV-E-AT vector after in vivo injection were modestly lower than those seen with the C-AT vector (FIG. 5B), with maximal levels of approximately 250 ng/ml at the 5×10[0068] 11 dose at and beyond 7 weeks in SCID mice. When the dose was further increased to 1×1012 particles, levels of approximately 1200 ng/ml were observed. These levels were stable for one year post-injection (FIG. 5C). Levels observed in SCID and immune competent C57B1/6 mice were similar.
  • EXAMPLE 3 Imunologic Studies
  • In studies in Balb/c mice, antibody levels against hAAT were high in 2 of 3 animals injected. The one which did not have circulating anti-hAAT was the only animal with levels of hAAT expression similar to those in the C57B1/6 and SCID groups. The high-dose C57C-AT injection group had detectable levels of antibody directed against VP3, but not hAAT. [0069]
  • In order to determine whether any cell-mediated immune responses were mounted, lymphocyte proliferation assays were performed using either hAAT or AAV-VP3 for antigenic stimulation of primary splenic lymphocytes harvested at the time of animal sacrifice, 16 weeks post-vector injection. Using this method, no immune responses were detectable in any of the mice. [0070]
  • EXAMPLE 4 Lack of Toxicity from Direct Vector Injection
  • In order to determine whether there was any direct toxicity, inflammation, or neoplastic change associated with vector injection, animals underwent complete necropsies. Histopathologic examination was performed on 5 μm sections taken from the site of vector injection and from a panel of other organs, including the brain, heart, lungs, trachea, pancreas, spleen, liver, kidney, and jejunum. No histologic abnormalities were observed in any of these sites, even among those mice which developed humanol immune responses against hAAT. [0071]
  • EXAMPLE 5 Molecular Evidence of AAV-AAT Vector Persistence
  • To confirm the presence of vector DNA, a vector-specific PCR ([0072] neo primers 5′-TATGGGATCGGCCATTGAAC-3′, and 5′-CCTGATGCTCTTC-GTCCAGA-3′, was performed on DNA extracted from 3 SCID mice 16 weeks after injection with the C-AT vector, and PCR products were analyzed by Southern blot analysis with a 32P-labeled vectorspecific probe (FIG. 8). The state of vector DNA was analyzed using the Hirt procedure (Carter et al, 1983) to separate the low molecular weight episomal DNA from the high molecular weight fraction, which would contain integrated forms and large concatemers. In each case, vector DNA was present in the high molecular weight DNA fraction, whereas in only one of the animals was there a signal in the episomal fraction. This result indicates that by 16 weeks most of the vector DNA in our animals was either integrated or in large concatemers.
  • EXAMPLE 6 In vivo Expression of HAAT from Murine Liver
  • Portal vein or tail vein injections were performed on 18 female C57BL/6 mice 8-10 weeks of age. The injection volume was 100 μl per mouse. [0073]
  • Each group had the following parameters: [0074]
  • 1. Group 1: 100 μl of PBS n=4. [0075]
  • 2. Group 2: 100 μl of p43CB-AT (3×10[0076] 10 IU/animal) n=3.
  • 3. Group 3: 100 μl of p43CB-AT (4×10[0077] 9 IU/animal) n=4.
  • 4. Group 4: 100 μl of C-AT (4×10[0078] 9 IU/animal) n—2.
  • 5. Group 5: 100 μl of E-AT (4×10[0079] 9 IU/animal) n=4.
  • 6. Group 6: EATM TV=100 μl by tail vein injection of E-AT (4×10[0080] 9 IU/animal) n—3.
  • 7. Group 0: 100 μl of PBS by tail vein injection n=2. [0081]
  • A total of 22 animals were used in this study. [0082]
  • All animals were anesthetized with 2-2-2 tribromoethanol (Avertin) using a working solution of 20 mg/ml at a dosage of 0.5 mg/g IP. A 2 cm ventral midline abdominal incision was made from the pubic symphysis extending cranially to the xyphoid process through skin and muscle layers. The portal vein was exposed by retracting the intestines and associated mesentery to the left side of the aninal. Additionally, the quadrate and right medial lobes of the liver were retracted cranially. Intestines and peritoneal cavity were continuously lavaged with 0.9% NaC.l [0083]
  • Virus or PBS was delivered into the portal vein using a 30 g needle attached to a 100 ul capillary pipette using mouth delivery via rubber tubing and a Drummond self-locking double layer 0.8 um filter. A small piece of Gel-Foam (0.5×0.5 cm) was applied to the injection site before the needle was removed from the portal vein. The needle was retracted from beneath the Gel-Foam and the piece was held in place with forceps while the intestines were replaced into the peritoneal cavity. [0084]
