WO1991006667A1 - Production of proteins using homologous recombination - Google Patents

Production of proteins using homologous recombination Download PDF

Info

Publication number
WO1991006667A1
WO1991006667A1 PCT/US1990/006436 US9006436W WO9106667A1 WO 1991006667 A1 WO1991006667 A1 WO 1991006667A1 US 9006436 W US9006436 W US 9006436W WO 9106667 A1 WO9106667 A1 WO 9106667A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
cells
amplifiable
region
primary
Prior art date
Application number
PCT/US1990/006436
Other languages
French (fr)
Inventor
Arthur I. Skoultchi
Original Assignee
Cell Genesys, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23714629&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1991006667(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Cell Genesys, Inc. filed Critical Cell Genesys, Inc.
Priority to DE69027526T priority Critical patent/DE69027526T3/en
Priority to CA002045175A priority patent/CA2045175C/en
Priority to EP91900640A priority patent/EP0452484B2/en
Publication of WO1991006667A1 publication Critical patent/WO1991006667A1/en
Priority to NO912642A priority patent/NO309238B1/en
Priority to GR960402466T priority patent/GR3021105T3/en
Priority to US10/787,959 priority patent/US20040265860A1/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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/59Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g. HCG; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • 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
    • C12N2795/00Bacteriophages
    • C12N2795/00011Details
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/20Pseudochromosomes, minichrosomosomes
    • C12N2800/206Pseudochromosomes, minichrosomosomes of yeast origin, e.g. YAC, 2u
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/44Vectors comprising a special translation-regulating system being a specific part of the splice mechanism, e.g. donor, acceptor

Definitions

  • the field of this invention is the expression of mammalian proteins.
  • the process of obtaining coding sequences and eliciting their expression has been a long and arduous one.
  • the identification of the coding sequence has frequently involved the construction of libraries, identification of fragments of the open reading frame, examining the flanking sequences, and the like.
  • the coding region has been only small fraction of the total nucleic acid associated with the gene.
  • pseudogenes or multi-membered gene families have obscured the ability to isolate a particular gene of interest. Nevertheless, as techniques have improved, there has been a continuous parade of successful identifications and isolation of genes of interest.
  • Expression of mammalian proteins of interest is achieved by employing homologous recombination for integration of an amplifiable gene and other regulatory sequences in proximity to a gene of interest without interruption of the production of a proper transcript.
  • the region comprising the amplifiable gene and the gene of interest may be amplified, the genome fragmented and directly -or indirectly transferred to an expression host for expression of the target protein. If not previously amplified, the target region is then amplified, and the cell population screened for cells producing the target protein. Cells which produce the target protein at high and stable levels are expanded and used for expression of the target protein.
  • FIG. 1 is a diagrammatic illustration of the plasmid pCG.l showing the sequence of the modified polylinker
  • FIG. 2 is a diagrammatic illustration of the construction of the plasmid pCG.HRl;
  • FIG. 3 is a diagrammatic illustration of the result of targeting the EPO locus by homologous recombination with the DNA from pCG.HRl cut with NotI.
  • FIG. 4 is a diagrammatic illustration of the PCR amplication fragment produced from cells in which a homologous recombination event has occurred.
  • Methods and compositions are provided for production of mammalian proteins of interest in culture.
  • the method employs homologous recombination in a host cell for integrating an amplifiable gene in the vicinity of a target gene, which target gene encodes the protein of interest.
  • the region comprising both the amplifiable gene and target gene will be referred to as the amplifiable region.
  • the resulting transformed primary cells may now be subjected to conditions which select for amplification, or the amplification may be performed subsequently.
  • Transform includes transform, transfeet, transduce, conjugation, fusion, electroporation or any other technique for introducing DNA into a viable cell.
  • the chromosomes or DNA of the transformed cells are then used to transfer the amplifiable region into the genome of secondary expression host cells, where the target region, if not previously amplified sufficiently or at all, is further amplified.
  • the resulting cell lines are screened for production of the target protein and secondary cell lines selected for desired levels of production, which cells may be expanded and used for production of the desired protein in culture.
  • the primary cell may be any mammalian cell of interest, particularly mammalian cells which do not grow readily in culture, more particularly primate cells, especially human cells, where the human cells may be normal cells, including embryonic or neoplastic cells, particularly normal cells.
  • fibroblasts particularly diploid skin fibroblasts, lymphocytes, epithelial cells, neurons, endothelial cells, or other somatic cells, or germ cells.
  • skin fibroblasts which can be readily propagated to provide for large numbers of normal cells, embryonic kidney cells, and the like. These cells may or may not be expressing the gene of interest. In those instances where the target gene is inducible or only expressed in certain differentiated cells, one may select cells in which the target gene is expressed, which may require immortalized cells capable of growth in culture.
  • Amplifiable genes include dihydrofolate reductase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc.
  • the amplifiable gene will have transcriptional signals which are functional in the secondary or expression host and desirably be functional in the primary host, particularly where amplification is employed in the primary host or the amplifiable gene is used as a marker.
  • the target genes may be any gene of interest, there already having been a large number of proteins of interest identified and isolated with continual additions to the list.
  • Proteins of interest include cytokines, such as interleukins 1-10; growth factors such as EGF, FGF, PDGF, and TGF; somatotropins; growth hormones; colony stimulating factors, such as G-, M-, and GM-CSF; erythropoietin; plasminogen activators, such as tissue and urine; enzymes, such as superoxide dismutase; interferons; T-cell receptors; surface membrane proteins; insulin; lipoproteins; ⁇ i-antitrypsin; CD proteins, such as CD3, 4, 8, 19; clotting factors, e.g., Factor VIIIc and von Willebrands factor; anticlotting factors, such as Protein C; atrial naturetic factor, tumor necrosis factor; transport proteins; homing receptors; addressins; regulatory proteins; etc.
  • cytokines such
  • constructs will be prepared where the amplifiable gene will be flanked on one or both sides with DNA homologous with the DNA of the target region.
  • the homologous DNA will generally be within 100 kb, usually 50 kb, preferably about 25 kb, of the transcribed region of the target gene, more preferably within 2 kb of the target gene.
  • gene is intended the coding region and those sequences required for transcription of a mature mRNA.
  • the homologous DNA may include the 5'-upstream region comprising any enhancer sequences, transcriptional initiation sequences, the region 5' of these sequences, or the like.
  • the homologous region may include a portion of the coding region, where the coding region may be comprised only of an open reading frame or combination of exons and introns.
  • the homologous region may comprise all or a portion of an intron, where all or a portion of one or more exons may also be present.
  • the homologous region may comprise the 3'-region, so as to comprise all or a portion of the transcription termination region, or the region 3' of this region.
  • the homologous regions may extend over all or a portion of the target gene or be outside the target gene comprising all or a portion of the transcriptional regulatory regions and/or the structural gene. For the most part, the homologous sequence will be joined to the amplifiable gene, proximally or distally.
  • a'sequence other than the wild-type sequence normally associated with the target gene will be used to separate the homologous sequence from the amplifiable gene on at least one side of the amplifiable gene. Some portion of the sequence may be the 5' or 3' sequence associated with the amplifiable gene, as a result of the manipulations associated with the amplifiable gene.
  • the homologous regions flanking the amplifiable gene need not be identical to the target region, where in vitro mutagenesis is desired. For example, one may wish to change the transcriptional initiation region for the target gene, so that a portion of the homologous region might comprise nucleotides different from the wild-type 5' region of the target gene.
  • a transcriptional initiation region different from the wild-type initiation region between the wild-type initiation region and the structural gene.
  • a signal leader sequence would be introduced in proper reading frame with the target gene to provide for secretion of the target protein expression product.
  • one may wish to introduce an enhancer in relation to the transcriptional initiation region which can be provided by, for example, integration of the amplifiable gene associated with the enhancer in a region upstream from the transcriptional initiation regulatory region or in an intron or even downstream from the target gene.
  • flanking sequences will be at least about 150 bp, and may be 12 kb or more, usually not more than about 8 kb.
  • the size of the flanking regions will be determined by the size of the known sequence, the number of sequences in the genome which may have homology to the site for integration, whether mutagenesis is involved and the extent of separation of the regions for mutagenesis, the particular site for integration, or the like.
  • the integrating constructs may be prepared in accordance with conventional ways, where sequences may be synthesized, isolated from natural sources, manipulated, cloned, ligated, subjected to in vitro mutagenesis, primer repair, or the like. At various stages, the joined sequences may be cloned, and analyzed by restriction analysis, sequencing, or the like. Usually the construct will be carried on a cloning vector comprising a replication system functional in a prokaryotic host, e.g., E. coli, and a marker for selection, e.g., biocide resistance, complementation to an auxotrophic host, etc. Other functional sequences may also be present, such as polylinkers, for ease of introduction and excision of the construct or portions thereof, or the like. A large number of cloning vectors are available such as pBR322, the pUC series, etc.
  • the construct may then be used for homologous recombination in the primary cell target.
  • Various techniques may be employed for integrating the construct into the genome of the primary cell without being joined to a replication system functional in the primary host. See for example, U.S. Patent No. 4,319,216, as well as the references cited in the Relevant Literature section.
  • the construct may be inserted into an appropriate vector, usually having a viral replication system, such as SV40, bovine papilloma virus, adenovirus, or the like.
  • the linear DNA sequence vector may also have a selectable marker for identifying transfected cells.
  • Selectable markers include the neo gene, allowing for selection with G418, the herpes tk gene for selection with HAT medium, qpt gene with mycophenolic acid, complementation of an auxotrophic host, etc.
  • the vector may or may not be capable of stable maintenance in the host. Where the vector is capable of stable maintenance, the cells will be screened for homologous integration of the vector into the genome of the host, where various techniques for curing the cells may be employed. Where the vector is not capable of stable maintenance, for example, where a temperature sensitive replication system is employed, one may change the temperature from the permissive temperature to the non-permissive temperature, so that the cells may be cured of the vector.
  • the selectable marker may select for the presence of the construct by means of the selectable marker.
  • the selectable marker may select for the presence of the construct by the amplifiable gene.
  • the selective medium may include from about 0.01-0.25 /xM of methotrexate.
  • the DNA will be introduced into the primary cells.
  • Techniques which may be used include calcium phosphate/DNA co-precipitates, microinjection of DNA into the nucleus, electroporation, bacterial protoplast fusion with intact cells, transfection, polycations, e.g., polybrene, polyornithine, etc., or the like.
  • the DNA may be single or double stranded » DNA, linear or circular.
  • Keown et al. Methods in Enzymoloqy (1989), Keown et al. , Methods and Enzvmoloqy (1990) Vol. 185, pp.
  • Upstream and/or downstream from the target region construct may be a gene which provides for identification of whether a double crossover has occurred.
  • the herpes simplex virus thymidine kinase gene may be employed since the presence of the thymidine kinase gene may be detected by the use of nucleoside analogs, such as acyclovir or gancyclovir, for their cytotoxic effects on cells that contain a functional HSV-tk gene.
  • nucleoside analogs such as acyclovir or gancyclovir
  • the presence of the marker gene as evidenced by resistance to a biocide or growth in a medium which selects for the presence of the marker gene establishes the presence and integration of the target construct into the host genome. No further selection need be made at this time, since the selection will be made in the secondary expression host, where expression of the amplified target gene may be detected. " If one wishes, one can determine whether homologous recombination has occurred by employing PCR and sequencing the resulting amplified DNA sequences. If desired, amplification may be performed at this time by stressing the primary cells with the appropriate amplifying reagent, so that multi-copies of the target gene are obtained. Alternatively, amplification may await transfer to the secondary cell expression host.
  • High molecular weight DNA greater than about 20kb, preferably greater than about 50kb DNA or preferably metaphase chromosomes are prepared from the primary recipient cell strain having the appropriate integration of the amplification vector. Preparation and isolation techniques are described by Nelson and Housman, In Gene Transfer (ed. R. Kucherlapati) Plenum Press, 1986. The DNA may then be introduced in the same manner as described above into the secondary host expression cells, using the same or different techniques than employed for the primary cells.
  • Various mammalian expression hosts are available and may be employed. These hosts include CHO cells, monkey kidney cells, C127 mouse fibroblasts, 3T3 mouse cells, Vero cells, etc. Desirably the hosts will have a negative background for the amplifiable gene or a gene which is substantially less responsive to the amplifying agent.
  • the transformed cells are grown in selective medium containing about 0.01-0.5 ⁇ ethotrexate and, where another marker is present, e.g., the neo gene, the medium may contain from about 0.1-1 mg/ml G418.
  • the resistant colonies are isolated and may then be analyzed for the presence of the construct in juxtaposition to the target gene. This may be as a result of detection of expression of the target gene product, where there will normally be a negative background for the target gene product, use of PCR, Southern hybridization, or the like.
  • the cells containing the construct are then expanded and subjected to selection and amplification with media containing progressively higher concentrations of the amplifying reagent, for example, 0.5-200 ⁇ M. of methotrexate for the DHFR gene, and may be analyzed at each selection step for production of the target product.
  • Expansion will include at least duplication and may result in at least 5 copies, preferably 10 copies or more in a tandem relationship.
  • protein production will be increased at least 1.5 fold from expression from a single copy, usually at least 3 fold, preferably at least 5 fold.
  • the various clones may then be screened for optimum stable production of the target product and these clones may then be expanded and used commercially for production in culture. In this manner, high yields of a product may be obtained, without the necessity of isolating the message and doing the various manipulations associated with genetic engineering or isolating the genomic gene, where very large genes can be a major research and development effort.
  • DHFR dihydrofolate reductase
  • CHO Chinese hamster ovary
  • DUKX-B11 cells Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 21:4216-4220 (1980)
  • secondary recipient are propagated in alpha-medium supplemented with 10% dialyzed fetal bovine serum.
  • the amplification vector is constructed from pUC19 (Yanisch-Perron et al. , Gene 33:103-119 (1985)).
  • a 1.8 kb Haell fragment containing a hygromycin B phosphotransferase gene (hph) driven by the herpes simplex virus thymidine kinase (HSV tk) promoter is isolated from pHyg (Sugden et al. , Mol. Cell. Biol. 5_:410-413 (1985)) by digestion with Haell and gel electrophoresis. Synthetic adaptors are added onto this fragment to convert the Haell ends into HindiII ends and the resulting fragment is joined to pUC19 digested with HindiII.
  • the resulting plasmid pUCH contains the hygromycin cassette such that transcription of hph and beta-lactamase are in the opposite orientation.
  • a 1.3 kb Sail fragment containing a DHFR gene driven by SV40 transcriptional signals is isolated from pTND (Connors et al. , DNA 2:651-661 (1988)) by digestion with Sail and gel electrophoresis. This fragment is ligated to pUCH digested with Sail.
  • the resulting plasmid pUCD contains the DHFR cassette such that DHFR and are transcribed in the same direction.
  • a 1.76 kb BamHI fragment from the phage F15 (Floppyner Degen et al. , J. Biol. Chem.
  • t-PA human tissue plasminogen activator
  • the resulting plasmid pCG contains a unique Notl site in the t-PA fragment which allows the plasmid to be linearized prior to transformation of the primary human diploid fibroblasts in order to increase the frequency of homologous recombination (Kucherlapati et al. , Proc. Natl. Acad. Sci. USA 81:3153-3157 (1984)).
  • the plasmid pCG linearized with Notl is introduced into the primary recipients by electroporation employing DNA at lOnM.
  • the resulting cells are then grown in selective medium (EEMEM with 200 ⁇ q/ ⁇ al hygromycin B) .
  • Resistant colonies are isolated and analyzed by PCR (Kim and Smithies, Nucleic Acids Res. 3 ⁇ :8887-8903 (1988)) using as primers the sequences GCGGCCTCGGCCTCTGCATA and CATCTCCCCTCTGGAGTGGA to distinguish homologous integrants from random ones.
  • Amplification of cellular DNA by PCR using these two primers yields a fragment of 1.9 kb only when DNA from correctly targeted cells is present.
  • Cells comprising the DHFR gene integrated into the t-PA region are expanded and used as a source of genetic material for preparation of secondary recipients.
  • Metaphase chromosomes are prepared Nelson et al. , J. Mol. Appl. Genet. 2 ⁇ :563-577 (1984)) from recipients demonstrating homologous recombination with the DHFR and are then transformed in DHFR-deficient
  • CHO cells by calcium phosphate mediated gene transfer (Nelson et al., J. Mol. Appl. Genet. s563 - 577 (1984)). The cells are then grown in selective medium (alpha-medium containing 200 ⁇ q/ml hygromycin B) . Resistant colonies are isolated and analyzed for expression of human t-PA (Kaufman et al. , Mol. Cell. Biol. 5:1750-1759 (1985)). The cell clones are then grown in selective medium containing progressively higher concentrations of methotrexate (.02-80 /xM, with steps of 4-fold increases in concentration) .
  • the cells are harvested and the human t-PA is analyzed employing an ELISA assay with a monoclonal antibody specific for t-PA (Weidle and Buckel, Gene 53,:31-41 (1987)). Clones providing for high levels of expression of t-PA are stored for subsequent use.
  • a clone was obtained by screening a human placental DNA genomic library (Clontech) in EMBL 3- SP6/T7 using two 36 bp oligonucleotide probes 5'- CTGGGTTGCTGAGTTCCGCAAAGTAGCTGGGTCTGG-3' and 5'- CGGGGGTCGGGGCTGTTATCTGCATGTGTGCGTGCG-3' to the presumed promoter region of human erythropoietin. From this clone two subclones were created in pSP72
  • a plasmid pCG.l was constructed by replacement of the polylinker of pBluescript SK(-) (Stratagene) between the Sad and Kpnl sites with a synthetic double stranded 72 base pair DNA fragment (FIG. 1). Referring to FIG. 2, into pCG.l was cloned between the Hindlll and Xbal sites a 678 bp fragment containing the enhancer and promoter of the immediate early gene of human cytomegalovirus (CMV, Boshart et al (1985) Cell 41, 521-530) obtained by a PCR amplification of the plasmid pUCH.CMV (gift of M. Calos, Stanford U.) using the oligonucleotide primers 5'-
  • the 620 bp BstEII-Xbal fragment from the pTD.2 was joined by the use of a BstEII-Xbal adapter to pCG.CMV restricted with Xbal to create the plasmid pCG.CMV/EPO, in which the BstEII site of the EPO fragment is next to the promoter end of the CMV fragment.
  • pCG.CMV/EPO was cloned successively a 1.94 kb fragment encoding methotrexate resistance from the plasmid pSV2dhfr (Subramani et al (1981) Mol. Cell. Biol.
  • neo gene was obtained as an Xhol-Sall fragment and the dhfr gene was obtained by PCR amplification using the primers 5'-
  • GGACGCGTGGATCCAGACATGATAAGATA-3' and 5'- GGACGCGTCAGCTGTGGAATGTGTGTCAG-3' designed to add Mlul sites at the ends of the resultant fragment.
  • the neo and dhfr genes were cloned into the Xhol and Mlul sites respectively of pCG.CMV/EPO to give the plasmids pCG.CMV/EPO/DHFR and pCG.CMV/EPO/Neo/DHFR such that their transcription is in the same orientation as that of CMV.
  • the 5 kb BamHI-Hindlll fragment from pTD.l was added via Clal adapters at the Clal site of pCG.CMV/EPO/Neo/DHFR to give pCG.HRl.
  • the 5' 5kb EPO fragment is in the same orientation as that of the 620 bp BstEII-Xbal fragment with respect to the original lambda clone.
  • BamHI-HindiII EPO fragment, the dhfr and G418 markers, the CMV enhancer/promoter and the 620 bp BstEII-Xbal EPO fragment can be released from pCG.HRl as a Notl or SacII fragment.
  • This Notl fragment can be used for homologous recombination as it is designed to serve as an omega structure in recombination having 5 kb and 620 bp of homology to facilitate the event (FIG. 3).
  • the DNA was first cut with Notl, then extracted with phenol/chloroform and precipitated by the addition of ethanol before centrifugation. The resultant DNA pellet was resuspended at a concentration of 2 mg/ml in a volume (10 ⁇ l ) of 10 mM Tris-HCl, 1 mM EDTA (TE) .
  • Transformed primary human 293 embryonal kidney cells (ATCC CRL 1573) were cultured in Cellgro DMEM HI6 (Mediatech) supplemented with 10% calf serum, glutamine (2 mM) and penicillin
  • DNA 50 ⁇ q
  • Notl as described above
  • the mixture was electroporated at 960 ⁇ F and 260 V with a BioRad Gene Pulser and then iced again for 10 min before plating onto a 10 cm dish.
  • the cells from a 10 cm dish were split equally among 5 24-well plates in media containig G418 at 0.6 mg/ml (effective concentration) . Under these electroporation conditions, 4-10 colonies/well survive drug selection af er 2 weeks.
  • pools of clones from the electroporated 293 cells from 4 wells each (representing about 16 colonies) were generated by trypsinizing wells and using 90% of each well for the pool. The remaining 10% of each well was then reseeded back into the well. Genomic DNA was then prepared from each pool as follows. The cells in each pool were pelleted by centrifugation for 2 min.
  • Proteinase K (10 ⁇ l, 10 mg/ml) was then added, and the samples were incubated for 4 hr at 50°C before extractions by vigorous vortexing with phenol/chloroform (200 ⁇ l each), then with chloroform (400 ⁇ l ) , the addition of ethanol (800 ⁇ l ) , and centrifugation at 25°C for 10 min.
  • the DNA pellets were washed with 70% ethanol, dried and resuspended in TE (20 ⁇ l ) . An average of 40 ⁇ q of genomic DNA was obtained from each sample. Approximately 1 ⁇ q from each sample of genomic DNA was used for PCR analysis. The DNA in a volume (10 ⁇ l ) of TE was boiled for 10 min.
  • the reaction (50 ⁇ l ) contained 10 mM Tris-HCl (pH 9.0 at 25°C), 50 mM KC1, 1.5 mM MgCl 2 , 0.01% gelatin, 0.1% Triton X-100, 200 ⁇ U dNTPs, 1 M each of the primers 5'-AAGCAGAGCTCGTTTAGTGAACCG-3' and 5'- TGAGCGTGAGTTCTGTGGAATGTG-3' , and 1.5 U of Tag DNA polymerase (Promega) .
  • metaphase chromosomes are prepared from the recipients demonstrating homologous recombination with DHFR and transformed in DHFR deficient CHO cells. After isolating resistant colonies and analyzing for expression of EPO, the cell clones are grown in selective medium containing progressively higher concentrations of methotrexate (.02-80 ⁇ K) with steps of 4-fold increases in concentration. The cells are then harvested, cloned and screened for production of EPO. Clones providing for at least 2-fold enhancement of EPO production are isolated.
  • the subject method provides for a novel approach to expression of a wide variety of mammalian genes of interest.
  • the method is simple, only requires the knowledge of a sequence of about 300 bp or more in the region of a target gene, and one may then use substantially conventional techniques for transferring the amplifiable region to an expression host, and production of the desired product in high yield.

