NZ253943A - Transfering polynucleotides into eukaryotic cells using co-lipofection complexes of a cationic lipid and the polynucleotide - Google Patents

Transfering polynucleotides into eukaryotic cells using co-lipofection complexes of a cationic lipid and the polynucleotide

Info

Publication number
NZ253943A
NZ253943A NZ25394393A NZ25394393A NZ253943A NZ 253943 A NZ253943 A NZ 253943A NZ 25394393 A NZ25394393 A NZ 25394393A NZ 25394393 A NZ25394393 A NZ 25394393A NZ 253943 A NZ253943 A NZ 253943A
Authority
NZ
New Zealand
Prior art keywords
gene
polynucleotide
yac
cells
dna
Prior art date
Application number
NZ25394393A
Inventor
Theodore Kyu Choi
Jeanne Frances Loring
Robert Maxwell Kay
Original Assignee
Genpharm Int
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 Genpharm Int filed Critical Genpharm Int
Publication of NZ253943A publication Critical patent/NZ253943A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Knock-in vertebrates, e.g. humanised vertebrates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0312Animal model for Alzheimer's disease
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Environmental Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Neurology (AREA)
  • Immunology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Description

New Zealand No. 253943 International No. PCT/US93/05873 TO BE ENTERED AFTER ACCEPTANCE AMD PUBLICATION Priority dates; International fifing date: Classification: ClO.Ni IS|oo,31, C.11WS |00,10\ Pik>\ V<31 ho", Qlo* K3S | IQ.
Publication date: 2 9 JAN W97 Journal No.: NIW ZEALAND PATENTS ACT 1953 COMPLETE SPECIFICATION Title of invention: Methods for producing transgenic non-human animals harboring a yeast" artificial chromosome Name, address and nationality of applicants) as in international application form: GENPHARM INTERNATIONAL, INC. of 297 North Bernardo Avenue, Mountain View, California 94043, United States of America I C Gol\OVkj<sc3 - ' 253943 METHODS FOR PRODUCING TRANSGENIC NON-HUMAN ANIMALS HARBORING A YEAST ARTIFICIAL CHROMOSOME TECHNICAL FIELD The invention relates to transgenic non-human animals capable of expressing xenogenic polypeptides, 15 transgenes used to produce such transgenic animals, transgenes capable of expressing xenogenic polypeptides, yeast artificial chromosomes comprising a polynucleotide sequence encoding a human protein such as a human immunoglobulin or amyloid precursor protein (APP), methods and transgenes for 20 transferring large polynucleotide sequences into cells, and methods for co-lipofection of discontinuous polynucleotide sequences into cells.
BACKGROUND OF THE INVENTION 25 Transferring exogenous genetic material into cells is the basis for modern molecular biology. The continuing development of novel methods for improving the efficiency, specificity, and/or size limitations of the transfer process has broadened the scope of research and product development by 30 enabling the production of polynucleotide clones and recombinant organisms that previously were impractical or impossible to construct. Calcium phosphate precipitation, electroporation, lipofection, ballistic transfer, DEAE-dextran transfection, microinjection, and viral-based transfer 35 methods, among others, have been described for introducing foreign DNA fragments into mammalian cells.
The art also has developed yeast artificial chromosome ("YAC") cloning vectors which are capable of propagating large (50 to more than 1000 kilobases) cloned 40 inserts (U.S. Patent 4,889,806) of xenogenic DNA. YAC clone WO 94/00569 PCT/US93/05873 libraries have been used to identify, map, and propagate large fragments of mammalian genomic DNA. YAC cloning is especially useful for isolating intact genes, particularly large genes having exons spanning several tens of kilobases or more, and 5 genes having distal regulatory elements located tens of kilobases or more upstream or downstream from the exonic sequences. YAC cloning is particularly advantageous for isolating large complex gene loci, such as unrearranged immunoglobulin gene loci, and genes which have been inexactly 10 mapped to an approximate chromosomal region (e.g., a Huntington's chorea gene). YAC cloning is also well-suited for making vectors for performing targeted homologous recombination in mammalian cells, since YACs allow the cloning of large contiguous sequences useful as recombinogenic 15 homology regions in homologous targeting vectors. Moreover, YACs afford a system for doing targeted homologous recombination in a yeast host cell to create novel, large transgenes (e.g., large minigenes, tandem gene arrays, etc.) in YAC constructs which could then be transferred to mammalian 2 0 host cells.
Unfortunately, manipulation of large polynucleotides is problematic. Large polynucleotides are susceptible to breakage by shearing forces and form highly viscous solutions even at relatively dilute concentrations, making in vitro 25 manipulation exceedingly difficult. For these reasons, and others, it is desirable to reduce the amount of manipulation that YAC clones and other large DNA fragments are subjected to in the process of constructing large transgene constructs or homologous recombination constructs. 3 0 More problematic is the fact that the transfer of large, intact polynucleotides into mammalian cells is typically inefficient or provides a restriction on the size of the polynucleotide transferred. For example, Schedl et al. (1992) Nucleic Acids Res. 20: 3073, describe transferring a 35 35 kilobase YAC clone into the mouse genome by pronuclear injection of murine embryos; however, the shear forces produced in the injection micropipette will almost certainly preclude the efficient transfer of significantly larger YAC clones in an intact form. Many large genes likely could not be transferred efficiently into mammalian cells by current microinjection methods.
Spheroplast fusion has been used to introduce YAC 5 DNA into fibroblasts, embryonal carcinoma cells, and CHO cells (Pachnie et al. (1990) Proc. Natl. Acad. Sci. (U.S.A.! 87; 5109; Payan et al. (1990) Mol. Cell. Biol. 10: 4163; Chirke et al. (1991) EMBO J. 10; 1629; Davies et al. (1992) Nucleic Acids Res. 20; 2693). Alternative transfection methods such 10 as calcium phosphate precipitation and lipofection have been used to transfer YAC DNA into mammalian cells (Eticciri et al. (1991) Proc. Natl. Acad. Sci. (U.S.A.) 88.: 2179; Strauss W and Jaenisch R (1992) EMBO J. 11: 417).
Thus, there exists a need in the art for an 15 efficient method for transferring large segments of DNA, such as large YAC clones, _nto mammalian cells, such as embryonic stem cells for making transgenic animals, with a minimum of manipulation and cloning procedures. In particular, it would be highly advantageous if it were possible to isolate a large 20 cloned mammalian genomic fragment from a YAC library, either linked to YAC yeast sequences or purified away from YAC yeast sequences, and transfer it intact into a mammalian host cell (e.g., an ES cell) with a second polynucleotide sequence (e.g., a selectable marker such as a neoR expression cassette) 25 without additional cloning or manipulation (e.g., ligation of the sequences to each other). Such a method would allow the efficient construction of transgenic cells, transgenic animals, and homologously targeted cells and animals. These transgenic/homologously targeted cells and animals could 30 provide useful models of, for example, human genetic diseases such._as Huntington's chorea and Alzheimer's disease, among others.
Alzheimer's Disease At present there is no known therapy for the various 35 forms of Alzheimer's disease (AD). However, there are several disease states for which effective treatment is available and which give rise to progressive intellectual deterioration closely resembling the dementia associated with Alzheimer's disease.
Alzheimer's disease is a progressive disease known generally as senile dementia. Broadly speaking the disease 5 falls into two categories, namely late onset and early onset. Late onset, which occurs in old age (65 + years), may be caused by the natural atrophy of the brain occurring at a faster rate and to a more severe degree than normal. Early onset Alzheimer's disease is much more infrequent but shows a 10 pathologically identical dementia with brain atrophy which develops well before the senile period, i.e.. between the ages of 35 and 60 years. There is evidence that one form of this type of Alzheimer's disease is inherited and is therefore known as familial Alzheimer's disease (FAD). 15 In both types of Alzheimer's disease the pathology is the same but the abnormalities tend to be more severe _and more widespread in cases beginning at an earlier age. The disease is characterized by four types of lesions in the brain, these are: amyloid plaques around neurons (senile 20 plaques), amyloid deposits around cerebral blood vessels, neurofibrillary tangles inside neurons, and neuronal cell death. Senile plaques are areas of disorganized neuropil up to 150/xm across with extracellular amyloid deposits at the center. Cerebrovascular amyloid deposits are amyloid material 25 surrounding cerebral blood vessels. Neurofibrillary tangles are intracellular deposits of amyloid protein consisting of two filaments twisted about each other in pairs.
The major protein subunit, amyloid /3 protein, is found in amyloid filaments of both the neurofibrillary tangle 3 0 and the senile plaque and is a highly aggregating small polypeptide of approximate relative molecular mass 4,000.
This protein is a cleavage product of a much larger precursor protein called amyloid precursor protein (APP).
The APP gene is known to be located on human 35 chromosome 21. A locus segregating with familial Alzheimer's disease has been mapped to chromosome 21 (St. George Hyslop et al (1987) Science 235: 885) close to the APP gene.
Recombinants between the APP gene and the AD locus hav6 been WO 94/00569 PCT/US93/05873 previously reported (Schellenberg et al. (1988) Science 241: 1507; Schellenberg et al. (1991) Am. J. Hum. Genetics 48: 563; Schellenberg et al. (1991) Am. J. Hum. Genetics 49: 511, incorporated herein by reference). The development of 5 experimental models of Alzheimer's disease that can be used to define further the underlying biochemical events involved in AD pathogenesis would be highly desirable. Such models could presumably be employed, in one application, to screen for agents that alter the degenerative course of Alzheimer's 10 disease. For example, a model system of Alzheimer's disease could be used to screen for environmental factors that induce or accelerate the pathogenesis of AD. In contradistinction, an experimental model could be used to screen for agents that inhibit, prevent, or reverse the progression of AD. 15 Presumably, such models could be employed to develop pharmaceuticals that are effective in preventing, arresting, or reversing AD.
Unfortunately, only humans and aged non-human primates develop any of the pathological features of AD; the 20 expense and difficulty of using primates and the length of time required for developing the AD pathology makes extensive research on such animals prohibitive. Rodents do not develop AD, even at an extreme age. It has been reported that the injection of /3-amyloid protein (/SAP) or cytotoxic ]3AP 25 fragments into rodent brain results in cell loss and induces an antigenic marker for neurofibrillary tangle components (Kowall et al. (1991) Proc. Natl. Acad. Sci. fU.S.A.I 88: 7247). Mice which carry an extra copy of the APP gene as a result of partial trisomy of chromosome 16 die before birth 30 (Coyle et al. (1988) Trends in Neurosci. 11: 390). Since the cloning of the APP gene, there have been several attempts to produce a mouse model for AD using nransgenes that include all or part of the APP gene, unfortunately much of the work remains unpublished since the mice were nonviable or failed to 35 show AD-like pathology; two published reports were retracted because of irregularities in reported results (Marx J Science 255: 1200).
WO 94/00569 PCT/US93/05873 6 Thus, there is also a need in the art for transgenic nonhuman animals harboring an intact human APP gene, either a wild-type allele, a disease-associated allele, or a combination of these, or a mutated rodent (e.g., murine) 5 allele which comprises sequence modifications which correspond to a human APP sequence. Cell strains and cell lines (e.g., astroglial cells) derived from such transgenic animals would also find wide application in the art as experimental models for developing AD therapeutics.
Nonhuman Transgenic Animals Expressing Human Immunoglobulin Making human monoclonal antibodies that bind predetermined antigens is difficult, requiring a source of viable lymphocytes from a human that has been immunized with an antigen of choice and which has made a substantial humoral 15 immune response to the immunogen. In particular, humans generally are incapable of making a substantial antibody response to a challenge with a human (self) antigen; unfortunately, many such human antigens are promising targets for therapeutic strategies involving human monoclonal 20 antibodies. One approach to making human antibodies that are specifically reactive with predetermined human antigens involves producing transgenic mice harboring unrearranged human immunoglobulin transgenes and having functionally disrupted endogenous immunoglobulin gene(s) (Lonberg and Kay, 25 WO 92/03918; Kucherlapati and Jakobovits, WO 91/10741).
However, efficiently transferring large DNA segments, such as those spanning significant portions of a human light or heavy chain immunoglobulin gene locus, presents a potential obstacle and/or reduces the efficiency of the process of generating the 30 transgenic animals.
Based on the foregoing, it is clear that a need exists for nonhuman cells and nonhuman animals harboring one or more large, intact transgenes, particularly a human APP gene or a human immunoglobulin transgene(s). Thus, it is an 35 object of the invention herein to provide methods and compositions for transferring large transgenes and large homologous recombination constructs, usually cloned as YACs, into mammalian cells, especially into embryonic stem cells. 3943 7 It is also an object of the invention to provide transgenic nonhuman cells and transgenic nonhuman animals harboring one or more APP transgenes of the invention.
The references discussed herein are provided solely for their disclosure prior to the filing dat* of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention. All citations are incorporated herein by reference.
SUMMARY OF THE INVENTION In a first aspect, the present invention provides a method for producing a co-lipofected mammalian cell having incorporated multiple heterologous DNA species, comprising the steps of: forming a co-lipofection complex comprising a cationic lipid, a first polynucleotide, and an unlinked second polynucleotide comprising at least one selectable marker gene expression cassette; contacting a mammalian cell with said co-lipofection complex under conditions whereby said first polynucleotide and said second polynucleotide are introduced into the same cell and are integrated into the genome.
In a further aspect, the present invention provides a co-lipofection complex, comprising cationic lipid noncovalently bound to a first DNA comprising a YAC clone containing xenogenic mammalian DNA and to a second DNA comprising a gene encoding a selectable marker.
The present invention further provides a co-lipofection complex comprising a polynucleotide of at least 50 kb, an unlinked selectable marker gene expression cassette, and a cationic lipid selected from the group consisting of: N[l-(2,3-dioleoyloxyl)propyl]-N,N,N-trimethylammonium chloride; N[1-2,3-dioleoyloxyl)propyl]-N,N,N-trimethy1ammonium methylsulfate; and dioctadecylamidoglycy1 spermidine. 253943 7a In a still further aspect, the present invention provides a composition comprising a mammalian cell and a co-lipofection complex of the invention.
In another aspect, the present invention provides a cotransfected mammalian cell produced by a method of the invention.
In a still further aspect, the present invention provides a transgenic, or chimeric transgenic non-human animal produced by a method of the invention.
The present invention further provides a transgenic mouse comprising a germline genome comprising a xenogenic polynucleotide sequence of at least 50 kb linked to a yeast-derived YAC sequence produced by cotransfecting a mouse ES cell with the composition of the invention.
The present invention also provides a transgenic mouse produced by cotransfecting a mouse ES cell with the composition of the invention comprising a YAC comprising an unrearranged human immunoglobulin gene having at least one V region gene, at least one J region gene, and at least one constant region gene.
In a still further aspect, the present invention provides a co-lipofection complex comprising a YAC containing an unrearranged human immunoglobulin gene comprising the 85 kb Spe I fragment, an unlinked selectable marker, and a cationic lipid.