  • The muscle and skin were closed in one layer using 2 simple interrupted 3-0 nylon sutures on an FS-1 cutting needle. Surgeries were performed on a thermoregulated operating board designed to maintain a temperature of 37 degrees. For recovery from anesthesia, the animals were placed under a heat lamp adjusted to maintain an ambient temperature of approximately 37 degrees and given subcutaneous fluid if there was a significant amount of blood loss during surgery. [0085]
  • Serum levels of hAAT in the mice were measured two weeks after injection. Serum levels of about 200-150 μg/ml HAAT were detected in mice receiving the p43CB-AT vector (FIG. 13). Studies using the E-AT vector show that injection of vector by portal vein led to greater levels of hAAT secretion as compared to E-AT administered by tail vein injection. [0086]
  • EXAMPLE 7 In vivo Expression of hAAT from Murine Lung
  • Mice were injected intratracheally with either C-AT or p43CB-AT vector. Serum levels of HAAT in the mice were measured at [0087] day 3, 14 and 31 after injection (FIG. 14). The p43CB-AT vector mediated high levels of expression of hAAT in lung.
  • It should be understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and the scope of the appended claims. [0088]
  • REFERENCES
  • U.S. Pat. No. 5,399,346 [0089]
  • U.S. Pat. No. 5,439,824 [0090]
  • U.S. Pat. No. 5,658,785 [0091]
  • U.S. Pat. No. 5,858,351 [0092]
  • Afione, S. A., Conrad, C. K., Kearns, W. G., Chunduru, S., Adams, R., Reynolds, T. C., Guggino, W. B., Cutting, G. R., Carter, B. J. and Flotte, T. R. (1996) [0093] J. Virol. 70:3235-3241.
  • Brantly, M. L., Wittes, J. T., Vogelmeier, C. F., Hubbard, R. C., Fells, G. A., Crystal, R. G. (1991) [0094] Chest 100:703-708.
  • Briggs, M. R., Kadonga, J. T., Bell, S. P., Tjian, R. (1986) [0095] Science 234:47-52.
  • Carter et al. (1983) [0096] Virology 126:505-515.
  • Deshpande, N., Chopra, A., Rangarajan, A., Shashidhara, L. S., Rodrigues, V., Krishna, S. (1997) [0097] J. Biol. Chem 272(16):10664-10668.
  • Garver, R. I., Jr., Chytil, A., Courtney, M., Crystal, R. G. (1987) [0098] Science 237:762-764.
  • Ferrari, F. K., Samulski, T., Shenk, T., Samulski, R. J. (1996) [0099] J. Virol. 70:3227-3234.
  • Ferrari, F. K., Xiao, X., McCarty, D. M., Sarnulski, R. J. (1997) [0100] Nature Med 3:1295-97.
  • Fisher, K. J., Gao, G. P., Weitzman, M. D., DeMatteo, R., Burda, J. F., Wilson, J. M. (1996) [0101] J. Virol. 70:520-532.
  • Flotte, T. R., Afione, S. A., Conrad, C., McGrath, S. A., Solow, R., Oka, H., Zeitlin, P. L., Guggino, W. B., Carter, B. J. (1993) [0102] Proc. Natl. Acad. Sci. USA 90:10613-10617.
  • Jooss, K., Yang, Y., Fisher, K. J., Wilson, J. M. (1998) [0103] J. Virol. 72:4212-4223.
  • Li, X, Eastrnan, E. M., Schwartz, R. J., Draghia-Akli, R. (1999) [0104] Nat. Biotechnol 17(3):241-245.
  • Li et al. (1997) [0105] J. Virol. 71:5236-43.
  • Kearns, W. G., Afione, S. A., Fulmer, S. B., Caruso, J., Flotte, T. R., Cutting, G. R. (1996) [0106] Gene Ther. 3:748-755.
  • Kessler, P. D., Podsakoff, G., Chen, X., McQuiston, S. A., Colosim, P. C., Matelis, L. A., Kurtzman, G. and Byrne, B. J. (1996) [0107] Proc. Natl. Acad. Sci. USA 93:14082-14087.
  • Klein, R. L. et al. (1998) [0108] Exp. Neurol. 150:183-194.
  • Kotin, R. M., Siniscalco, M., Samulski, R. J., Zhu, X. D., Hunter, L., Laughlin, C. A., McLaughlin, S., Muzyczka, N., Rocchi, M., Berns, K. I. (1990) [0109] Proc. Natl. Acad. Sci. USA 87:2211-2215.
  • Mitchell, P. J., Tjian, R. (1989) [0110] Science 245:371-378.
  • Murphy, J. E., Zhou, S., Giese, K., Williams, L. T., Escobedo, J. A., Dwarki, V. J. (1997) [0111] Proc. Natl. Acad. Sci. USA 94:13921-13926, 1997.
  • Nettelbeck, D. M., Jerome V., Muller R. (1998) [0112] Gene Ther 5(12)1656-1664.
  • Pitluk, Z. W., Ward, D. C. (1991) [0113] J. Virol. 65:6661-6670.
  • Stewart, A. F., Richard, III, C. W., Suzow, J., Stephan D., Weremowicz, S., Morton, C. C., Andra, C. N. (1996) [0114] Genomics 37(1):68-76.
  • Wei, C-F. etal (1993) [0115] J. Biol Chem 258(22):13506-13512.
  • Xiao, X., Li, J., Samulski, R. J. (1996) [0116] J. Virol. 70:8098-8108.
  • Zolotukhin, S., Potter, M., Hauswirth, W., Guy, J., Muzyczka, N. (1996) [0117] J. Virol. 70(7):4646-4654.
  • Zolotukhin, S. et al. (1999) [0118] Gene Ther. (6).