Abstract

Methods and compositions are provided for expression of mammalian genes in culture. An amplifiable gene is introduced by homologous recombination in juxtaposition to a target gene, the resulting combination of amplifiable gene and target gene transferred to a convenient host and the target gene amplified by means of the amplifiable gene. The resulting expression host may then be grown in culture with enhanced expression of the target gene.

Description

PRODUCTION OF PROTEINS USING HOMOLOGOUS RECOMBINATION
INTRODUCTION
Technical Field
The field of this invention is the expression of mammalian proteins.
Background
The discoveries of restriction enzymes, cloning, sequencing, reverse transcriptase, and monoclonal antibodies has resulted in extraordinary capabilities in isolating, identifying, and manipulating nucleic acid sequences. As a result of these capabilities, numerous genes and their transcriptional control elements have been identified and manipulated. The genes have been used for producing large amounts of a desired protein in heterologous hosts (bacterial and eukaryotic host cell systems) .
In many cases, the process of obtaining coding sequences and eliciting their expression has been a long and arduous one. The identification of the coding sequence, either cDNA or genomic DNA, has frequently involved the construction of libraries, identification of fragments of the open reading frame, examining the flanking sequences, and the like. In mammalian genes where introns are frequently encountered, in many instances, the coding region has been only small fraction of the total nucleic acid associated with the gene. In other cases, pseudogenes or multi-membered gene families have obscured the ability to isolate a particular gene of interest. Nevertheless, as techniques have improved, there has been a continuous parade of successful identifications and isolation of genes of interest.
In many situations one is primarily interested in a source of the protein product. The cell type in the body which produces the protein is frequently an inadequate source, since the protein may be produced in low amounts, the protein may only be produced in a differentiated host cell which is only difficultly grown in culture, or the host cell, particularly a human cell, is not economic or efficient in a culture process for production of the product. There is, therefore, significant interest in developing alternative techniques for producing proteins of interest in culture with cells which provide for economic and efficient production of the desired protein and, when possible, appropriate processing of the protein product.
Relevant Literature Mansour et al. , Nature, 336;348-352 (1988), describe a general strategy for targeting mutations to non-selectable genes. Weidle et al. , Gene, 6:193- 203, (1988), describe amplification of tissue-type plasminogen activator with a DHFR gene and loss of amplification in the absence of selective pressure. Murnane and Yezzi, Somatic Cell and Molecular Genetics, 3^:273-286, (1988), describe transformation of a human cell line with an integrated selectable gene marker lacking a transcriptional promoter, with tandem duplication and amplification of the gene marker. Thomas and Capecchi, Cell, 51:503-512, (19871, describe site-directed mutagenesis by gene targeting in mouse embryo-derived stem cells. Song et al., Proc. Natl. Acad. Sci. USA, 84:6820-6824, (1987), describe homologous recombination in human cells by a two staged integration. Liskay et al., "Homologous Recombination Between Repeated Chromosomal Sequences in Mouse Cells," Cold Spring Harbor, Symp. Quant. Biol. 4_9_:13-189, (1984), describe integration of two different mutations of the same gene and homologous recombination between the mutant genes. Rubnitz and Subramani, Mol. and Cell. Biol. 4_:2253- 2258, (1984), describe the minimum amount of homology required for homologous recombination in mammalian cells. Kim and Smithies, Nucl. Acids. Res. 16:8887- 8903, (1988), describe an assay for homologous recombination using the polymerase chain reaction.
SUMMARY OF THE INVENTION
Expression of mammalian proteins of interest is achieved by employing homologous recombination for integration of an amplifiable gene and other regulatory sequences in proximity to a gene of interest without interruption of the production of a proper transcript. The region comprising the amplifiable gene and the gene of interest may be amplified, the genome fragmented and directly -or indirectly transferred to an expression host for expression of the target protein. If not previously amplified, the target region is then amplified, and the cell population screened for cells producing the target protein. Cells which produce the target protein at high and stable levels are expanded and used for expression of the target protein.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 is a diagrammatic illustration of the plasmid pCG.l showing the sequence of the modified polylinker;
FIG. 2 is a diagrammatic illustration of the construction of the plasmid pCG.HRl;
FIG. 3 is a diagrammatic illustration of the result of targeting the EPO locus by homologous recombination with the DNA from pCG.HRl cut with NotI.; FIG. 4 is a diagrammatic illustration of the PCR amplication fragment produced from cells in which a homologous recombination event has occurred.
DESCRIPTION OF SPECIFIC EMBODIMENTS
Methods and compositions are provided for production of mammalian proteins of interest in culture. The method employs homologous recombination in a host cell for integrating an amplifiable gene in the vicinity of a target gene, which target gene encodes the protein of interest. The region comprising both the amplifiable gene and target gene will be referred to as the amplifiable region. The resulting transformed primary cells may now be subjected to conditions which select for amplification, or the amplification may be performed subsequently. "Transform" includes transform, transfeet, transduce, conjugation, fusion, electroporation or any other technique for introducing DNA into a viable cell. The chromosomes or DNA of the transformed cells are then used to transfer the amplifiable region into the genome of secondary expression host cells, where the target region, if not previously amplified sufficiently or at all, is further amplified. The resulting cell lines are screened for production of the target protein and secondary cell lines selected for desired levels of production, which cells may be expanded and used for production of the desired protein in culture. The primary cell may be any mammalian cell of interest, particularly mammalian cells which do not grow readily in culture, more particularly primate cells, especially human cells, where the human cells may be normal cells, including embryonic or neoplastic cells, particularly normal cells. Various cell types may be employed as the primary cells, including fibroblasts, particularly diploid skin fibroblasts, lymphocytes, epithelial cells, neurons, endothelial cells, or other somatic cells, or germ cells. Of particular interest are skin fibroblasts, which can be readily propagated to provide for large numbers of normal cells, embryonic kidney cells, and the like. These cells may or may not be expressing the gene of interest. In those instances where the target gene is inducible or only expressed in certain differentiated cells, one may select cells in which the target gene is expressed, which may require immortalized cells capable of growth in culture.
A number of amplifiable genes exist, where by appropriate use of a selection agent, a gene integrated in the genome will be amplified with adjacent flanking DNA. Amplifiable genes include dihydrofolate reductase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc. The amplifiable gene will have transcriptional signals which are functional in the secondary or expression host and desirably be functional in the primary host, particularly where amplification is employed in the primary host or the amplifiable gene is used as a marker.
The target genes may be any gene of interest, there already having been a large number of proteins of interest identified and isolated with continual additions to the list. Proteins of interest include cytokines, such as interleukins 1-10; growth factors such as EGF, FGF, PDGF, and TGF; somatotropins; growth hormones; colony stimulating factors, such as G-, M-, and GM-CSF; erythropoietin; plasminogen activators, such as tissue and urine; enzymes, such as superoxide dismutase; interferons; T-cell receptors; surface membrane proteins; insulin; lipoproteins; αi-antitrypsin; CD proteins, such as CD3, 4, 8, 19; clotting factors, e.g., Factor VIIIc and von Willebrands factor; anticlotting factors, such as Protein C; atrial naturetic factor, tumor necrosis factor; transport proteins; homing receptors; addressins; regulatory proteins; etc.
For homologous recombination, constructs will be prepared where the amplifiable gene will be flanked on one or both sides with DNA homologous with the DNA of the target region. The homologous DNA will generally be within 100 kb, usually 50 kb, preferably about 25 kb, of the transcribed region of the target gene, more preferably within 2 kb of the target gene. By gene is intended the coding region and those sequences required for transcription of a mature mRNA. The homologous DNA may include the 5'-upstream region comprising any enhancer sequences, transcriptional initiation sequences, the region 5' of these sequences, or the like. The homologous region may include a portion of the coding region, where the coding region may be comprised only of an open reading frame or combination of exons and introns. The homologous region may comprise all or a portion of an intron, where all or a portion of one or more exons may also be present. Alternatively, the homologous region may comprise the 3'-region, so as to comprise all or a portion of the transcription termination region, or the region 3' of this region. The homologous regions may extend over all or a portion of the target gene or be outside the target gene comprising all or a portion of the transcriptional regulatory regions and/or the structural gene. For the most part, the homologous sequence will be joined to the amplifiable gene, proximally or distally.
Usually a'sequence other than the wild-type sequence normally associated with the target gene will be used to separate the homologous sequence from the amplifiable gene on at least one side of the amplifiable gene. Some portion of the sequence may be the 5' or 3' sequence associated with the amplifiable gene, as a result of the manipulations associated with the amplifiable gene. The homologous regions flanking the amplifiable gene need not be identical to the target region, where in vitro mutagenesis is desired. For example, one may wish to change the transcriptional initiation region for the target gene, so that a portion of the homologous region might comprise nucleotides different from the wild-type 5' region of the target gene. Alternatively, one could provide for insertion of a transcriptional initiation region different from the wild-type initiation region between the wild-type initiation region and the structural gene. Similarly, one might wish to introduce various mutations into the structural gene, so that the homologous region would comprise mismatches, resulting in a change in the encoded protein. For example, a signal leader sequence would be introduced in proper reading frame with the target gene to provide for secretion of the target protein expression product. Alternatively, one might change the 3' region, e.g., untranslated region, polyadenylation site, etc. of the target gene. Therefore, by homologous recombination, one can provide for maintaining the integrity of the target gene, so as to express the wild-type protein under the transcriptional regulation of the wild-type promoter or one may provide for a change in transcriptional regulation, processing or sequence of the target gene. In some instances, one may wish to introduce an enhancer in relation to the transcriptional initiation region, which can be provided by, for example, integration of the amplifiable gene associated with the enhancer in a region upstream from the transcriptional initiation regulatory region or in an intron or even downstream from the target gene.
In order to prepare the subject constructs,, it will be necessary to know the sequence which is targeted for homologous recombination. While it is reported that a sequence of 14 bases complementary to a sequence in a genome may provide for homologous recombination, normally the individual flanking sequences will be at least about 150 bp, and may be 12 kb or more, usually not more than about 8 kb. The size of the flanking regions will be determined by the size of the known sequence, the number of sequences in the genome which may have homology to the site for integration, whether mutagenesis is involved and the extent of separation of the regions for mutagenesis, the particular site for integration, or the like.
The integrating constructs may be prepared in accordance with conventional ways, where sequences may be synthesized, isolated from natural sources, manipulated, cloned, ligated, subjected to in vitro mutagenesis, primer repair, or the like. At various stages, the joined sequences may be cloned, and analyzed by restriction analysis, sequencing, or the like. Usually the construct will be carried on a cloning vector comprising a replication system functional in a prokaryotic host, e.g., E. coli, and a marker for selection, e.g., biocide resistance, complementation to an auxotrophic host, etc. Other functional sequences may also be present, such as polylinkers, for ease of introduction and excision of the construct or portions thereof, or the like. A large number of cloning vectors are available such as pBR322, the pUC series, etc.
Once the construct is prepared, it may then be used for homologous recombination in the primary cell target. Various techniques may be employed for integrating the construct into the genome of the primary cell without being joined to a replication system functional in the primary host. See for example, U.S. Patent No. 4,319,216, as well as the references cited in the Relevant Literature section. Alternatively, the construct may be inserted into an appropriate vector, usually having a viral replication system, such as SV40, bovine papilloma virus, adenovirus, or the like. The linear DNA sequence vector may also have a selectable marker for identifying transfected cells. Selectable markers include the neo gene, allowing for selection with G418, the herpes tk gene for selection with HAT medium, qpt gene with mycophenolic acid, complementation of an auxotrophic host, etc. The vector may or may not be capable of stable maintenance in the host. Where the vector is capable of stable maintenance, the cells will be screened for homologous integration of the vector into the genome of the host, where various techniques for curing the cells may be employed. Where the vector is not capable of stable maintenance, for example, where a temperature sensitive replication system is employed, one may change the temperature from the permissive temperature to the non-permissive temperature, so that the cells may be cured of the vector. In this case, only those cells having integration of the construct comprising the amplifiable gene and, when present, the selectable marker, will be able to survive selection. Where a selectable marker is present, one may select for the presence of the construct by means of the selectable marker. Where the selectable marker is not present, one may select for the presence of the construct by the amplifiable gene. For the neo gene or the herpes tk gene, one could employ a medium for growth of the transformants of about 0.1-1 g/ml of G418 or HAT medium respectively. Where DHFR is the amplifiable gene, the selective medium may include from about 0.01-0.25 /xM of methotrexate. In carrying out the homologous recombination, the DNA will be introduced into the primary cells. Techniques which may be used include calcium phosphate/DNA co-precipitates, microinjection of DNA into the nucleus, electroporation, bacterial protoplast fusion with intact cells, transfection, polycations, e.g., polybrene, polyornithine, etc., or the like. The DNA may be single or double stranded » DNA, linear or circular. For various techniques for transforming mammalian cells, see Keown et al. , Methods in Enzymoloqy (1989), Keown et al. , Methods and Enzvmoloqy (1990) Vol. 185, pp. 527-537 and Mansour et al. , Nature, 336:348-352, (1988). Upstream and/or downstream from the target region construct may be a gene which provides for identification of whether a double crossover has occurred. For this purpose, the herpes simplex virus thymidine kinase gene may be employed since the presence of the thymidine kinase gene may be detected by the use of nucleoside analogs, such as acyclovir or gancyclovir, for their cytotoxic effects on cells that contain a functional HSV-tk gene. The absence of sensitivity to these nucleoside analogs indicates the absence of the thymidine kinase and, therefore, where homologous recombination has occurred, that a double crossover event has also occurred.
The presence of the marker gene as evidenced by resistance to a biocide or growth in a medium which selects for the presence of the marker gene, establishes the presence and integration of the target construct into the host genome. No further selection need be made at this time, since the selection will be made in the secondary expression host, where expression of the amplified target gene may be detected." If one wishes, one can determine whether homologous recombination has occurred by employing PCR and sequencing the resulting amplified DNA sequences. If desired, amplification may be performed at this time by stressing the primary cells with the appropriate amplifying reagent, so that multi-copies of the target gene are obtained. Alternatively, amplification may await transfer to the secondary cell expression host.
High molecular weight DNA, greater than about 20kb, preferably greater than about 50kb DNA or preferably metaphase chromosomes are prepared from the primary recipient cell strain having the appropriate integration of the amplification vector. Preparation and isolation techniques are described by Nelson and Housman, In Gene Transfer (ed. R. Kucherlapati) Plenum Press, 1986. The DNA may then be introduced in the same manner as described above into the secondary host expression cells, using the same or different techniques than employed for the primary cells. Various mammalian expression hosts are available and may be employed. These hosts include CHO cells, monkey kidney cells, C127 mouse fibroblasts, 3T3 mouse cells, Vero cells, etc. Desirably the hosts will have a negative background for the amplifiable gene or a gene which is substantially less responsive to the amplifying agent.
The transformed cells are grown in selective medium containing about 0.01-0.5 μϋ ethotrexate and, where another marker is present, e.g., the neo gene, the medium may contain from about 0.1-1 mg/ml G418. The resistant colonies are isolated and may then be analyzed for the presence of the construct in juxtaposition to the target gene. This may be as a result of detection of expression of the target gene product, where there will normally be a negative background for the target gene product, use of PCR, Southern hybridization, or the like.
The cells containing the construct are then expanded and subjected to selection and amplification with media containing progressively higher concentrations of the amplifying reagent, for example, 0.5-200 μM. of methotrexate for the DHFR gene, and may be analyzed at each selection step for production of the target product. Expansion will include at least duplication and may result in at least 5 copies, preferably 10 copies or more in a tandem relationship. Thus protein production will be increased at least 1.5 fold from expression from a single copy, usually at least 3 fold, preferably at least 5 fold.
The various clones may then be screened for optimum stable production of the target product and these clones may then be expanded and used commercially for production in culture. In this manner, high yields of a product may be obtained, without the necessity of isolating the message and doing the various manipulations associated with genetic engineering or isolating the genomic gene, where very large genes can be a major research and development effort.
The following examples are offered by way of illustration and not by way of limitation.
EXPERIMENTAL
Cells
Normal human diploid skin fibroblasts, ("primary recipient") are propagated in EEMEM medium supplemented with 20% fetal calf serum. Dihydrofolate reductase (DHFR) deficient Chinese hamster ovary (CHO) DUKX-B11 cells (Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 21:4216-4220 (1980)) ( "secondary recipient") are propagated in alpha-medium supplemented with 10% dialyzed fetal bovine serum.