In yet another aspect, the present invention provides a method of introducing a heterologous polynucleotide containing a complete structural gene into a mammalian cell, comprising the steps of: forming a lipofection complex comprising a cationic lipid and a heterologous first polynucleotide comprising a complete structural gene and an unlinked second polynucleotide comprising a selectable marker; contacting a mammalian cell with said lipofection complex under conditions whereby said heterologous first and second polynucleotides are introduced into the mammalian cell and are integrated into the genome. 7b 253943 BRIEF DESCRIPTION OF THE INVENTION In accordance with the foregoing objects, in one aspect of the invention methods for transferring large transgenes and large homologous targeting constructs, typically propagated as YACs and preferably spanning at least one complete transcriptional complex, into mammalian cells., such as ES cells, are provided. In one aspect, the methods provide for transferring the large transgenes and large homologous targeting constructs by a lipofection method, such as co-lipofection, wherein a second unlinked polynucleotide is transferred into the mammalian cells along with the large transgene and/or large homologous targeting construct. Preferably, the second polynucleotide confers a selectable phenotype (e.g., resistance to G418 selection) to cells which have taken up and integrated the polynucleotide sequence(s). Usually, the large transgene or homologous targeting construct is transferred with yeast-derived YAC sequences in polynucleotide linkage, but yeast-derived YAC sequences may be removed by restriction enzyme digestion and separation (e.g, pulsed gel electrophoresis). The large transgene(s) and/or homologous targeting construct(s) are generally mixed with the unlinked second polynucleotide (e.g., a neoR expression cassette to confer a selectable phenotype) and contacted with a cationic lipid (e.g., DOGS, DOTMA, DOTAP) to form cationic lipid-DNA complexes which are contacted with mammalian cells (e.g., ES cells) in conditions suitable for uptake of the DNA into the cells (e.g., culture medium, physiological phosphate-buffered saline, serum-free ES medium). Generally, cells 8 harbarisig the large transgene or large homologous targeting constatiict concomitantly harbor at least one copy of the second polynucleotide, so that selection for-cells harboring the second polynucleotide have a significant probability of also 5 harboring at least one copy of the large transgene or large homologous targeting construct, generally as an integrated or homologously recombined segment of an endogenous chromosomal locus. Hence selection for the second polynucleotide (e.g., neoR expression cassette) generally also selects cells 10 harboring the large transgene or large homologous targeting construct without requiring cumbersome polynucleotide linkage (i.e., ligation) of the large transgene or large homologous targeting construct to the second polynucleotide prior to lipofection. According to the co-lipofection methods of the 15 invention, large segments of xenogenic DNA are rapidly and efficiently transferred into mammalian cells (e.g., murine ES cells) without requiring linkage of a selectable marker gene and subsequent cloning.
The invention also provides mammalian cells, 20 preferably ES cells, harboring at least one copy of integrated or homologously recombined large xenogenic (preferably heterologous) mammalian genomic DNA sequences linked to yeast-derived YAC sequences. Preferably, the large xenogenic (preferably heterologous) mammalian genomic DNA sequences 25 "comprise a complete structural gene, more preferably a complete transcriptional unit, and in one embodiment a complete human APP gene. Typically, the resultant transgenic mammalian cells also comprise at least one integrated copy of the unlinked second polynucleotide (e.g., the selectable 30 marker), which is usually nonhomologously integrated into at least one chromosomal locus, sometimes at a chromosomal locus distinct from that at which the large transgene (s) or large homologous targeting construct(s) has been incorporated.
During the transfection process and shortly thereafter, novel 35 mammalian cells, such as ES cells, comprising large foreign DNA sequences, an unlinked selectable marker gene, and a suitable cationic lipid are formed, such novel mammalian cells are one aspect of the present invention.
PCT /US93/05873 The invention also provides transgenic nonhuman animals comprising a genome having at least one copy of integrated or homologously recombined large xenogenic (preferably heterologous) mammalian genomic DNA sequences 5 linked to yeast-derived YAC sequences. Preferably, the large xenogenic (preferably heterologous) mammalian genomic DNA sequences comprise a complete structural gene, more preferably a complete transcriptional unit, and in one embodiment a complete human APP gene. Typically, the resultant transgenic 10 nonhuman mammal also comprises a genome having- at least one integrated copy of the unlinked second polynucleotide (e.g., the selectable marker), which is usually nonhomologously integrated into at least one chromosomal locus, sometimes at a chromosomal locus/loci distinct from that at which the large 15 transgene(s) or large homologous targeting construct(s) has/have been incorporated. Preferably, the large transgene and/or large homologous targeting construct which has been incorporated into a chromosomal locus (or loci) of the nonhuman animal is expressed, more preferably is expressed 20 similarly to the naturally-occurring homolog gene in the non-human animal species (e.g., in a similar tissue-specific pattern arid/or developmental pattern) .
The invention also provides compositions for co-lipofection: transgenesis compositions and homologous 25 targeting compositions for transferring xenogenic, typically heterologous, large (i.e., 50 kb or more) polynucleotides into mammalian cells, such as ES cells for making transgenic nonhuman animals harboring at least one copy of at least one integrated large foreign transgene and/or harboring at least 30 one homologously targeted construct in its genome. A transgenesis composition comprises: (1) at least one large transgene species, (2) at least one unlinked second polynucleotide species (such as an expression cassette containing the selectable marker gene neoR), and (3) at least 35 one species of suitable cationic lipid. A homologous targeting composition comprises: (1) at least one large homologous targeting construct species, (2) at least one unlinked second polynucleotide species (such as an expression cassette containing the selectable marker gene neoR), and (3) at least one species of suitable cationic lipid. Preferably the large transgene or large homologous targeting construct spans an entire transcriptional unit. One preferred 5 embodiment of a co-lipofection composition is a composition comprising: (1) a human APP gene sequence (or a modified murine or rat APP gene having a non-naturally occurring sequence corresponding to a human APP sequence) linked to yeast.-derived YAC sequences, (2) an expression cassette 10 encoding a selectable marker, and (3) a suitable cationic lipid. Another preferred embodiment of a co-lipofection composition is a composition comprising: (l) a human unrearranged immunoglobulin gene sequence (heavy or light chain gene sequence comprising at least two V gene complete 15 segment, at least one complete D segment (if heavy chain gene) , at least one complete J segment, and at J.east one constant region gene) linked to yeast-derived YAC sequences, (2) an expression cassette encoding a selectable marker, and (3) a suitable cationic lipid.
In one aspect of the invention, multiple species of unlinked polynucleotide sequences are co-lipofected into murine embryonic stem cells and/or other mammalian cells, wherein at least one species of the unlinked polynucleotide sequences comprises a selectable marker gene which confers a 25 selectable phenotype to cells which have incorporated it. The resultant cells are selected for the presence of the selectable marker; such selected cells have a significant probability of comprising at least one integrated copy of the other species of polynucleotide sequence(s) introduced into 30 the cells.
BRIEF DESCRIPTION OF THE FIGURES Fig. 1: Chemical structures of representative cationic lipids for forming co-lipofection complexes of the 35 present invention.
Fig. 2: PCR analysis of ES clones co-lipofected with the human APP transgene. Shaded circles denote wells which were not used. Row pools (A-P) contained 18 (A-H) or 16 (I-P) WO 94/00569 PCT/US93/05873 11 clones each. Column pools (P1-P18) contained 16 (P1-P12) or 8 (P13-P18) clones each.
Fig. 3: PCR analysis of ES clones co-lipofected with the human APP transgene. Pools P3, P4, P9, P10, Pll, and P12 5 and pools G, H, K, M, N, O, and P were candidates for containing clones with both promoter and exon 17 sequences.
Fig. 4: PCR analysis of ES clones co-lipofected with the human APP transgene.
Fig. 5: Southern blot analysis of YAC clone DNA 10 using a human Alu sequence probe.
Fig. 6: Partial restriction digest mapping of human APP YAC.
Fig. 7: PCR analysis of RNA transcripts expressed from integrated human APP transgene.
Fig. 8: Quantitative RNase protection assay for detecting APP RNA transcripts from human APP transgene. Definitions Unless defined otherwise, all technical and scientific terms usee herein have the same meaning as commonly 20 understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are described. For purposes of the 25 present invention, the following terms are defined below.
The term "corresponds to" is used herein to mean that a polynucleotide sequence is homologous (i.e., is identical, not strictly evolutionarily related) to all or a portion of a reference polynucleotide sequence, or that a 30 polypeptide sequence is identical to a reference polypeptide sequence. In contradistinction, the term "complementary to" is used herein to mean that the complementary sequence is homologous to all or a portion of a reference polynucleotide sequence. For illustration, the nucleotide sequence "TATAC" 35 corresponds to a reference sequence "TATAC" and is complementary to a reference sequence "GTATA." The terms "substantially corresponds to", "substantially homologous", or "substantial identity" as used WO 94/00569 PCT/US93/05873 12 herein denotes a characteristic of a nucleic acid sequence, wherein a nucleic acid sequence has at least 70 percent sequence identity as compared to a reference sequence, typically at least 85 percent sequence identity, and 5 preferably at least 95 percent sequence identity as compared to a reference sequence. The percentage of sequence identity is calculated excluding small deletions or additions which total less than 25 percent of the reference sequence. The reference sequence may be a subset of a larger sequence, such 10 as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome. However, the reference sequence is at least 18 nucleotides long, typically at least 30 nucleotides long, and preferably at least 50 to 100 nucleotides long. "Substantially complementary" as used 15 herein refers to a sequence that is complementary to a sequence that substantially corresponds to a reference sequence.
Specific hybridization is defined herein as the formation of hybrids between a targeting transgene sequence 20 (e.g., a polynucleotide of the invention which may include substitutions, deletion, and/or additions) and a specific target DNA sequence (e.g., a human APP gene sequence or human immunoglobulin gene sequence), wherein a labeled targeting transgene sequence preferentially hybridizes to the target 25 such that, for example, a single band corresponding to a restriction fragment of a gene can be identified on a Southern blot of DNA prepared from cells using said labeled targeting transgene sequence as a probe. It is evident that optimal hybridization conditions will vary depending upon the sequence 30 composition and length(s) of the targeting transgene(s) and endogenous target(s), and the experimental method selected by the practitioner. Various guidelines may be used to select appropriate hybridization conditions (see. Maniatis et al., Molecular Cloning: A Laboratory Manual (1989), 2nd Ed., Cold 35 Spring Harbor, N.Y. and Berger and Kimmel, Methods in Enzvmologv. Volume 152. Guide to Molecular Cloning Technioues (1987), Academic Press, Inc., San Ditgo, CA., which are incorporated herein by reference.
WO 94/00569 PCT/US93/05873 13 The term "naturally-occurring" as used herein as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) 5 that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring. As used herein, laboratory strains of rodents which may have been selectively bred according to classical genetics are considered naturally-occurring animals. 10 The term "cognate" as used herein refers to a gene sequence that is evolutionarily and functionally related between species. For example but not limitation, in the human genome, the human immunoglobulin heavy chain gene locus is the cognate gene to the mouse immunoglobulin heavy chain gene 15 locus, since the sequences and structures of these two genes indicate that they are highly homologous and both genes encode a protein which functions to bind antigens specifically.
As used herein, the term "xenogenic" is defined in relation to a recipient mammalian host cell or nonhuman animal 20 and means that an amino acid sequence or polynucleotide sequence is not encoded by or present in, respectively, the naturally-occurring genome of the recipient mammalian host cell or nonhuman animal. Xenogenic DNA sequences are foreign DNA sequences; for example, human APP genes or immunoglobulin 25 genes are xenogenic with respect to murine ES cells; also, for illustration, a human cystic fibrosis-associated CFTR allele is xenogenic with respect to a human cell line that is homozygous for wild-type (normal) CFTR alleles. Thus, a cloned murine nucleic acid sequence that has been mutated 30 (e.g., by site directed mutagenesis) is xenogenic with respect to the murine genome from which the sequence was originally derived, if the mutated sequence does not naturally occur in the murine genome.
As used herein, a "heterologous gene" or 35 "heterologous polynucleotide sequence" is defined in relation to the transgenic nonhuman organism producing such a gene product. A heterologous polypeptide also referred to as a xenogeneic polypeptide, is defined as a polypeptide having an 14 amino acid sequence or an encoding DNA sequence corresponding to that of a cognate gene found in an organism not consisting of the transgenic nonhuman animal. Thus, a transgenic mouse harboring a human APP gene can be described as harboring a 5 heterologous APP gene. A transgenic mouse harboring a human immunoglobulin gene can be described as harboring a heterologous immunoglobulin gene. A transgene containing various gene segments encoding a heterologous protein sequence may be readily identified, e.g. by hybridization or DNA 10 sequencing, as being from a species of organism other than the transgenic animal. For example, expression of human APP amino acid sequences may be detected in the transgenic nonhuman animals of the invention with antibodies specific for human APP epitopes,encoded by human AP gene segments. A cognate 15 heterologous gene refers to a corresponding gene from another species; thus, if murine APP is the reference, human APF- is a cognate heterologous gene (as is porcine, ovine, or rat. APP, along with AP genes from other species).
As used herein, the term "targeting construct" 20 refers to a polynucleotide which comprises: (1) at least one homology region having a sequence that is substantially identical to or substantially complementary to a sequence present in a host cell endogenous gene locus, and (2) a targeting region which becomes integrated into a host cell 25 endogenous gene locus by homologous recombination between a targeting construct homology region and said endogenous gene locus sequence. If the targeting construct is a "hit-and-run" or Hin-and-out,, type construct (Valancius and Smithies (1991) Mol. Cell. Biol. 11: 1402; Donehower et al. (1992) Nature 356: 30 215; (1991) J. NIH Res. 3.: 59; Hasty et al. (1991) Nature 350: 243, which are incorporated herein by reference), the targeting region is only transiently incorporated into the endogenous gene locus and is eliminated from the host genome by selection. A targeting region may comprise a sequence that 35 . is substantially homologous to an endogenous gene sequence and/or may comprise a nonhomologous sequence, such as a selectable marker (e.g., neo, tk, gpt). The term "targeting construct" does not necessarily indicate that the polynucleotide comprises a gene which becomes integrated into the host genome, nor does it necessarily indicate that the polynucleotide comprises a complete structural gene sequence. As used in the art, the term "targeting construct" is 5 synonymous with the term "targeting transgene" as used herein.
