US20060251626A1 - Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors - Google Patents

Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors Download PDF

Info

Publication number
US20060251626A1
US20060251626A1 US11/449,778 US44977806A US2006251626A1 US 20060251626 A1 US20060251626 A1 US 20060251626A1 US 44977806 A US44977806 A US 44977806A US 2006251626 A1 US2006251626 A1 US 2006251626A1
Authority
US
United States
Prior art keywords
aav
body weight
infused
per gram
cardiomyocytes
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
US11/449,778
Inventor
Jeffrey Leiden
Eric Svensson
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.)
Arch Development Corp
Original Assignee
Arch Development Corp
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 Arch Development Corp filed Critical Arch Development Corp
Priority to US11/449,778 priority Critical patent/US20060251626A1/en
Publication of US20060251626A1 publication Critical patent/US20060251626A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • 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

  • this invention relates to the use of recombinant adeno-associated virus (rAAV) vectors to transduce cardiomyocytes in vivo by infusing the rAAV into a coronary artery or coronary sinus.
  • rAAV adeno-associated virus
  • coronary artery perfusion of mouse hearts with a rAAV encoding the LacZ gene produced efficient transduction of cardiomyocytes which was stable for at least 8 weeks.
  • rAAV infection is not associated with detectable myocardial inflammation or myocyte necrosis.
  • rAAV is a useful vector for the stable expression of therapeutic genes in the myocardium and can be used to deliver genes for inducing angiogenesis, inhibiting angiogenesis, stimulating cell proliferation, inhibiting cell proliferation and/or treating or ameliorating other cardiovascular conditions.
  • Myocardial gene therapy can be used for the treatment of a number of cardiovascular diseases, including ischemic cardiomyopathies, congestive heart failure, and malignant arrhythmias (Nabel (1995) Circulation 91:541-548).
  • a useful vector for myocardial gene delivery will allow efficient and stable transduction of cardiomyocytes with a variety of transgenes after either direct intramyocardial injection or infusion into the coronary arteries or sinuses.
  • plasmid DNA vectors injected directly into the left ventricular myocardium have been expressed for ⁇ 6 months by cardiomyocytes adjacent to the area of injection (Lin et al. (1990a) Circulation 82:2217-2221; Kass et al. (1993) Proc.
  • rAAV vectors have been shown to program efficient and stable recombinant gene expression in skeletal muscle and liver in both rodents and primates (Fisher et al. (1997) Nat. Med. 3:306-312; Kessler et al. (1996) Proc. Natl. Acad. Sci. USA 93:14082-14087; and Snyder et al. (1997) Nat. Genet. 16: 270-276) and in cardiac muscle directly injected with rAAV (U.S. Pat. No. 5,858,351 to Podsakoff et al.).
  • rAAV vectors used in gene therapy applications unlike RDAd, do not encode viral proteins, the rAAV vectors have not been associated with immune responses to foreign transgene proteins.
  • this invention establishes that by infusing rAAV in much higher amounts proportional to body weight of the animal and for particular time periods, then rAAV provides unexpected efficient and stable gene transfer into the heart, opening up use of rAAV vectors to deliver therapeutically-effective molecules to cardiomyocytes in amounts useful for treating or ameliorating cardiac diseases or conditions.
  • the present invention is directed to a method of treating a cardiovascular condition by infusing an rAAV vector into a coronary artery or a coronary sinus for a time and in an amount sufficient to stably and efficiently transduce the cardiomyocytes perfused by the artery or sinus.
  • the rAAV vector encodes at least one nucleic acid which is operably linked to a control region and which encodes a therapeutically-effective molecule. After infusion and transduction of the cardiomyocytes, the therapeutically-effective molecule is expressed in an amount effective to treat or ameliorate the cardiovascular condition.
  • this method provides a means of delivering AAV vectors in a stable and efficient manner.
  • the vector can be infused by any convenient means and in conjunction with surgery or other cardiac procedure, if desired.
  • FIG. 1 Schematic of AAV CMV-LacZ .
  • ITR indicates inverted terminal repeats; BGH pA, bovine growth hormone polyadenylation signal; CMV Pr, CMV immediate-early promoter; LacZ, bacterial LacZ gene.
  • This invention relates to treating cardiovascular conditions using rAAV vectors.
  • an rAAV vector encoding a therapeutically effective molecule is infused into a coronary artery or a coronary sinus to deliver the vector to the heart in a manner which stably and efficiently transduces cardiomyocytes. It has unexpectedly been found that the ability to obtain stable and efficient transduction of cardiomyocytes by rAAV depends upon the duration of the infusion period and the amount of virus infused relative to body weight
  • rAAV displays significant advantages for myocardial gene transfer compared with plasmid DNA or adenovirus vectors.
  • rAAV when delivered as described herein, allows efficient transduction of cardiomyocytes.
  • rAAV vectors program stable expression of foreign transgenes in immunocompetent hosts. The stability of transgene expression observed with rAAV even after expression of a foreign transgene protein likely reflects the fact that rAAV vectors, unlike their adenovirus counterparts, do not express any viral gene products and are therefore significantly less immunogenic. This lack of immunogenicity represents a major advantage of rAAV for myocardial gene transfer.
  • the invention is directed to a method of treating a cardiovascular condition which comprises infusing an rAAV vector into a coronary artery or sinus of an animal for a time and in an amount sufficient to stably and efficiently transduce cardiomyocytes perfused by the artery or sinus, wherein that vector encodes at least one nucleic acid, i.e., the transgene, encoding a therapeutically-effective molecule; and expressing the therapeutically-effective molecule in an amount effective to treat or ameliorate the cardiovascular condition.