DNA Vector
The amplification vector is constructed from pUC19 (Yanisch-Perron et al. , Gene 33:103-119 (1985)). A 1.8 kb Haell fragment containing a hygromycin B phosphotransferase gene (hph) driven by the herpes simplex virus thymidine kinase (HSV tk) promoter is isolated from pHyg (Sugden et al. , Mol. Cell. Biol. 5_:410-413 (1985)) by digestion with Haell and gel electrophoresis. Synthetic adaptors are added onto this fragment to convert the Haell ends into HindiII ends and the resulting fragment is joined to pUC19 digested with HindiII. The resulting plasmid pUCH contains the hygromycin cassette such that transcription of hph and beta-lactamase are in the opposite orientation. A 1.3 kb Sail fragment containing a DHFR gene driven by SV40 transcriptional signals is isolated from pTND (Connors et al. , DNA 2:651-661 (1988)) by digestion with Sail and gel electrophoresis. This fragment is ligated to pUCH digested with Sail. The resulting plasmid pUCD contains the DHFR cassette such that DHFR and are transcribed in the same direction. A 1.76 kb BamHI fragment from the phage F15 (Friezner Degen et al. , J. Biol. Chem. 2 1:6972-6985 (1986)) which contains 1.45 kb of DNA flanking the transcriptional start of human tissue plasminogen activator (t-PA) in addition to the first exon and part of the first intron is isolated by gel electrophoresis after BamHI digestion. This fragment is joined to pUCD following digestion of the latter with BamHI. The resulting plasmid pUCG has the promoter of the t-PA fragment oriented opposite to that of the DHFR cassette. The t-PA fragment contains a single NeoI site, which is not unique to pUCG. A partial NeoI digest is carried out and a Notl linker is inserted. The resulting plasmid pCG contains a unique Notl site in the t-PA fragment which allows the plasmid to be linearized prior to transformation of the primary human diploid fibroblasts in order to increase the frequency of homologous recombination (Kucherlapati et al. , Proc. Natl. Acad. Sci. USA 81:3153-3157 (1984)).
Preparation of Primary Recipients
The plasmid pCG linearized with Notl is introduced into the primary recipients by electroporation employing DNA at lOnM. The resulting cells are then grown in selective medium (EEMEM with 200 μq/τal hygromycin B) . Resistant colonies are isolated and analyzed by PCR (Kim and Smithies, Nucleic Acids Res. 3^:8887-8903 (1988)) using as primers the sequences GCGGCCTCGGCCTCTGCATA and CATCTCCCCTCTGGAGTGGA to distinguish homologous integrants from random ones. Amplification of cellular DNA by PCR using these two primers yields a fragment of 1.9 kb only when DNA from correctly targeted cells is present. Cells comprising the DHFR gene integrated into the t-PA region are expanded and used as a source of genetic material for preparation of secondary recipients.
Preparation of Secondary Recipients
Metaphase chromosomes are prepared Nelson et al. , J. Mol. Appl. Genet. 2^:563-577 (1984)) from recipients demonstrating homologous recombination with the DHFR and are then transformed in DHFR-deficient
CHO cells by calcium phosphate mediated gene transfer (Nelson et al., J. Mol. Appl. Genet. s563-577 (1984)). The cells are then grown in selective medium (alpha-medium containing 200 μq/ml hygromycin B) . Resistant colonies are isolated and analyzed for expression of human t-PA (Kaufman et al. , Mol. Cell. Biol. 5:1750-1759 (1985)). The cell clones are then grown in selective medium containing progressively higher concentrations of methotrexate (.02-80 /xM, with steps of 4-fold increases in concentration) . After this amplification procedure, the cells are harvested and the human t-PA is analyzed employing an ELISA assay with a monoclonal antibody specific for t-PA (Weidle and Buckel, Gene 53,:31-41 (1987)). Clones providing for high levels of expression of t-PA are stored for subsequent use. Isolation of a Genomic Clone Containing Sequences for Targeting Erythropoietin A clone was obtained by screening a human placental DNA genomic library (Clontech) in EMBL 3- SP6/T7 using two 36 bp oligonucleotide probes 5'- CTGGGTTGCTGAGTTCCGCAAAGTAGCTGGGTCTGG-3' and 5'- CGGGGGTCGGGGCTGTTATCTGCATGTGTGCGTGCG-3' to the presumed promoter region of human erythropoietin. From this clone two subclones were created in pSP72
(Krieg and Melton (1987) Meth. Enzymol. 155, 397-415), one containing a 5 kb BamHI-HindiII fragment from the region upstream to the coding region of EPO (pTD.l) and one containing a 5 kb Hindlll-BamHI fragment coding for EPO (pTD.2).
Construction of DNA Fragment for
Targeting Erythropoietin A plasmid pCG.l was constructed by replacement of the polylinker of pBluescript SK(-) (Stratagene) between the Sad and Kpnl sites with a synthetic double stranded 72 base pair DNA fragment (FIG. 1). Referring to FIG. 2, into pCG.l was cloned between the Hindlll and Xbal sites a 678 bp fragment containing the enhancer and promoter of the immediate early gene of human cytomegalovirus (CMV, Boshart et al (1985) Cell 41, 521-530) obtained by a PCR amplification of the plasmid pUCH.CMV (gift of M. Calos, Stanford U.) using the oligonucleotide primers 5'-
CGCCAAGCTTGGCCATTGCATACGTT-3' and 5'-
GAGGTCTAGACGGTTCACTAAACGAGCTCT-3' in order to engineer Hindlll and Xbal sites respectively onto the ends of the resultant fragment. The resultant plasmid pCG.CMV was used for further constructions.
The 620 bp BstEII-Xbal fragment from the pTD.2 was joined by the use of a BstEII-Xbal adapter to pCG.CMV restricted with Xbal to create the plasmid pCG.CMV/EPO, in which the BstEII site of the EPO fragment is next to the promoter end of the CMV fragment. Into pCG.CMV/EPO was cloned successively a 1.94 kb fragment encoding methotrexate resistance from the plasmid pSV2dhfr (Subramani et al (1981) Mol. Cell. Biol. 1 , 854-864) and a 1.15 kb fragment encoding G418 resistance from the plasmid pMClneo polyA (Thomas and Capecchi (1987) Cell 5_1, 503-512). The neo gene was obtained as an Xhol-Sall fragment and the dhfr gene was obtained by PCR amplification using the primers 5'-
GGACGCGTGGATCCAGACATGATAAGATA-3' and 5'- GGACGCGTCAGCTGTGGAATGTGTGTCAG-3' designed to add Mlul sites at the ends of the resultant fragment. The neo and dhfr genes were cloned into the Xhol and Mlul sites respectively of pCG.CMV/EPO to give the plasmids pCG.CMV/EPO/DHFR and pCG.CMV/EPO/Neo/DHFR such that their transcription is in the same orientation as that of CMV. Finally, the 5 kb BamHI-Hindlll fragment from pTD.l was added via Clal adapters at the Clal site of pCG.CMV/EPO/Neo/DHFR to give pCG.HRl. In pCG.HRl, the 5' 5kb EPO fragment is in the same orientation as that of the 620 bp BstEII-Xbal fragment with respect to the original lambda clone. A 9.54 kb fragment containing the 5' 5kb
BamHI-HindiII EPO fragment, the dhfr and G418 markers, the CMV enhancer/promoter and the 620 bp BstEII-Xbal EPO fragment can be released from pCG.HRl as a Notl or SacII fragment. This Notl fragment can be used for homologous recombination as it is designed to serve as an omega structure in recombination having 5 kb and 620 bp of homology to facilitate the event (FIG. 3). For electroporation, the DNA was first cut with Notl, then extracted with phenol/chloroform and precipitated by the addition of ethanol before centrifugation. The resultant DNA pellet was resuspended at a concentration of 2 mg/ml in a volume (10 μl ) of 10 mM Tris-HCl, 1 mM EDTA (TE) . Introduction of DNA into cells
Transformed primary human 293 embryonal kidney cells (ATCC CRL 1573) were cultured in Cellgro DMEM HI6 (Mediatech) supplemented with 10% calf serum, glutamine (2 mM) and penicillin
(100 U/ml)/streptomycin (0.1 mg/ml) and grown at 37°C in 5% CO,. At 90% confluency, cells were prepared for electroporation by trypsinization, concentration by
7 brief centrifugation and resuspension in PBS at 10 cells/0.8 ml. The cells were equilibrated at 4°C, and
DNA (50 μq) restricted with Notl (as described above) was added. The mixture was electroporated at 960 μF and 260 V with a BioRad Gene Pulser and then iced again for 10 min before plating onto a 10 cm dish.
After incubation at 37°C for 48 hr, the cells from a 10 cm dish were split equally among 5 24-well plates in media containig G418 at 0.6 mg/ml (effective concentration) . Under these electroporation conditions, 4-10 colonies/well survive drug selection af er 2 weeks.
Detection of Homologous Recombination by PCR Analysis Using Notl restricted DNA from pCG.HRl, successful homologous recombination is obtained by insertion of the 3.8 kb construct at the targeted EPO locus while simultaneously deleting 1.2 kb of genomic sequence (FIG. 3). PCR is used to detect unique targeting events versus random integration of the DNA as diagrammed in FIG. 4. Two primers are synthesized, one to the 3' end of CMV and the other to the region 3' to the Xbal site used for the 620 bp BstEII-Xbal fragment in the targeting DNA. A homologous recombination event generates a DNA target in the genome from which these primers produce an amplification product of 860 bp.
In order to detect the targeting event, pools of clones (from the electroporated 293 cells) from 4 wells each (representing about 16 colonies) were generated by trypsinizing wells and using 90% of each well for the pool. The remaining 10% of each well was then reseeded back into the well. Genomic DNA was then prepared from each pool as follows. The cells in each pool were pelleted by centrifugation for 2 min. in a 1.5 ml microcentrifuge tube, resuspended in PBS (20 μl ) , and treated for 1 hr at 37°C with a solution (400 μl) containing 10 mM Tris-HCl (pH7.5), 100 mM NaCl, 5 mM EDTA, 1% SDS and RNase A (40 μq/ml ) . Proteinase K (10 μl, 10 mg/ml) was then added, and the samples were incubated for 4 hr at 50°C before extractions by vigorous vortexing with phenol/chloroform (200 μl each), then with chloroform (400 μl ) , the addition of ethanol (800 μl ) , and centrifugation at 25°C for 10 min. The DNA pellets were washed with 70% ethanol, dried and resuspended in TE (20 μl ) . An average of 40 μq of genomic DNA was obtained from each sample. Approximately 1 μq from each sample of genomic DNA was used for PCR analysis. The DNA in a volume (10 μl ) of TE was boiled for 10 min. prior to the addition of PCR mix (40 μl ) . The reaction (50 μl ) contained 10 mM Tris-HCl (pH 9.0 at 25°C), 50 mM KC1, 1.5 mM MgCl2, 0.01% gelatin, 0.1% Triton X-100, 200 μU dNTPs, 1 M each of the primers 5'-AAGCAGAGCTCGTTTAGTGAACCG-3' and 5'- TGAGCGTGAGTTCTGTGGAATGTG-3' , and 1.5 U of Tag DNA polymerase (Promega) . Following an initial incubation of 94°C for 3 min, the samples were subjected to 45 cycles of'denaturation at 94°C for 1 min., annealing at 66°C for 1.5 min. and extension at 72°C for 2 min. At the end of the 45 cycles, the samples were incubated an additional 5 min. at 72°C. A portion (20 μl ) of each sample was analyzed on a 1% agarose gel run in TBE and stained with ethidium bromide. Out of the 90 pools analyzed from 3 electroporations, two samples were identified which exhibited the correct size fragment by ethidium bromide staining. The DNA from the PCR reaction was recovered and subjected to restriction mapping with Xbal. The correct amplification product should upon treatment with Xbal yield two fragments, 669bp and 191bp. The samples from the two pools both yield fragments of the correct sizes. In addition, the sample from pool 1 exhibits other bands in the uncut material.
Following the procedure described previously, metaphase chromosomes are prepared from the recipients demonstrating homologous recombination with DHFR and transformed in DHFR deficient CHO cells. After isolating resistant colonies and analyzing for expression of EPO, the cell clones are grown in selective medium containing progressively higher concentrations of methotrexate (.02-80 μK) with steps of 4-fold increases in concentration. The cells are then harvested, cloned and screened for production of EPO. Clones providing for at least 2-fold enhancement of EPO production are isolated.
It is evident from the above results, that the subject method provides for a novel approach to expression of a wide variety of mammalian genes of interest. The method is simple, only requires the knowledge of a sequence of about 300 bp or more in the region of a target gene, and one may then use substantially conventional techniques for transferring the amplifiable region to an expression host, and production of the desired product in high yield. All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A method for producing mammalian proteins comprising: growing mammalian secondary expression host cells comprising multiple copies of an amplifiable region comprising a target gene heterologous to said secondary expression host and expressing a protein of interest and an amplifiable gene, whereby said target gene is expressed and said protein is produced; wherein said secondary host expression cells are produced by the method comprising: transforming primary mammalian cells comprising said target gene with a construct comprising an amplifiable gene and at least one flanking region of a total of at least about 150 bp homologous with a DNA sequence at the locus of the coding region of said target gene to provide amplification of said target gene, wherein said amplifiable gene is at a site which does not interfere with the expression of said target gene, whereby said construct becomes homologously integrated into the genome of said primary cells to define an amplifiable region; selecting for primary cells comprising said construct by means of said amplifiable gene or other marker present in said construct; isolating DNA portions of said genome from said primary cells, wherein said portions are large enough to include all of said amplifiable region; transforming secondary expression host cells with said primary cell DNA portions and cloning said transformed secondary expression host cells to produce clones of said secondary expression host cells differing in said DNA portions present in said secondary expression host cells; selecting clones of said mammalian secondary expression host cells comprising said amplifiable region; and amplifying said amplifiable region by means of an amplifying agent, wherein said amplifying is prior to said isolating or after said selecting and prior to said growing.
2. A method according to Claim 1, wherein said amplifiable gene is a mammalian DHFR gene.
3. A method according to Claim 1, wherein said portions are metaphase chromosomes.
4. A method according to Claim 1, wherein said portions are restriction fragments.
5. A method according to Claim 1, wherein said primary cells are human cells.
6. A method according to Claim 5, wherein said human cells are fibroblast cells.
7. A method according to Claim 1, wherein said construct comprises a biocidal marker providing resistance to a biocide for said primary host cells.
8. A method for producing mammalian proteins comprising: transforming mammalian primary mammalian cells comprising said target gene with a construct comprising an amplifiable gene and at least one flanking region of at least about 150 bp homologous with a DNA sequence within 50 kb of the coding region of said target gene, wherein said amplifiable gene is at a site which does not interfere with the expression of said target gene, whereby said construct becomes homologously integrated into the genome of said primary cells to define an amplifiable region comprising said amplifiable gene and said target gene in said genome; selecting for primary cells comprising said construct by means of said amplifiable gene or other marker present in said construct; isolating DNA portions of said genome from said primary cells, wherein said portions are large enough to include all of said amplifiable region; transforming mammalian secondary expression host cells with said primary cell DNA portions, wherein said secondary expression host cells are of a different species from said primary host cells, and cloning said transformed secondary expression host cells to produce clones of said secondary expression host cells differing in said DNA portions present in said secondary expression host cells; selecting clones of said mammalian secondary expression host cells comprising said amplifiable region; amplifying said amplifiable region by means of an amplifying agent, wherein said amplifying is prior to said isolating or after said selecting; and groing said secondary expression host cells comprising multiple copies of said amplifiable region, whereby said target gene is expressed and said protein is produced.
9. A method according to Claim 8, wherein said amplifying is with said secondary expression host cells.
10. A method according to Claim 8, wherein said primary cells are human cells.
11. A method according to Claim 10, wherein said human cells are diploid fibroblast cells.
12. A method according to Claim 8, wherein said amplifiable gene is a mutated DHFR gene having a higher Km than the wild-type gene.
13. A method according to Claim 12, wherein said secondary host expression cell is DHFR deficient.
14. A method according to Claim 8, wherein said construct further comprises a marker gene separated from said amplifiable region by an homologous flanking region.
15. A human cell comprising an amplifiable gene at other than its wild-type site in the human genome and within the locus of a target gene expressing a protein to provide amplification of said target gene.
16. A human cell according to Claim 14, wherein said cell is a normal cell.
17. A human cell according to Claim 14, wherein said cell is a neoplastic cell.
18. A human cell according to Claim 14, wherein said amplifiable gene is a DHFR gene.
19. A mammalian cell other than a human cell for expression of mammalian proteins in culture comprising an amplifiable region comprising an amplifiable gene within lOkb of a human wild-type gene expressing a protein, wherein said two genes are separated by substantially solely human wild-type sequence associated with said target gene and the flanking sequence associated with the amplifiable gene.
20. A method for producing, cells for expression of a heterologous protein in culture, said method comprising: transforming mammalian primary cells comprising said target gene with a construct comprising an amplifiable gene and at least one flanking region of at least about 150bp homologous with a DNA sequence within lOkb of the coding region of said target gene, wherein said amplifiable gene is at a site which does not interfere with the expression of said target gene, whereby said construct becomes homologously integrated into the genome of said primary cells to define an amplifiable region comprising said amplifiable gene and said target gene in said genome; selecting for primary cells comprising said construct by means of said amplifiable gene or other marker present in said construct; isolating DNA portions of said genome from said primary cells, wherein said portions are large enough to include all of said amplifiable region; transforming mammalian secondary expression host cells with said primary cell DNA portions, wherein said secondary expression host cells are of a different species from said primary host cells, and cloning said transformed secondary expression host cells to produce clones of said secondary expression host cells differing in said DNA portions present in said secondary expression host cells; selecting clones of said mammalian secondary expression host cells comprising said amplifiable region; and amplifying said amplifiable region by means of an amplifying agent, wherein said amplifying is either prior to said isolating or after said selecting.
21. A method according to Claim 20, wherein said amplifying is with said secondary expression host cells.
22. A method according to Claim 20, wherein said primary cells are human cells.
23. A method according to Claim 22, wherein said human cells are diploid fibroblast cells.
24. A method according to Claim 20, wherein said amplifiable gene is a mutated DHFR gene having a higher Km than the wild-type gene.
25. A method according to Claim 24, wherein said secondary host expression cell is DHFR deficient.
PCT/US1990/006436 1989-11-06 1990-11-06 Production of proteins using homologous recombination WO1991006667A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
DE69027526T DE69027526T3 (en) 1989-11-06 1990-11-06 PREPARATION OF PROTEINS BY HOMOLOGOUS RECOMBINATION
CA002045175A CA2045175C (en) 1989-11-06 1990-11-06 Production of proteins using homologous recombination
EP91900640A EP0452484B2 (en) 1989-11-06 1990-11-06 Production of proteins using homologous recombination
NO912642A NO309238B1 (en) 1989-11-06 1991-07-05 Production of proteins using homologous recombination
GR960402466T GR3021105T3 (en) 1989-11-06 1996-09-19 Production of proteins using homologous recombination
US10/787,959 US20040265860A1 (en) 1990-11-06 2004-02-27 Production of proteins using homologous recombination