The terms "homology region" and "homology clamp" as used herein refer to a segment (i.e., a portion) of a targeting construct having a sequence that substantially corresponds to, or is substantially complementary to, a 10 predetermined endogenous gene sequence, which can include sequences flanking said gene. A homology region is generally at least about 100 nucleotides long, preferably at least about 250 to 500 nucleotides long, typically at least about 1000 nucleotides long or longer. Although there is no demonstrated 15 theoretical minimum length for a homology clamp to mediate homologous recombination, it is believed that homologous recombination efficiency generally increases with the length of the homology clamp. Similarly, the recombination efficiency increases with the degree of sequence homology 20 between a targeting construct homology region and the endogenous target sequence, with optimal recombination efficiency occurring when a homology clamp is isogenic with the endogenous target sequence. The terms "homology clamp" and "homology region" are interchangeable as used herein, and 25 the alternative terminology is offered for clarity, in view of the inconsistent usage of similar terms in the art. A homology clamp does not necessarily connote formation of a base-paired hybrid structure with an endogenous sequence. Endogenous gene sequences that substantially correspond to, or 30 are substantially complementary to, a transgene homology region are referred to herein as "crossover target sequences" or "endogenous target sequences." As used herein, the term "minigene" or "minilocus" refers to a heterologous gene construct wherein one or more 35 nonessential segments of a gene are deleted with respect to the naturally-occurring gene. Typically, deleted segments are intronic sequences of at least about 100 basepairs to several kilobases, and may span up to several tens of kilobases or 16 more. Isolation and manipulation of large (i.e., greater than about 50 kilobases) targeting constructs is frequently difficult and may reduce the efficiency of transferring the targeting construct into a host cell. Thus, it is frequently 5 desirable to reduce the size of a targeting construct by deleting one or more nonessential portions of the gene. Typically, intronic sequences that do not encompass essential regulatory elements may be deleted. For example, a human immunoglobulin heavy chain minigene may comprise a deletion of 10 an intronic segment between the J gene segments and the /i constant region exons of the human heavy chain immunoglobulin gene locus. Frequently, if convenient restriction sites bound a nonessential intronic sequence of a cloned gene sequence, a deletion of the intronic sequence may be produced by: (1) 15 digesting the cloned DNA with the appropriate restriction enzymes, (2) separating the restriction fragments (e.g., by electrophoresis), (3) isolating the restriction fragments encompassing the essential exons and regulatory elements, and (4) ligating the isolated restriction fragments to form a 2 0 minigene wherein the exons are in the same linear order as is present in the germline copy of the naturally-occurring gene. Alternate methods for producing a minigene will be apparent to those of skill in the art (e.g., ligation of partial genomic clones which encompass essential exons but which lack portions 25 of intronic sequence). Most typically, the gene segments comprising a minigene will be arranged in the same linear order as is present in the germline gene, however, this will not always be the case. Some desired regulatory elements (e.g., enhancers, silencers) may be relatively position- 3 0 insensitive, so that the regulatory element will function correctly even if positioned differently in a minigene than in the corresponding germline gene. For example, an enhancer may be located at a different distance from a promoter, in a different orientation, and/or in a different linear order. For 35 example, an enhancer that is located 3' to a promoter in germline configuration might be located 5• to the promoter in a minigene. Similarly, some genes may have exons which are alternatively spliced at the RNA level, and thus a minigene WO 94/00569 PCT/US93/05873 17 may have fewer exons and/or exons in a different linear order than the corresponding germline gene and still encode a functional gene product. A cDNA encoding a gene product may also be used to construct a minigene. However, since it is 5 generally desirable that the heterologous minigene be expressed similarly to the cognate naturally-occurring nonhuman gene, transcription of a cDNA minigene typically is driven by a linked gene promoter and enhancer from the naturally-occurring gene.
As used herein, the term "large transgene" or "large homologous targeting construct" generally refers to polynucleotides that are larger than 50 kb, usually larger than 100 kb, frequently larger than 260 kb, occasionally as large as 500 kb, and sometimes as large as 1000 kb or larger. 15 As used herein, the term "transcriptional unit" or "transcriptional complex" refers to a polynucleotide sequence that comprises a structural gene (exons), a cis-acting linked promoter and other cis-acting sequences necessary for efficient transcription of the structural sequences, distal 20 regulatory elements necessary for appropriate tissue-specific and developmental transcription of the structural sequences, and additional cis sequences important for efficient transcription and translation (e.g., polyadenylation site, mRNA stability controlling sequences).
As used herein, "linked" means in polynucleor.de linkage (i.e., phosphodiester linkage). "Unlinked" means not linked to another polynucleotide sequence; hence, two sequences are unlinked if each sequence has a free 5' terminus and a free 3' terminus.
DETAILED DESCRIPTION OF THE INVENTION Generally, the nomenclature used hereafter and the laboratory procedures in cell culture, molecular genetics, and nucleic acid chemistry and hybridization described below are 35 those well known and commonly employed in the art. Standard techniques are used for recombinant nucleic acid methods, polynucleotide synthesis, cell culture, and transgene incorporation (e.g., lipofection protocols). Generally 18 enzymatic reactions, oligonucleotide synthesis, and purification steps are performed according to the manufacturer's specifications. The techniques and procedures are generally performed according to conventional methods in 5 the art and various general references which are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference. 10 Chimeric targeted mice are derived according to Hogan, et al., Manipulating the Mouse Embryo: A Laboratory Manual. Cold Spring Harbor Laboratory (1988) and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. E.J. Robertson, ed., IRL Press, Washington, D.C., 15 (1987) which are incorporated herein by reference.
Embryonic stem cells are manipulated according to published procedures (Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. E.J. Robr tson, ed., IRL Press, Washington, D.C. (1987); Zjilstra et al., Nature 342:435-438 20 (1989); and Schwartzberg et al., Science 246:799-803 (1989), each of which is incorporated herein by reference).
Oligonucleotides can be synthesized on an Applied Bio Systems oligonucleotide synthesizer according to specifications provided by the manufacturer. 25 It has often been observed that cDNA-based transgenes are poorly expressed or inappropriately regulated. Genomic DNA-based transgenes (i.e., constructed from cloned genomic DNA sequences) which substantially retain the content and organization of the naturally-occurring gene locus are 3 0 more likely to be correctly expressed, but are limited in size by the cloning capacity of bacteriophage and plasmid/cosmid vectors. The yeast artificial chromosome (YAC) is a recently developed cloning vehicle with a capacity of approximately 2 megabases (Mb) (Burke et al. (1987) Science 236: 806). The 35 ability to reproducibly and efficiently introduce YACs into transgenic mice can significantly surpass current transgene size limits.
WO 94/00569 PCT/US93/05873 19 In general, the invention is based on the unexpected finding that large (i.e., greater than about 50 kb) cloned polynucleotides can be efficiently transferred into mammalian cells, such as ES cells, and are incorporated into at least 5 one chromosomal location and stably replicated as a segment of a chromosome. Further, it was found that large cloned polynucleotides comprising a complete transcriptional unit can be transferred into mammalian cells (e.g., ES cells), incorporated into a chromosomal location, and transcribed to 10 produce a detectable concentration of RNA transcripts of the structural gene sequences. It was also found that unrearranged immunoglobulin genes cloned in YACs can be introduced into ES cells and developed to form a transgenic animal in which productive VDJ rearrangement occurs, and 15 expression of immunoglobulin chains also occurs. It has also been found that large transgenes can be cloned in YACs and, after isolation from the host yeast cells, efficiently transferred into mammalian cells (e.g., ES cells) without prior separation of the desired transgene sequences from 20 yeast-derived YAC sequences, and that the presence of such yeast-derived YAC sequences can be non-interfering (i.e., compatible with efficient transgene integration and transcription of a transgene transcriptional unit). Unexpectedly, it also has been found that large transgenes, 25 with or without linked yeast-derived YAC sequences, can be efficiently co-transfected into mammalian cells (e.g., ES cells) with unlinked polynucleotides containing a selectable marker, such as, for example, a JieoR expression cassette; and that selection for cells harboring the selectable marker gene 30 and expressing the selectable marker are are highly likely to also harbor the large transgene species which has been co-lipofected, thus allowing efficient selection for large transgene DNA sequences without requiring prior ligation (and cloning) of a selectable marker gene. The finding that large 35 DNA segments, such as YAC clones, can be efficiently co-lipofected with a selectable marker gene permits, for the first time, the construction of transgenic mammalian cells and transgenic nonhuman animals harboring large xenogenic DNA segments that are typically difficult to manipulate. Thus, large polynucleotides, typically 50 to 100 kb in size, frequently more than 250 kb in size, occasionally more than about 500 kb, and sometimes 1000 kb or larger, may be 5 efficiently introduced into mammalian cells. The mammalian cells may be ES cells, such as murine ES cells (e.g., the AB-l line), so that the resultant transgenic cells can be injected into blastocysts to generate transgenic nonhuman animals, such as transgenic mice or transgenic rats, harboring large DNA 10 transgenes, which are preferably expressed in cthe nonhuman transgenic animals. The present methods may also-be carried out with somatic cells, such as epithelial cells _(e...g..-, keratinocytes), endothelial cells, hematopoietic cells., and myocytes, for example.
Embryonic Stem Cells If embryonic stem (ES) cells are used as the transgene recipients, it is possible to develop a transgenic animal harboring the targeted gene(s) which comprise the integrated targeting transgene(s). Briefly, this technology 20 involves the introduction of a gene, by nonhomologous integration or homologous recombination, in a pluripotent cell line (e.g., a murine ES cell line) that is capable of differentiating into germ cell tissue.
A large transgene can be nonhomologously integrated 25 into a chromosomal location of the host genome. Alternatively, a homologous targeting construct (which may comprise a transgene) that contains at least one altered copy of a portion of a germline gene or a xenogenic cognate gene (including heterologous genes) can be introduced into the 30 genome of embryonic stem cells. In a portion of the cells, the introduced DNA is either nonhomologously integrated into a chromosomal location or homologously recombines with the endogenous (i.e., naturally occurring) copy of the mouse gene, replacing it with the altered construct. Cells containing the 35 newly engineered genetic sequence(s) are injected into a host mouse blastocyst, which is reimplanted into a recipient female. Some of these embryos develop into chimeric mice that possess a population of germ cells partially derived from the 21 mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (reviewed by Capecchi et al. (1989) Science 244; 1288, incorporated herein by reference). 5 For homologous targeting constructs, targeting efficiency generally increases with the length of the targeting transgene portion (i.e., homology region) that is . substantially complementary to a reference sequence present in the target DNA (i.e., crossover target sequence). In general, 10 targeting efficiency is optimized with the use of isogenic DNA homology regions, although it is recognized that the presence of recombinases in certain ES cell clones may reduce the degree of sequence identity required for efficient recombination.
The invention also provides transgenes which encode a gene product that is xenogenic (e.g., heterologous) to a nonhuman host species. Such transgenes typically comprise a structural gene sequence expression cassette, wherein a linked promoter and, preferably, an enhancer drive expression of 2 0 structural sequences encoding a xenogenic (e.g., heterologous protein). For example, the invention provides transgenes which comprise a mammalian enhancer and at least one human APP promoter linked to structural sequences that encode a human APP protein. Transgenic mice harboring such transgenes 25 express human APP mRNA(s). Preferably, the polynucleotide sequence encoding the xenogenic (e.g., heterologous) protein is operably linked to cis-acting transcriptional regulatory regions (e.g., promoter, enhancer) so that a heterologous protein is expressed in a manner similar to the expression of 30 the cognate endogenous gene in the naturally-occurring nonhuman animal. Thus, it is generally preferable to operably link a transgene structural encoding sequence to transcriptional regulatory elements which naturally occur in or near the cognate endogenous gene. However, transgenes 35 encoding heterologous proteins may be targeted by employing a homologous gene targeting construct targeted adjacent to the endogenous transcriptional regulatory sequences, so that the operable linkage of a regulatory sequence occurs upon WO 94/00569 PCT/US93/05873 22 integration of the transgene into a targeted endogenous chromosomal location of the ES cell.
Selectable Marker Genes A selectable marker gene expression cassette 5 typically comprises a promoter which is operational in the targeted host cell (e.g., ES cell) linked to a structural sequence that encodes a protein or polypeptide that confers a selectable phenotype on the targeted host cell, and a polyadenylation signal. A promoter included in an expression 10 cassette may be constitutive, cell type-specific, stage-specific, and/or modulatable (e.g., by hormones such as glucocorticoids; MMTV promoter), but is expressed prior to and/or during selection. An expression cassette can optionally include one or more enhancers, typically linked 15 upstream of the promoter and within about 3-10 kilobases. However, when the selectable marker is contained in a homologous targeting construct, homologous recombination at the targeted endogenous site(s) can be chosen to place the selectable marker structural sequence downstream of a 2 0 functional endogenous promoter, and it may be possible for the targeting construct replacement region to comprise only a structural sequence encoding the selectable marker, and rely upon an endogenous promoter to drive transcription (Doetschman et al. (1988) Proc. Natl. Acad. Sci. (U.S.A.) 85: 8583, 25 incorporated herein by reference). Similarly, an endogenous enhancer located near a targeted endogenous site may be relied on to enhance transcription of selectable marker gene sequences in enhancerless constructs. Preferred expression cassettes of the invention encode and express a selectable 3 0 drug resistance marker and/or a HSV thymidine kinase enzyme.
Suitable drug resistance genes include, for example: gpt (xanthine-guanine phosphoribosyltransferase), which can be selected for with mycophenolic acid; neo (neomycin phosphotransferase), which can be selected for with G418, 35 hygromycin, or puromycin; and DFHR (dihydrofolate reductase), which can be selected for with methotrexate (Mulligan and Berg (1981) Proc. Natl. Acad. Sci. (U.S.A.1 78: 2072; Southern and Berg (1982) J. Mol. AppI. Genet. 3.: 327; which are WO 94/00569 PCT/US93/05873 23 incorporated herein by reference). Other suitable selectable markers will be apparent to those in the art.
Selection for correctly co-lipofected recombinants will generally employ at least positive selection, wherein a 5 selectable marker gene expression cassette encodes and expresses a functional protein (e.g., neo or gpt) that confers a selectable phenotype to targeted cells harboring the endogenously integrated expression cassette, so that, by addition of a selection agent (e.g., G418, puromycin, or 10 mycophenolic acid) such targeted cells have a growth or survival advantage over cells which do not have an integrated expression cassette. Further guidance regarding selectable marker genes is available in several publications, including Smith and Berg (1984) Cold Spring Harbor Svmp. Quant. Biol. 15 49.: 171; Sedivy and Sharp (1989) Proc. Natl. Acad. Sci.
(U.S.A.) 86: 227; Thomas and Capecchi (1987) op.cit.. which are incorporated herein by reference.
Large Xenogenic Polynucleotides Large polynucleotides are usually cloned in YAC 2 0 vectors. For example, human genomic DNA libraries in YAC cloning vectors can be screened (e.g., by PCR or labeled polynucleotide probe hybridization) to isolate YAC clones spanning complete genes of interest (e.g., a human APP gene, a human immunoglobulin heavy chain locus or light chain locus), 25 or significant portions of such genes which comprise a complete transcriptional unit. Methods for making YAC libraries, isolating desired YAC clones, and purifying YAC DNA are described in the art (U.S. Patent 4,889,806; Burke et al. (1987) Science 236: 806; Murry et al. (1986) Cell 45: 529, 30 incorporated herein by reference).
Once a desired YAC clone is isolated, and preferably deproteinized, yeast-derived YAC sequences may optionally be completely or partially removed by digestion with one or more restriction enzymes which cut outside the desired cloned large 35 transgene sequence; yeast-derived sequences are separated from the cloned insert sequences by, for example, pulsed gel electrophoresis. Preferably, a complete unrearranged YAC WO 94/00569 PCT/US93/05873 24 clone is used as a large transgene or large homologous targeting construct in the methods of the invention.
In one aspect, preferred YAC clones are those which completely or partially span structural gene sequences 5 selected from the group consisting of: human APP gene, human immunoglobulin heavy chain locus, human immunoglobulin light chain locus, human al-antitrypsin gene, human Duchenne muscular dystrophy gene, human Huntington's chorea-associated loci, and other large structural genes, preferably human 10 genes.