Claims (12)

  1. 41. A recombinant adeno-associated viral vector comprising a polynucleotide that encodes an α-1-antitrypsin polypeptide.
  2. 42. The vector of claim 41, wherein said vector comprises a promoter operably linked to said polynucleotide.
  3. 43. The vector of claim 42, wherein said promoter is selected from the group consisting of a CMV promoter, a hybrid CMV enhancer/β-actin promoter, an EF1 promoter, an U1a promoter and an U1b promoter.
  4. 44. The vector of claim 42, wherein said promoter is an inducible promoter selected from the group consisting of a Tet-inducible promoter and a VP16-LexA promoter.
  5. 45. The vector of claim 42, wherein said vector further comprises an enhancer.
  6. 46. The vector of claim 45, wherein said vector further comprises a CMV enhancer.
  7. 47. The vector of claim 45, wherein said enhancer comprises a synthetic enhancer.
  8. 48. The vector of claim 47, wherein said synthetic enhancer comprises a muscle-specific enhancer.
  9. 49. The vector of claim 41, wherein said vector comprises at least a first intron sequence.
  10. 50. The vector of claim 49, wherein said intron sequence comprises intron II from a mammalian gene encoding α-1-antitrypsin.
  11. 51. The vector of claim 41, wherein said polynucleotide encodes a human α-1-antitrypsin polypeptide.
  12. 52. The vector of claim 41, wherein said vector is selected from the group consisting of DE-AT (SEQ ID NO:3)) E-AT (SEQ ID NO:2), C-AT (SEQ ID NO:1), C-AT2(SEQ ID NO:7), p43C-AT (SEQ ID NO:4), p43CB-AT (SEQ ID NO:6), p43C-AT-IN (SEQ ID
US10/267,117 1998-04-24 2002-10-08 rAAV compositions for gene therapy Abandoned US20030082162A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/267,117 US20030082162A1 (en) 1998-04-24 2002-10-08 rAAV compositions for gene therapy
US10/340,112 US20030095949A1 (en) 1998-04-24 2003-01-10 Adeno-associated viral vectors for the treatment and prevention of diabetes
US11/932,912 US20090186002A1 (en) 1998-04-24 2007-10-31 rAAV-delivered alpha-1-antitrypsin compositions and method for the treatment and prevention of diabetes