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US43206989A 1989-11-06 1989-11-06
US432,069 1989-11-06

Publications (1)

Publication Number Publication Date
WO1991006667A1 true WO1991006667A1 (en) 1991-05-16

Family

ID=23714629

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1990/006425 WO1991006666A1 (en) 1989-11-06 1990-11-06 Production of proteins using homologous recombination
PCT/US1990/006436 WO1991006667A1 (en) 1989-11-06 1990-11-06 Production of proteins using homologous recombination

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US1990/006425 WO1991006666A1 (en) 1989-11-06 1990-11-06 Production of proteins using homologous recombination

Country Status (14)

Country Link
US (3) US5578461A (en)
EP (3) EP0747485B1 (en)
JP (2) JPH04505104A (en)
KR (1) KR100233781B1 (en)
AT (2) ATE174058T1 (en)
AU (1) AU635844B2 (en)
CA (1) CA2045175C (en)
DE (2) DE69032809T2 (en)
DK (2) DK0452484T3 (en)
ES (2) ES2127458T3 (en)
GR (2) GR3021105T3 (en)
NO (1) NO309238B1 (en)
SG (2) SG122763A1 (en)
WO (2) WO1991006666A1 (en)

Cited By (159)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0584214A1 (en) * 1991-05-06 1994-03-02 Cell Genesys, Inc. Gene manipulation and expression using genomic elements
WO1995031560A1 (en) * 1994-05-13 1995-11-23 Transkaryotic Therapies, Inc. Dna construct for effecting homologous recombination and uses thereof
WO1996029411A1 (en) * 1995-03-17 1996-09-26 Transkaryotic Therapies, Inc. Protein production and delivery
EP0750044A2 (en) * 1991-11-05 1996-12-27 Transkaryotic Therapies, Inc. DNA constructs and transfection of cells therewith for homologous recombination
US5686263A (en) * 1991-05-31 1997-11-11 Genentech, Inc. Method for enhancing gene expression
US5733761A (en) * 1991-11-05 1998-03-31 Transkaryotic Therapies, Inc. Protein production and protein delivery
US5733753A (en) * 1992-12-22 1998-03-31 Novo Nordisk A/S Amplification of genomic DNA by site specific integration of a selectable marker construct
US5789215A (en) * 1991-08-20 1998-08-04 Genpharm International Gene targeting in animal cells using isogenic DNA constructs
US5925544A (en) * 1996-11-18 1999-07-20 Novo Nordisk A/S Method of homologous recombination followed by in vivo selection of DNA amplification
US6025157A (en) * 1997-02-18 2000-02-15 Genentech, Inc. Neurturin receptor
US6048729A (en) * 1987-05-01 2000-04-11 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6054288A (en) * 1991-11-05 2000-04-25 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6063630A (en) * 1991-11-05 2000-05-16 Transkaryotic Therapies, Inc. Targeted introduction of DNA into primary or secondary cells and their use for gene therapy
EP1054018A1 (en) 1999-05-18 2000-11-22 Target Quest B.V. Fab fragment libraries and methods for their use
US6294655B1 (en) 1998-04-03 2001-09-25 Hyseq, Inc. Anti-interleukin-1 receptor antagonist antibodies and uses thereof
US6337072B1 (en) 1998-04-03 2002-01-08 Hyseq, Inc. Interleukin-1 receptor antagonist and recombinant production thereof
US6372453B1 (en) 1997-02-18 2002-04-16 Genetech, Inc. Neurturin receptor
US6399350B1 (en) 1996-08-29 2002-06-04 The Regents Of The University Of California KUZ, a novel family of metalloproteases
WO2002049673A2 (en) 2000-12-20 2002-06-27 F. Hoffmann-La Roche Ag Conjugates of erythropoietin (pep) with polyethylene glycol (peg)
US6426191B1 (en) 1998-04-03 2002-07-30 Hyseq, Inc. Assays involving an IL-1 receptor antagonist
US6524818B1 (en) 1997-09-26 2003-02-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US6528313B1 (en) 1989-03-20 2003-03-04 Institut Pasteur Procedure for specific replacement of a copy of a gene present in the recipient genome by the integration of a gene different from that where the integration is made
US6531124B1 (en) 1992-07-10 2003-03-11 Transkaryotic Therapies, Inc. In vivo production and delivery of insulinotropin for gene therapy
WO2003020702A2 (en) 2001-08-31 2003-03-13 The Rockefeller University Phosphodiesterase activity and regulation of phosphodiesterase 1-b-mediated signaling in brain
US6541623B1 (en) 1998-04-03 2003-04-01 Hyseq, Inc. Interleukin—1 receptor antagonist and uses thereof
US6566089B1 (en) * 1996-09-04 2003-05-20 Tularik Inc. Cell-based drug screens for regulators of gene expression
US6610296B2 (en) 1995-10-26 2003-08-26 Genentech, Inc. Methods of enhancing cognitive function using an AL-1 neurotrophic factor immunoadhesin
US6670178B1 (en) 1992-07-10 2003-12-30 Transkaryotic Therapies, Inc. In Vivo production and delivery of insulinotropin for gene therapy
EP1375666A2 (en) * 1989-12-22 2004-01-02 Applied Research Systems ARS Holding N.V. Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
US6692737B1 (en) 1991-11-05 2004-02-17 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6740503B1 (en) 1997-09-26 2004-05-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US6803211B2 (en) 2000-08-25 2004-10-12 Pfizer Inc. Methods and compositions for diagnosing and treating disorders involving angiogenesis
WO2005042581A2 (en) 2003-11-01 2005-05-12 Biovation Ltd. Modified anti-cd52 antibody
EP1619250A1 (en) 1996-01-08 2006-01-25 Genentech, Inc. WSX receptor and ligands
US7005299B1 (en) 1993-04-21 2006-02-28 The University Of Edinburgh Expression of heterologous genes according to a targeted expression profile
WO2006028967A2 (en) 2004-09-02 2006-03-16 Yale University Regulation of oncogenes by micrornas
US7067484B1 (en) 1994-10-27 2006-06-27 Genentech, Inc. AL-1 neurotrophic factor treatments
EP1714661A2 (en) 2000-05-19 2006-10-25 The Center for Blood Research, INC. Methods for diagnosing and treating hemostatic disorders by modulating p-selectin activity
US7235360B2 (en) 1997-03-14 2007-06-26 Biogen Idec Inc. Method for integrating genes at specific sites in mammalian cells via homologous recombination and vectors for accomplishing the same
WO2007112005A2 (en) 2006-03-24 2007-10-04 Syntonix Pharmaceuticals, Inc. Pc5 as a factor ix propeptide processing enzyme
EP1925671A1 (en) 2001-06-06 2008-05-28 Bioriginal Food & Science Corp. Flax (Linum usitatissimum L.) seed-specific promoters
WO2008140449A1 (en) 2007-05-11 2008-11-20 Thomas Jefferson University Methods of treatment and prevention of neurodegenerative diseases and disorders
US7459435B2 (en) 2002-08-29 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
US7459436B2 (en) 2002-11-22 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
WO2009014835A2 (en) 2007-06-21 2009-01-29 Angelica Therapeutics, Inc. Modified toxins
US7510858B2 (en) 2004-08-02 2009-03-31 Janssen Pharmaceutica N.V. IRAK 1c splice variant and its use
WO2009042538A2 (en) 2007-09-24 2009-04-02 Cornell University Immunogenic proteins from genome-derived outer membrane of leptospira and compositions and methods based thereon
US7514072B1 (en) 1998-12-14 2009-04-07 Hannelore Ehrenreich Method for the treatment of cerebral ischaemia and use of erythropoietin or erythropoietin derivatives for the treatment of cerebral ischaemia
US7527962B2 (en) 2001-12-19 2009-05-05 Millenium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
EP1025253B2 (en) 1997-10-20 2009-07-29 Roche Diagnostics GmbH Positive-negative selection for homologous recombination
WO2009114185A2 (en) 2008-03-12 2009-09-17 The Rockefeller University Methods and compositions for translational profiling and molecular phenotyping
EP2112229A2 (en) 2002-11-25 2009-10-28 Sequenom, Inc. Methods for identifying risk of breast cancer and treatments thereof
US7645733B2 (en) 2003-09-29 2010-01-12 The Kenneth S. Warren Institute, Inc. Tissue protective cytokines for the treatment and prevention of sepsis and the formation of adhesions
EP2143796A2 (en) 2000-02-29 2010-01-13 Millennium Pharmaceuticals, Inc. 1983, 52881, 2398, 45449, 50289, and 52872, G protein-coupled receptors and uses therefor
WO2010036353A2 (en) 2008-09-25 2010-04-01 Suregene Llc Genetic markers for optimizing treatment for schizophrenia
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
EP2184359A1 (en) 2001-06-06 2010-05-12 Bioriginal Food & Science Corp. Flax (Linum usitatissimum L.) seed-specific promoters
EP2186407A1 (en) 2002-02-13 2010-05-19 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
US7723055B2 (en) 2002-11-21 2010-05-25 University Of Massachusetts Diagnosing and treating hematopoietic cancers
US7745391B2 (en) 2001-09-14 2010-06-29 Compugen Ltd. Human thrombospondin polypeptide
EP2218732A1 (en) 1997-11-28 2010-08-18 Merck Serono Biodevelopment Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
US7785588B2 (en) 1997-02-18 2010-08-31 Genentech, Inc. Anti-neurturin receptor-A antibody compositions comprising cytokines or neurotrophic factors
EP2236623A1 (en) 2006-06-05 2010-10-06 Cancer Care Ontario Assessment of risk for colorectal cancer
EP2243834A1 (en) 2007-03-05 2010-10-27 Cancer Care Ontario Assessment of risk for colorectal cancer
EP2281828A2 (en) 2000-12-29 2011-02-09 The Kenneth S. Warren Institute, Inc. Compositions comprising modified erythropoietin
US7893197B2 (en) 2004-08-25 2011-02-22 Janssen Pharmaceutica N.V. Relaxin-3 chimeric polypeptides and their preparation and use
EP2314673A1 (en) 2001-02-14 2011-04-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2316463A1 (en) 2001-02-14 2011-05-04 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
EP2319530A1 (en) 2006-01-27 2011-05-11 Hannelore Ehrenreich Method for treating and/or preventing multiple sclerosis, and use of erythropoietin in the production of a drug for intermittent treatment and/or intermittent prevention of multiple sclerosis
WO2011057027A2 (en) 2009-11-04 2011-05-12 Janssen Pharmaceutica Nv Method for treating heart failure with stresscopin-like peptides
EP2322558A1 (en) 1999-06-30 2011-05-18 Millennium Pharmaceuticals, Inc. Antibodies directed against glycoprotein VI and uses thereof
US7976836B2 (en) 2000-12-06 2011-07-12 Anthrogenesis Corporation Treatment of stroke using placental stem cells
US8014957B2 (en) 2005-12-15 2011-09-06 Fred Hutchinson Cancer Research Center Genes associated with progression and response in chronic myeloid leukemia and uses thereof
EP2365086A1 (en) 2005-02-09 2011-09-14 Bioriginal Food & Science Corporation Novel omega-3 fatty acid desaturase family members and uses thereof
US8021833B2 (en) 2003-02-12 2011-09-20 Functional Genetics, Inc. Method for reducing HIV viral budding by administering a VPS28-specfic antibody that disrupts Gag-TSG101-VPS28 binding interactions
US8057789B2 (en) 2002-02-13 2011-11-15 Anthrogenesis Corporation Placental stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
EP2390311A1 (en) 2002-04-12 2011-11-30 Celgene Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US8138317B2 (en) 2007-07-17 2012-03-20 Hoffmann-La Roche Inc. Purification of pegylated polypeptides
WO2012035037A1 (en) 2010-09-14 2012-03-22 F. Hoffmann-La Roche Ag Method for purifying pegylated erythropoietin
EP2441830A2 (en) 2005-11-03 2012-04-18 Momenta Pharmaceuticals, Inc. Heparan sulfate glycosaminoglycan lyase and uses thereof
WO2012072268A2 (en) 2010-12-02 2012-06-07 Fraunhofer Gesellschaft Zur Förderung Der Angewadten Forschung E.V. Tolerance induction or immunosupression to prevent in particular graft-versus-host-disease (gvhd) by short-term pre-incubation of transplanted cell suspensions, tissues or organs coated with ligands to cell surface molecules
WO2012075337A2 (en) 2010-12-01 2012-06-07 Spinal Modulation, Inc. Directed delivery of agents to neural anatomy
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
EP2514840A1 (en) 2008-01-02 2012-10-24 Suregene Llc Genetic markers of mental illness
WO2013003697A1 (en) 2011-06-30 2013-01-03 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
US8354384B2 (en) 2005-06-23 2013-01-15 Yale University Anti-aging micrornas
WO2013020044A1 (en) 2011-08-03 2013-02-07 The Charlotte-Mecklenburg Hospital Authority D/B/A Carolinas Medical Center Treatment of fibrosis using microrna-19b
EP2561887A2 (en) 2003-01-14 2013-02-27 Dana Farber Cancer Institute, Inc. Cancer therapy sensitizer
EP2581455A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
WO2013054200A2 (en) 2011-10-10 2013-04-18 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
WO2013067451A2 (en) 2011-11-04 2013-05-10 Population Diagnostics Inc. Methods and compositions for diagnosing, prognosing, and treating neurological conditions
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8530629B2 (en) 2009-01-30 2013-09-10 Ab Biosciences, Inc. Lowered affinity antibodies and uses therefor
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
EP2666854A1 (en) 2007-12-04 2013-11-27 Proteon Therapeutics, Inc. Recombinant elastase proteins and methods of manufacturing and use thereof
US8617535B2 (en) 2002-11-26 2013-12-31 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US8691217B2 (en) 2005-12-29 2014-04-08 Anthrogenesis Corporation Placental stem cell populations
US8795533B2 (en) 2007-07-17 2014-08-05 Hoffmann-La Roche Inc. Chromatographic methods
US8895256B2 (en) 2005-10-13 2014-11-25 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US8962247B2 (en) 2008-09-16 2015-02-24 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non invasive prenatal diagnoses
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9029383B2 (en) 2007-05-11 2015-05-12 The Trustees Of The University Of Pennsylvania Methods of treatment of skin ulcers
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
WO2015081290A1 (en) 2013-11-27 2015-06-04 Inis Biotech Llc Methods for modulating angiogenesis of cancers refractory to anti-vegf treatment
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9180165B2 (en) 2007-07-19 2015-11-10 Hannelore Ehrenreich Use of EPO receptor activation or stimulation for the improvement of the EDSS score in patients with multiple sclerosis
US9198938B2 (en) 2008-11-19 2015-12-01 Antrhogenesis Corporation Amnion derived adherent cells
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
US9216200B2 (en) 2007-09-28 2015-12-22 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
WO2016044271A2 (en) 2014-09-15 2016-03-24 Children's Medical Center Corporation Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
WO2016118832A1 (en) 2015-01-22 2016-07-28 University Of Massachusetts Cancer immunotherapy
US9494597B2 (en) 2012-04-02 2016-11-15 Ab Biosciences, Inc. Human control antibodies and uses therefor
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9605313B2 (en) 2012-03-02 2017-03-28 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
EP3153521A1 (en) 2010-03-26 2017-04-12 Trustees of Dartmouth College Vista regulatory t cell mediator protein, vista binding agents and use thereof
US9624290B2 (en) 2010-01-28 2017-04-18 Ab Biosciences, Inc. Lowered affinity antibodies and methods of making the same
EP3168232A1 (en) 2009-11-13 2017-05-17 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for the diagnosis, prognosis, monitoring, treatment and modulation of post-transplant lymphoproliferative disorders and hypoxia associated angiogenesis disorders using galectin-1
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
WO2018011381A1 (en) 2016-07-15 2018-01-18 F. Hoffmann-La Roche Ag Method for purifying pegylated erythropoietin
US9920361B2 (en) 2012-05-21 2018-03-20 Sequenom, Inc. Methods and compositions for analyzing nucleic acid
US9926593B2 (en) 2009-12-22 2018-03-27 Sequenom, Inc. Processes and kits for identifying aneuploidy
US9976180B2 (en) 2012-09-14 2018-05-22 Population Bio, Inc. Methods for detecting a genetic variation in subjects with parkinsonism
US10040838B2 (en) 2008-11-04 2018-08-07 Janssen Pharmaceutica Nv CRHR2 peptide agonists and uses thereof
US10059750B2 (en) 2013-03-15 2018-08-28 Angelica Therapeutics, Inc. Modified toxins
US10059997B2 (en) 2010-08-02 2018-08-28 Population Bio, Inc. Compositions and methods for discovery of causative mutations in genetic disorders
US10066001B2 (en) 2013-03-15 2018-09-04 Apotex Inc. Enhanced liquid formulation stability of erythropoietin alpha through purification processing
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US10210306B2 (en) 2006-05-03 2019-02-19 Population Bio, Inc. Evaluating genetic disorders
US10233495B2 (en) 2012-09-27 2019-03-19 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
US10240205B2 (en) 2017-02-03 2019-03-26 Population Bio, Inc. Methods for assessing risk of developing a viral disease using a genetic test
WO2019129877A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Process for providing pegylated protein composition
WO2019129878A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Process for providing pegylated protein composition
WO2019129876A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Process for providing pegylated protein composition
US10370455B2 (en) 2014-12-05 2019-08-06 Immunext, Inc. Identification of VSIG8 as the putative VISTA receptor (V-R) and use thereof to produce VISTA/VSIG8 agonists and antagonists
US10407724B2 (en) 2012-02-09 2019-09-10 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
US10522240B2 (en) 2006-05-03 2019-12-31 Population Bio, Inc. Evaluating genetic disorders
US10724096B2 (en) 2014-09-05 2020-07-28 Population Bio, Inc. Methods and compositions for inhibiting and treating neurological conditions
US10738358B2 (en) 2008-09-16 2020-08-11 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
US10961585B2 (en) 2018-08-08 2021-03-30 Pml Screening, Llc Methods for assessing risk of developing a viral of disease using a genetic test
US11009509B2 (en) 2015-06-24 2021-05-18 Janssen Pharmaceutica Nv Anti-VISTA antibodies and fragments
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11060145B2 (en) 2013-03-13 2021-07-13 Sequenom, Inc. Methods and compositions for identifying presence or absence of hypermethylation or hypomethylation locus
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
US11332791B2 (en) 2012-07-13 2022-05-17 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
US11365447B2 (en) 2014-03-13 2022-06-21 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
WO2022159414A1 (en) 2021-01-22 2022-07-28 University Of Rochester Erythropoietin for gastroinfestinal dysfunction
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
US11529416B2 (en) 2012-09-07 2022-12-20 Kings College London Vista modulators for diagnosis and treatment of cancer