Preferred YAC cloning vectors are: a modified pYAC3 vector (Burke et al. (1987) op.cit.. incorporated herein by reference), pYACneo (Traver et al. (1989) Proc. Natl. Acad. Sci. (U.S.A.) 86: 5898, incorporated herein by reference), and 15 pCGS966 (Smith et al. (1990) Proc. Natl. Acad. Sci. (U.S.A.) 87: 8242, incorporated herein by reference).
Cationic Lipids Lipofection, and various variations of its basic methodology, have been described previously in the art (U.S. 20 Patents 5,049,386; 4,946,787; and 4,897,355) and lipofection reagents are now sold commercially (e.g., "Transfectam" and "Lipofectin").
Cationic and neutral lipids that are suitable for efficient lipofection of DNA have been described in the art. 25 Lipofection may be accomplished by forming lipid complexes with DNA made according to Feigner (W091/17424, incorporated herein by reference) and/or cationic lipidization (W091/16024; incorporated herein by reference). Various lipofection protocols described in the art may be adapted for co-30 lipofection according to the invention; for example but not limitation, general lipofection protocols are described in the following references which are incorporated herein: Behr et al. (1989) Proc. Natl. Acad. Sci. (U.S.A.) 86: 6982; Demeneix et al. (1991) Int. J. Dev. Biol. 35: 481; Loeffler et al. 35 (1990) J. Neurochem. 54; 1812; Bennett et al. (1992) Mol.
Pharmacol. 41: 1023; Bertling et al. (1991) Biotechnol. AppI. Biochem. 13.: 390; Feigner et al. (1987) Proc. Natl. Acad. Sci. (U.S.A.) 84.: 7413; Feigner and Ringold (1989) Nature 337: 3G7; Gareis et al. (1991) Cell. Mol. Biol. 37: 191; Jarnagin et al. (1992) Nucleic Acids Res. 20: 4205; Jiao et al. (1992) Exp. Neurol. 115: 400; Lim et al. (1991) Circulation 83.: 2007; Malone et al. (1989) Proc. Natl. Acad. Sci. (U.S.A.) 86: 6077; Powell et al. (1992) Eur. J. Vase. Surg. 6: 130; Strauss and Jaenisch (1992) EMBO J. 11: 417; and Leventis and Silvius (1990) Biochim. Biophvs. Acta 1023: 124.
Newer polycationic lipospermines compounds exhibit broad cell ranges (Behr et al., (1989) op.cit.) and DNA is 10 coated by these compounds. In addition, a combination of neutral and cationic lipid has been shown to be highly efficient at transfection of animal cells and showed a broad spectrum of effectiveness in a variety of cell lines (Rose et al., (1991) BioTechniaues 10:520) A lipofection complex (or a cationic lipidized DNA complex) is defined as the product made by mixing a suitable cationic lipid composition with one or more polynucleotide species, such as a large transgene and a selectable marker gene expression cassette. Such a co-lipofection complex is 20 characterized by an interaction between the polynucleotides and lipid components that results in the formation of a co-lipofection complex that, when contacted with mammalian cells under suitable conditions (e.g., buffered saline or ES cell medium with or without serum, 20-45°C), results in 25 incorporation of t-:ie polynucleotides into the mammalian cells; preferably the mammalian cells are ES cells, such as murine ES cells.
Various suitable cationic lipids may be used, either alone or in combination with one or more other cationic lipid 30 species or neutral lipid species. Generally, suitable cationic lipids comprise a positively charged head group (one or more charges) and a covalently linked fatty acid tail. A suitable cationic lipid composition is "Transfectam" (ProMega, Madison, WI) comprising the cationic lipid-polyamine 35 dioctadecylamidoglycyl spermidine (DOGS). DOTMA is a preferred lipid known as N-(2,3-di(9-(Z)—octadecenyloxy))-prop—1—N,N,N- trimethylammonium chloride. DNA-DOTMA complexes made essentially from DOTMA and DNA. Other examples al WO 94/00569 PCT/US93/05873 26 suitable cationic lipids are: dioleoylphosphatidylethanolamine (PtdEtn, DOPE) , dioctadecylamidoglycyl, N-trimethylammonium chloride, N-trimethylammonium methylsulfate, DORI and DORI-ether (DORIE). DORI is N-[1-(2,3-dioleoyl)propyl]-N,N-5 dimethyl-N-hydroxyethylammonium acetate and DORIE is N-[l-(2, 3-dioleyloxy) propyl ] -N, N-dimethy 1-N-hydroxyethylammonium acetate. DOTAP is N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl sulfate; this lipid has ester rather than ether linkages and can be metabolized by cells. 10 Optionally, one or more co-lipids may be combined with a suitable cationic lipid. An optional co-lipid is to be understood as a structure capable of producing a stable DNA-lipid complex, alone with DNA, or in combination with other lipid components and DNA, and is preferably neutral, although 15 it can alternatively be positively or negatively charged. Examples of optional co-lipids are phospholipid-related materials, such as lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin, 20 cephalin, cardiolipin, phosphatidic acid, cerebrosides, dicetylphosphate, dioleoylphosphatidylcholine (DOPC), dipalmitoyl-phosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-25 phosphatidylethanolamine (DOPE), palmitoyloleoy-lphosphatidylcholine (POPC),palmitoyloleoyl-phosphatidylethanolamine (POPE) and dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethy1)-cyclohexane-l-carboxylate (DOPE-mal). Additional 30 non-phosphorous containing lipids are, e.g.,stearylamine, dodecylamine, hexadecylamine, acetyl palmitate, glycerolricinoleate, hexadecyl stereate, isopropyl myristate, amphoteric acrylic polymers, triethanolamine-lauryl sulfate, alkyl-aryl sulfate polyethyloxylated fatty acid amides, 35 dioctadecyldimethyl ammonium bromide and the like.
Generally, to form a lipofection complex, the polynucleotide(s) is/are combined according to the teachings in the art and herein with a suitable cationic lipid, in the WO 94/00569 PCT/US93/05873 27 presence or absence of one or more co-lipids, at about pH 7.4-7.8 and 20-30°C. When more than one species of polynucleotide are combined in a lipofection complex it may be referred to herein as a co-lipofection complex. A co-lipofection complex 5 generally comprises a large polynucleotide (a transgene or homologously targeting construct) and a selectable marker gene expression cassette. The co-lipofection complex is administered to a cell culture, preferably murine ES cells, under lipofection conditions as described in the art and 10 herein.
General Methods A preferred method of the invention is to transfer a substantially intact YAC clone comprising a large heterologous transgene into a pluripotent stem cell line which can be used 15 to generate transgenic nonhuman animals following injection into a host blastocyst. A particularly preferred embodiment of the invention is a human APP gene targeting construct co-lipofected with an unlinked positive (e.g., neo) selection expression cassette. The human APP transgene is transferred 20 into mouse ES cells (e.g., by co-lipofection with neo) under conditions suitable for the continued viability of the co-lipofected ES cells. The lipofected ES cells are cultured under selective conditions for positive selection (e.g., a. selective concentration of G418). Selected cells are then 25 verified as having the correctly targeted transgene recombination by PCR analysis according to standard PCR or Southern blotting methods known in the art (U.S. Patent 4,683,202; Erlich et al., (1991) Science 252: 1643, which are incorporated herein by reference).
Correctly targeted ES cells are then transferred into suitable blastocyst hosts for generation of chimeric transgenic animals according to methods known in the art (Capecchi, M. (1989) TIG 5:70; Capecchi, M. (1989) Science 244:1288. incorporated herein by reference). Several studies 35 have already used PCR to successfully identify the desired transfected cell lines (Zimmer and Gruss (1989) Nature 338; 150; Mouellic et al. (1990) Proc. Natl. Acad. Sci. (U.S.A.) 87: 4712; Shesely et al. (1991) Proc. Natl. Acad. Sci. USA 88 WO 94/00569 PCT/US93/05873 28 4294, which are incorporated he&e&n by reference). This approach is very effective when number of cells receiving exogenous targeting transgene(s) is high (i.e., with electroporation or lipofection) and the treated cell 5 populations are allowed to expand (Capecchi, M. (1989) op.cit., incorporated herein by reference).
For making transgenic non-human animals (which include homologously targeted non-human animals), embryonal stem cells (ES cells) are preferred. Murine ES cells, such 10 as AB-1 line grown on mitotically inactive SNL76/7 cell feeder layers (McMahon and Bradley, Cell 62:1073-1085 (1990)) essentially as described (Robertson, E.J. (1987) in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. E.J. Robertson, ed. (Oxford: IRL Press), p. 71-112) 15 may be used for homologous gene targeting. Other suitable ES lines include, but are not limited to, the E14 line (Hooper et al. (1987) Nature 326: 292-295), the D3 line (Doetschman et al. (1985) J. Embrvol. Exp. Morph. 87: 27-45), and the CCE line (Robertson et al. (1986) Nature 323: 445-448). Rat, 20 hamster, bovine, and porcine ES cell lines are also available in the art for producing non-murine transgenic non-human animals bearing a human APP gene sequence. The success of generating a mouse line from ES cells bearing a large transgene or specifically targeted genetic alteration depends 25 on the pluripotence of the ES cells (i.e., their ability, once injected into a host blastocyst, to participate in embryogenesis and contribute to the germ cells of the resulting animal). The blastocysts containing the injected ES cells are allowed to develop in the uteri of pseudopregnant 3 0 nonhuman females and are born as chimeric mice. The resultant transgenic mice are chimeric for cells having the large transgene(s)/homologous targeting constructs and are backcrossed and screened for the presence of the transgene(s) and/or YAC sequences by PCR or Southern blot analysis on tail 35 biopsy DNA of offspring so as to identify transgenic mice heterozygous for the transgene(s)/homologous targeting constructs. By performing the appropriate crosses, it is possible to produce a transgenic nonhuman animal homozygous 29 for multiple large transgenes/homologous recombination constructs, and optionally also for a transgene encoding a different heterologous protein. Such transgenic animals are satisfactory experimental models for various diseases linked 5 to the transferred transgene(s) .
For performing certain types of studies, transgenic rats harboring and expressing a human APP sequence may be preferred.
EXPERIMENTAL EXAMPLES 10 EXAMPLE 1 Materials and Transfection Calibration Pilot experiments to determine toxicity levels, optimum DNA:lipid ratios, etc. were performed with ] (2kb PGKneo cassette in pUC) and with PYPNN (a modified pYACneo 15 vector containing a PGKneo cassette in place of the SV40-neo cassette in the acentric arm.
The YAC used in these calibration experiments was an 85kb human IgH gene fragment cloned into a modified pYACneo vector (EcoRI->NotI cloning site alteration). The YAC was 20 thus lOOkb in length including the vector arms.
DOTMA (Lipofectin, BRL, Bethesda, MD) and DOGS (Transfectam, ProMega, Madison, WI) were tested as cationic lipids. Fig. 1 shows chemical structures of representative cationic lipids which can be used to form co-lipofection 25 complexes. ES cell toxicity curves were performed for each. Toxic effects could be seen with DOTMA at the 3 0/ig/ml level. DOGS showed no toxic effects at the 60/zg/ml level.
Optimal DNA:lipid ratios were determined for both lipids, using pGKneo as reporter. Optima for DOTMA and DOGS 30 were at 1:10 and 1:50 (DNA:lipid, wt:wt), respectively.
Optimal ES cell number for each lipid was determined. Optimal incubation times and conditions were determined (how long and in what buffer gave maximal transfection with both DNA:lipid complexes). Pilot studies 35 indicated that 3-5 x 106 ES cells incubated with a 1:50 mixture of DNA:DOGS in serum free DMEM for 3-4 hours at 37°C yielded the maximal number of transfectants. These conditions were routinely used for the YAC lipofection experiments.
WO 94/00569 PCT/US93/05873 Neor was provided by an unlinked plasmid carrying a PGKneo cassette, either eem or pYPNN, in a co-lipofection procedure. DNA:plasmid molar ratios varied from 1:4 to 1:20. An equal weight of carrier DNA (sheared herring sperm) was 5 also added.
Yeast blocks were prepared at 3.5 x 109 cells/ml in 0.67% low gel temp agarose. The YAC was isolated by PFGE. Outer lanes were stained with EtBr and aligned with the unstained portion. A thin slice containing the YAC was 10 isolated using a brain knife. Approximately ljxg of YAC was recovered in approximately lOmls of gel. The gel was washed extensively in gelase buffer (40mM bis-Tris pH 6.0, IjiM EDTA, 40mM NaCl), melted at 70°C, cooled to 40°C, and incubated with 10U gelase (Epicentre Technologies, Madison, WI) overnight. 15 After digestion, pYPNN or picenter were added at a typical molar ratio of 1:4 (YAC:plasmid). An equal weight of sheared herring sperm DNA was added as carrier. The agarase digestion mix containing approximately 100 mg of the YAC was directly incubated with Transfectam at optimal DNA:lipid 20 ratios for 30 minutes at room temperatrres. No polyamines were added.
ES cells were washed, trypsinized, and resuspended in serum free DMEM. Nine mis of cell suspension containing 3 x 106 ES cells (and about 105 feeder cells) were plated onto a 25 60mm petri plate (not tissue culture plastic). About 1 ml of the DNA-lipid mix was added to the cells in DMEM and incubated at 37°C for 3-4 hours. The cells were then collected in DMEM + FBS, and plated at 106 per 100mm tissue culture dish. G418 selection was applied 24 hours later, and colonies picked 30 after about 10 days.
Typically, l-2|ig of YAC was used per experiment. Thus, about 10-20 separate lipofections were performed on a given day. Generally, several hundred G418 resistant clones were picked, of which at least 1-2% contained specific YAC 35 derived sequence. Of these, approximately 10% carry the intact YAC, as determined by fine structure southern blotting using probes covering the entire YAC insert, PFGE southern analysis, and PCR analysis.
WO 94/00569 PCT/US93/05873 31 EXAMPLE 2 Production of mice carrying a YAC encoding human Amvloid Precursor Protein Preparation of the APP YAC DNA: A 650kb human 5 genomic fragment containing the full length APP gene was isolated as a yeast artificial chromosome (YAC) in a yeast host strain (clone #B142F9) from the Washington University YAC library (available from Center for Genetics in Medicine Librarian, Washington University School of Medicine, St. 10 Louis, Missouri). The yeast strain was grown to late log phase in AHC medium, resuspended in 0.67% low gelling temperature agarose (SeaPlaque, FMC Corp.) at 3.5 x 109 cells/ml, and cooled in block formers (Bio-Rad). Intact yeast chromosomal DNA was prepared as follows. Thirty 250/xl blocks 15 of B142F9 cells were swirled in a 150 mm petri dish containing 50mls of YSS (YSS: 4mg/ml Novozyme 234 (Novo Nordisk), 1M sorbitol, lOOmM EDTA, 50mM Potassium Phosphate, pH 5.5) at 37°C for 30 minutes. The blocks were washed once in TE (10 mM Tris pH 7.5, ImM EDTA) and swirled in 50 mis of LDS (LDS: 1% 20 lithium dodecyl sulfate, 1% sarcosyl, 100 mM EDTA) for 3 0 to 60 minutes at 37°C. The LDS was removed with a sterile 50ml pipette, and the blocks swirled in 50mls of fresh LDS overnight. The blocks were rinsed several times in 50mM EDTA, and stored at 4°C in 50mM EDTA. lOOjil segments of the 25 prepared blocks were loaded into each well of a 1% low gelling temp agarose gel in 0.25X TBE (14 x 25cm CHEF gel, 10 well gel comb, Bio-Rad). The yeast chromosomes were separated by pulsed field gel electrophoresis (CHEF-DRIL, Bio-Rad) using a 60 second switch time at 200V and 14°C for 48 hours. The end 30 lanes of the gel were removed, stained for 2 hours in 0.5/xg/ml ethidium bromide, and the separated chromosomes visualized on a UV transluminator. Under these conditio .-s, the 650kb YAC was separated from the nearest endogenous yeast chromosome by 3-5mm. The gel segments were notched to indicate the location 35 of 650kb YAC, and the segments realigned with the remainder of the gel. A 2 mm wide slice of the gel containing the 650kb YAC was isolated using a brain knife (Roboz Surgical Instrument Co) and stored in 50mM EDTA at 4°C. Approximately WO 94/00569 PCT/US93/05873 32 nq of YAC DNA was isolated in approximately 10 mis of gel. The agarose slice containing the YAC DNA was equilibrated in gelase buffer (40mM bis-Tris pH 6.0, lmM EDTA, 40mM NaCl), melted at 70°C for 20 minutes until completely liquid, and 5 cooled to 45°C. Gelase (25 U? Epicentre Technologies, Madison, WI) was added and the molten agarose mixture was incubated at 45°C for 90 minutes to liquify the DNA-agarose mixture.