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US8302598P 1998-04-24 1998-04-24
US09/299,141 US6461606B1 (en) 1998-04-24 1999-04-23 Materials and methods for gene therapy
US10/267,117 US20030082162A1 (en) 1998-04-24 2002-10-08 rAAV compositions for gene therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/299,141 Continuation US6461606B1 (en) 1998-04-24 1999-04-23 Materials and methods for gene therapy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/340,112 Continuation US20030095949A1 (en) 1998-04-24 2003-01-10 Adeno-associated viral vectors for the treatment and prevention of diabetes

Publications (1)

Publication Number Publication Date
US20030082162A1 true US20030082162A1 (en) 2003-05-01

Family

ID=22175690

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/299,141 Expired - Lifetime US6461606B1 (en) 1998-04-24 1999-04-23 Materials and methods for gene therapy
US10/267,117 Abandoned US20030082162A1 (en) 1998-04-24 2002-10-08 rAAV compositions for gene therapy
US10/340,112 Abandoned US20030095949A1 (en) 1998-04-24 2003-01-10 Adeno-associated viral vectors for the treatment and prevention of diabetes
US11/932,912 Abandoned US20090186002A1 (en) 1998-04-24 2007-10-31 rAAV-delivered alpha-1-antitrypsin compositions and method for the treatment and prevention of diabetes

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/299,141 Expired - Lifetime US6461606B1 (en) 1998-04-24 1999-04-23 Materials and methods for gene therapy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/340,112 Abandoned US20030095949A1 (en) 1998-04-24 2003-01-10 Adeno-associated viral vectors for the treatment and prevention of diabetes
US11/932,912 Abandoned US20090186002A1 (en) 1998-04-24 2007-10-31 rAAV-delivered alpha-1-antitrypsin compositions and method for the treatment and prevention of diabetes

Country Status (11)

Country Link
US (4) US6461606B1 (en)
EP (1) EP1071806B1 (en)
AT (1) ATE269412T1 (en)
AU (1) AU3663699A (en)
CA (1) CA2326327C (en)
DE (1) DE69918090T2 (en)
DK (1) DK1071806T3 (en)
ES (1) ES2219011T3 (en)
HK (1) HK1035205A1 (en)
NZ (1) NZ507308A (en)
PT (1) PT1071806E (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210361778A1 (en) * 2020-04-06 2021-11-25 Homology Medicines, Inc. Adeno-associated virus compositions for ids gene transfer and methods of use thereof

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6759237B1 (en) * 1998-11-05 2004-07-06 The Trustees Of The University Of Pennsylvania Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing same
US6821775B1 (en) * 2000-02-11 2004-11-23 Genvec, Inc. Viral vector encoding pigment epithelium-derived factor
KR20030072214A (en) * 2000-05-26 2003-09-13 스미또모 세이야꾸 가부시키가이샤 Novel recombinant adenovirus vector with relieved side effects
WO2002095006A2 (en) * 2001-05-24 2002-11-28 Genzyme Corporation Muscle-specific expression vectors
US7312202B2 (en) * 2003-02-18 2007-12-25 Board Of Regents, The University Of Texas System Rationally designed and chemically synthesized promoter for genetic vaccine and gene therapy
ES2247942B1 (en) * 2004-08-27 2006-10-01 Instituto Nacional De Investigacion Y Tecnologia Agraria Y Alimentaria (Inia) GENE CONSTRUCTION, VECTOR AND DNA VACCINE FOR THE VACCINATION OF AQUATIC ANIMALS.
US7479550B2 (en) * 2006-06-02 2009-01-20 The Board Of Regents Of The University Of Texas System Amyloid β gene vaccines
RU2502800C2 (en) * 2008-01-15 2013-12-27 Эббви Инк. Improved expression vectors of mammals and their use
WO2013063601A1 (en) 2011-10-28 2013-05-02 University Of Florida Research Foundation, Inc. Chimeric promoter for cone photoreceptor targeted gene therapy
MX365403B (en) 2012-01-09 2019-05-31 Serpin Pharma Llc Peptides and methods of using same.
FI3253406T3 (en) 2015-02-05 2024-01-18 Canem Holdings Llc Compositions for treating granulomatosis with polyangiitis
PT3265571T (en) 2015-03-03 2022-06-29 Fond Telethon Multiple vector system and uses thereof
JP6929272B2 (en) 2015-08-28 2021-09-01 セルピン ファーマ リミテッド ライアビリティ カンパニー Methods for disease treatment
WO2017055370A1 (en) 2015-09-28 2017-04-06 Fondazione Telethon Treatment of bone growth disorders
CA3085696A1 (en) * 2017-12-12 2019-06-20 Kamada Ltd Methods of inducing immune tolerance and reducing anti-drug antibody response
US20220249703A1 (en) * 2019-02-15 2022-08-11 Allen Institute Artificial expression constructs for selectively modulating gene expression in selected neuronal cell populations
WO2023213817A1 (en) 2022-05-02 2023-11-09 Fondazione Telethon Ets Gene therapy for gyrate atrophy of the choroid and retina