  • the nucleic acid is operably linked to a control region, e.g., promoters, enhancers, termination signals and the like, to permit expression of the molecule.
  • each can be controlled separately by individual control regions or, any group of them, or all of them, can be controlled in an operon, i.e, with one control region driving expression of multiple genes on a single transcript.
  • rAAV vectors useful in the present invention can be any rAAV vector with one or more transgenes (or nucleic acids of interest) inserted therein in a manner allowing expression of the transgene under control of appropriate regulatory elements such as promoters, enhancers, transcription terminators and the like.
  • rAAV vectors are well known in the art and can be prepared by standard methodology know to those of ordinary skill in the art. For example, U.S. Pat. No.
  • a “transgene” or “nucleic acid of interest” or the “nucleic acid encoded in the rAAV vector” as used herein refers to any nucleotide sequence which encodes a therapeutically-effective molecule that can be used to treat a cardiovascular condition.
  • Such transgenes may normally be foreign to the animal being treated or may be a gene normally found in that animal for which altered expression (e.g., temporal, spatial or amount of expression) is desired to achieve a particular therapeutic effect.
  • the therapeutically-effective molecule encoded by the transgene is protein or an anti-sense RNA that imparts a benefit to the animal or subject undergoing treatment or amelioration of a cardiac condition or disease in accordance with this invention.
  • Proteins that can be administered to treat or ameliorate cardiovascular conditions are numerous and include, but are not limited to, molecules competent to induce angiogenesis, e.g., angiogenesis factors; anti-angiogenesis factors; proteins capable of inhibiting vascular smooth muscle cell proliferation; proteins useful for treating atherosclerosis; proteins useful for treating restenosis, proteins useful for stimulating cardiomyocyte activity; proteins capable of secretion from cardiomyocytes that exert their effect in the heart or capable of transport to other locales for treatment of a cardiovascular condition or disease; hormones, cytokines or growth factors useful for treating cardiac conditions or diseases; and proteins capable of stimulating vascular smooth muscle cell proliferation.
  • Other genes encoding proteins useful in this invention include ion channel genes, contractile protein genes, phospholamban encoding genes and genes encoding ⁇ adrenergic receptors or ⁇ adrenergic kinases.
  • Angiogenic factors include, but are not limited to FGF-1, FGF-2, FGF-5, VEGF, HIF-1 and the like.
  • Proteins useful for treating restenosis include thymidine kinase, cytosine deaminase, p21, p27, p53, Rb, and NF- ⁇ B.
  • this invention can be used to deliver any protein via an rAAV vector that has a therapeutic benefit for treating or ameliorating a cardiovascular condition or disease.
  • a protein competent to induce angiogenesis or an “angiogenesis factor” as used herein is a protein or substance that causes proliferation of new blood vessels and includes fibroblast growth factors, endothelial cell growth factors or other proteins with such biological activity.
  • Particular proteins known to induce angiogenesis are FGF-1, FGF-2, FGF-5, VEGF and active fragment thereof, and HIF-1.
  • Proteins competent to inhibit angiogenesis or “anti-angiogenesis factors” are proteins or substances that inhibit the formation of new blood vessels.
  • Anti-sense RNA that can be administered to treat or ameliorate cardiovascular conditions have one of the same activities as proteins useful in the invention.
  • Such RNA include, but are not limited to, c-myb, c-myc and others.
  • Anti-sense RNA molecules including how to design and use such molecules in expression vectors are well know in the art and can be contructed by routine methodology.
  • a strand of RNA whose sequence of bases is complementary to the sense, or translated, RNA strand can form a duplex to block translation or degradation of a particular mRNA or otherwise control or alter expression of the desired mRNA.
  • control region or “regulatory element” refers to polyadenylation signals, upstream regulatory domains, promoters, enhancers, transcription termination sequences and the like which regulate the transcription and translation of a nucleic acid sequence.
  • operably linked refers to an arrangement of elements wherein the components are arranged so as to perform their usual function.
  • control regions or regulatory elements operably linked to a coding sequence are capable of effecting the expression of the coding sequence.
  • the control elements need not be contiguous with the coding sequence, so long as they function to direct the expression thereof.
  • intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered “operably linked” to the coding sequence.
  • the regulatory elements of the invention can be derived from any source, e.g., viruses, mammals, insects or even synthetic, provided that they function in cardiomyocytes.
  • any promoter can used to control expression of the transgene.
  • Such promoters can be promiscous, i.e., active in many cell types, such as the SV40 early promoter, the mouse mammary tumor virus LTR promoter, the adenovirus major late promoter (Ad MLP), a herpes simplex promoter, a CMV promoter such as the CMV immediate early promoter, a rous sarcoma virus (RSV) promoter.
  • the promoter can be tissue-specific for expression in cardiomyocytes.
  • the rAAV is delivered to cardiac myocytes by infusion into a coronary artery or coronary sinus.
  • This mode of delivery has also been referred to as intraluminal delivery through a coronary artery, intracoronary delivery or intraarterial delivery.
  • infusion into a coronary artery includes intracoronary perfusion.
  • the rAAV vector can be infused when the heart is in situ, i.e., in the body cavity or when the heart or heart tissue (cardiac tissue) has been removed from the body as might occur when the heart is being donated for transplant into a recipient.