Introduction of the APP YAC into ES cells 10 Embryonic stem cells (AB-1) were washed in PBS, trypsinized, and resuspended in serum free ES medium (DMEM, IX glutamine, pen/strep, 1 mM 2-mercaptoethanol, IX NEAA). Approximately 5 x 106 cells in 9 ml of serum-free ES cell suspension were placed into each of ten 60mm petri (non-tissue 15 culture treated) dishes. A linearized plasmid containing a selectable marker (PGKneoA+R, containing the PGK promoter fused to the neomycin resistance gene; Rudnicki et al. (1988) Mol. Cell. Biol. 8.: 406.; Rudnicki et al. (1989) Biochem. Cell. Biol. 67: 590, incorporated herein by reference) was added to 20 l ml of gelase treated YAC DNA at a 2:1 (plasmid:YAC) molar ratio. A cationic lipid (Transfectam, ProMega, Madison, WI) was added at a 50:1 (Transfectam:DNA) weight:weight ratio, the mixture was gently inverted once to mix and incubated at room temperature for approximately 3 0 minutes. One ml of the 25 DNA:lipid mixture was then added to each 60mm dish of ES cells and incubated for 4 hours in a 37°C C02 incubator. The cells were then transferred to a sterile 250ml bottle, an equal volume of ES medium (as above, but including 15% fetal calf serum) was added. Cells were removed from the dishes with 3 0 gentle pipetting and combined with an equal volume of ES medium containing 15 percent fetal calf serum. This cell suspension was transferred 15 ml aliquots to 100mm tissue culture plates containing mix., tically inactivated SNL76/7 fibroblast feeder cells (McMahon and Bradley (1990) Cell 62.: 35 1073, incorporated herein by reference) and returned to the tissue culture incubator for 24 hours. After 24 hours, the medium was changed to ES medium containing 10 percent fetal "calf serum and 400/jg/ml G418, and refed every 48 hours. After 33 7 days, a total of 366 G418 resistant colonies were counted. Each of 24 0 colonies were individually transferred to a well of a 96-well microtitre dish containing 50 fil of 0.25 percent trypsin in calcium-free magnesium-free PBS. After 15 minutes, 5 50 Ml of serum-containing medium was added, the colony dissociated by trituration, and the cell suspension was transferred to duplicate 96-well plates containing culture medium and feeder layers (as supra). After 4-5 days, one set of dishes was frozen according to conventional methods 10 (Ramirez-Solis et al. Guide to Techniques in Mouse Development (1992) Methods in Enzvmoloav. incorporated herein by reference) . Cells were dissociated in 50 /il trypsin and mixed with 50 fil of Freezing Medium (20% DMSO, 20% fetal calf serum in DMEM) ; 100 nl of sterile silicon oil was layered on top of 15 the cell suspension in each well, and the plates were placed in Styrofoam containers and frozen at -80°C.
Identification of ES clones containing APP sequences The other set of microtitre dishes containing lipofectant ES clones was used to prepare DNA for PCR 20 analysis. 50^il of lysis buffer (50mM Tris pH 8.0, 200mM NaCl, 25mM EDTA, 0.2% SDS, lmg/ml Proteinase K) was added to each well (Ramirez-Solis et al. (1992) Anal. Biochem. 201: 331, incorporated herein by reference). After an overnight incubation at 55°C, 5/xl of 2.5M NaCl and 95 /xl of 100% EtOH 25 were added to each well. The dishes were gently swirled at room temperature for 60 minutes to precipitate the DNA. The wells were then rinsed 5 times with 70% EtOH, and dried at 37°C. The DNAs were resuspended overnight in 100/xl of H20 at 37°C in a humidified incubator.
The individual DNA samples were pooled in rows and columns for PCR (Fig. 2), and the pools analyzed for APP sequences by PCR, using the following primers (adapted from Fidani et al., Human Molecular Genetics 1, 165-168, 1992): APP-PA: 51-GCT TTT GAC GTT GGG GGT TA-3' APP-PB2: 5•-TTC GTG AAC AGT GGG AGG GA-3• APP-17A: 5'-ATA ACC TCA TCC AAA TGT CCC C-3' APP-17B: 5-GTA ACC CAA GCA TCA TGG AAG C-3' WO 94/00569 PCT/US93/05873 34 APP—PA/PB2 denote primers specific for the promoter region of the human APP gene, APP7A/7B are specific for exon 7, and APP17A/17B are specific for exon 17.
PCR analysis of the pools indicated 42 clones which 5 potentially carried both promoter and exon 17 sequences (Fig. 3). PCR analysis of the 42 clones individually indicated 6 clones (#s 23, 24, 176, 213, 219, 230) containing both promoter and exon 17 sequences (Fig. 4). These clones were expanded in culture, and frozen in vials in liquid 10 nitrogen. These cells were mounted in agarose blocks for PFGE analysis, and harvested for RNA isolation.
Structural Analysis of ES clones containing APP sequences The integrity of the APP YAC carried by ES clones was first estimated using a rare cutter fingerprint technique 15 as follows. Restriction enzymes which infrequently cut human DNA were used to define patterns of fragments which hybridized to a human alu fragment probe. Rare cutters often contain the dinucleotide CpG, and mammalian cells often methylate CpG dinucleotides rendering most restriction sites containing them 20 refractory to digestion. However, yeast cells do not methylate CpGs, and thus the pattern of CpG containing restriction sites in a given fragment will depend on whether the fragment is propagated as a YAC in yeast or within a mammalian cell line. Thus, only rare cutter enzymes without 25 CpG in their recognition sequence were used to generate a diagnostic pattern of alu-containing fragments from the YAC.
B142F9 agarose blocks were digested completely with the restriction enzymes Sfi I, Pac I, Swa I, Pme I, and Apa I, and analyzed by PFGE Southern blotting using total human DNA 30 as a probe for Alu fragments (Fig. 5). The pattern of bands generated by Sfi I digestion was used as a reference pattern, since there was an even distribution of bands from 30 kb to 220 kb. If a YAC were to integrate intact into ES cells, Sfi I digestion would be expected to generate a similar pattern of 35 bands, with the exception of the terminal fragments. The terminal fragments could be easily identified by reprobing the Sfi I digest with pBR322 sequences. In this case, the entire set of fragments were ordered by partial digest mapping.
Briefly, B142F9 blocks were digested with a range of Sfi I concentrations, separated by PFGE, and probed with either the 2.5 kb (trp arm specific) or the 1.6 kb (ura arm specific) Bam HI-Pvu II fragment of pBR322, such that at a particular level 5 of partial digestion, a ladder of bands were generated. Each band differed from its naarest neighbor by the distance to the neighboring Sfi I sites (Fig. 6).
The six ES lines were digested to completion with Sfi I and probed under high stringency conditions with total 10 human DNA. Of the six lines, only three (24, 176, 230) showed a pattern of bands consistent with the reference pattern from the APP YAC. The rare cutter fingerprinting approach does not require any knowledge of the sequence of the fragment cloned in the YAC, and is thus applicable to the analysis of any YAC 15 containing human DNA. Further, if probes for repetitive elements from other species which were not found in the target mammalian cell line were available, this approach could be used to analyze the structure of YACs containing other foreign DNAs into other mammalian cell lines. 2 0 Transcriptional analysis of APP YAC containing ES clones The six ES lines were analyzed for transcription by PCR. Total RNA was prepared from each cell line by standard guanidium isothiocyanate/lithium chloride procedures (Sambrook et al., Molecular Cloning). Complementary DNA was prepared 25 using an oligo-dT primer, and the cDNAs were analyzed by PCR for splice products of the human APP gene using the PCR primers derived from exons 6 and 9 as depicted below (adapted from Golde et al., Neuron 4 253-267, 1990). To exclude inappropriate amplification of mouse APP cDNAs, the 3' end 3 0 nucleotide of each oligo was chosen such that it was specific for the human cDNA sequence and not the corresponding mouse cDNA sequence. PCR oligos specific for mouse APP cDNA were also prepared. human APP specific oligos: APP—HAS1: 5'-CAG GAA TTC CAC CAC AGA GTC TGT GGA A-31 APP-HAS2: 5•-CAG GAT CCG TGT CTC GAG ATA CTT GTC A-3• 36 mouse APP specific oligos: APP-MAS1: 5'-CAG GAA TTC CAC CAC TGA GTC CGT GGA G-3• APP-MAS2: 5'-CAG GAT CCG TGT CTC CAG GTA CTT GTC G-3' Clones 24, 176, and 230 showed the expected PCR 5 bands indicative of alternatively spliced human APP transcripts encoding the 770, 751, and 695 amino acid forms of the protein (Fig. 7). Clones 23, 213, and 219 did not contain PCR detectable transcript, and also served as a negative control, indicating that the human APP specific oligos did not 10 amplify bands from mouse APP transcripts endogenous to the ES cell lines. Further, the RT-PCR analysis confirmed and validated the results of the rare cutter fingerprint analysis which predicted that clones 24, 176, and 230 contained the intact YAC whereas clones 23, 213, and 219 did not. 15 Quantitative analysis of APP transcript in ES cells Since RT-PCR analysis is qualitative, RNase protection assays are used to quantitate the alternatively spliced human APP transcripts in the ES lines. The RNase probe was generated by cloning the 310 bp RT-PCR product as an 20 Eco RI-Bam HI fragment into the vector pSP72 (Fig. 8). The resultant plasmid, pHAPP, is linearized at the Hpa I site, and antisense transcript is generated from the SP6 promoter.
RNase protection assays are performed according to standard protocols (Sambrook et al., Molecular Cloning). 25 Alternatively, SI nuclease protection analysis is used to quantitate the transcripts. pHAPP is digested with Xho I and Hpa I to release a 446 bp fragment. The double stranded fragment is end-labelled with Klenow, denatured, hybridized to RNA samples from the ES lines carrying human APP 3 0 sequences, and SI analysis performed according to standard methods (Sambrook, et al., Molecular Cloning).
In addition, expression of human APP protein can be determined by immunoprecipitation of human APP using antibodies specific for human APP protein from ES cell lines 35 and tissue of transgenic animals. Such antibodies may also permit direct detection of human APP by standard immunohistochemical analysis of tissue sections.
Analysis of human APP expression in transgenic mice WO 94/00569 PCT/US93/05873 37 The qualitative and quantitative assays described above are also applicable to the analysis of the human APP gene in tissues of the transgenic mice derived from these ES lines.
Production of chimeric founders and germline transmission of the APP VAC Clones 23, 213 and 219 were injected into blastocysts to generate chimeric founder animals as described (Robertson, ed. Teratocarcinomas and Embryonic Stem Cells). 10 Founders are bred to wild type mice to generate F1 animals carrying the APP YAC.
Mouse models of Alzheimer's Disease Overexpression of the wild type human (or mouse) APP protein may result in phenotypes characteristic of Alzheimer's 15 Disease, including neurofibrillary tangle formation, plaque formation, and neurological dysfunction. Toward that end, different mouse lines expressing the APP YAC can be interbred to increase the number, and hence expression, of human APP genes.
Alternatively, mutations identified as associated with Familial Alzheimer's Disease (codon 717:V->I, F, or G) and/or Hereditary Cerebral Hemorrhage with Amyloidosis, Dutch type (HCHWA-D, codon 693:glu->gln) may be introduced into the human APP gene contained on the YAC using standard yeast 25 molecular genetic techniques such as insertion/eviction of a plasmid carrying a subcloned fragment of the APP gene containing the mutation, or by oligonucleotide directed transformation of yeast (Guthrie and Fink, Guide to Yeast Molecular Genetics and Molecular Biology) . jTACs carrying 30 these mutated APP genes can be introduced into t.ansn^nic mice using procedures described above. Other naturally— ring human APP disease allele sequences also may used, -.^eluding but not limited to those described at codon (Ala -> Gly), codon 692 (Glu -> Gin or Gly), and codon 713 (Ala -> Val) and 35 others that are described (Hendricks et al. (1932) Nature Genet, l: 218; Jones et al. (1992) Nature Genet. 1: 306; Hardy et al. (1992) Nature Genet. 1: 233; Mullan et al. (1992) PCT/Ul-jSP J/05873 38 Nature Genet.. 1: 505; Levy et. al. (1990) Science 248; 1124, incorporated herein by reference).
EXAMPLE 3 Transgenic Mice Expressing a Human Immunoglobulin Gene Cloned 5 in a Yeast Artificial Chromosome An 85 kb fragment of the human heavy chain immunoglobulin gene was cloned as a YAC, and embryonic stem cell lines carrying substantially intact, integrated YACs were derived by co-lipofection of the YAC and an unlinked 10 selectable marker. Chimeric founder animals were produced by blastocyst injection and offspring transgenic for the YAC clone were obtained. Analysis of serum from these offspring for the presence of human heavy chain demonstrated expression of the YAC borne immunoglobulin gene fragment. Unlike fusion 15 of yeast spheroplasts with mammalian cells, no yeast chromosomal DNA need be introduced by the co-lipofection method as the YAC(s) are typically first isolated from yeast chromosomes by a separation method, such as pulsed-field gel electrophoresis (PFGE). The YAC was introduced into ES cells 20 by co-lipofection with an unlinked selectable marker plasmid. The co-lipofection strategy differs from lipofection of modified YACs in that retrofitting vectors do not need to be constructed or recombined into the YAC, and YACs carried in recombination deficient hosts can be used. In contrast to 25 microinjection approaches, it is likely that larger YACs can be introduced by co-lipofection than microinjection due to the technical hurdles in purification of intact YAC DNA and because of the high shear forces imparted on the DNA during microinjection. Furthermore, unlike fusion of yeast 30 spheroplasts with mammalian cells where some of the yeast chromosomes integrate with the YAC5' 6, no yeast chromosomal DNA is introduced in co-lipofection since the YAC is first isolated by pulsed field gel electrophoresis.
Transgenic mice were produced by blastocyst 35 injection of ES cells carrying an intact YAC. The YAC was maintained intact through the germline, and human heavy chain antibody subunits were detected in the serum of transgenic offspring.
WO 94/00569 PCT/US93/05873 39 Human Heavy Chain Gene Fragment The 85 kb Spe I fragment of the unrearranged human immunoglobulin heavy chain locus was isolated. The 85kb Spe I fragment of the human heavy chain immunoglobulin (H) chain 5 gene contains at least one of each element required for correct rearrangement and expression of a human IgM heavy chain molecule.
Cloning of J1.3P An 85kb Spe I restriction fragment of the human 10 heavy chain immunoglobulin gene contains VH6, the functional diversity (D) segments, all six joining (J) segments, and the Cfi constant region segment (Hofker et al. (1989) Proc. natl. Acad. Sci. (U.S.A.) 86: 5587; Berman et al. (1988) EMBO J. 7: 727; Shin et al. (2.991) EMBO J. 10: 3641). Fresh human sperm 15 was harvested and genomic DNA prepared in agarose blocks as described in Strauss et al. (1992) Mamm. Genome 2: 150). A size selected (50-100kb) Spe I complete digest YAC library was prepared in the yeast host strain AB1380 in pYACneo15, using the Spe I site near the centromere as the cloning site. A 20 size selected (50-100kb) Spe I complete digest YAC library was produced in the YAC vector pYACneo15 and screened by colony hybridization with a probe specific for human C/x (Traver et al. (1989) Proc. Natl. Acad. Sci. (U.S.A.) 86: 5898). One positive clone (Jl) was identified among approximately 18,000 25 primary transformants. Because yeast mitochondrial DNA often obscured the YAC on pulsed field gel electrophoresis, a r* petite variant lacking mitochondrial DNA was selected by EtBr treatment, and denoted J1.3P. One subclone, J1.3P, was mounted in agarose blocks at 3.5 x 109 cells/ml and intact 30 yeast chromosomal DNA was prepared (Smith et al. (1990) proc.
Natl. Acad. Sci. (U.S.A.) 87: 8242). The YAC DNA was isolated in a 3-4mm wide gel slice from a low melting point preparative CHEF gel (Biorad). The gel slice was equilibrated in b-agarase buffer (Gelase, Epicentre Technologies), melted at 35 70"C for 20 minutes, cooled to 45*C, and digested with 10 units of agarase overnight at 45*C.
Characterization of YAC J1.3P • WO 94/00569 PCT/US93/05873 40 The authenticity of the J1.3P insert was determined by restriction mapping and Southern analysis. The ends of the insert were subcloned, using the bacterial selectable markers in the centromeric and acentromeric arms of pYACneo. Fine 5 structure restriction analyses of the terminal fragments were entirely consistent with published maps and sequences of the region (Fox et al. Analysis and manipulation of veast mitochondrial genes. In Guide to Yeast Genetics and Molecular-Biology (1991) eds. Guthrie C and Fink G, Academic Press, San 10 Diego, California; Word et al. (1989) Int. Immunol. J.: 296) and defined the orientation of the insert with respect to the vector arms. The orientation was further verified by PCR analysis of the acentromeric insert for VH6 sequences, and hybridization of the centromeric insert with the C/i probe. 15 Southern analysis of the Cfi region was consistent with published maps and restriction analyses (Hofker et al. (1989) Proc. Natl. Acad. Sci. (U.S.A.) 86: 5587). The functional diversity segments of the human heavy chain are contained in a 35 kb span containing a four-fold polymorphic repeat of D 20 segments. Southern analysis of the J1.3P YAC produced a "restriction fragment fingerprint" of the D region in which iill of the D specific bands in the YAC were present in human genomic DNA.
Co-lipofection of J1.3P YAC into ES cells 25 The J1.3P YAC was co-lipofected with an unlinked linearized plasmid carrying the neor gene driven by the mouse PGK promoter (Soriano et al. (1991) Cell 64: 893).
Selectable marker plasmids Plasmid is a 5 kb plasmid containing an expression 3 0 cassette consisting of the neo gene under the transcriptional control of the mouse phosphoglycerate kinase-1 promoter and the PGK-1 poly (A) site (Tybulewicz et al. (1991) Cell 40; 271). The plasmid pYPNN is a variant of pYACneo containing the PGKneo cassette in place of the SV40 promoter-neor 3 5 cassette, constructed by exchange of a 4.5kb Sal I-Apa I fragment of pYACneo for a 1.5kb Sal I-Apa I fragment of a containing the PGK promotor, neor coding region, and the WO 94/00569 PCT/US93/05873 41 PGKp(A) signal. The plasmids were linearized with Sal I (a) or Not I (pYPNN).
Lipofection of YAC DNA into ES cells.