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4797368A (en) * 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
US5139941A (en) * 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5399346A (en) * 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5439824A (en) * 1993-05-06 1995-08-08 The United States Of America Increased expression of α-1-antitrypsin in expression vectors through the inclusion of intron II
US5587308A (en) * 1992-06-02 1996-12-24 The United States Of America As Represented By The Department Of Health & Human Services Modified adeno-associated virus vector capable of expression from a novel promoter
US5658776A (en) * 1993-11-09 1997-08-19 Targeted Genetics Corporation Generation of high titers of recombinant AAV vectors
US5658785A (en) * 1994-06-06 1997-08-19 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5693531A (en) * 1993-11-24 1997-12-02 The United States Of America As Represented By The Department Of Health And Human Services Vector systems for the generation of adeno-associated virus particles
US5846528A (en) * 1996-01-18 1998-12-08 Avigen, Inc. Treating anemia using recombinant adeno-associated virus virions comprising an EPO DNA sequence
US5858351A (en) * 1996-01-18 1999-01-12 Avigen, Inc. Methods for delivering DNA to muscle cells using recombinant adeno-associated virus vectors
US5863541A (en) * 1994-06-30 1999-01-26 University Of Pittsburgh AAV capsid vehicles for molecular transfer
US6174527B1 (en) * 1995-02-24 2001-01-16 The Trustees Of The University Of Pennsylvania Methods and compositions for gene therapy for the treatment of defects in lipoprotein metabolism
US6261834B1 (en) * 1991-11-08 2001-07-17 Research Corporation Technologies, Inc. Vector for gene therapy
US6723707B1 (en) * 1997-08-25 2004-04-20 The Trustees Of The University Of Pennsylvania Use of insulin-like growth factor-I in muscle

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4797368A (en) * 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
US5139941A (en) * 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5399346A (en) * 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US6261834B1 (en) * 1991-11-08 2001-07-17 Research Corporation Technologies, Inc. Vector for gene therapy
US5866696A (en) * 1992-06-02 1999-02-02 The United States Of America As Represented By The Department Of Health And Human Services Modified adeno-associated virus vector capable of expression from a novel promoter
US5587308A (en) * 1992-06-02 1996-12-24 The United States Of America As Represented By The Department Of Health & Human Services Modified adeno-associated virus vector capable of expression from a novel promoter
US5439824A (en) * 1993-05-06 1995-08-08 The United States Of America Increased expression of α-1-antitrypsin in expression vectors through the inclusion of intron II
US5658776A (en) * 1993-11-09 1997-08-19 Targeted Genetics Corporation Generation of high titers of recombinant AAV vectors
US5693531A (en) * 1993-11-24 1997-12-02 The United States Of America As Represented By The Department Of Health And Human Services Vector systems for the generation of adeno-associated virus particles
US5658785A (en) * 1994-06-06 1997-08-19 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5863541A (en) * 1994-06-30 1999-01-26 University Of Pittsburgh AAV capsid vehicles for molecular transfer
US6174527B1 (en) * 1995-02-24 2001-01-16 The Trustees Of The University Of Pennsylvania Methods and compositions for gene therapy for the treatment of defects in lipoprotein metabolism
US5858351A (en) * 1996-01-18 1999-01-12 Avigen, Inc. Methods for delivering DNA to muscle cells using recombinant adeno-associated virus vectors
US5846528A (en) * 1996-01-18 1998-12-08 Avigen, Inc. Treating anemia using recombinant adeno-associated virus virions comprising an EPO DNA sequence
US6723707B1 (en) * 1997-08-25 2004-04-20 The Trustees Of The University Of Pennsylvania Use of insulin-like growth factor-I in muscle