  • the vector can be infused through any artery or vein attached thereto, by contacting with or soaking the heart in an appropriately concentrated solution of the vector, or by a combination of both.
  • infusion includes delivering rAAV to a heart or heart tissue ex vivo by the means disclosed herein. If necessary, the infusion can be repeated at intervals such as 3 months, 6 months, one year, or as appropriately determined.
  • treating cardiac conditions include treating cardiac or cardiovascular diseases.
  • cardiac conditions subject to treatment or amelioration according to the method of the present invention include, but are not limited to, myocardial ischemia, myocardial infarction, congestive heart failure, dilated and hypertrophic cardiomyopathy, cardiac arrythmia, cardiac hypertrophy, cardiac transplantation and rejection.
  • the cardiac condition such as ischemia
  • the rAAV vector used in accordance with the method of this invention would encode an angiogenesis factor.
  • the rAAV vector is infused into a coronary artery for a time and in an amount sufficient to stably and efficiently transduce cardiac tissue perfused by the artery, wherein the AAV vector encodes a therapeutically-effective molecule which is expressed in the cardiac tissue in an amount effective to treat or ameliorate a cardiovascular condition including, but not limited to, inducing angiogenesis, inhibiting angiogenesis, stimulating or inhibiting cell proliferation, treating restenosis, treating atherosclerosis, treating congestive heart failure, treating ischemic cardiomyopathies or treating malignant arrhythmias, myocardial infarction, congestive heart failure, or dilated and hypertrophic cardiomyopathy.
  • a cardiovascular condition including, but not limited to, inducing angiogenesis, inhibiting angiogenesis, stimulating or inhibiting cell proliferation, treating restenosis, treating atherosclerosis, treating congestive heart failure, treating ischemic cardiomyopathies or treating malignant arrhythmias, myocardial infarction, congestive heart failure, or
  • the method of the present invention can be used with any animal, including but not limited to, mammals such as rodents, dogs, cats, cattle, primates and humans.
  • the method is used for gene therapy to treat human acquired or inherited cardiac conditions or diseases.
  • the present invention thus provides a method of treating and/or ameliorating a cardiovascular condition by infusing an rAAV vector for a time and in and amount sufficient to stable and efficiently transduce cardiomyocytes which was heretofore unachievable by methods known in the art.
  • stable and efficient transduction means that significant number of cardiomyocytes are transduced and are capable of expressing the protein for a prolonged period of time.
  • Stable and efficient transduction occurs over a period of time and can actually be observed over time as an increase in the percentage of transduced cardiomyocytes, as continued expression of the transgene, or as continued observation of the therapeutic effect at a molecular, microscopic or macroscopic level.
  • stable and efficient transduction can be manifested by ongoing development and or growth of new blood vessels, by observing the improved blood flow to the heart, or by determining measuring the level of ischemia in the heart tissue.
  • efficient transduction occurs when at least about 10%, and preferably more, of the cardiomyocytes have been transduced, i.e., infected by, the rAAV.
  • the rAAV transduced i.e., infected by, the rAAV.
  • about 25%, about 40% or even about 50% of the cardiomyocytes will be transduced.
  • about 10% of the cardiomyocytes can be transduced using only rAAV, this percentage can be increased by co-infusing adenovirus as a helper virus without adverse effects.
  • the time of infusion contributes to acheiving stable and efficient transduction of the cardiomyocytes as well.
  • the infusion time ranges from about 2 minutes to about 30 minutes, more preferably from about 5 minutes to about 20 minutes and most perferably is about fifteen minutes.
  • the amount of rAAV infused into the animal is proportional to the body weight of the animal.
  • stable and efficient transduction occurs when the amount of rAAV infused ranges from about 1 ⁇ 10 5 IU (infectious units) of AAV per gram body weight to about 1 ⁇ 10 9 IU AAV per gram body weight, and preferably from about 1 ⁇ 10 6 IU AAV per gram body weight to about 1 ⁇ 10 8 IU AAV per gram body weight, and most preferably is about 5-6 ⁇ 10 7 IU AAV per gram body weight.
  • the example described below demonstrates efficient and stable transduction of cardiac myocytes in vivo after intracoronary infusion of an rAAV vector.
  • Ad CMV-LacZ The structure of pAAV CMV-LacZ is shown in FIG. 1 .
  • Ad CMV-LacZ and the E3-deleted adenovirus, Ad d1309 were propagated and purified as described (Barr 1994).
  • rAAV was prepared as described (Rolling et al. (1995) Mol. Biotechnol. 3:9-15) and purified by cesium chloride gradient centrifugation. Viral titer was assessed by a dot blot hybridization assay to determine the number of viral genomes per milliliter and by infecting HeLa cells with the virus and staining with X-gal 24 hours after infection. All viral preparations had titers of 1 to 2 ⁇ 10 11 genomes/mL, and 2 to 3 ⁇ 10 9 infectious units (IU)/mL.
  • Freshly isolated hearts were fixed in PBS plus 1.25% glutaraldehyde for 10 minutes at room temperature, stained overnight with X-gal (Lin 1990a), and counterstained with eosin.
  • Cardiac homogenates were assayed for ⁇ -galactosidase ( ⁇ -gal) activity and protein concentrations. ⁇ -Gal activities were normalized for total protein and for the number of infectious rAAV or RDAd particles injected.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention relates to the use of recombinant adeno-associated virus (rAAV) vectors to transduce cardiomyocytes in vivo by infusing the rAAV into a coronary artery or coronary sinus. rAAV infection is not associated with detectable myocardial inflammation or myocyte necrosis. Thus, rAAV is a useful vector for the stable expression of therapeutic genes in the myocardium and can be used to deliver genes for inducing angiogenesis, inhibiting angiogenesis, stimulating cell proliferation, inhibiting cell proliferation and/or treating or ameliorating other cardiovascular conditions.