Families Citing this family (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100233781B1 (en) * 1989-11-06 1999-12-01 스테펜 에이. 쉐어윈 Production of proteins using homologous recombination
US5272071A (en) * 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
GB9028062D0 (en) * 1990-12-24 1991-02-13 Agricultural & Food Res Production of transgenic animals
US20020108136A1 (en) * 1997-03-21 2002-08-08 Sri Transgenic animals produced by homologous sequence targeting
WO1993024642A1 (en) * 1992-06-04 1993-12-09 Exemplar Corporation Insertion of heterologous dna outside of known chromosomal genes
US6001968A (en) 1994-08-17 1999-12-14 The Rockefeller University OB polypeptides, modified forms and compositions
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US6350730B1 (en) 1994-08-17 2002-02-26 The Rockefeller University OB polypeptides and modified forms as modulators of body weight
US6471956B1 (en) 1994-08-17 2002-10-29 The Rockefeller University Ob polypeptides, modified forms and compositions thereto
US6048837A (en) * 1994-08-17 2000-04-11 The Rockefeller University OB polypeptides as modulators of body weight
US6124439A (en) * 1994-08-17 2000-09-26 The Rockefeller University OB polypeptide antibodies and method of making
US6124448A (en) * 1994-08-17 2000-09-26 The Rockfeller University Nucleic acid primers and probes for the mammalian OB gene
US7148004B1 (en) 1997-01-16 2006-12-12 The Rockefeller University Oligonucleotides of the OB-R isoforms and methods of diagnosing body weight
US7063958B1 (en) 1996-01-16 2006-06-20 The Rockefeller University Nucleic acids db, the receptor for leptin
US7084252B1 (en) 1996-01-16 2006-08-01 The Rockefeller University DB, the receptor for leptin
US7619079B2 (en) 1996-02-14 2009-11-17 The Rockefeller University Db, the receptor for leptin, nucleic acids encoding the receptor, and uses thereof
US5792210A (en) * 1996-06-10 1998-08-11 Environmental Behavior Modification Inc. Electrical tongue stimulator and method for addiction treatment
JP4436934B2 (en) * 1996-09-13 2010-03-24 ノボザイムス,インコーポレイティド Cells with DNA insertion mutations that produce altered amounts of polypeptides
JPH10117771A (en) * 1996-10-23 1998-05-12 Natl Food Res Inst Frozen dough resistant-practical bread yeast
EP1007732B1 (en) 1997-01-17 2006-07-26 Maxygen, Inc. EVOLUTION OF procaryotic WHOLE CELLS BY RECURSIVE SEQUENCE RECOMBINATION
US6326204B1 (en) * 1997-01-17 2001-12-04 Maxygen, Inc. Evolution of whole cells and organisms by recursive sequence recombination
US7148054B2 (en) * 1997-01-17 2006-12-12 Maxygen, Inc. Evolution of whole cells and organisms by recursive sequence recombination
US5948653A (en) 1997-03-21 1999-09-07 Pati; Sushma Sequence alterations using homologous recombination
US6172211B1 (en) 1997-07-11 2001-01-09 Boehringer Ingelheim International Gmbh Nucleic acid encoding tag7 polypeptide
KR100641969B1 (en) 1997-07-23 2006-11-06 로셰 디아그노스틱스 게엠베하 Production of erythropoietin by endogenous gene activation
US6548296B1 (en) 1997-07-23 2003-04-15 Roche Diagnostics Gmbh Methods for identifying human cell lines useful for endogenous gene activation, isolated human lines identified thereby, and uses thereof
ES2281133T3 (en) 1997-07-23 2007-09-16 Roche Diagnostics Gmbh IDENTIFICATION OF HUMAN CELLULAR LINES FOR THE PRODUCTION OF HUMAN PROTEINS THROUGH THE ENDOGENA ACTIVATION OF GENES.
KR100622203B1 (en) 1997-07-23 2006-09-07 로셰 디아그노스틱스 게엠베하 Identification of human cell lines for the production of human proteins by endogenous gene activation
CA2298015C (en) * 1997-07-23 2015-01-27 Roche Diagnostics Gmbh Production of erythropoietin by endogenous gene activation
AU750025B2 (en) * 1997-09-26 2002-07-11 Athersys, Inc. Expression of endogenous genes by non-homologous recombination of a vector construct with cellular DNA
AU757930B2 (en) 1997-12-01 2003-03-13 Roche Diagnostics Gmbh Optimization of cells for endogenous gene activation
JPH11243944A (en) * 1998-03-04 1999-09-14 Natl Food Res Inst Dryness-resistant practical bread yeast for baking
KR100795626B1 (en) 1998-07-21 2008-01-17 코브라 바이오매뉴팩쳐링 피엘씨. A polynucleotide comprising a ubiquitous chromatin opening element UCOE
AU1447600A (en) * 1998-10-22 2000-05-08 Thomas J. Gardella Bioactive peptides and peptide derivatives of parathyroid hormone (pth) and parathyroid hormone-related peptide (pthrp)
US6773911B1 (en) 1998-11-23 2004-08-10 Amgen Canada Inc. Apoptosis-inducing factor
TW570977B (en) * 1998-12-07 2004-01-11 Li-Wei Hsu An expression system for producing recombinant human erythropoietin, method for purifying secreted human erythropoietin and uses thereof
US6495360B1 (en) * 1999-05-28 2002-12-17 Photogen, Inc. Method for enhanced protein stabilization and for production of cell lines useful for production of such stabilized proteins
US20050129669A1 (en) * 1999-06-21 2005-06-16 Transkaryotic Therapies, Inc., A Massachusetts Corporation DNA construct for effecting homologous recombination and uses thereof
AU7734800A (en) * 1999-09-29 2001-04-30 General Hospital Corporation, The Polypeptide derivatives of parathyroid hormone (pth)
ATE412670T1 (en) 1999-09-29 2008-11-15 Gen Hospital Corp PARATHYROID HORMONE (PTH) POLYPEPTIDE DERIVATIVES
AU1647501A (en) 1999-12-10 2001-06-18 Cytos Biotechnology Ag Replicon based activation of endogenous genes
EP1266016A2 (en) * 2000-01-13 2002-12-18 Novozymes Biotech, Inc. Methods for producing a polypeptide using a crippled translational initiator sequence
US6686188B2 (en) 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
GB0018876D0 (en) 2000-08-01 2000-09-20 Applied Research Systems Method of producing polypeptides
CA2634294A1 (en) * 2000-08-03 2002-02-14 Therapeutic Human Polyclonals, Inc. Production of humanized antibodies in transgenic animals
JP4656814B2 (en) * 2000-12-20 2011-03-23 エフ.ホフマン−ラ ロシュ アーゲー Erythropoietin conjugate
CA2454275C (en) * 2001-07-23 2012-10-23 The General Hospital Corporation Conformationally constrained parathyroid hormone (pth) analogs
KR100982922B1 (en) * 2002-01-17 2010-09-20 론자 바이올로직스 피엘씨 Glutamine-auxothrophic human cells capable of producing proteins and capable of growing in a glutamine-free medium
US20060052320A1 (en) * 2002-05-06 2006-03-09 Limin Li Mammalian genes involved in rapamycin resistance and tumorgenesis annexin XIII genes
WO2003100007A2 (en) 2002-05-24 2003-12-04 Schering-Plough Ltd. Eta-1 gene and methods for use
AU2003268135A1 (en) * 2002-08-15 2004-03-19 Functional Genetics, Inc. Mammalian genes involved in rapamycin resistance and tumorgenesis: rapr6 genes
AU2003265525A1 (en) * 2002-08-15 2004-03-19 Functional Genetics, Inc. Mammalian genes involved in rapamycin resistance and tumorgenesis: rapr7 genes
JP2006517101A (en) * 2003-01-13 2006-07-20 エス. ラオ、マヘンドラ Sustained expression of candidate molecules in proliferating stem and progenitor cells to deliver therapeutic products
US20050048041A1 (en) * 2003-01-13 2005-03-03 Rao Mahendra S. Persistent expression of candidate molecule in proliferating stem and progenitor cells for delivery of therapeutic products
WO2004093902A1 (en) * 2003-03-19 2004-11-04 The General Hospital Corporation CONFORMATIONALLY CONSTRAINED PARATHYROID HORMONES WITH α-HELIX STABILIZERS
EP1653985A4 (en) 2003-07-17 2009-08-05 Gen Hospital Corp Conformationally constrained parathyroid hormone (pth) analogs
US7287952B2 (en) * 2003-07-23 2007-10-30 Lockheed Martin Corporation Feeder load automation system and method of use
US11311574B2 (en) 2003-08-08 2022-04-26 Sangamo Therapeutics, Inc. Methods and compositions for targeted cleavage and recombination
US8409861B2 (en) 2003-08-08 2013-04-02 Sangamo Biosciences, Inc. Targeted deletion of cellular DNA sequences
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
SI1696947T1 (en) * 2003-12-19 2014-05-30 F. Hoffmann-La Roche Ag Use of erythropoietin in the treatment of disturbances of iron distribution in chronic inflammatory intestinal diseases
ATE409741T1 (en) * 2004-01-19 2008-10-15 Nsgene As NERVE GROWTH FACTOR SECRETING HUMAN THERAPEUTIC CELLS
AU2005299413A1 (en) 2004-10-22 2006-05-04 Revivicor, Inc. Ungulates with genetically modified immune systems
US7619007B2 (en) 2004-11-23 2009-11-17 Adamas Pharmaceuticals, Inc. Method and composition for administering an NMDA receptor antagonist to a subject
CA2595676A1 (en) * 2005-01-25 2006-08-03 Apollo Life Sciences Limited Molecules and chimeric molecules thereof
ATE481096T1 (en) 2005-04-06 2010-10-15 Adamas Pharmaceuticals Inc METHODS AND COMPOSITIONS FOR TREATING CNS DISEASES
KR101304081B1 (en) 2006-08-04 2013-09-05 프로롱 파마슈티컬스, 엘엘씨 Modified erythropoietin
JP2009545320A (en) * 2006-08-04 2009-12-24 ザ ジェネラル ホスピタル コーポレイション Polypeptide derivative of parathyroid hormone (PTH)
ES2378407T3 (en) 2006-09-01 2012-04-12 Therapeutic Human Polyclonals, Inc. Increased expression of human or humanized immunoglobulin in non-human transgenic animals
WO2008051568A2 (en) 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US7745148B2 (en) 2006-10-30 2010-06-29 Functional Genetics, Inc. Random homozygous gene perturbation to enhance antibody production
US8568737B2 (en) 2007-08-01 2013-10-29 The General Hospital Corporation Screening methods using G-protein coupled receptors and related compositions
WO2009111375A2 (en) * 2008-03-01 2009-09-11 Abraxis Bioscience, Llc Treatment, diagnostic, and method for discovering antagonist using sparc specific mirnas
CA2723716A1 (en) * 2008-05-07 2009-11-12 Abraxis Bioscience, Llc Enhancement of drug therapy by mirna
WO2010037224A1 (en) 2008-10-03 2010-04-08 St. Michael's Hospital Method for preventing and treating cardiovascular diseases with brca1
US20100120063A1 (en) * 2008-10-10 2010-05-13 Discoverx Corporation GPCR Arrestin Assays
EP2348827B1 (en) 2008-10-27 2015-07-01 Revivicor, Inc. Immunocompromised ungulates
JP2012515532A (en) 2009-01-20 2012-07-12 ラモット アット テル アビブ ユニバーシティ, リミテッド MIR-21 promoter driven target cancer treatment
US8211655B2 (en) * 2009-02-12 2012-07-03 Discoverx Corporation Wild-type receptor assays
US9260517B2 (en) 2009-11-17 2016-02-16 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
WO2011143406A2 (en) 2010-05-13 2011-11-17 The General Hospital Corporation Parathyroid hormone analogs and uses thereof
ES2828663T3 (en) * 2012-04-18 2021-05-27 Univ Leland Stanford Junior Nondisruptive Genetic Direction
CN103864913A (en) 2012-12-18 2014-06-18 联亚生技开发股份有限公司 Recombinant protein
CN106460009A (en) 2014-03-21 2017-02-22 小利兰·斯坦福大学托管委员会 Genome editing without nucleases
EP3015475A1 (en) 2014-10-31 2016-05-04 Novartis AG Mammalian cells expressing cytomegalovirus antigens
EP3031822A1 (en) 2014-12-08 2016-06-15 Novartis AG Cytomegalovirus antigens
EP3048114A1 (en) 2015-01-22 2016-07-27 Novartis AG Cytomegalovirus antigens and uses thereof
EP3047856A1 (en) 2015-01-23 2016-07-27 Novartis AG Cmv antigens and uses thereof
JP7157981B2 (en) 2016-03-07 2022-10-21 チャールストンファーマ, エルエルシー anti-nucleolin antibody
WO2018144807A2 (en) * 2017-02-02 2018-08-09 Massachusetts Institute Of Technology Engineered yeast as a method for bioremediation
WO2021014385A1 (en) 2019-07-24 2021-01-28 Glaxosmithkline Biologicals Sa Modified human cytomegalovirus proteins
WO2023144665A1 (en) 2022-01-28 2023-08-03 Glaxosmithkline Biologicals Sa Modified human cytomegalovirus proteins

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4656134A (en) * 1982-01-11 1987-04-07 Board Of Trustees Of Leland Stanford Jr. University Gene amplification in eukaryotic cells

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH634437A5 (en) 1979-03-14 1983-01-31 Tokyo Shibaura Electric Co DISCHARGE RESISTANCE.
US4634665A (en) * 1980-02-25 1987-01-06 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
WO1984002077A1 (en) * 1982-11-30 1984-06-07 Us Health Process for producing poxvirus recombinants for expression of foreign genes
EP0290799B9 (en) * 1983-01-13 2004-09-01 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Transgenic dicotyledonous plant cells and plants
US4740461A (en) * 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
EP0273085A1 (en) * 1986-12-29 1988-07-06 IntraCel Corporation A method for internalizing nucleic acids into eukaryotic cells
US4950599A (en) * 1987-01-29 1990-08-21 Wolf Bertling Method for exchanging homologous DNA sequences in a cell using polyoma encapsulated DNA fragments
ZA88319B (en) * 1987-02-06 1988-08-12 Lubrizol Enterprises, Inc. Ocs enhancer
EP0317509A3 (en) * 1987-11-16 1990-01-31 Ciba-Geigy Ag Targetted incorporation of genes into a plant genome
GB8807683D0 (en) * 1988-03-31 1988-05-05 Ici Plc Regulation of gene expression
AU644352B2 (en) * 1988-12-08 1993-12-09 Damon Biotech Inc. Expression induction method employing mutant dhfr gene
FR2646438B1 (en) * 1989-03-20 2007-11-02 Pasteur Institut A METHOD FOR SPECIFIC REPLACEMENT OF A COPY OF A GENE PRESENT IN THE RECEIVER GENOME BY INTEGRATION OF A GENE DIFFERENT FROM THAT OR INTEGRATION
WO1990014092A1 (en) * 1989-05-18 1990-11-29 Cell Genesys, Inc. Single-strand site-directed modification of mammalian genes in vivo
NL8901931A (en) * 1989-07-26 1991-02-18 Mogen International N V En Rij METHOD FOR PLACE-ORGANIZED INTRODUCTION OF FOREIGN DNA INTO THE NAME OF PLANTS.
KR100233781B1 (en) * 1989-11-06 1999-12-01 스테펜 에이. 쉐어윈 Production of proteins using homologous recombination
US5272071A (en) * 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
ATE264916T1 (en) * 1989-12-22 2004-05-15 Applied Research Systems DNA CONSTRUCTS TO ACTIVATE AND ALTER THE EXPRESSION OF ENDOGENE GENES
NZ245015A (en) * 1991-11-05 1995-12-21 Transkaryotic Therapies Inc Delivery of human growth hormone through the administration of transfected cell lines encoding human growth hormone, which are physically protected from host immune response; the transfected cells and their production

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4656134A (en) * 1982-01-11 1987-04-07 Board Of Trustees Of Leland Stanford Jr. University Gene amplification in eukaryotic cells

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Molecular and Cellular Biology, Volume 4, issued 1984, RUBNITZ et al, "The minimum amount of homology required for homologous recombination in Mammalian Cells, pages 2253-2257, See entire document. *
Proc. Nalt Acad. Sci, Volume 84, issued August 1987, (USA), KAMARCK et al, "Carcino embryonic antigen fanid: Expression in a mouse L-cell transfectant and characterization of a partial cDNA in bactenophage, pages 5350-5354, See entire document but in particular page 5350, Col 2, paragraphs 1&2; page 5351, Results, first paragraph. *
Proc. Natl. Acad Sci., Volume 77, No. 7, issued July 1980, (USA), G. URLAULS et al., "Isolation of Cluneese hamster cell mutants deficient in dihydrofolate reductase activity, pages 4216-4220, See entire document. *
Proc. Natl. Acad. Sci, Volume 84, issued October 1987, (USA), SONG et al, "Accurate Modification of a Chromosomal Plasmidly Homologous recombination in human cells, pages 6820-6824, See entire document. *