The digested agarose/DNA mixture was divided into 1 5 ml (approximately 100 ng) portions in polystyrene tubes (Falcon) and 100 ng pYPNN or 20 ng , and l fig sheared herring sperm DNA (Sigma) was mixed in each tube, and cationic lipid (Transfectam, ProMega) was then added at a 10:1 ratio (wt:wt) and gently mixed into the DNA solution. The mixture 10 was incubated for 30 min at room temperature to allow formation of DNA-lipid complexes. Rapidly growing confluent cultures of AB-1 embryonic stem (ES) cells on mitotically inactivated SNL 76/7 fibroblast feeder layers were trypsinized to yield a single cell suspension, washed with serum-15 containing medium, and resuspended in serum-free DMEM (Gibco). For each lipofection, 9 ml of cell suspension containing 3 x 106 ES cells and about 1 x 105 feeder cells were mixed with 1 ml of the DNA-lipid mixture in a 60 mm petri dish (Falcon 1007; Becton Dickinson) and incubated for 4 hours at 37° c in 20 a humidified 5% C02 atmcsphere. Dishes were swirled gently during the incubation to minimize cell attachment. After incubation, cells were diluted with serum-containing ES cell medium, dispersed gently, and plated at 1 x 106 on 100 mm culture dishes containing feeder layers. Cells were selected 25 in G418 (400 /ig/ml powder, Gibco) for 9-12 days, beginning 24 hours after plating. Two different plasmids were tested: pYPNN (a 12 kb derivative of pYACneo carrying the PGKneo cassette in place of the SV40-neo cassette) and ickensian (a 5 kb plasmid carrying the same PGKneo cassette). The YAC:plasmid molar 30 ratio was 1:8 for pYPNN and 1:4 for ickensian. Two cationic lipid formulations were tested, DOGS (Transfectam; ProMega) and DOTMA (Lipofectin; BRL). Similar transfection efficiencies were obtained for DOGS and DOTMA with linearized plasmids, but DOGS was ultimately chosen for the YAC 3 5 experiments because its cationic moiety is spermine, obviating the need for exogenously added spermine as a DNA protectant, and because DOGS was not toxic to ES cells at the concentrations used. Because the DNA:lipid ratio was found to WO 94/00569 PCT/US93/05873 42 be important to the transfection efficiency, and precise measurement of the YAC DNA concentration was difficult, each lipofection contained an estimated 10-fold excess (1 ng) of sheared herring sperm carrier DNA to provide a baseline level 5 of DNA.
Analysis of ES clones G418-resistant clones were dispersed with trypsin and the cells from each clone were divided into one well of a 96-well plate that was frozen and a second 96-well or 24-well 10 plate used for preparation of DNA for screening by Southern analysis. Positive clones were thawed and expanded for further analysis.
Southern blot hybridization and PCR Genomic DNA was prepared from ES cells and tail 15 biopsies by rapid preparation methods (Laird et al. (1991) Nucleic Acids Res. 19; 4293) and subjected to Southern _ analysis by standard methods. For pulsed field gel electrophoresis, ES cells were embedded in agarose blocks at 107 cells/ml, prepared for restriction digestion, and digested 20 overnight with Spe I. For Southern analysis of pulsed field gels, the DNA was acid-nicked, then transferred to GeneScreen Plus (DuPont) in denaturing solution (0.4N NaOH, 1.5 M NaCl). Oligonucleotides suitable for PCR amplification of the VH6 region were prepared from published sequences. Primers used 25 were 5 ' CAGGTACAGCTGCAGCAGTCA3 ' and 5 ' TCCGGAGTCACAGAGTTCAGC3 ' , which amplified a diagnostic 275 bp product.
Production and analysis of transgenic mice Clones containing intact YAC sequences were injected into blastocysts to produce chimeric founder animals, which 3 0 were bred with C57BL/6 wild type mice and JH~ mice, which carry targeted inactivations of both copies of the mouse heavy chain gene. Thymic cells from transgenic offspring were mounted in agarose blocks for pulsed field gel electrophoresis and Southern analysis to confirm transmission of the intact 35 • YAC.
ELXSA assays Human mu chain was detected using a 2-site ELISA "assay. Polyvinyl chloride microtiter plates were coated with WO 94/00569 PCT/US93/05873 43 mouse monoclonal anti-human IgM clone CH6 (The Binding Site, San Diego, CA) at 1.25 /zg/ml in 100 /xl PBS by overnight incubation at 4 C. Plates were blocked by 1 hr incubation with 5% chicken serum (JRH, Lexana, KS) in PBS. Following 6 5 washes with PBS, 0.5% tween-20, serum samples and standards were diluted in 100 jul PBS, 0.5% Tween-20, 5% chicken serum (PTCS) and incubated in the wells for 1 hr at room "up. Purified human myeloma-derived IgM, kappa (Calbiochem, La Jolla, CA) was used as a standard. Plates were then washed 6 10 times with PBS, 0.5% tween-20 before addition of peroxidase conjugated rabbit anti-human IgM, Fc5u fragment specific antibody diluted 1/1000 in 100 n1 PTCS. After another 1 hr incubation at room temperature, the wells were washed 6 times and developed for 1/2 hr with 10 0 Ml ABTS substrate (Sigma) . 15 Assay plates were read at 415-49 0 nm on a Vmax _croplate reader (Molecular Devices, Menlo Park, CA), ana IgM concentration determined from a 4-parameter logistic curve fit of the standard values. A level of 4.89 ng/ml in serum samples is routinely detected by this assay and differentiated 20 from background by at least 3 standard deviations.
Results Approximately eight ng of J1.3PYAC DNA were lipofected in eight separate experiments (Table 1). Of the 1221 G418 resistant clones screened, 15 contained the two 25 diagnostic Eco RI C/i fragments (Table 2) . Two of the clones (#s 195 and 553) contained only one of the two C/j, bands, which may have arisen from fragmentation of the YAC within the missing Eco RI fragment (Table 2). The two selectable marker plasmids, pYPNN and ace, produced, respectively, frequencies 30 of 0.5 and 13.5 G418 resistant clones per 106 transfected cells; the efficiency of pYPNN selection was much lower even though it was used at twice the molarity of ace. It is unclear why the plasmids differed, since they both contained the same neor cassette, but it may be a consequence of the 35 extent of sequence homology between the plasmid and the vector arms, or different efficiencies of neor expression from the two plasmids.
Expt. # Selection plasmid Table 1. Summary of J1.3PYAC Lipofections YAC: neo /of § of G418r #C/i molar ratio lipofections clones positive Clone # 9 I •«< o 8 o\ «0 1 2 3 4 6 7 8 pYPNN PYPNN pYPNN pYPNN pYPNN mpt at at 1:8 1:8 1:8 1:8 1:8 1:4 1:4 1:4 8 9 9 9 9 4 16 18 24 19 12 8 10 44 480 624 3 1 0 0 0 0 3 10 14, 18, 21 12 86, 266, 480 25, 35, 164, Table 1. Co-lipofection of J1.3PYAC DNA with selectable marker plasmid. Approximately 100 ng of YAC J 1.3 was combined with the PGKneo-containing plasmids pYPNN or at and carrier DNA. Each experiment consisted of 4 to 18 separate lipofections of 100 ng YAC DNA each using the same solution of DNA/lipid complexes. A human Cfi probe was used for initial screening of clones. c ca) VO u> e (SI oo --I » . 0 <c o Table 2. Summary of ES clone structural analysis Clone 12 14 18 21 86 16 19 19 22 26 37 46 48 55 56 VH6 (PCR): -* -* + -* - + + - nd - + - + + nd - + D region: - - + - - - + - + - + nd + + nd - + Cfi analysis: + + + + + + + + + - + + + + + - + 3' end: - - + - + + nd - + - - nd + + nd - + PGFE analysis: - - + - nd nd - nd - nd - nd + + nd nd - Table 2. ES clones which were positive for the C/i probe were analyzed for VH6 . sequences by PCR, D region integrity by Southern blot, and 3' end integrity by Southern blot. "+11 in the VH6 analysis denotes amplification of a diagnostic 275 bp PCR product using VH6 specific primers. I,+" in the D region fingerprint denotes a completely parental banding pattern. + in the Cp analysis signifies that 2 diagnostic bands were present when hybridized with probe, while indicates the presence of a single band. "+" in the 3' end analysis denotes the reduction in size of the flanking Xhol band to 4.1 kb in a Xhol/Spel double digest, as well as the presence of a 4.5 kb internal Xhol band in both digests. "+" in the pulsed field gel electrophoresis analysis denotes the presence of a single 85 kb Spel band when probed with the D region probe, nd: not determined. Clones carrying intact YAC inserts are marked in bold. *Clones 12, 14, 18, and 21 were analyzed for VH6 by probing Apa I and Eco RI digests using the 275 bp PCR product DNA as probe. Only clone 18 was positive for the diagnostic VH6 bands. & C C/) 00 w PCT/US93/0S873 46 Analysis of YAC structure in ES cells.
The four C/li+ clones from the pYPNN co-lipofections (#s 12,14,18,21) were analyzed for D region structure by the restriction fragment fingerprint assay described above, of 5 the four clones, only clone 18 retained the fingerprint of the parent YAC. Clones 14 and 21 contained fewer bands than the parent, suggesting that YAC sequences may have been lost, while clone 12 contained several additional bands, consistent with integration of more than one copy of the YAC in this ES 10 line.
The integrity of the 3' end of the insert region in the four ES lines was assessed by Southern analysis using the 10.5Jcb Nde I-Spe I terminal fragment isolated by vector recircularization as probe. Three bands are expected from a 15 Xho X digest of the parent YAC: a very large D-J-C/x band (>3 0 kb) , a 4.5 kb Cjx-CS band, and an 8.9 kb C£-vector band. A double digest with Xho I and Spe I is expected to reduce the size of the 8.9 kb band to 4.1 kb. The 4.5 kb and 8.9 bands are present in the Xho I digests, while the 4.5 kb and 4.1 kb 20 bands are present in the Xho I~Spe I digests of the parent YAC. Among the four ES lines, only line 18 contained the parental YAC banding pattern indicative of an intact 3'-end. The presence of an 8.9 kb band is consistent with the retention of the vector arm Xho I site in the ES line, 25 suggesting that very little of the telomeric region had been lost in this clone. Loss of YAC terminal sequences would be expected to result in aberrant Xho I bands. Among the other three ES lines, clone 14 lacked the 4.5 kb Xho I band, while clones 12 and 21 contained aberrantly short Xho I bands, 30 indicating rearranged or deleted 3' end regions in these ES clones. A similar analysis of 5* end integrity was not possible due to repetitive elements in the region. However, PCR and Southern analysis using the VH6 PCR product as probe indicated that clone 18 contained VH6 sequences, while clones 35 14, 12, and 21 did not (Table 2).
Of the 13 Cn+ ES cell lines from the d co-lipof ections, one was lost during clonal expansion, and one (# 2 66) was eliminated because it lacked Vh6 sequence. The WO 94/00569 PCT/US93/05873 Al remainder were analyzed for D region structure, 3*-end integrity and/or VH6 sequence (Table 2). Of the 11 lines analyzed for D region fingerprint, six (#s 86, 191, 220, 371, 463, 567) showed an intact D region while five had aberrant 5 patterns. 3' end analysis of five of the six lines with intact D regions revealed that all but one (#220) contained an intact 31 end. PCR analysis revealed that five of the six lines with intact D regions (#86, 220, 371, 463, 567) contained VH6 sequences, while only one of five lines without 10 intact D regions (#35) contained VH6 sequences.
Ten of the ES cell lines were examined for full length insert by pulsed field .Southern analysis using the D region or Cju probe (Table 2). Only clones 18, 371, and 463 contained an 85 kb Spe I fragment indicative of a full length 15 insert; all of the other clones had a smaller Spe I fragment. The Spe I digest of clone 18 was screened with both D and C/x probes and a probe for VH6; all three probes hybridized to a single band of 85 kb.
The pulsed field Southern analysis, taken together 20 with the D region, 3' end and VH6 fine structure analyses, indicate that the YAC insert was transferred intact in three ES l.xnes: #18, #371, and #463. A high degree of internal rearrangement, deletion or fragmentation was generally seen in the ES lines carrying disrupted YAC sequences, although subtle 25 alterations of structure were also detected (e.g., #567).
Overall, the frequency of intact YAC transfer was low, l in 400 G418r clones (3/1221). However, the isolation of the clone DNAs and the primary screen for C/i sequences (which eliminated 1206 of the 1221 clones from further analysis) were 3 0 rapidly performed using the microtitre plate protocols described in Methodology. Thus, only 15 clones required extensive analysis (Table 2).
Molecular analysis of YAC structure in ES cells is greatly facilitated by a low, preferably single, copy of the 35 YAC. The D region, pulsed field gel analysis, and 3' end analyses of the ES lines are consistent with a low or single copy integration of the YAC. Analysis of clones 18, 371, and 4 63 for a diagnostic 31 end flanking band showed that clones WO 94/00569 PCT/US93/05873 48 18 and 371 carried a single copy of "the YAC insert, while 463 may h?ive an additional intact Mor partially intact copy. Production of chimeras and germline transmission of the YAC Blastocysts were injected with ES lines 18, 371, and 5 463. Chimeric founder animals ranging from 10% to 95% ES cell contribution to coat color were derived from all three lines. The oldest animal, a 40% chimeric male derived from ES line 18, transmitted the ES cell genotype to 20 of 73 offspring. Eleven of the 20 agouti offspring were positive for an intact 10 D region fingerprint, consistent with Mendelian segregation of a hemizygous YAC transgene allele. In addition, pulsed field Southern analysis using the D region probe demonstrated a single 85 kb Spe I band in transgenic offspring, indicating that the YAC was stably maintained through the germline. 15 Thus, co-lipofection of YACs into ES cells does not abrogate ES cell totipotency.
Southern analysis of integration sites for the co-lipofected selectable marker indicated integration of 2 to 10 plasmid copies. Because it is possible that the marker 20 plasmids could be a source of mutations if they were to insert at multiple loci, the integration sites of the plasmid were tracked by Southern analysis for plasmid sequences. Since pYPNN and the YAC vector arms lack Eco RI sites and contain pBR322 sequences, each Eco RI band which hybridized to a 25 pBR322 probe represents the integration of a separate intact or fragmented copy of pYPNN or the YAC vector arms. Analysis of ES cell clone 18 DNA revealed eight Eco RI bands ranging in size from 5.5 kb to 20 kb, and the offspring of a hemizygous transgenic animal bred with non-transgenic mates were analyzed 30 for segregation of the Eco RI bands. Among 14 offspring, all eight Eco RI bands were detected in tail DNAs of the 9 transgenic pups, and none were detected in tail DNAs of the 5 non-transgenic pups. Thus, all detectable marker plasmids segregated with the YAC, indicating that they had inserted at 35 or near the YAC integration site. Co-integration of different DNAs have been observed in transgenic mice produced by microinjection of zygotes, and it is expected that co-integration of plasmid DNAs would be no more mutagenic for co- WO 94/00569 PCT/US93/05873 49 lipofection than for zygote microinjection. Presumably, the herring sperm carrier DNA had also co-integrated with the YAC, and may be a source of Eco RI sites in the Southern analysis. Since co-integrated carrier DNA may potentially adversely 5 affect YAC transgene function, it is frequently preferable to omit carrier DNA. Preliminary experiments with a 650 kb YAC indicate that carrier DNA is not required for efficient lipofection of intact YACs into ES cells. This preliminary work also suggests that the size limit of YACs which can be 10 successfully co-lipofected into ES cells is at least 650 kb. Serum expression of hunan immunoglobulins in transgenic mice Line 18 transgenic mice were assayed for human mu chain in the serum by ELISA. Human mu heavy chain was detected in the serum of transgenic offspring (Table 3). 15 Although the human mu serum levels in the transgenics were clearly within the detectable range, they were very low compared to serum levels of endogenous mouse IgM. The low level of transgene expression is due in part to competition from the endogenous heavy chain gene. The transgene was 20 introduced into a background in which the endogenous heavy chain alleles are inactivated, and in this mouse, the human mu serum levels were elevated approximately 10 fo^d (Table 3).
• WO 94/00569 50 ?able 3. Detection of serum human IaM bv ELISA Genotype YAC YAC YAC YAC YAC YAC YAC 18+ 18+ 18+ 18+ 18+ 18+ 18+ Sex Age at assay (wk) F 3, 9, 20 M 17 F 10 F 6, 17 F 4 F 6 M 6 Human IgM < 5 ng/ml 12.2 ng/ml 27.0 ng/ml < 5 ng/ml 5.8 ng/ml 10.5 ng/ml 10.4 ng/ml YAC 18+/JH- M , 8 165 ng/ml Wild type F 6 < 5 ng/ml Wild type F 6 < 5 ng/ml Wild type F 6 < 5 ng/ml Wild type M 6 < 5 ng/ml Wild type F 34 < 5 ng/ml Wild type F 34 < 5 ng/ml Wild type M 34 < 5 ng/ml Wild type M 34 < 5 ng/ml Table 3. Blood samples from transgenic animals and controls were analyzed by ELISA for human IgM at the ages indicated. All of the transgenic animals are derived from a single clone 18 founder chimera, and are hemizygous for the YAC (YAC 18+). Five of the seven animals in a wild type background had detectable human IgM in their serum. The level of detection of the ELISA was 5 ng human IgM/ml serum. The serum human IgM level was elevated approximately 10-fold when the YAC transgene was bred into a background lacking functional endogenous mouse heavy chain genes (YAC18+/JH-). 51 FACS Analysis of YAC+/Jn~ Mice Fluorescence-activated cell-sorting (FACS) analysis was performed on mice positive for the YAC containing the 85kb heavy chain gene fragment and homozygous for a functionally 5 disrupted ("knocked-out") endogenous murine immunoglobulin heavy chain gene by disruption of the JH region by homologous gene targeting- The mice had a single copy of the YAC transgene and lacked functional murine heavy chain alleles. The FACS analysis used antibodies to detect human mu chains, 10 among others, and showed that about 60 cells per 10,000 total peripheral lymphocytes from the mice expressed a human mu chain immunoglobulin. This level is approximately 1-2 percent of the number of cells that express murine mu chains in a wild-type (non-transgenic/non-knockout) mouse spleen. 15 FACS detected human mu chain expression in cells obtained from the spleen and peritoneal cavity of the YAC+/JH" mice.
Although the foregoing invention has been described in some detail by way of illustration and example, for 20 purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims. 2 5 3 9 4 3 PCT/US93/0S873