Description

  • This application is a continuation-in-part application of provisional application U.S. Ser. No. 60/113,923, filed Dec. 28, 1998, which is incorporated by reference herein in its entirety.
  • This work was supported in part by grants from the National Institutes of Health (DK-48997, AR-42895, and HL-54592) to Jeffrey M. Leiden.
  • FIELD OF THE INVENTION
  • The ability to stably and efficiently program recombinant gene expression in cardiomyocytes facilitates gene therapy approaches for a variety of cardiovascular diseases and conditions. Accordingly, this invention relates to the use of recombinant adeno-associated virus (rAAV) vectors to transduce cardiomyocytes in vivo by infusing the rAAV into a coronary artery or coronary sinus. For example, coronary artery perfusion of mouse hearts with a rAAV encoding the LacZ gene produced efficient transduction of cardiomyocytes which was stable for at least 8 weeks. Moreover, rAAV infection is not associated with detectable myocardial inflammation or myocyte necrosis. Thus, rAAV is a useful vector for the stable expression of therapeutic genes in the myocardium and can be used to deliver genes for inducing angiogenesis, inhibiting angiogenesis, stimulating cell proliferation, inhibiting cell proliferation and/or treating or ameliorating other cardiovascular conditions.
  • BACKGROUND OF THE INVENTION
  • Myocardial gene therapy can be used for the treatment of a number of cardiovascular diseases, including ischemic cardiomyopathies, congestive heart failure, and malignant arrhythmias (Nabel (1995) Circulation 91:541-548). A useful vector for myocardial gene delivery will allow efficient and stable transduction of cardiomyocytes with a variety of transgenes after either direct intramyocardial injection or infusion into the coronary arteries or sinuses. For example, plasmid DNA vectors injected directly into the left ventricular myocardium have been expressed for ≧6 months by cardiomyocytes adjacent to the area of injection (Lin et al. (1990a) Circulation 82:2217-2221; Kass et al. (1993) Proc. Natl. Acad. Sci. USA 90:11498-11502; and Guzman et al. (1993) Circ. Res. 73:1202-1207). However, the therapeutic usefulness of this approach has been limited by the low efficiency of cardiomyocyte transduction (0.1% to 1.0% of cardiomyocytes in the area of injection).
  • Both intramyocardial injection and intracoronary infusion of replication-defective adenovirus (RDAd) vectors have been used to efficiently transduce cardiomyocytes in rodents, rabbits, and pigs in vivo. However, the feasibility of adenovirus-mediated gene transfer has been limited by immune responses to viral and foreign transgene proteins, which cause significant myocardial inflammation, eliminate virus-transduced cells within 30 days of infection, and thereby result in transient recombinant gene expression in immunocompetent hosts (Guzman et al. (1993) Circ. Res. 73:1202-1207; French et al. (1994) Circulation 90:2414-2424; and Barr et al. (1994) Gene Ther. 1:51-58).
  • Recently, rAAV vectors have been shown to program efficient and stable recombinant gene expression in skeletal muscle and liver in both rodents and primates (Fisher et al. (1997) Nat. Med. 3:306-312; Kessler et al. (1996) Proc. Natl. Acad. Sci. USA 93:14082-14087; and Snyder et al. (1997) Nat. Genet. 16: 270-276) and in cardiac muscle directly injected with rAAV (U.S. Pat. No. 5,858,351 to Podsakoff et al.). However, since rAAV vectors used in gene therapy applications, unlike RDAd, do not encode viral proteins, the rAAV vectors have not been associated with immune responses to foreign transgene proteins.
  • While a previous report showed that rAAV can transduce cardiomyocytes in vivo, the efficiency of rAAV-mediated transgene expression in the heart was both low (about 0.2%) and localized (Kaplitt et al. (1996) Ann. Thorac. Surg. 62:1669-1676). In that study, pigs hearts were rapidly perfused with a low titer of rAAV (less than 104 expressing units AAV per gram of body weight). Based on those results, infusing rAAV into the heart would have severely limited use as a vector for myocardial gene therapy. However, as demonstrated herein, this invention establishes that by infusing rAAV in much higher amounts proportional to body weight of the animal and for particular time periods, then rAAV provides unexpected efficient and stable gene transfer into the heart, opening up use of rAAV vectors to deliver therapeutically-effective molecules to cardiomyocytes in amounts useful for treating or ameliorating cardiac diseases or conditions.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to a method of treating a cardiovascular condition by infusing an rAAV vector into a coronary artery or a coronary sinus for a time and in an amount sufficient to stably and efficiently transduce the cardiomyocytes perfused by the artery or sinus. The rAAV vector encodes at least one nucleic acid which is operably linked to a control region and which encodes a therapeutically-effective molecule. After infusion and transduction of the cardiomyocytes, the therapeutically-effective molecule is expressed in an amount effective to treat or ameliorate the cardiovascular condition.
  • Thus, this method provides a means of delivering AAV vectors in a stable and efficient manner. The vector can be infused by any convenient means and in conjunction with surgery or other cardiac procedure, if desired.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Schematic of AAVCMV-LacZ. ITR indicates inverted terminal repeats; BGH pA, bovine growth hormone polyadenylation signal; CMV Pr, CMV immediate-early promoter; LacZ, bacterial LacZ gene.
  • FIG. 2. Gene transfer into cardiomyocytes in vivo with AAVCMV-LacZ. Gross sections (left) and photomicrographs (right) of mouse hearts after coronary artery perfusion with 1.5×109 IU of AAVCMV-LacZ and staining with X-gal. Bar=25 microns.
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention relates to treating cardiovascular conditions using rAAV vectors. In accordance with the invention, an rAAV vector encoding a therapeutically effective molecule is infused into a coronary artery or a coronary sinus to deliver the vector to the heart in a manner which stably and efficiently transduces cardiomyocytes. It has unexpectedly been found that the ability to obtain stable and efficient transduction of cardiomyocytes by rAAV depends upon the duration of the infusion period and the amount of virus infused relative to body weight
  • Moreover, rAAV displays significant advantages for myocardial gene transfer compared with plasmid DNA or adenovirus vectors. For example, rAAV, when delivered as described herein, allows efficient transduction of cardiomyocytes. Further, rAAV vectors program stable expression of foreign transgenes in immunocompetent hosts. The stability of transgene expression observed with rAAV even after expression of a foreign transgene protein likely reflects the fact that rAAV vectors, unlike their adenovirus counterparts, do not express any viral gene products and are therefore significantly less immunogenic. This lack of immunogenicity represents a major advantage of rAAV for myocardial gene transfer.
  • Hence, the invention is directed to a method of treating a cardiovascular condition which comprises infusing an rAAV vector into a coronary artery or sinus of an animal for a time and in an amount sufficient to stably and efficiently transduce cardiomyocytes perfused by the artery or sinus, wherein that vector encodes at least one nucleic acid, i.e., the transgene, encoding a therapeutically-effective molecule; and expressing the therapeutically-effective molecule in an amount effective to treat or ameliorate the cardiovascular condition. Further, the nucleic acid is operably linked to a control region, e.g., promoters, enhancers, termination signals and the like, to permit expression of the molecule. When more than one nucleic acid is present on the rAAV vector, each can be controlled separately by individual control regions or, any group of them, or all of them, can be controlled in an operon, i.e, with one control region driving expression of multiple genes on a single transcript.