Cited By (293)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6303379B1 (en) 1987-05-01 2001-10-16 Transkaryotic Therapies, Inc. Vivo protein production and delivery system for gene therapy
US6048729A (en) * 1987-05-01 2000-04-11 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6638768B1 (en) 1989-03-20 2003-10-28 Institut Pasteur Procedure for specific replacement of a copy of a gene present in the recipient genome by the integration of a gene different from that where the integration is made
US6528314B1 (en) 1989-03-20 2003-03-04 Institut Pasteur Procedure for specific replacement of a copy of a gene present in the recipient genome by the integration of a gene different from that where the integration is made
US6528313B1 (en) 1989-03-20 2003-03-04 Institut Pasteur Procedure for specific replacement of a copy of a gene present in the recipient genome by the integration of a gene different from that where the integration is made
EP1375666A2 (en) * 1989-12-22 2004-01-02 Applied Research Systems ARS Holding N.V. Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
EP1482053A2 (en) * 1989-12-22 2004-12-01 Applied Research Systems ARS Holding N.V. Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
EP1484412A2 (en) * 1989-12-22 2004-12-08 Applied Research Systems ARS Holding N.V. Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
EP1375666A3 (en) * 1989-12-22 2007-09-26 Applied Research Systems ARS Holding N.V. Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
EP1482053A3 (en) * 1989-12-22 2007-10-17 Laboratoires Serono SA Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
EP1484412A3 (en) * 1989-12-22 2007-10-17 Laboratoires Serono SA Eukaryotic host cell line in which an endogenous gene has been activated, and uses thereof
EP0584214A1 (en) * 1991-05-06 1994-03-02 Cell Genesys, Inc. Gene manipulation and expression using genomic elements
EP0584214A4 (en) * 1991-05-06 1995-04-19 Cell Genesys Inc Gene manipulation and expression using genomic elements.
US5686263A (en) * 1991-05-31 1997-11-11 Genentech, Inc. Method for enhancing gene expression
US5789215A (en) * 1991-08-20 1998-08-04 Genpharm International Gene targeting in animal cells using isogenic DNA constructs
US5733761A (en) * 1991-11-05 1998-03-31 Transkaryotic Therapies, Inc. Protein production and protein delivery
US6692737B1 (en) 1991-11-05 2004-02-17 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US7410799B2 (en) 1991-11-05 2008-08-12 Shire Human Genetic Therapies, Inc. In vivo production and delivery of erythropoietin or insulinotropin for gene therapy
US6048724A (en) * 1991-11-05 2000-04-11 Transkaryotic Therapies Inc. Method of producing clonal cell strains which express exogenous DNA encoding glucagon-like peptide 1
US6054288A (en) * 1991-11-05 2000-04-25 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6063630A (en) * 1991-11-05 2000-05-16 Transkaryotic Therapies, Inc. Targeted introduction of DNA into primary or secondary cells and their use for gene therapy
EP0750044A2 (en) * 1991-11-05 1996-12-27 Transkaryotic Therapies, Inc. DNA constructs and transfection of cells therewith for homologous recombination
US6214622B1 (en) 1991-11-05 2001-04-10 Transkaryotic Therapies, Inc. Targeted introduction of DNA into primary or secondary cells and their use for gene therapy
US6270989B1 (en) 1991-11-05 2001-08-07 Transkaryotic Therapies, Inc. Protein production and delivery
US6537542B1 (en) 1991-11-05 2003-03-25 Transkaryotic Therapies, Inc. Targeted introduction of DNA into primary or secondary cells and their use for gene therapy and protein production
US5994127A (en) * 1991-11-05 1999-11-30 Transkaryotic Therapies, Inc. In vivo production and delivery of erythropoietin or insulinotropin for gene therapy
US5641670A (en) * 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
US6355241B1 (en) 1991-11-05 2002-03-12 Transkaryotic Therapies, Inc. In vivo production and delivery of erythropoietin
EP0750044A3 (en) * 1991-11-05 1997-01-15 Transkaryotic Therapies, Inc. DNA constructs and transfection of cells therewith for homologous recombination
US5968502A (en) * 1991-11-05 1999-10-19 Transkaryotic Therapies, Inc. Protein production and protein delivery
US6565844B1 (en) 1991-11-05 2003-05-20 Transkaryotic Therapies, Inc. Protein production and protein delivery
US6048524A (en) * 1991-11-05 2000-04-11 Transkaryotic Therapies, Inc. In vivo production and delivery of erythropoietin for gene therapy
US6846676B2 (en) 1991-11-05 2005-01-25 Transkaryotic Therapies, Inc. In Vivo production and delivery of erythropoietin or insulinotropin for gene therapy
US6670178B1 (en) 1992-07-10 2003-12-30 Transkaryotic Therapies, Inc. In Vivo production and delivery of insulinotropin for gene therapy
US6531124B1 (en) 1992-07-10 2003-03-11 Transkaryotic Therapies, Inc. In vivo production and delivery of insulinotropin for gene therapy
US5733753A (en) * 1992-12-22 1998-03-31 Novo Nordisk A/S Amplification of genomic DNA by site specific integration of a selectable marker construct
EP1403376A3 (en) * 1993-04-21 2007-08-29 The University Court Of The University Of Edinburgh Expression of heterologous genes according to a targeted expression profile
US7005299B1 (en) 1993-04-21 2006-02-28 The University Of Edinburgh Expression of heterologous genes according to a targeted expression profile
WO1995031560A1 (en) * 1994-05-13 1995-11-23 Transkaryotic Therapies, Inc. Dna construct for effecting homologous recombination and uses thereof
AU709058B2 (en) * 1994-05-13 1999-08-19 Transkaryotic Therapies, Inc. Dna construct for effecting homologous recombination and uses thereof
US7067484B1 (en) 1994-10-27 2006-06-27 Genentech, Inc. AL-1 neurotrophic factor treatments
WO1996029411A1 (en) * 1995-03-17 1996-09-26 Transkaryotic Therapies, Inc. Protein production and delivery
US6610296B2 (en) 1995-10-26 2003-08-26 Genentech, Inc. Methods of enhancing cognitive function using an AL-1 neurotrophic factor immunoadhesin
EP1619250A1 (en) 1996-01-08 2006-01-25 Genentech, Inc. WSX receptor and ligands
US6399350B1 (en) 1996-08-29 2002-06-04 The Regents Of The University Of California KUZ, a novel family of metalloproteases
US7105313B2 (en) 1996-09-04 2006-09-12 Amgen Inc. Cell-based drug screens for regulators of gene expression
US6566089B1 (en) * 1996-09-04 2003-05-20 Tularik Inc. Cell-based drug screens for regulators of gene expression
US5925544A (en) * 1996-11-18 1999-07-20 Novo Nordisk A/S Method of homologous recombination followed by in vivo selection of DNA amplification
US7785588B2 (en) 1997-02-18 2010-08-31 Genentech, Inc. Anti-neurturin receptor-A antibody compositions comprising cytokines or neurotrophic factors
US6025157A (en) * 1997-02-18 2000-02-15 Genentech, Inc. Neurturin receptor
US6372453B1 (en) 1997-02-18 2002-04-16 Genetech, Inc. Neurturin receptor
US7235360B2 (en) 1997-03-14 2007-06-26 Biogen Idec Inc. Method for integrating genes at specific sites in mammalian cells via homologous recombination and vectors for accomplishing the same
US7785831B2 (en) 1997-09-26 2010-08-31 Abt Holding Company Compositions and methods for non-targeted activation of endogenous genes
US6524818B1 (en) 1997-09-26 2003-02-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US7842792B2 (en) 1997-09-26 2010-11-30 Abt Holding Company Compositions and methods for non-targeted activation of endogenous genes
US6740503B1 (en) 1997-09-26 2004-05-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US7569220B2 (en) 1997-09-26 2009-08-04 Abt Holding Company Compositions and methods for non-targeted activation of endogenous genes
US7033782B2 (en) 1997-09-26 2006-04-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US6602686B1 (en) 1997-09-26 2003-08-05 Athersys, Inc. Compositions and method for non-targeted activation of endogenous genes
US6623958B1 (en) 1997-09-26 2003-09-23 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US6541221B1 (en) 1997-09-26 2003-04-01 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
EP1025253B2 (en) 1997-10-20 2009-07-29 Roche Diagnostics GmbH Positive-negative selection for homologous recombination
EP2218731A1 (en) 1997-11-28 2010-08-18 Merck Serono Biodevelopment Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
EP2218730A1 (en) 1997-11-28 2010-08-18 Merck Serono Biodevelopment Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
EP2218732A1 (en) 1997-11-28 2010-08-18 Merck Serono Biodevelopment Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
EP2228384A1 (en) 1997-11-28 2010-09-15 Merck Serono Biodevelopment Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
EP2228385A1 (en) 1997-11-28 2010-09-15 Merck Serono Biodevelopment Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
US6337072B1 (en) 1998-04-03 2002-01-08 Hyseq, Inc. Interleukin-1 receptor antagonist and recombinant production thereof
US6294655B1 (en) 1998-04-03 2001-09-25 Hyseq, Inc. Anti-interleukin-1 receptor antagonist antibodies and uses thereof
US6541623B1 (en) 1998-04-03 2003-04-01 Hyseq, Inc. Interleukin—1 receptor antagonist and uses thereof
US6426191B1 (en) 1998-04-03 2002-07-30 Hyseq, Inc. Assays involving an IL-1 receptor antagonist
US7514072B1 (en) 1998-12-14 2009-04-07 Hannelore Ehrenreich Method for the treatment of cerebral ischaemia and use of erythropoietin or erythropoietin derivatives for the treatment of cerebral ischaemia
EP1054018A1 (en) 1999-05-18 2000-11-22 Target Quest B.V. Fab fragment libraries and methods for their use
EP2067788A2 (en) 1999-05-18 2009-06-10 Dyax Corp. Fab fragment libraries and methods for their use
EP2322558A1 (en) 1999-06-30 2011-05-18 Millennium Pharmaceuticals, Inc. Antibodies directed against glycoprotein VI and uses thereof
EP2902414A1 (en) 1999-06-30 2015-08-05 Millennium Pharmaceuticals, Inc. Antibodies directed against glycoprotein VI and uses thereof
EP2143796A2 (en) 2000-02-29 2010-01-13 Millennium Pharmaceuticals, Inc. 1983, 52881, 2398, 45449, 50289, and 52872, G protein-coupled receptors and uses therefor
EP1714661A2 (en) 2000-05-19 2006-10-25 The Center for Blood Research, INC. Methods for diagnosing and treating hemostatic disorders by modulating p-selectin activity
US6803211B2 (en) 2000-08-25 2004-10-12 Pfizer Inc. Methods and compositions for diagnosing and treating disorders involving angiogenesis
US8580563B2 (en) 2000-12-06 2013-11-12 Anthrogenesis Corporation Placental stem cells
US7976836B2 (en) 2000-12-06 2011-07-12 Anthrogenesis Corporation Treatment of stroke using placental stem cells
US9149569B2 (en) 2000-12-06 2015-10-06 Anthrogenesis Corporation Treatment of diseases or disorders using placental stem cells
WO2002049673A2 (en) 2000-12-20 2002-06-27 F. Hoffmann-La Roche Ag Conjugates of erythropoietin (pep) with polyethylene glycol (peg)
EP2281828A2 (en) 2000-12-29 2011-02-09 The Kenneth S. Warren Institute, Inc. Compositions comprising modified erythropoietin
US9139813B2 (en) 2001-02-14 2015-09-22 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
EP2336299A1 (en) 2001-02-14 2011-06-22 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2316918A1 (en) 2001-02-14 2011-05-04 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP3246396A1 (en) 2001-02-14 2017-11-22 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
EP2316919A1 (en) 2001-02-14 2011-05-04 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2336300A1 (en) 2001-02-14 2011-06-22 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2314673A1 (en) 2001-02-14 2011-04-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2336301A1 (en) 2001-02-14 2011-06-22 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2316463A1 (en) 2001-02-14 2011-05-04 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
EP2338983A1 (en) 2001-02-14 2011-06-29 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
EP2184359A1 (en) 2001-06-06 2010-05-12 Bioriginal Food & Science Corp. Flax (Linum usitatissimum L.) seed-specific promoters
EP1925671A1 (en) 2001-06-06 2008-05-28 Bioriginal Food & Science Corp. Flax (Linum usitatissimum L.) seed-specific promoters
WO2003020702A2 (en) 2001-08-31 2003-03-13 The Rockefeller University Phosphodiesterase activity and regulation of phosphodiesterase 1-b-mediated signaling in brain
US7745391B2 (en) 2001-09-14 2010-06-29 Compugen Ltd. Human thrombospondin polypeptide
US8334111B2 (en) 2001-12-19 2012-12-18 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
US7527962B2 (en) 2001-12-19 2009-05-05 Millenium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
EP2436393A1 (en) 2001-12-19 2012-04-04 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
US7910346B2 (en) 2001-12-19 2011-03-22 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
US8057789B2 (en) 2002-02-13 2011-11-15 Anthrogenesis Corporation Placental stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
EP2292091A1 (en) 2002-02-13 2011-03-09 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
EP2186407A1 (en) 2002-02-13 2010-05-19 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
EP2301343A1 (en) 2002-02-13 2011-03-30 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
EP2390311A1 (en) 2002-04-12 2011-11-30 Celgene Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US7459435B2 (en) 2002-08-29 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
US7723055B2 (en) 2002-11-21 2010-05-25 University Of Massachusetts Diagnosing and treating hematopoietic cancers
US7459436B2 (en) 2002-11-22 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
EP2112229A2 (en) 2002-11-25 2009-10-28 Sequenom, Inc. Methods for identifying risk of breast cancer and treatments thereof
US8617535B2 (en) 2002-11-26 2013-12-31 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
EP2561887A2 (en) 2003-01-14 2013-02-27 Dana Farber Cancer Institute, Inc. Cancer therapy sensitizer
US8021833B2 (en) 2003-02-12 2011-09-20 Functional Genetics, Inc. Method for reducing HIV viral budding by administering a VPS28-specfic antibody that disrupts Gag-TSG101-VPS28 binding interactions
US7645733B2 (en) 2003-09-29 2010-01-12 The Kenneth S. Warren Institute, Inc. Tissue protective cytokines for the treatment and prevention of sepsis and the formation of adhesions
WO2005042581A2 (en) 2003-11-01 2005-05-12 Biovation Ltd. Modified anti-cd52 antibody
US7510858B2 (en) 2004-08-02 2009-03-31 Janssen Pharmaceutica N.V. IRAK 1c splice variant and its use
US7893197B2 (en) 2004-08-25 2011-02-22 Janssen Pharmaceutica N.V. Relaxin-3 chimeric polypeptides and their preparation and use
EP2338993A1 (en) 2004-09-02 2011-06-29 Yale University Regulation of oncogenes by microRNAs
US7893034B2 (en) 2004-09-02 2011-02-22 Yale University Regulation of oncogenes by microRNAs
EP2338994A1 (en) 2004-09-02 2011-06-29 Yale University Regulation of oncogenes by microRNAs
US7741306B2 (en) 2004-09-02 2010-06-22 Yale University Regulation of oncogenes by microRNAs
WO2006028967A2 (en) 2004-09-02 2006-03-16 Yale University Regulation of oncogenes by micrornas
EP2298897A1 (en) 2004-09-02 2011-03-23 Yale University Regulation of oncogenes by microRNAs
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
US10603357B2 (en) 2004-11-29 2020-03-31 Bristol-Myers Squibb Company Therapeutic TREM-1 peptides
EP2365086A1 (en) 2005-02-09 2011-09-14 Bioriginal Food & Science Corporation Novel omega-3 fatty acid desaturase family members and uses thereof
US8354384B2 (en) 2005-06-23 2013-01-15 Yale University Anti-aging micrornas
US8895256B2 (en) 2005-10-13 2014-11-25 Anthrogenesis Corporation Immunomodulation using placental stem cells
US9539288B2 (en) 2005-10-13 2017-01-10 Anthrogenesis Corporation Immunomodulation using placental stem cells
EP2441830A2 (en) 2005-11-03 2012-04-18 Momenta Pharmaceuticals, Inc. Heparan sulfate glycosaminoglycan lyase and uses thereof
EP2450441A2 (en) 2005-11-03 2012-05-09 Momenta Pharmaceuticals, Inc. Heparan sulfate glycosaminoglycan lyase and uses thereof
US8014957B2 (en) 2005-12-15 2011-09-06 Fred Hutchinson Cancer Research Center Genes associated with progression and response in chronic myeloid leukemia and uses thereof
US8455250B2 (en) 2005-12-29 2013-06-04 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US10383897B2 (en) 2005-12-29 2019-08-20 Celularity, Inc. Placental stem cell populations
US9078898B2 (en) 2005-12-29 2015-07-14 Anthrogenesis Corporation Placental stem cell populations
US8691217B2 (en) 2005-12-29 2014-04-08 Anthrogenesis Corporation Placental stem cell populations