Claims (48)

52 CLAIMS
1. A method for producing a co-lipofected mammalian cell having incorporated multiple heterologous DNA species, comprising the steps of: 5 forming a co-lipofection complex comprising a cationic lipid, a first polynucleotide, and an unlinked second polynucleotide comprising at least one selectable marker gene expression cassette; contacting a mammalian cell with said co-lipofection 10 complex under conditions whereby said first polynucleotide and said second polynucleotide are introduced into the same cell and are integrated into the genome.
2. A method according to Claim 1, wherein said cationic 15 lipid is selected from the group consisting of: N[l-(2,3- dioleoyloxyl)propyl]-N,N,N-trimethylammonium chloride; N[l-2,3-dioleoyloxyl)propyl]-N,N,N-trimethylammonium methylsulfate; N-(2,3-di(9-(Z)-octadecenyloxy))—prop-l-N,N,N-trimethylammonium chloride; dioleoylphosphatidylethanolamine 20 (PtdEtn, DOPE); and dioctadecylamidoglycyl spermidine.
3. A method according to Claim 1, wherein the first polynucleotide is at least 500 kb. 25
4. A method according to Claim 1, wherein at least one of said selectable marker is a drug resistance gene.
5. A method according to Claim 4, wherein said selectable marker is a gene encoding neomycin resistance. 30
6 . A method according to Claim 1, further comprising the step of selecting for cells having said selectable marker.
7. A method according to Claim 1, wherein said mammalian 35 cell is a nonhuman embryonal stem cell.
8. A method according to Claim 7, wherei embryonal stem cell is a mouse ES cell. WO 94/00569 253943 PCT/US93/05873 53
9. A method according to Claim 8, wherein said cationic lipid is dioctadecylamidoglycyl spermidine (DOGS), said first polynucleotide contains a human APP gene sequence, and said unlinked second polynucleotide contains a neomycin resistance 5 gene.
10. A method according to Claim 1, wherein said first polynucleotide comprises xenogenic mammalian DNA sequences linked to yeast- derived YAC sequences. 10
11. A co-lipofection complex, comprising cationic lipid noncovalently bound to a first DNA comprising a YAC clone containing xenogenic mammalian DNA and to a second DNA comprising a gene encoding a selectable marker. 15
12. A co-lipofection complex according to Claim 11, wherein the cationic lipid is selected from the group consisting ofj_ N [ 1- (2 ,3-dioleoyloxy 1) propyl ] -N, N, N-trimethylammonium chloride; N[1-2,3-dioleoyloxyl)propyl]-N,N,N-trimethylammonium methylsulfate; and dioctadecylamidoglycyl spermidine. 20
13. A co-lipofection complex according to Claim 11, wherein the YAC DNA comprises a human APP gene sequence or a human immunoglobulin gene sequence. 25
14. A co-lipofection complex according to Claim 13, wherein said cationic lipid is dioctadecylamidoglycyl spermidine.
15. A co-lipofection complex according to Claim 14, wherein said selectable marker gene is neoR. 30
16. A co-lipofection complex comprising a polynucleotide of at least 50 kb, an unlinked selectable marker gene expression cassette, and a cationic lipid selected from the group consisting of: N[1-(2,3-dioleoyloxyl)propyl]-N,N,N- 35 trimethylammonium chloride; N[1-2,3-dioleo; N,N,N-trimethylammonium methylsulfate; and dioctadecylamidoglycyl spermidine. 25 3 9 4 3- WO 94/00569 PCT/US93/05873 54
17. A composition comprising a mammalian cell and a co-lipofection complex of Claim 16.
18. A composition according to Claim 17, wherein the 5 mammalian cell is a nonhuman embryonal stem cell.
19. A composition according to Claim 17, wherein the nonhuman embryonal stem cell is a mouse ES cell. 10
20. A cotransfected mammalian cell produced by a method of Claim 1.
21. A cotransfected mammalian cell produced by a method of Claim 10. 15
22. A chimeric transgenic nonhuman animal produced by the method of Claim 1.
23. A transgenic nonhuman animal produced by the method of 2 0 Claim 9.
24. A transgenic mouse comprising a germline genome comprising a xenogenic polynucleotide sequence of at least 50 kb linked to a yeast-derived YAC sequence produced by cotransfecting a mouse ES cell 2 5 with the composition of Claim 11.
25. A transgenic mouse according to Claim 24, wherein the xenogenic polynucleotide is at least about 500 kb.
26. A transgenic mouse according to Claim 25, wherein the 3 0 xenogenic polynucleotide sequence encodes a human APP protein or a human immunoglobulin.
27. A transgenic mouse according to Claim 26, further comprising a neoR gene expression cassette. 35
28. A co-lipofection complex comprising a YAGycbntaini-ljJV an unrearranged human immunoglobulin gene comprising the 85^)%d Spe I fragment, an unlinked selectable marker, and a cationic lipid. 1 ■ 2 5 3 9 4 3 WO 94/00569 PCT/US93/0S873 \ 55
29. A transgenic mouse produced by cotransfecting a mouse ES cell with the composition of Claim 11 comprising a YAC comprising an unrearranged human immunoglobulin gene having at least one V region gene, at least one J region gene, and at least one constant region gene. 5
30. A transgenic mouse of claim 29, wherein the transgenic mouse expresses a human immunoglobulin chain.
31. A transgenic mouse of claim 29, wherein the YAC comprises 10 a 85 kb Spe I fragment of the human heavy chain locus.
32. A method of introducing a heterologous polynucleotide containing a complete structural gene into a mammalian cell, comprising the steps of: forming a lipofection complex comprising a cationic lipid and a heterologous fu st polynucleotide comprising a complete structural gene and an unlinked second polynucleotide comprising a selectable marker; 10 contacting a mammalian cell with said lipofection complex under conditions whereby said heterologous first and second polynucleotides are introduced into the mammalian cell and are integrated into the genome. 15 20
33. A method of claim 32, wherein said heterologous polynucleotide comprises a complete transcription unit comprising a cis-acting linked promoter which naturally occurs adjacent to said structural gene in germline DNA.
34. A method of claim 32, wherein said heterologous polynucleotide is a yeast artificial chromosome. 25
35. A method of claim 32, wherein said polynucleotide is at least 650 kb. 56 253943
36. A method of claim 32, wherein said mammalian cells are pluripotent mouse embryonic stem cells.
37. A method of claim 36, comrpising the further step of selecting mouse embryonic stem cells which have incorporated the heterologous polynucleotide, introducing said cells into mouse blastocysts to produce transgenic mice harboring a germline copy of the heterologous polynucleotide.
38- A method of claim 37, wherein the lipofection complex comprises a cationic lipid selected from the group consisting of: N[1-(2,3-dioleoyloxyl)propyl]-N, N,N-trimethylammonium chloride? N[l-2,3-dioleoyloxyl)propyl]-N,N,N-trimethylammonium methylsulfate ; N- (2 , 3-di (9 - (Z) -octadecenyloxy)) -prop-i-N, N, N-trimethylammonium chloride; dioleoylphosphatidylethanolamine (PtdEtn, DOPE); and dioctadecylamidoglycyl spermidine.
39. A method as defined in claim 1 for producing a co-lipofected mammalian cell having incorporated multiple heterologous DNA species substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
40. A co-lipofection complex as defined in claim 11 or claim 16 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
41. A composition as defined in claim 17 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
42. A cotransfected mammalian cell as claimed jjjr^laim 20 oroCT^im 21 substantially as herein described with reference to anyjfexample thereof \nd S'-4 ~ j with or without reference to the accompanying drawings. » «\V^ v i f. " ^ S 57 2 5 3 9 4 3
43. A chimeric transgenic nonhuman animal as claimed in claim 22 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
44. A transgenic nonhuman animal as claimed in claim 23 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
45. A transgenic mouse as defined in claim 24 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
46. A co-lipofection complex as claimed in claim 28 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
47. A transgenic mouse as defined in claim 29 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
48. A method as defined in claim 32 of introducing a heterologous polynucleotide containing a complete structural gene into a mammalian cell substantially as herein described with reference to any example? thereof and with or without reference to the accompanying drawings. GlSNfWfN rfrn^AT^ALj,, (MS.. By .uuTorised agents A. J Rark &£oo
NZ25394393A 1992-06-18 1993-06-18 Transfering polynucleotides into eukaryotic cells using co-lipofection complexes of a cationic lipid and the polynucleotide NZ253943A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US90097292A 1992-06-18 1992-06-18
US149393A 1993-01-07 1993-01-07