  • rAAV vectors useful in the present invention can be any rAAV vector with one or more transgenes (or nucleic acids of interest) inserted therein in a manner allowing expression of the transgene under control of appropriate regulatory elements such as promoters, enhancers, transcription terminators and the like. rAAV vectors are well known in the art and can be prepared by standard methodology know to those of ordinary skill in the art. For example, U.S. Pat. No. 5,858,351 and the references cited therein describe a variety of rAAV vectors suitable for use in gene therapy as well as how to make and propagate those vectors (see, e.g., Kotin (1994) Human Gene Therapy 5:793-801 or Berns, “Parvoviridae and their Replication” in Fundamental Virology, 2nd Edition, (Fields & Knipe, eds.)).
  • A “transgene” or “nucleic acid of interest” or the “nucleic acid encoded in the rAAV vector” as used herein refers to any nucleotide sequence which encodes a therapeutically-effective molecule that can be used to treat a cardiovascular condition. Such transgenes may normally be foreign to the animal being treated or may be a gene normally found in that animal for which altered expression (e.g., temporal, spatial or amount of expression) is desired to achieve a particular therapeutic effect. The therapeutically-effective molecule encoded by the transgene is protein or an anti-sense RNA that imparts a benefit to the animal or subject undergoing treatment or amelioration of a cardiac condition or disease in accordance with this invention.
  • Proteins that can be administered to treat or ameliorate cardiovascular conditions are numerous and include, but are not limited to, molecules competent to induce angiogenesis, e.g., angiogenesis factors; anti-angiogenesis factors; proteins capable of inhibiting vascular smooth muscle cell proliferation; proteins useful for treating atherosclerosis; proteins useful for treating restenosis, proteins useful for stimulating cardiomyocyte activity; proteins capable of secretion from cardiomyocytes that exert their effect in the heart or capable of transport to other locales for treatment of a cardiovascular condition or disease; hormones, cytokines or growth factors useful for treating cardiac conditions or diseases; and proteins capable of stimulating vascular smooth muscle cell proliferation. Other genes encoding proteins useful in this invention include ion channel genes, contractile protein genes, phospholamban encoding genes and genes encoding β adrenergic receptors or β adrenergic kinases.
  • Angiogenic factors include, but are not limited to FGF-1, FGF-2, FGF-5, VEGF, HIF-1 and the like. Proteins useful for treating restenosis include thymidine kinase, cytosine deaminase, p21, p27, p53, Rb, and NF-κB. Hence, this invention can be used to deliver any protein via an rAAV vector that has a therapeutic benefit for treating or ameliorating a cardiovascular condition or disease.
  • A protein competent to induce angiogenesis or an “angiogenesis factor” as used herein is a protein or substance that causes proliferation of new blood vessels and includes fibroblast growth factors, endothelial cell growth factors or other proteins with such biological activity. Particular proteins known to induce angiogenesis are FGF-1, FGF-2, FGF-5, VEGF and active fragment thereof, and HIF-1. Proteins competent to inhibit angiogenesis or “anti-angiogenesis factors” are proteins or substances that inhibit the formation of new blood vessels.
  • Anti-sense RNA that can be administered to treat or ameliorate cardiovascular conditions have one of the same activities as proteins useful in the invention. Such RNA include, but are not limited to, c-myb, c-myc and others. Anti-sense RNA molecules, including how to design and use such molecules in expression vectors are well know in the art and can be contructed by routine methodology. Thus a strand of RNA whose sequence of bases is complementary to the sense, or translated, RNA strand can form a duplex to block translation or degradation of a particular mRNA or otherwise control or alter expression of the desired mRNA.
  • As used herein, a “control region” or “regulatory element” refers to polyadenylation signals, upstream regulatory domains, promoters, enhancers, transcription termination sequences and the like which regulate the transcription and translation of a nucleic acid sequence.
  • The term “operably linked” refers to an arrangement of elements wherein the components are arranged so as to perform their usual function. Thus, control regions or regulatory elements operably linked to a coding sequence are capable of effecting the expression of the coding sequence. The control elements need not be contiguous with the coding sequence, so long as they function to direct the expression thereof. Thus, for example, intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered “operably linked” to the coding sequence.
  • The regulatory elements of the invention can be derived from any source, e.g., viruses, mammals, insects or even synthetic, provided that they function in cardiomyocytes. For example, any promoter can used to control expression of the transgene. Such promoters can be promiscous, i.e., active in many cell types, such as the SV40 early promoter, the mouse mammary tumor virus LTR promoter, the adenovirus major late promoter (Ad MLP), a herpes simplex promoter, a CMV promoter such as the CMV immediate early promoter, a rous sarcoma virus (RSV) promoter. Alternatively the promoter can be tissue-specific for expression in cardiomyocytes.
  • The rAAV is delivered to cardiac myocytes by infusion into a coronary artery or coronary sinus. This mode of delivery has also been referred to as intraluminal delivery through a coronary artery, intracoronary delivery or intraarterial delivery. As used herein, infusion into a coronary artery includes intracoronary perfusion. In accordance with the invention, the rAAV vector can be infused when the heart is in situ, i.e., in the body cavity or when the heart or heart tissue (cardiac tissue) has been removed from the body as might occur when the heart is being donated for transplant into a recipient. In the case of a heart being prepared for transplantation or for heart tissue, the vector can be infused through any artery or vein attached thereto, by contacting with or soaking the heart in an appropriately concentrated solution of the vector, or by a combination of both. Thus as described herein, infusion includes delivering rAAV to a heart or heart tissue ex vivo by the means disclosed herein. If necessary, the infusion can be repeated at intervals such as 3 months, 6 months, one year, or as appropriately determined.
  • As used herein, treating cardiac conditions include treating cardiac or cardiovascular diseases. Examples of cardiac conditions subject to treatment or amelioration according to the method of the present invention include, but are not limited to, myocardial ischemia, myocardial infarction, congestive heart failure, dilated and hypertrophic cardiomyopathy, cardiac arrythmia, cardiac hypertrophy, cardiac transplantation and rejection. For example, if the cardiac condition, such as ischemia, can be treated or improved by inducing angiogenesis, then the rAAV vector used in accordance with the method of this invention would encode an angiogenesis factor.