US9254312B2 (en) 2006-01-27 2016-02-09 Hannelore Ehrenreich Method for the treatment and/or prophylaxis of multiple sclerosis, and use of erythropoietin for the manufacture of a medicament for the intermittent treatment and/or intermittent prophylaxis of multiple sclerosis
EP2319530A1 (en) 2006-01-27 2011-05-11 Hannelore Ehrenreich Method for treating and/or preventing multiple sclerosis, and use of erythropoietin in the production of a drug for intermittent treatment and/or intermittent prevention of multiple sclerosis
US7566565B2 (en) 2006-03-24 2009-07-28 Syntonix Pharmaceuticals, Inc. PC5 as a factor IX propeptide processing enzyme
US8021880B2 (en) 2006-03-24 2011-09-20 Syntonix Pharmaceuticals, Inc. PC5 as a factor IX propeptide processing enzyme
US7795400B2 (en) 2006-03-24 2010-09-14 Syntonix Pharmaceuticals, Inc. PC5 as a factor IX propeptide processing enzyme
EP2650305A1 (en) 2006-03-24 2013-10-16 Biogen Idec Hemophilia Inc. PC5 as a factor IX propeptide processing enzyme
WO2007112005A2 (en) 2006-03-24 2007-10-04 Syntonix Pharmaceuticals, Inc. Pc5 as a factor ix propeptide processing enzyme
US10529441B2 (en) 2006-05-03 2020-01-07 Population Bio, Inc. Evaluating genetic disorders
US10210306B2 (en) 2006-05-03 2019-02-19 Population Bio, Inc. Evaluating genetic disorders
US10522240B2 (en) 2006-05-03 2019-12-31 Population Bio, Inc. Evaluating genetic disorders
EP2236623A1 (en) 2006-06-05 2010-10-06 Cancer Care Ontario Assessment of risk for colorectal cancer
US8916146B2 (en) 2007-02-12 2014-12-23 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
EP2243834A1 (en) 2007-03-05 2010-10-27 Cancer Care Ontario Assessment of risk for colorectal cancer
WO2008140449A1 (en) 2007-05-11 2008-11-20 Thomas Jefferson University Methods of treatment and prevention of neurodegenerative diseases and disorders
US9029383B2 (en) 2007-05-11 2015-05-12 The Trustees Of The University Of Pennsylvania Methods of treatment of skin ulcers
EP2977452A2 (en) 2007-05-11 2016-01-27 Thomas Jefferson University Methods of treatment and prevention of neurodegenerative diseases and disorders
US8252897B2 (en) 2007-06-21 2012-08-28 Angelica Therapeutics, Inc. Modified toxins
WO2009014835A2 (en) 2007-06-21 2009-01-29 Angelica Therapeutics, Inc. Modified toxins
US8138317B2 (en) 2007-07-17 2012-03-20 Hoffmann-La Roche Inc. Purification of pegylated polypeptides
US8889837B2 (en) 2007-07-17 2014-11-18 Hoffman-La Roche Inc. Purification of pegylated polypeptides
US8795533B2 (en) 2007-07-17 2014-08-05 Hoffmann-La Roche Inc. Chromatographic methods
US9180165B2 (en) 2007-07-19 2015-11-10 Hannelore Ehrenreich Use of EPO receptor activation or stimulation for the improvement of the EDSS score in patients with multiple sclerosis
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
WO2009042538A2 (en) 2007-09-24 2009-04-02 Cornell University Immunogenic proteins from genome-derived outer membrane of leptospira and compositions and methods based thereon
US9216200B2 (en) 2007-09-28 2015-12-22 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
EP2666854A1 (en) 2007-12-04 2013-11-27 Proteon Therapeutics, Inc. Recombinant elastase proteins and methods of manufacturing and use thereof
EP3176262A1 (en) 2007-12-04 2017-06-07 Proteon Therapeutics, Inc. Recombinant elastase proteins and methods of manufacturing and use thereof
EP2570494A1 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570501A2 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570497A2 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570495A1 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570500A2 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570493A1 (en) 2008-01-02 2013-03-20 Suregene LLC Genetic markers of mental illness
EP2570499A2 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570496A2 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2570498A2 (en) 2008-01-02 2013-03-20 Suregene Llc Genetic markers of mental illness
EP2514840A1 (en) 2008-01-02 2012-10-24 Suregene Llc Genetic markers of mental illness
EP2581458A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
EP2581460A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
EP2581456A2 (en) 2008-01-17 2013-04-17 Suregene Llc Genetic Markers of Mental Illness
EP2581453A2 (en) 2008-01-17 2013-04-17 Suregene Llc Genetic Markers of Mental Illness
EP2581454A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
EP2581459A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
EP2581455A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
EP2581457A2 (en) 2008-01-17 2013-04-17 Suregene LLC Genetic markers of mental illness
EP3369827A1 (en) 2008-03-12 2018-09-05 The Rockefeller University Methods and compositions for translational profiling and molecular phenotyping
WO2009114185A2 (en) 2008-03-12 2009-09-17 The Rockefeller University Methods and compositions for translational profiling and molecular phenotyping
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US10612086B2 (en) 2008-09-16 2020-04-07 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
US10738358B2 (en) 2008-09-16 2020-08-11 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
US8962247B2 (en) 2008-09-16 2015-02-24 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non invasive prenatal diagnoses
EP2631303A1 (en) 2008-09-25 2013-08-28 SureGene LLC Genetic markers for assessing risk of developing Schizophrenia
WO2010036353A2 (en) 2008-09-25 2010-04-01 Suregene Llc Genetic markers for optimizing treatment for schizophrenia
EP2868753A1 (en) 2008-09-25 2015-05-06 SureGene LLC Genetic markers for optimizing treatment for Schizophrenia
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
US11261251B2 (en) 2008-09-26 2022-03-01 Dana-Farber Cancer Institute, Inc. Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
EP3530672A1 (en) 2008-09-26 2019-08-28 Dana Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses thereof
US9102727B2 (en) 2008-09-26 2015-08-11 Emory University Human anti-PD-1 antibodies and uses therefor
US10011656B2 (en) 2008-09-26 2018-07-03 Emory University Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
US10370448B2 (en) 2008-09-26 2019-08-06 Emory University Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
US10040838B2 (en) 2008-11-04 2018-08-07 Janssen Pharmaceutica Nv CRHR2 peptide agonists and uses thereof
US9198938B2 (en) 2008-11-19 2015-12-01 Antrhogenesis Corporation Amnion derived adherent cells
US8530629B2 (en) 2009-01-30 2013-09-10 Ab Biosciences, Inc. Lowered affinity antibodies and uses therefor
WO2011057027A2 (en) 2009-11-04 2011-05-12 Janssen Pharmaceutica Nv Method for treating heart failure with stresscopin-like peptides
EP3168232A1 (en) 2009-11-13 2017-05-17 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for the diagnosis, prognosis, monitoring, treatment and modulation of post-transplant lymphoproliferative disorders and hypoxia associated angiogenesis disorders using galectin-1
US11180799B2 (en) 2009-12-22 2021-11-23 Sequenom, Inc. Processes and kits for identifying aneuploidy
US9926593B2 (en) 2009-12-22 2018-03-27 Sequenom, Inc. Processes and kits for identifying aneuploidy
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9624290B2 (en) 2010-01-28 2017-04-18 Ab Biosciences, Inc. Lowered affinity antibodies and methods of making the same
EP3153521A1 (en) 2010-03-26 2017-04-12 Trustees of Dartmouth College Vista regulatory t cell mediator protein, vista binding agents and use thereof
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10781254B2 (en) 2010-03-26 2020-09-22 The Trustees Of Dartmouth College VISTA regulatory T cell mediator protein, VISTA binding agents and use thereof
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US9464274B2 (en) 2010-07-13 2016-10-11 Anthrogenesis Corporation Methods of generating natural killer cells
US10059997B2 (en) 2010-08-02 2018-08-28 Population Bio, Inc. Compositions and methods for discovery of causative mutations in genetic disorders
US11788142B2 (en) 2010-08-02 2023-10-17 Population Bio, Inc. Compositions and methods for discovery of causative mutations in genetic disorders
WO2012035037A1 (en) 2010-09-14 2012-03-22 F. Hoffmann-La Roche Ag Method for purifying pegylated erythropoietin
US10273277B2 (en) 2010-09-14 2019-04-30 Hoffmann-La Roche Inc. Method for purifying PEGylated erythropoietin
WO2012075337A2 (en) 2010-12-01 2012-06-07 Spinal Modulation, Inc. Directed delivery of agents to neural anatomy
US10227564B2 (en) 2010-12-02 2019-03-12 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti CD4 antibodies to prevent in particular graft-versus-host-disease (GvHD)
WO2012072268A2 (en) 2010-12-02 2012-06-07 Fraunhofer Gesellschaft Zur Förderung Der Angewadten Forschung E.V. Tolerance induction or immunosupression to prevent in particular graft-versus-host-disease (gvhd) by short-term pre-incubation of transplanted cell suspensions, tissues or organs coated with ligands to cell surface molecules
US9745552B2 (en) 2010-12-02 2017-08-29 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti CD4 antibodies to prevent in particular graft-versus-host-disease (GvHD)
US10577588B2 (en) 2010-12-02 2020-03-03 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti CD4 antibodies to prevent in particular graft-versus-host-disease (GVHD)
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US11090339B2 (en) 2011-06-01 2021-08-17 Celularity Inc. Treatment of pain using placental stem cells
WO2013003697A1 (en) 2011-06-30 2013-01-03 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
WO2013020044A1 (en) 2011-08-03 2013-02-07 The Charlotte-Mecklenburg Hospital Authority D/B/A Carolinas Medical Center Treatment of fibrosis using microrna-19b
US11339439B2 (en) 2011-10-10 2022-05-24 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
WO2013054200A2 (en) 2011-10-10 2013-04-18 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
WO2013067451A2 (en) 2011-11-04 2013-05-10 Population Diagnostics Inc. Methods and compositions for diagnosing, prognosing, and treating neurological conditions
US11180807B2 (en) 2011-11-04 2021-11-23 Population Bio, Inc. Methods for detecting a genetic variation in attractin-like 1 (ATRNL1) gene in subject with Parkinson's disease
US11174516B2 (en) 2012-02-09 2021-11-16 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
US10407724B2 (en) 2012-02-09 2019-09-10 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
US10906965B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation wtih antibodies that bind peptidoglycan recognition protein 1
US10906975B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation with antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US10189904B2 (en) 2012-02-15 2019-01-29 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10150809B2 (en) 2012-02-15 2018-12-11 Bristol-Myers Squibb Company Antibodies that bind peptidoglycan recognition protein 1
US9605313B2 (en) 2012-03-02 2017-03-28 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
US10738359B2 (en) 2012-03-02 2020-08-11 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
US11312997B2 (en) 2012-03-02 2022-04-26 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
US9494597B2 (en) 2012-04-02 2016-11-15 Ab Biosciences, Inc. Human control antibodies and uses therefor
US9920361B2 (en) 2012-05-21 2018-03-20 Sequenom, Inc. Methods and compositions for analyzing nucleic acid
US11306354B2 (en) 2012-05-21 2022-04-19 Sequenom, Inc. Methods and compositions for analyzing nucleic acid
US11752189B2 (en) 2012-06-22 2023-09-12 The Trustees Of Dartmouth College Vista antagonist and methods of use
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
US11332791B2 (en) 2012-07-13 2022-05-17 Sequenom, Inc. Processes and compositions for methylation-based enrichment of fetal nucleic acid from a maternal sample useful for non-invasive prenatal diagnoses
US11529416B2 (en) 2012-09-07 2022-12-20 Kings College London Vista modulators for diagnosis and treatment of cancer
US11008614B2 (en) 2012-09-14 2021-05-18 Population Bio, Inc. Methods for diagnosing, prognosing, and treating parkinsonism
US9976180B2 (en) 2012-09-14 2018-05-22 Population Bio, Inc. Methods for detecting a genetic variation in subjects with parkinsonism
US10597721B2 (en) 2012-09-27 2020-03-24 Population Bio, Inc. Methods and compositions for screening and treating developmental disorders
US11618925B2 (en) 2012-09-27 2023-04-04 Population Bio, Inc. Methods and compositions for screening and treating developmental disorders
US10233495B2 (en) 2012-09-27 2019-03-19 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US11060145B2 (en) 2013-03-13 2021-07-13 Sequenom, Inc. Methods and compositions for identifying presence or absence of hypermethylation or hypomethylation locus
US10059750B2 (en) 2013-03-15 2018-08-28 Angelica Therapeutics, Inc. Modified toxins
US10066001B2 (en) 2013-03-15 2018-09-04 Apotex Inc. Enhanced liquid formulation stability of erythropoietin alpha through purification processing
WO2015081290A1 (en) 2013-11-27 2015-06-04 Inis Biotech Llc Methods for modulating angiogenesis of cancers refractory to anti-vegf treatment
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
US11365447B2 (en) 2014-03-13 2022-06-21 Sequenom, Inc. Methods and processes for non-invasive assessment of genetic variations
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
US10724096B2 (en) 2014-09-05 2020-07-28 Population Bio, Inc. Methods and compositions for inhibiting and treating neurological conditions
US11549145B2 (en) 2014-09-05 2023-01-10 Population Bio, Inc. Methods and compositions for inhibiting and treating neurological conditions
WO2016044271A2 (en) 2014-09-15 2016-03-24 Children's Medical Center Corporation Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
US11390885B2 (en) 2014-09-15 2022-07-19 Children's Medical Center Corporation Methods and compositions to increase somatic cell nuclear transfer (SCNT) efficiency by removing histone H3-lysine trimethylation
US10370455B2 (en) 2014-12-05 2019-08-06 Immunext, Inc. Identification of VSIG8 as the putative VISTA receptor (V-R) and use thereof to produce VISTA/VSIG8 agonists and antagonists
WO2016118832A1 (en) 2015-01-22 2016-07-28 University Of Massachusetts Cancer immunotherapy
US11009509B2 (en) 2015-06-24 2021-05-18 Janssen Pharmaceutica Nv Anti-VISTA antibodies and fragments
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
US11603403B2 (en) 2016-04-15 2023-03-14 Immunext, Inc. Anti-human vista antibodies and use thereof
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
US11649283B2 (en) 2016-04-15 2023-05-16 Immunext, Inc. Anti-human vista antibodies and use thereof
US11603402B2 (en) 2016-04-15 2023-03-14 Immunext, Inc. Anti-human vista antibodies and use thereof
WO2018011381A1 (en) 2016-07-15 2018-01-18 F. Hoffmann-La Roche Ag Method for purifying pegylated erythropoietin
US10240205B2 (en) 2017-02-03 2019-03-26 Population Bio, Inc. Methods for assessing risk of developing a viral disease using a genetic test
US11913073B2 (en) 2017-02-03 2024-02-27 Pml Screening, Llc Methods for assessing risk of developing a viral disease using a genetic test
US10941448B1 (en) 2017-02-03 2021-03-09 The Universite Paris-Saclay Methods for assessing risk of developing a viral disease using a genetic test
US10544463B2 (en) 2017-02-03 2020-01-28 Pml Screening, Llc Methods for assessing risk of developing a viral disease using a genetic test
US10563264B2 (en) 2017-02-03 2020-02-18 Pml Screening, Llc Methods for assessing risk of developing a viral disease using a genetic test
WO2019129878A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Process for providing pegylated protein composition
WO2019129876A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Process for providing pegylated protein composition
WO2019129877A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Process for providing pegylated protein composition
US11518781B2 (en) 2017-12-29 2022-12-06 Hoffmann-La Roche Inc. Process for providing PEGylated protein composition
US10961585B2 (en) 2018-08-08 2021-03-30 Pml Screening, Llc Methods for assessing risk of developing a viral of disease using a genetic test
US11913074B2 (en) 2018-08-08 2024-02-27 Pml Screening, Llc Methods for assessing risk of developing a viral disease using a genetic test
WO2022159414A1 (en) 2021-01-22 2022-07-28 University Of Rochester Erythropoietin for gastroinfestinal dysfunction