Publications (1)

Publication Number Publication Date
NZ253943A true NZ253943A (en) 1997-01-29

Family

ID=26669098

Family Applications (1)

Application Number Title Priority Date Filing Date
NZ25394393A NZ253943A (en) 1992-06-18 1993-06-18 Transfering polynucleotides into eukaryotic cells using co-lipofection complexes of a cationic lipid and the polynucleotide

Country Status (6)

Country Link
EP (1) EP0648265A4 (en)
JP (1) JPH07508410A (en)
AU (1) AU4541093A (en)
CA (1) CA2135313A1 (en)
NZ (1) NZ253943A (en)
WO (1) WO1994000569A1 (en)

Families Citing this family (238)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6150584A (en) * 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) * 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
US6657103B1 (en) 1990-01-12 2003-12-02 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1991010741A1 (en) * 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
US6610493B1 (en) 1993-06-17 2003-08-26 Brigham And Women's Hospital Screening compounds for the ability to alter the production of amyloid-β peptide
ATE381614T1 (en) * 1992-07-24 2008-01-15 Amgen Fremont Inc FORMATION OF XENOGENE ANTIBODIES
WO1994023049A2 (en) * 1993-04-02 1994-10-13 The Johns Hopkins University The introduction and expression of large genomic sequences in transgenic animals
JPH09507746A (en) 1993-10-27 1997-08-12 アテナ ニューロサイエンシズ,インコーポレイティド Transgenic animal containing APP allele with Swedish mutation
FR2714830B1 (en) * 1994-01-10 1996-03-22 Rhone Poulenc Rorer Sa Composition containing nucleic acids, preparation and uses.
US5777153A (en) * 1994-07-08 1998-07-07 Gilead Sciences, Inc. Cationic lipids
FR2732895B1 (en) * 1995-04-11 1997-05-16 Pasteur Merieux Serums Vacc USE OF A CATIONIC AMPHIPATHIC COMPOUND AS A TRANSFECTING AGENT, AS A VACCINE ADDITIVE, OR AS A MEDICINAL PRODUCT
CA2761116A1 (en) * 1995-04-27 1996-10-31 Amgen Fremont Inc. Human antibodies derived from immunized xenomice
US6531586B1 (en) 1995-04-28 2003-03-11 The Hospital For Sick Children Genetic sequences related to Alzheimer's Disease
US5986054A (en) 1995-04-28 1999-11-16 The Hospital For Sick Children, Hsc Research And Development Limited Partnership Genetic sequences and proteins related to alzheimer's disease
US6210919B1 (en) 1995-04-28 2001-04-03 Hsc Research And Development Limited Partnership Genetic sequences and proteins related to alzheimer's disease
EP0830368A1 (en) 1995-06-07 1998-03-25 Genta Incorporated Novel carbamate-based cationic lipids
US6717031B2 (en) 1995-06-07 2004-04-06 Kate Dora Games Method for selecting a transgenic mouse model of alzheimer's disease
US6632976B1 (en) 1995-08-29 2003-10-14 Kirin Beer Kabushiki Kaisha Chimeric mice that are produced by microcell mediated chromosome transfer and that retain a human antibody gene
EP0876483A1 (en) * 1996-01-26 1998-11-11 HSC Research and Development Limited Partnership Nucleic acids and proteins related to alzheimer's disease, and uses therefor
US6093816A (en) 1996-06-27 2000-07-25 Isis Pharmaceuticals, Inc. Cationic lipids
CA2183901A1 (en) * 1996-08-22 1998-02-23 Johanna E. Bergmann Targets for therapy and diagnosis of alzheimer's disease and down syndrome in humans
KR20080059467A (en) 1996-12-03 2008-06-27 아브게닉스, 인크. Transgenic mammals having human ig loci including plural vh and vk regions and antibodies produced therefrom
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
KR100849443B1 (en) 1998-12-23 2008-07-31 화이자 인크. Human monoclonal antibodies to ctla-4
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
US6833268B1 (en) 1999-06-10 2004-12-21 Abgenix, Inc. Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
US6900367B2 (en) 2000-09-29 2005-05-31 Novartis Transgenic Drosophila melanogaster expressing a β42 in the eye
EP1383541A4 (en) * 2001-03-22 2009-11-04 Chromos Molecular Systems Inc Methods for delivering nucleic acid molecules into cells and assessment thereof
US20030119104A1 (en) 2001-05-30 2003-06-26 Edward Perkins Chromosome-based platforms
DE60327608D1 (en) 2002-02-25 2009-06-25 Genentech Inc NEW TYPE 1 CYTOKIN RECEPTOR GLM-R
CA2508375C (en) 2002-12-02 2014-05-27 Abgenix, Inc. Antibodies directed to tumor necrosis factor and uses thereof
WO2004080406A2 (en) 2003-03-07 2004-09-23 Alnylam Pharmaceuticals Therapeutic compositions
CA2519528C (en) 2003-03-19 2016-01-26 Abgenix, Inc. Antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen and uses thereof
DK1620544T3 (en) 2003-04-17 2019-01-14 Alnylam Pharmaceuticals Inc MODIFIED iRNA AGENTS
EP2457586A1 (en) 2003-06-27 2012-05-30 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
WO2005016111A2 (en) 2003-08-08 2005-02-24 Abgenix, Inc. Antibodies directed to parathyroid hormone (pth) and uses thereof
CN1274814C (en) * 2003-09-23 2006-09-13 李宁 Method for raising efficiency for producing transgenic animal
WO2005092926A2 (en) 2004-03-19 2005-10-06 Amgen Inc. Reducing the risk of human and anti-human antibodies through v gene manipulation
EP1791868B1 (en) 2004-07-01 2011-02-23 Novo Nordisk A/S Antibodies binding to receptors kir2dl1, -2, 3 but not kir2ds4 and their therapeutic use
JP2008517608A (en) 2004-10-22 2008-05-29 レビビコア, インコーポレイテッド An ungulate having a genetically modified immune system
US20080026457A1 (en) 2004-10-22 2008-01-31 Kevin Wells Ungulates with genetically modified immune systems
AU2005307831A1 (en) 2004-11-17 2006-05-26 Amgen, Inc. Fully human monoclonal antibodies to IL-13
EP1838737B1 (en) 2004-12-20 2011-04-06 Amgen Fremont Inc. Binding proteins specific for human matriptase
WO2006068953A2 (en) 2004-12-21 2006-06-29 Astrazeneca Ab Antibodies directed to angiopoietin-2 and uses thereof
EP1851245B1 (en) 2005-01-26 2012-10-10 Amgen Fremont Inc. Antibodies against interleukin-1 beta
AU2006315580A1 (en) 2005-11-10 2007-05-24 Curagen Corporation Method of treating ovarian and renal cancer using antibodies against T cell immunoglobulin domain and mucin domain 1 (TIM-1) antigen
CA2633956C (en) 2005-12-13 2016-12-06 Olivia Raeber Binding proteins specific for insulin-like growth factors and uses thereof
AR056857A1 (en) 2005-12-30 2007-10-24 U3 Pharma Ag DIRECTED ANTIBODIES TO HER-3 (RECEIVER OF THE HUMAN EPIDERMAL GROWTH FACTOR-3) AND ITS USES
EP1984403A2 (en) 2006-01-12 2008-10-29 Alexion Pharmaceuticals, Inc. Antibodies to ox-2/cd200 and uses thereof
BRPI0709481A2 (en) 2006-04-07 2011-07-19 Government Of The Us Secretary Dept Of Health And Human Services isolated monoclonal antibody, isolated human monoclonal antibody, pharmaceutical composition, isolated recombinant anti-igf-i and anti-igf-ii antibody or antigen-binding fragment thereof, method for detecting factor i and factor ii of human insulin growth in a sample , method for detecting human insulin growth factor i in a sample, isolated nucleic acid, recombinant cell, host cell, method for preparing an antibody, method for preparing an antibody, method for treating neoplastic disease in a mammalian individual, method for to diagnose neoplastic disease in a mammal and method to classify a drug candidate compound
TW200813091A (en) 2006-04-10 2008-03-16 Amgen Fremont Inc Targeted binding agents directed to uPAR and uses thereof
CL2007002225A1 (en) 2006-08-03 2008-04-18 Astrazeneca Ab SPECIFIC UNION AGENT FOR A RECEIVER OF THE GROWTH FACTOR DERIVED FROM PLATES (PDGFR-ALFA); NUCLEIC ACID MOLECULA THAT CODIFIES IT; VECTOR AND CELL GUESTS THAT UNDERSTAND IT; CONJUGADO UNDERSTANDING THE AGENT; AND USE OF THE AGENT OF A
JP5362563B2 (en) 2006-08-03 2013-12-11 アストラゼネカ・アクチエボラーグ Antibodies against αVβ6 and uses thereof
NZ580245A (en) 2007-03-22 2012-01-12 Biogen Idec Inc Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
CN101802211B (en) 2007-08-02 2014-10-01 诺维莫尼公司 Anti-RANTES antibodies and methods of use thereof
JOP20080381B1 (en) 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
EP2615113A3 (en) 2007-08-23 2013-11-13 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
SI2207809T1 (en) 2007-09-26 2013-11-29 U3 Pharma Gmbh Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
CN101939336B (en) 2007-11-12 2014-05-14 U3制药有限公司 AXL antibodies
EP4223299A3 (en) 2007-12-04 2023-08-30 Alnylam Pharmaceuticals, Inc. Carbohydrate conjugates as delivery agents for oligonucleotides
WO2009126933A2 (en) 2008-04-11 2009-10-15 Alnylam Pharmaceuticals, Inc. Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
WO2010032060A1 (en) 2008-09-19 2010-03-25 Medimmune Llc Antibodies directed to dll4 and uses thereof
DK2348827T3 (en) 2008-10-27 2015-07-20 Revivicor Inc IMMUNICIPLY COMPROMATED PETS
AU2009313203B2 (en) 2008-11-10 2015-08-27 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
CA2748158A1 (en) 2008-12-23 2010-07-01 Astrazeneca Ab Targeted binding agents directed to .alpha.5.beta.1 and uses thereof
SG175276A1 (en) 2009-05-05 2011-11-28 Novimmune Sa Anti-il-17f antibodies and methods of use thereof
JP5635617B2 (en) 2009-11-09 2014-12-03 アレクシオン ファーマシューティカルズ, インコーポレイテッド Reagents and methods for detecting PNH type II leukocytes and their identification as risk factors for thrombotic disorders
EP3670539A1 (en) 2009-11-13 2020-06-24 Daiichi Sankyo Europe GmbH Material and methods for treating or preventing her-3 associated diseases
NZ599405A (en) 2009-11-24 2014-09-26 Medimmune Ltd Targeted binding agents against b7-h1
SG182408A1 (en) 2010-01-11 2012-08-30 Alexion Pharma Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
WO2011109427A2 (en) 2010-03-01 2011-09-09 Alnylam Pharmaceuticals, Inc. Improving the biological activity of sirna through modulation of its thermodynamic profile
CN105963694B (en) 2010-04-30 2019-11-05 阿雷克森制药公司 Anti- C5A antibody and the method for using the antibody
SG186397A1 (en) 2010-06-22 2013-01-30 Univ Colorado Regents Antibodies to the c3d fragment of complement component 3
AU2011286407A1 (en) 2010-08-06 2013-02-21 Amgen Use of HER3 binding agents in prostate treatment
SG10201600531TA (en) 2011-01-24 2016-02-26 Univ Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
AR085141A1 (en) 2011-02-03 2013-09-11 Alexion Pharma Inc A METHOD FOR EXTENDING THE SURVIVAL OF A RENAL PAD, USE OF AN ANTIBODY ANTI-CD200 TO EXTEND THE SURVIVAL OF PADS
JP5908972B2 (en) 2011-04-07 2016-04-26 アムジエン・インコーポレーテツド Novel antigen binding protein
US9150644B2 (en) 2011-04-12 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies that bind insulin-like growth factor (IGF) I and II
JOP20200043A1 (en) 2011-05-10 2017-06-16 Amgen Inc Methods of treating or preventing cholesterol related disorders
CN107936121B (en) 2011-05-16 2022-01-14 埃泰美德(香港)有限公司 Multispecific FAB fusion proteins and methods of use thereof
KR102395085B1 (en) 2011-06-21 2022-05-09 알닐람 파마슈티칼스 인코포레이티드 Angiopoietin-like 3(angptl3) irna compostions and methods of use thereof
WO2013012855A1 (en) 2011-07-18 2013-01-24 Amgen Inc. Apelin antigen-binding proteins and uses thereof
WO2013067057A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
AU2012332587B2 (en) 2011-11-01 2017-02-23 Bionomics, Inc. Antibodies and methods of treating cancer
WO2013067055A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Methods of blocking cancer stem cell growth
WO2013067060A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
EP2780040A1 (en) 2011-11-16 2014-09-24 Amgen Inc. Methods of treating epidermal growth factor deletion mutant viii related disorders
SG10201912170WA (en) 2011-11-18 2020-02-27 Alnylam Pharmaceuticals Inc Rnai Agents, Compositions And Methods Of Use Thereof For Treating Transthyretin (TTR) Associated Diseases
KR102689177B1 (en) 2011-11-18 2024-07-30 알닐람 파마슈티칼스 인코포레이티드 MODIFIED RNAi AGENTS
MX360772B (en) 2012-02-06 2018-11-15 Inhibrx Inc Cd47 antibodies and methods of use thereof.
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
EA039663B1 (en) 2012-05-03 2022-02-24 Амген Инк. Use of an anti-pcsk9 antibody for lowering serum cholesterol ldl and treating cholesterol related disorders
CN104540961A (en) 2012-06-11 2015-04-22 安姆根公司 Dual receptor antagonistic antigen-binding proteins and uses thereof
AU2013296321B2 (en) 2012-08-03 2019-05-16 Alnylam Pharmaceuticals, Inc. Modified RNAi agents
PL2929031T4 (en) 2012-12-05 2022-06-27 Alnylam Pharmaceuticals, Inc. Pcsk9 irna compositions and methods of use thereof
JO3519B1 (en) 2013-01-25 2020-07-05 Amgen Inc Antibody constructs for CDH19 and CD3
ES2728936T3 (en) 2013-01-25 2019-10-29 Amgen Inc Antibodies directed against CDH19 for melanoma
EA036963B1 (en) 2013-02-06 2021-01-20 Инхибркс, Инк. Use of a cd47 epitope to produce a cd47 antibody
ES2649490T3 (en) 2013-03-14 2018-01-12 Alnylam Pharmaceuticals, Inc. Compositions of RNAi against component C5 of the complement and methods for its use
EP3653642A1 (en) 2013-03-15 2020-05-20 Amgen Research (Munich) GmbH Single chain binding molecules comprising n-terminal abp
US9505849B2 (en) 2013-03-15 2016-11-29 Amgen Research (Munich) Gmbh Antibody constructs for influenza M2 and CD3
EP2999785B1 (en) 2013-05-22 2018-04-04 Alnylam Pharmaceuticals, Inc. Serpina1 irna compositions and methods of use thereof
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
CA2916259C (en) 2013-06-28 2024-02-20 Amgen Inc. Methods for treating homozygous familial hypercholesterolemia
US10077304B2 (en) 2013-08-14 2018-09-18 The Governing Council Of The University Of Toronto Antibodies against frizzled receptor
AR097738A1 (en) 2013-09-23 2016-04-13 Alnylam Pharmaceuticals Inc METHODS TO TREAT OR PREVENT DISEASES ASSOCIATED WITH TRANSTIRETINE (TTR)
IL282401B (en) 2013-12-12 2022-08-01 Alnylam Pharmaceuticals Inc Complement component irna compositions and methods of use thereof
US10119136B2 (en) 2014-01-09 2018-11-06 Alnylam Pharmaceuticals, Inc. RNAi agents modified at the 4′-C position
KR20230152154A (en) 2014-02-11 2023-11-02 알닐람 파마슈티칼스 인코포레이티드 KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
NZ724815A (en) 2014-04-04 2017-11-24 Bionomics Inc Humanized antibodies that bind lgr5
CA2948381C (en) 2014-05-22 2023-11-07 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
TWI695011B (en) 2014-06-18 2020-06-01 美商梅爾莎納醫療公司 Monoclonal antibodies against her2 epitope and methods of use thereof
AR101669A1 (en) 2014-07-31 2017-01-04 Amgen Res (Munich) Gmbh ANTIBODY CONSTRUCTS FOR CDH19 AND CD3
CA2956471A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs
AU2015295242B2 (en) 2014-07-31 2020-10-22 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
NZ730296A (en) 2014-08-20 2023-09-29 Alnylam Pharmaceuticals Inc Modified double-stranded rna agents
WO2016040589A1 (en) 2014-09-12 2016-03-17 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
US20170304459A1 (en) 2014-10-10 2017-10-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhalation delivery of conjugated oligonucleotide
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2016130806A2 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
MX2017012610A (en) 2015-04-08 2018-03-16 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the lect2 gene.
MX2017013348A (en) 2015-04-17 2018-08-01 Amgen Res Munich Gmbh Bispecific antibody constructs for cdh3 and cd3.
AU2016257996A1 (en) 2015-05-06 2017-10-26 Alnylam Pharmaceuticals, Inc. Factor XII (Hageman Factor) (F12), Kallikrein B, Plasma (Fletcher Factor) 1 (KLKB1), and Kininogen 1 (KNG1) iRNA compositions and methods of use thereof
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
TW202346349A (en) 2015-07-31 2023-12-01 德商安美基研究(慕尼黑)公司 Antibody constructs for dll3 and cd3
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
TWI717375B (en) 2015-07-31 2021-02-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cd70 and cd3
JP7028764B2 (en) 2015-09-02 2022-03-02 アルナイラム ファーマシューティカルズ, インコーポレイテッド Programmed cell death 1 Ligand 1 (PD-L1) iRNA composition and its usage
EP3350328A1 (en) 2015-09-14 2018-07-25 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
WO2017100542A1 (en) 2015-12-10 2017-06-15 Alnylam Pharmaceuticals, Inc. Sterol regulatory element binding protein (srebp) chaperone (scap) irna compositions and methods of use thereof
PL3411402T3 (en) 2016-02-03 2022-02-28 Amgen Research (Munich) Gmbh Bcma and cd3 bispecific t cell engaging antibody constructs
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
AU2017216237B2 (en) 2016-02-03 2024-04-04 Amgen Research (Munich) Gmbh PSMA and CD3 bispecific T cell engaging antibody constructs
KR102413037B1 (en) 2016-03-15 2022-06-23 메르사나 테라퓨틱스, 인코포레이티드 NAPI2B Targeted Antibody-Drug Conjugates and Methods of Using Same
AU2017233121B2 (en) 2016-03-15 2023-12-21 Itabmed (Hk) Limited Multispecific Fab fusion proteins and use thereof
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
JOP20170091B1 (en) 2016-04-19 2021-08-17 Amgen Res Munich Gmbh Administration of a bispecific construct binding to CD33 and CD3 for use in a method for the treatment of myeloid leukemia
JP2019518028A (en) 2016-06-10 2019-06-27 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Complement component C5i RNA composition and its use for treating paroxysmal nocturnal hemoglobinuria (PNH)
WO2018049261A1 (en) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus expressing immune checkpoint modulators
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
TWI790217B (en) 2016-12-16 2023-01-21 美商阿尼拉製藥公司 METHODS FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES USING TRANSTHYRETIN (TTR) iRNA COMPOSITIONS
JOP20190189A1 (en) 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
BR112019021852A2 (en) 2017-04-18 2020-06-02 Alnylam Pharmaceuticals, Inc. RNAI AGENT AND A VACCINE AGAINST HBV, USE OR METHOD AND KIT FOR TREATMENT
AR111773A1 (en) 2017-05-05 2019-08-21 Amgen Inc PHARMACEUTICAL COMPOSITION THAT INCLUDES CONSTRUCTS OF BISPECTIFIC ANTIBODIES FOR STORAGE AND ADMINISTRATION
EP3704252A1 (en) 2017-11-01 2020-09-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
CA3082507A1 (en) 2017-12-11 2019-06-20 Amgen Inc. Continuous manufacturing process for bispecific antibody products
EP3728281A1 (en) 2017-12-21 2020-10-28 Alnylam Pharmaceuticals Inc. Chirally-enriched double-stranded rna agents
UY38041A (en) 2017-12-29 2019-06-28 Amgen Inc CONSTRUCTION OF BIESPECFIC ANTIBODY DIRECTED TO MUC17 AND CD3
MA53325A (en) 2018-07-30 2021-06-09 Amgen Inc PROLONGED ADMINISTRATION OF CD33 AND CD3-BINDING BISPECIFIC ANTIBODY CONSTRUCTION
JOP20210022A1 (en) 2018-08-03 2021-01-28 Amgen Inc Antibody constructs for cldn18.2 and cd3
FI3843757T3 (en) 2018-08-27 2024-06-06 Affimed Gmbh Cryopreserved nk cells preloaded with an antibody construct
JP2022500003A (en) 2018-09-18 2022-01-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA composition and its usage
EP3863670A1 (en) 2018-10-11 2021-08-18 Amgen Inc. Downstream processing of bispecific antibody constructs
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
US20220056444A1 (en) 2018-12-05 2022-02-24 Empirico Inc. Process to inhibit or eliminate eosinophilic diseases of the airway and related conditions
US20220056455A1 (en) 2018-12-20 2022-02-24 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of kcnt1 related disorders
WO2020150265A1 (en) 2019-01-15 2020-07-23 Empirico Inc. Prodrugs of alox-15 inhibitors and methods of using the same
AU2020279101A1 (en) 2019-05-17 2021-11-18 Alnylam Pharmaceuticals, Inc. Oral delivery of oligonucleotides
MX2021014644A (en) 2019-06-13 2022-04-06 Amgen Inc Automated biomass-based perfusion control in the manufacturing of biologics.
AU2020323901A1 (en) 2019-07-26 2022-02-24 Amgen Inc. Anti-IL13 antigen binding proteins
EP4013870A1 (en) 2019-08-13 2022-06-22 Alnylam Pharmaceuticals, Inc. Small ribosomal protein subunit 25 (rps25) irna agent compositions and methods of use thereof
WO2021050640A1 (en) 2019-09-10 2021-03-18 Amgen Inc. Purification method for bispecific antigen-binding polypeptides with enhanced protein l capture dynamic binding capacity
JP2023500661A (en) 2019-11-01 2023-01-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
CA3156683A1 (en) 2019-11-13 2021-05-20 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
WO2021102373A1 (en) 2019-11-22 2021-05-27 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
IL293824A (en) 2019-12-13 2022-08-01 Alnylam Pharmaceuticals Inc Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
CA3164129A1 (en) 2019-12-20 2021-06-24 Amgen Inc. Mesothelin-targeted cd40 agonistic multispecific antibody constructs for the treatment of solid tumors
AU2020414409A1 (en) 2019-12-27 2022-06-16 Affimed Gmbh Method for the production of bispecific FcyRIIl x CD30 antibody construct
US20230093169A1 (en) 2020-01-22 2023-03-23 Amgen Research (Munch) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021178607A1 (en) 2020-03-05 2021-09-10 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
WO2021183861A1 (en) 2020-03-12 2021-09-16 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
IL296601A (en) 2020-03-19 2022-11-01 Amgen Inc Antibodies against mucin 17 and uses thereof
JP2023519274A (en) 2020-03-26 2023-05-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド CORONAVIRUS iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
EP4133076A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Angiotensin-converting enzyme 2 (ace2) irna compositions and methods of use thereof
WO2021222065A1 (en) 2020-04-27 2021-11-04 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
WO2021231691A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rsi)
WO2021231698A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
EP4150076A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
EP4150090A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
WO2021231685A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150088A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
CA3183756A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
TW202210101A (en) 2020-05-29 2022-03-16 美商安進公司 Adverse effects-mitigating administration of a bispecific construct binding to cd33 and cd3
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
WO2022011262A1 (en) 2020-07-10 2022-01-13 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and compositions for treating epilepsy
WO2022066847A1 (en) 2020-09-24 2022-03-31 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
WO2022076291A1 (en) 2020-10-05 2022-04-14 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
EP4225792A1 (en) 2020-10-08 2023-08-16 Affimed GmbH Trispecific binders
AU2021365822A1 (en) 2020-10-21 2023-06-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
AR124019A1 (en) 2020-11-06 2023-02-01 Amgen Res Munich Gmbh POLYPEPTIDE CONSTRUCTIONS THAT BIND CD3
CN116323671A (en) 2020-11-06 2023-06-23 安进公司 Multi-targeting bispecific antigen binding molecules with increased selectivity
PE20231516A1 (en) 2020-11-06 2023-09-28 Amgen Res Munich Gmbh POLYPEPTIDE CONSTRUCTIONS THAT SELECTIVELY BIND CLDN6 AND CD3
KR20230098335A (en) 2020-11-06 2023-07-03 암젠 인크 Antigen binding domains with reduced clipping ratio
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
JP2024508714A (en) 2021-02-12 2024-02-28 アルナイラム ファーマシューティカルズ, インコーポレイテッド Superoxide dismutase 1 (SOD1) iRNA composition and method for using the same for treating or preventing superoxide dismutase 1- (SOD1-) related neurodegenerative diseases
WO2022182864A1 (en) 2021-02-25 2022-09-01 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
IL307239A (en) 2021-03-29 2023-11-01 Alnylam Pharmaceuticals Inc Huntingtin (htt) irna agent compositions and methods of use thereof
AR125290A1 (en) 2021-04-02 2023-07-05 Amgen Inc MAGEB2 JOINING CONSTRUCTIONS
JP2024519293A (en) 2021-04-29 2024-05-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド Signal Transducer and Activator of Transcription 6 (STAT6) iRNA Compositions and Methods of Use Thereof
MX2023012931A (en) 2021-05-06 2023-11-13 Amgen Res Munich Gmbh Cd20 and cd22 targeting antigen-binding molecules for use in proliferative diseases.
EP4341401A1 (en) 2021-05-18 2024-03-27 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
EP4341405A1 (en) 2021-05-20 2024-03-27 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
MX2023014168A (en) 2021-06-04 2024-02-27 Alnylam Pharmaceuticals Inc HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF.
EP4363574A1 (en) 2021-06-29 2024-05-08 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023003995A1 (en) 2021-07-23 2023-01-26 Alnylam Pharmaceuticals, Inc. Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
AU2022370009A1 (en) 2021-10-22 2024-05-16 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023076450A2 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
AU2022382368A1 (en) 2021-11-03 2024-05-02 Affimed Gmbh Bispecific cd16a binders
WO2023079493A1 (en) 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
AU2022421244A1 (en) 2021-12-22 2024-07-11 Camp4 Therapeutics Corporation Modulation of gene transcription using antisense oligonucleotides targeting regulatory rnas
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023218027A1 (en) 2022-05-12 2023-11-16 Amgen Research (Munich) Gmbh Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2023240277A2 (en) 2022-06-10 2023-12-14 Camp4 Therapeutics Corporation Methods of modulating progranulin expression using antisense oligonucleotides targeting regulatory rnas
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
WO2024119145A1 (en) 2022-12-01 2024-06-06 Camp4 Therapeutics Corporation Modulation of syngap1 gene transcription using antisense oligonucleotides targeting regulatory rnas

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4873191A (en) * 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4736866B1 (en) * 1984-06-22 1988-04-12 Transgenic non-human mammals
US5175385A (en) * 1987-09-03 1992-12-29 Ohio University/Edison Animal Biotechnolgy Center Virus-resistant transgenic mice
US5175384A (en) * 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice

Also Published As

Publication number Publication date
JPH07508410A (en) 1995-09-21
WO1994000569A1 (en) 1994-01-06
CA2135313A1 (en) 1994-01-06
AU4541093A (en) 1994-01-24
EP0648265A1 (en) 1995-04-19
EP0648265A4 (en) 1996-12-04

Similar Documents

Publication Publication Date Title
US5981175A (en) Methods for producing recombinant mammalian cells harboring a yeast artificial chromosome
NZ253943A (en) Transfering polynucleotides into eukaryotic cells using co-lipofection complexes of a cationic lipid and the polynucleotide
Choi et al. Transgenic mice containing a human heavy chain immunoglobulin gene fragment cloned in a yeast artificial chromosome
US5643763A (en) Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
US5612205A (en) Homologous recombination in mammalian cells
Tang et al. A Cre/loxP‐deleter transgenic line in mouse strain 129S1/SvImJ
JP3732407B2 (en) Chromosome modification method
US6461864B1 (en) Methods and vector constructs for making non-human animals which ubiquitously express a heterologous gene
WO1993016177A1 (en) Homogenotization of gene-targeting events
US6090554A (en) Efficient construction of gene targeting vectors
Huxley Exploring gene function: use of yeast artificial chromosome transgenesis
WO1996034097A1 (en) GENE-TARGETED NON-HUMAN MAMMALS DEFICIENT IN SOD-1 GENE AND EXPRESSING HUMANIZED Aβ SEQUENCE WITH SWEDISH FAD MUTATION
US5777194A (en) Gene-targeted mice with humanized Aβ sequence and Swedish FAD mutation
Camus et al. Unexpected behavior of a gene trap vector comprising a fusion between the Sh ble and the lacZ genes
WO1999060108A2 (en) Transgenic animals produced by homologous sequence targeting
JP4102435B2 (en) How to promote enzyme diversification
US20090007283A1 (en) Transgenic Rodents Selectively Expressing Human B1 Bradykinin Receptor Protein
JP2001231403A (en) Nonhuman animal retaining varied alien chromosome or its fragment
WO2005080574A1 (en) Transgenic mouse model and method for evaluating glucocerebrosidase deficiencies
Tucker Creating targeted mutations in mouse embryonic stem cells using yeast artificial chromosomes
US20050235370A1 (en) Spatiotemporally controlled adult somatic mutagenesis system
WO2001013716A1 (en) α-TOCOPHEROL TRANSPORT PROTEIN KNOCKOUT ANIMAL