  • Thus the rAAV vector is infused into a coronary artery for a time and in an amount sufficient to stably and efficiently transduce cardiac tissue perfused by the artery, wherein the AAV vector encodes a therapeutically-effective molecule which is expressed in the cardiac tissue in an amount effective to treat or ameliorate a cardiovascular condition including, but not limited to, inducing angiogenesis, inhibiting angiogenesis, stimulating or inhibiting cell proliferation, treating restenosis, treating atherosclerosis, treating congestive heart failure, treating ischemic cardiomyopathies or treating malignant arrhythmias, myocardial infarction, congestive heart failure, or dilated and hypertrophic cardiomyopathy.
  • The method of the present invention can be used with any animal, including but not limited to, mammals such as rodents, dogs, cats, cattle, primates and humans. Preferably the method is used for gene therapy to treat human acquired or inherited cardiac conditions or diseases.
  • The present invention thus provides a method of treating and/or ameliorating a cardiovascular condition by infusing an rAAV vector for a time and in and amount sufficient to stable and efficiently transduce cardiomyocytes which was heretofore unachievable by methods known in the art. For this invention, stable and efficient transduction means that significant number of cardiomyocytes are transduced and are capable of expressing the protein for a prolonged period of time. Stable and efficient transduction occurs over a period of time and can actually be observed over time as an increase in the percentage of transduced cardiomyocytes, as continued expression of the transgene, or as continued observation of the therapeutic effect at a molecular, microscopic or macroscopic level. For example, with angiogenesis, stable and efficient transduction can be manifested by ongoing development and or growth of new blood vessels, by observing the improved blood flow to the heart, or by determining measuring the level of ischemia in the heart tissue.
  • Alternatively, efficient transduction occurs when at least about 10%, and preferably more, of the cardiomyocytes have been transduced, i.e., infected by, the rAAV. By following the methods of the invention and by observing at particular times after transduction ranging over a few to many weeks, about 25%, about 40% or even about 50% of the cardiomyocytes will be transduced. While about 10% of the cardiomyocytes can be transduced using only rAAV, this percentage can be increased by co-infusing adenovirus as a helper virus without adverse effects.
  • The time of infusion contributes to acheiving stable and efficient transduction of the cardiomyocytes as well. Thus the infusion time ranges from about 2 minutes to about 30 minutes, more preferably from about 5 minutes to about 20 minutes and most perferably is about fifteen minutes.
  • The amount of rAAV infused into the animal is proportional to the body weight of the animal. Hence in accordance with the invention, stable and efficient transduction occurs when the amount of rAAV infused ranges from about 1×105 IU (infectious units) of AAV per gram body weight to about 1×10 9 IU AAV per gram body weight, and preferably from about 1×106 IU AAV per gram body weight to about 1×108 IU AAV per gram body weight, and most preferably is about 5-6×107 IU AAV per gram body weight.
  • The example described below demonstrates efficient and stable transduction of cardiac myocytes in vivo after intracoronary infusion of an rAAV vector.
  • Throughout this application, various publications, patents, and patent applications have been referred to. The teachings and disclosures of these publications, patents, and patent applications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which the present invention pertains.
  • It is to be understood and expected that variations in the principles of invention herein disclosed in an exemplary embodiment may be made by one skilled in the art and it is intended that such modifications, changes, and substitutions are to be included within the scope of the present invention.
  • EXAMPLE Intracoronary Infusion of rAAV
  • I. Methods:
  • Plasmids and Viruses
  • The structure of pAAVCMV-LacZ is shown in FIG. 1. AdCMV-LacZ and the E3-deleted adenovirus, Add1309, were propagated and purified as described (Barr 1994).
  • Propagation and Purification of rAAV
  • rAAV was prepared as described (Rolling et al. (1995) Mol. Biotechnol. 3:9-15) and purified by cesium chloride gradient centrifugation. Viral titer was assessed by a dot blot hybridization assay to determine the number of viral genomes per milliliter and by infecting HeLa cells with the virus and staining with X-gal 24 hours after infection. All viral preparations had titers of 1 to 2×1011 genomes/mL, and 2 to 3×109 infectious units (IU)/mL.
  • Intracoronary Perfusion with rAAV
  • Adult C57BL/6 mouse hearts were perfused via the left carotid artery with cardioplegia solution (110 mmol/L NaCl, 25 mmol/L KCl, 22 mmol/L NaHCO3, 16 mmol/L MgCl2, 0.8 mmol/L CaCl2, 40 mmol/L glucose) at 4° C. until they stopped beating. They were then perfused ex vivo for 15 minutes with 1.5×109 IU of AAVCMV-LacZ in 0.5 mL of PBS at a rate of 33 μL/min at 4° C. After perfusion, the hearts were transplanted into the neck of a syngeneic host with anastomosis of the donor aorta to the right common carotid artery of the host and anastomosis of the donor pulmonary artery to the right external jugular vein (Lin et al. (1990b) J. Heart Transplant. 9:720-723) (n=3 for each time point).
  • X-Gal Staining
  • Freshly isolated hearts were fixed in PBS plus 1.25% glutaraldehyde for 10 minutes at room temperature, stained overnight with X-gal (Lin 1990a), and counterstained with eosin.
  • β-Galactosidase Activity
  • Cardiac homogenates were assayed for β-galactosidase (β-gal) activity and protein concentrations. β-Gal activities were normalized for total protein and for the number of infectious rAAV or RDAd particles injected.
  • II. Results
  • Many clinical applications of myocardial gene therapy may require the stable and efficient transduction of cardiomyocytes distributed throughout large areas of myocardium. Coronary artery infusions of RDAd have been shown to result in the efficient transduction of cardiomyocytes throughout the region of perfused myocardium (Barr 1994). To test whether rAAV is similarly capable of transducing cardiomyocytes after coronary artery perfusion, hearts from C57BL/6 mice were explanted and perfused with 1.5×109 IU of AAVCMV-LacZ for 15 minutes at 4° C. via a catheter placed in the left common carotid artery. These perfused hearts were then transplanted into syngeneic hosts, and the arterial circulation was reestablished by anastomosis of the transplanted aorta to the recipient carotid artery. Such transplanted and revascularized hearts resumed beating and continued to do so until the recipient mice were killed 2, 4, or 8 weeks after perfusion. Two weeks after perfusion, small numbers (<1%) of β-gal-positive cardiomyocytes were detected throughout the myocardium of the rAAV-perfused hearts (FIG. 2). By 4 weeks after perfusion, ≈40% of the cardiomyocytes were β-gal positive. This high level of transduction was stable at weeks after perfusion, with >50% of the cardiomyocytes continuing to express β-gal. Similar increases in recombinant gene expression over the first several weeks after rAAV infection have been observed in skeletal muscle (Fisher 1997; Kessler 1998). It has been postulated that such increases may reflect the gradual process of conversion of the single-stranded AAV genome into a double-stranded DNA molecule that is competent for transcription of the transgene (Ferrari et al. (1996) J. Virol. 70:3227-3234). Thus, rAAV delivered by coronary artery perfusion can be used to stably transduce cardiomyocytes throughout the myocardium.