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210361778A1 (en) * 2020-04-06 2021-11-25 Homology Medicines, Inc. Adeno-associated virus compositions for ids gene transfer and methods of use thereof

Also Published As

Publication number Publication date
CA2326327C (en) 2009-06-16
WO1999055564A1 (en) 1999-11-04
HK1035205A1 (en) 2001-11-16
WO1999055564A8 (en) 1999-12-02
AU3663699A (en) 1999-11-16
EP1071806A2 (en) 2001-01-31
US6461606B1 (en) 2002-10-08
ATE269412T1 (en) 2004-07-15
NZ507308A (en) 2003-01-31
DK1071806T3 (en) 2004-07-19
US20030095949A1 (en) 2003-05-22
US20090186002A1 (en) 2009-07-23
CA2326327A1 (en) 1999-11-04
PT1071806E (en) 2004-09-30
ES2219011T3 (en) 2004-11-16
DE69918090T2 (en) 2005-06-16
DE69918090D1 (en) 2004-07-22
WO1999055564A9 (en) 2000-04-06
EP1071806B1 (en) 2004-06-16

Similar Documents

Publication Publication Date Title
US20090186002A1 (en) rAAV-delivered alpha-1-antitrypsin compositions and method for the treatment and prevention of diabetes
US7662627B2 (en) Adeno-associated virus materials and methods
EP1923466A2 (en) Vectors encoding recombinant AAV genomes
US20030143202A1 (en) Anemia
US20100278791A1 (en) Adeno-associated virus serotype i nucleic acid sequences, vectors and host cells containing same
AU2021236481A1 (en) Evasion of neutralizing antibodies by a recombinant adeno-associated virus
JP2001500497A (en) Methods of gene therapy directed by recombinant adeno-associated virus
US6037177A (en) Method for increasing the efficiency of recombinant AAV production
WO2003064664A1 (en) Physiologically regulated erythropoietin- exprressing vector for the treatment of anaemia
JP2002529098A (en) Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing the same
JP2018526003A (en) Chimeric AAV-anti-VEGF for treating canine cancer
JP2021514201A (en) Hybrid adjustment element
US20010034062A1 (en) Antibody gene therapy with adeno-associated viral vectors
US20210180085A1 (en) Transgene cassettes designed to express a human mecp2 gene
BR112021002202A2 (en) mini-gde for the treatment of glycogen storage disease iii
US6797505B2 (en) Recombinant AAV vectors for gene therapy of hemophilia A
JP2023535956A (en) AAV-mediated therapy for improved X-linked retinoschisis
US20220233655A1 (en) Improved delivery of gene therapy vectors to retinal cells using a glycoside hydrolase enzyme
EP1845163A2 (en) Adeno-associated virus serotype l nucleic acid sequences, vetors and host cells containing same
RU2760301C1 (en) Aav5-based vaccine for induction of specific immunity to sars-cov-2 virus and/or prevention of coronavirus infection caused by sars-cov-2
US20230151390A1 (en) Vectors for the treatment of acid ceramidase deficiency
WO1999055564A2 (en) Materials and methods for gene therapy
ES2384975T3 (en) AAV vectors for in vivo genetic therapy of rheumatoid arthritis
WO2023102518A1 (en) Gnao1 gene therapy vectors and uses thereof
US20040092008A1 (en) Recombinant AAV vectors for gene therapy of hemophilia A

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF FLORIDA;REEL/FRAME:021437/0481

Effective date: 20030429