Claims (23)

1. A method of treating a cardiovascular condition which comprises:
infusing a recombinant adeno-associated virus (AAV) vector into a coronary artery or a coronary sinus for a time and in an amount sufficient to stably and efficiently transduce cardiomyocytes perfused by said artery or said sinus, wherein said AAV vector encodes at least one nucleic acid operably linked to a control region, said nucleic acid encoding a therapeutically-effective molecule; and
expressing said therapeutically-effective molecule in an amount effective to treat or ameliorate said cardiovascular condition.
2. The method of claim 1, wherein said AAV transduces at least about 10% of said cardiomyocytes.
3. The method of claim 1, wherein said AAV transduces at least about 40% of said cardiomyocytes.
4. The method of claim 1, wherein said AAV transduces at least about 50% of said cardiomyocytes.
5. The method of claim 1, wherein said AAV is infused for at least about 2 minutes to about 30 minutes.
6. The method of claim 1, wherein said AAV is infused for at least about 5 minutes to about 20 minutes.
7. The method of claim 1, wherein said AAV is infused for about 15 minutes.
8. The method of claim 1, wherein said amount of AAV is about 1×105 IU AAV per gram body weight to about 1×109 IU AAV per gram body weight.
9. The method of claim 9, wherein said amount of AAV is about 1×106 IU AAV per gram body weight to about 1×108 IU AAV per gram body weight.
10. The method of claim 9, wherein said amount of AAV is about 6×107 IU AAV per gram body weight.
11. The method of claim 1, wherein about 1×105 IU AAV per gram body weight to about 1×109 IU AAV per gram body weight is infused for about 2 to about 30 minutes.
12. The method of claim 11, wherein about 1×106 IU AAV per gram body weight to about 1×108 IU AAV per gram body weight is infused.
13. The method of claim 11, wherein about 6×107 IU AAV per gram body weight is infused.
14. The method of any one of claims 11, 12 or 13, wherein said AAV is infused for about 5 to about 20 minutes.
15. The method of any one of claims 11, 12 or 13, wherein said AAV is infused for about 15 minutes.
16. The method of claim 11, wherein about 6×107 IU AAV per gram body weight is infused for about 15 minutes.
17. The method of claim 1, wherein said coronary artery is infused ex vivo or in vivo.
18. The method of claim 1, wherein said therapeutically-effective molecule is an anti-sense RNA or a protein.
19. The method of claim 1 wherein therapeutically-effective molecule is an ion channel gene, a contractile protein, a phospholamban, a β adrenergic receptor, a β adrenergic kinase, a growth factor, an angiogenic factor, a protein or nucleic acid capable of inducing angiogenesis, or a protein or nucleic acid capable of inhibiting angiogenesis.
20. The method of claim 1, wherein said therapeutically-effective molecule is FGF-1, FGF-2, FGF-5, VEGF, or HIF-1.
21. The method of claim 1, wherein said therapeutically-effective molecule is thymidine kinase, p21, p27, p53, Rb or NF-κB.
22. The method of claim 1, wherein said cardiovascular condition is restenosis, atherosclerosis, congestive heart failure, ischemic cardiomyopathy, malignant arrhythmia, myocardial infarction, congestive heart failure, or dilated and hypertrophic cardiomyopathy.
23. The method of claim 1, wherein treating or ameliorating said cardiovascular condition is for inducing angiogenesis, inhibiting angiogenesis, stimulating or inhibiting cell proliferation, treating restenosis, treating atherosclerosis, treating congestive heart failure, treating ischemic cardiomyopathy or treating malignant arrhythmia.
US11/449,778 1998-12-28 2006-06-09 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors Abandoned US20060251626A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/449,778 US20060251626A1 (en) 1998-12-28 2006-06-09 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11392398P 1998-12-28 1998-12-28
US09/473,830 US7078387B1 (en) 1998-12-28 1999-12-28 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors
US11/449,778 US20060251626A1 (en) 1998-12-28 2006-06-09 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/473,830 Continuation US7078387B1 (en) 1998-12-28 1999-12-28 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors

Publications (1)

Publication Number Publication Date
US20060251626A1 true US20060251626A1 (en) 2006-11-09

Family

ID=36659079

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/473,830 Expired - Fee Related US7078387B1 (en) 1998-12-28 1999-12-28 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors
US11/449,778 Abandoned US20060251626A1 (en) 1998-12-28 2006-06-09 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/473,830 Expired - Fee Related US7078387B1 (en) 1998-12-28 1999-12-28 Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors

Country Status (1)

Country Link
US (2) US7078387B1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8221738B2 (en) 2008-02-19 2012-07-17 Celladon Corporation Method for enhanced uptake of viral vectors in the myocardium

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7745416B2 (en) * 1995-04-11 2010-06-29 The Regents Of The University Of California Method for in vivo regulation of cardiac muscle contractility
WO2002064157A2 (en) * 2001-01-23 2002-08-22 Boston Scientific Corporation Localized myocardial injection method for treating ischemic myocardium
AU2006315601A1 (en) * 2005-11-14 2007-05-24 Enterprise Partners Venture Capital Stem cell factor therapy for tissue injury
US9517250B2 (en) 2010-04-28 2016-12-13 The J. David Gladstone Institutes Methods for generating cardiomyocytes
EP2580328A2 (en) 2010-06-11 2013-04-17 Cellartis AB Micrornas for the detection and isolaton of human embryonic stem cell-derived cardiac cell types
GB2537000C (en) 2012-05-25 2019-10-09 Univ California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP3760719A1 (en) 2013-11-18 2021-01-06 CRISPR Therapeutics AG Crispr-cas system materials and methods
US9994831B2 (en) 2013-12-12 2018-06-12 The Regents Of The University Of California Methods and compositions for modifying a single stranded target nucleic acid
BR112016027897A2 (en) 2014-06-18 2017-10-24 Albert Einstein College Medicine Inc multimeric polypeptide, nucleic acid, recombinant expression vector, genetically modified host cell, composition, method, and method of treating an infection in an individual
SG10202008443UA (en) 2015-03-13 2020-10-29 Jackson Lab A three-component crispr/cas complex system and uses thereof
WO2016164371A1 (en) 2015-04-07 2016-10-13 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Methods for inducing cell division of postmitotic cells
WO2016196655A1 (en) 2015-06-03 2016-12-08 The Regents Of The University Of California Cas9 variants and methods of use thereof
US20190048340A1 (en) 2015-09-24 2019-02-14 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
PT3380613T (en) 2015-11-23 2022-12-02 Univ California Tracking and manipulating cellular rna via nuclear delivery of crispr/cas9
KR20180132070A (en) 2016-03-03 2018-12-11 큐 바이오파마, 인크. T-cell modulated multimeric polypeptides and methods for their use
BR112018073606A2 (en) 2016-05-18 2019-02-26 Cue Biopharma, Inc. T-cell modulating multimeric polypeptides and methods of using them
JP2019522466A (en) 2016-05-18 2019-08-15 アルバート アインシュタイン カレッジ オブ メディシン,インコーポレイティド Mutant PD-L1 polypeptide, T cell regulatory multimeric polypeptide, and methods of use thereof
WO2018053037A1 (en) 2016-09-13 2018-03-22 The Jackson Laboratory Targeted enhanced dna demethylation
US11332713B2 (en) 2016-11-16 2022-05-17 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
CN110325205B (en) 2016-12-22 2023-08-15 库尔生物制药有限公司 T cell modulating multimeric polypeptides and methods of use thereof
EP3565829A4 (en) 2017-01-09 2021-01-27 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
CN110869498A (en) 2017-05-10 2020-03-06 加利福尼亚大学董事会 CRISPR/CAS9 directed editing of cellular RNA via nuclear delivery
CA3074839A1 (en) 2017-09-07 2019-03-14 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptide with conjugation sites and methods of use thereof
JP2021506251A (en) 2017-12-14 2021-02-22 クリスパー セラピューティクス アーゲー New RNA programmable endonuclease system, as well as its use in genome editing and other applications
CN111886241A (en) 2018-01-09 2020-11-03 库尔生物制药有限公司 Multimeric T cell modulating polypeptides and methods of use thereof
EP3765092A4 (en) 2018-03-15 2022-01-12 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
BR112020018658A2 (en) 2018-03-15 2020-12-29 KSQ Therapeutics, Inc. GENE REGULATION COMPOSITIONS AND METHODS FOR IMPROVED IMU-NOTERAPY
JP2021518139A (en) 2018-03-19 2021-08-02 クリスパー セラピューティクス アーゲー New RNA Programmable Endonuclease System and Its Use
WO2020018166A1 (en) 2018-07-16 2020-01-23 The Regents Of The University Of Michigan Nuclease-mediated nucleic acid modification
TW202039542A (en) 2018-12-19 2020-11-01 美商庫爾生物製藥有限公司 Multimeric t-cell modulatory polypeptides and methods of use thereof
EP3935080A4 (en) 2019-03-06 2023-04-05 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
CA3132630A1 (en) 2019-03-12 2020-09-17 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
EP4003508A1 (en) 2019-07-31 2022-06-01 Memorial Sloan Kettering Cancer Center Perfusion modulated tumor dose sculpting with single dose radiotherapy
US20230295615A1 (en) 2020-08-07 2023-09-21 The Jackson Laboratory Targeted Sequence Insertion Compositions and Methods
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
AU2022290382A1 (en) 2021-06-11 2023-11-23 Bayer Aktiengesellschaft Type v rna programmable endonuclease systems
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2024068996A1 (en) 2022-09-30 2024-04-04 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
WO2024129743A2 (en) 2022-12-13 2024-06-20 Bluerock Therapeutics Lp Engineered type v rna programmable endonucleases and their uses

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5661133A (en) * 1991-11-12 1997-08-26 The Regents Of The University Of Michigan Expression of a protein in myocardium by injection of a gene
US5705388A (en) * 1994-12-23 1998-01-06 Ribozyme Pharmaceuticals, Inc. CETP Ribozymes
US5792453A (en) * 1995-02-28 1998-08-11 The Regents Of The University Of California Gene transfer-mediated angiogenesis therapy
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
US6100242A (en) * 1995-02-28 2000-08-08 The Regents Of The University Of California Gene therapies for enhancing cardiac function

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6162796A (en) 1995-09-27 2000-12-19 The Rockefeller University Method for transferring genes to the heart using AAV vectors
US5962313A (en) 1996-01-18 1999-10-05 Avigen, Inc. Adeno-associated virus vectors comprising a gene encoding a lyosomal enzyme
CA2247099A1 (en) 1996-03-04 1997-09-12 Targeted Genetics Corporation Methods for transducing cells in blood vessels using recombinant aav vectors
ATE270324T1 (en) 1997-04-14 2004-07-15 Cell Genesys Inc METHODS FOR INCREASE THE EFFICIENCY OF RECOMBINANT AAV PRODUCTS
US6037177A (en) 1997-08-08 2000-03-14 Cell Genesys, Inc. Method for increasing the efficiency of recombinant AAV production

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5661133A (en) * 1991-11-12 1997-08-26 The Regents Of The University Of Michigan Expression of a protein in myocardium by injection of a gene
US5661133B1 (en) * 1991-11-12 1999-06-01 Univ Michigan Collateral blood vessel formation in cardiac muscle by injecting a dna sequence encoding an angiogenic protein
US5705388A (en) * 1994-12-23 1998-01-06 Ribozyme Pharmaceuticals, Inc. CETP Ribozymes
US5792453A (en) * 1995-02-28 1998-08-11 The Regents Of The University Of California Gene transfer-mediated angiogenesis therapy
US6100242A (en) * 1995-02-28 2000-08-08 The Regents Of The University Of California Gene therapies for enhancing cardiac function
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

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8221738B2 (en) 2008-02-19 2012-07-17 Celladon Corporation Method for enhanced uptake of viral vectors in the myocardium
US8636998B2 (en) 2008-02-19 2014-01-28 Celladon Corporation Method for enhanced uptake of viral vectors in the myocardium

Also Published As

Publication number Publication date
US7078387B1 (en) 2006-07-18

Similar Documents

Publication Publication Date Title
US7078387B1 (en) Efficient and stable in vivo gene transfer to cardiomyocytes using recombinant adeno-associated virus vectors
US6936243B2 (en) Adeno-associated viral vector-mediated delivery of DNA to cells of the liver
Bish et al. Adeno-associated virus (AAV) serotype 9 provides global cardiac gene transfer superior to AAV1, AAV6, AAV7, and AAV8 in the mouse and rat
US6100242A (en) Gene therapies for enhancing cardiac function
US5792453A (en) Gene transfer-mediated angiogenesis therapy
US7238674B2 (en) Methods for delivering DNA to muscle cells using recombinant adeno-associated virus vectors
Chu et al. Direct comparison of efficiency and stability of gene transfer into the mammalian heart using adeno-associated virus versus adenovirus vectors
US20060292117A1 (en) Improved rAAv vectors
US20070128163A1 (en) Adeno-associated virus-mediated delivery of angiogenic factors
US5780447A (en) Recombinant adeno-associated viral vectors
JP2004313198A (en) Adenovirus-mediated gene transfer to cardiac or vascular smooth muscle
WO1998024479A9 (en) Adeno-associated viral vector-mediated delivery of dna to cells of the liver
AU763049B2 (en) Efficient and stable (in vivo) gene transfer to cardiomyocytes using recombinantadeno-associated virus vectors
US20020159978A1 (en) Muscle-directed gene transfer by use of recombinant AAV-1 and AAV-6 virions
Giacca Virus-mediated gene transfer to induce therapeutic angiogenesis: where do we stand?
EP1695719A1 (en) Combination of a nucleic acid and a vasoactive agent for enhanced gene delivery
Wang et al. AAV gene transfer to the heart
AU2002226400B2 (en) Sequences upstream of the carp gene, vectors containing them and uses thereof
US20060099183A1 (en) Use of recombinant adeno-associated virus vector (rAAV) for the prevention of smooth muscle cell proliferation in a vascular graft
AU2002226400A1 (en) Sequences upstream of the carp gene, vectors containing them and uses thereof
AU706050C (en) Gene transfer-mediated angiogenesis therapy
Herzog et al. Improved Hepatic Gene Transfer by Using
ZACCHIGNA et al. IN VIVO GENE TRANSFER WITH AAV VECTORS AS A TOOL TO UNDERSTAND GENE FUNCTION IN BLOOD VESSEL FORMATION
AU2006200170A1 (en) Combination of a nucleic acid and a vasoactive agent for enhanced gene delivery

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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