Also Published As

Publication number Publication date
ES2127458T3 (en) 1999-04-16
DK0452484T3 (en) 1996-10-14
JP2001186897A (en) 2001-07-10
CA2045175C (en) 2003-03-18
EP0945515A3 (en) 2002-08-21
EP0452484A4 (en) 1992-05-13
EP0747485B1 (en) 1998-12-02
EP0452484A1 (en) 1991-10-23
ATE139574T1 (en) 1996-07-15
DE69027526T3 (en) 2005-03-24
GR3029328T3 (en) 1999-05-28
NO912642D0 (en) 1991-07-05
EP0452484B2 (en) 2004-07-28
DE69027526D1 (en) 1996-07-25
US5578461A (en) 1996-11-26
DK0747485T3 (en) 1999-08-16
AU7740791A (en) 1991-05-31
SG88714A1 (en) 2002-05-21
SG122763A1 (en) 2006-06-29
US6015708A (en) 2000-01-18
KR100233781B1 (en) 1999-12-01
WO1991006666A1 (en) 1991-05-16
AU635844B2 (en) 1993-04-01
DE69032809T2 (en) 1999-07-08
EP0945515A2 (en) 1999-09-29
ES2090297T3 (en) 1996-10-16
NO309238B1 (en) 2001-01-02
ATE174058T1 (en) 1998-12-15
DE69032809D1 (en) 1999-01-14
EP0747485A1 (en) 1996-12-11
CA2045175A1 (en) 1991-05-07
EP0452484B1 (en) 1996-06-19
GR3021105T3 (en) 1996-12-31
JPH04505104A (en) 1992-09-10
US5981214A (en) 1999-11-09
DE69027526T2 (en) 1996-12-05
ES2090297T5 (en) 2005-03-01
NO912642L (en) 1991-09-05

Similar Documents

Publication Publication Date Title
AU635844B2 (en) Production of proteins using homologous recombination
EP0491875B1 (en) Recombinant dna method and vectors for use therein
AU2008339985B2 (en) Mammalian expression vector
CN101688222A (en) Promoter
EP0795030A1 (en) Method for making recombinant yeast artificial chromosomes
EP1017803A1 (en) Expression of endogenous genes by non-homologous recombination of a vector construct with cellular dna
WO2010140387A1 (en) Expression vector for establishing hyper-producing cells, and hyper-producing cells
KR100259933B1 (en) Gene manipulation and expression using genomic elements
KR20170132784A (en) Eukaryotic expression vectors containing regulatory elements of the globin gene cluster
JPH05503015A (en) mammalian expression vector
US5747308A (en) Recombinant DNA method
EP1967585B1 (en) Method for gene amplification
US20040265860A1 (en) Production of proteins using homologous recombination
WO1989010959A1 (en) Supertransformants for high expression rates in eukaryotic cells
EP1913143A1 (en) Improved protein expression
KR20050084867A (en) High expression locus vector based on ferritin heavy chain gene locus

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP NO

AL Designated countries for regional patents

Kind code of ref document: A1

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

WWE Wipo information: entry into national phase

Ref document number: 2045175

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1991900640

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1991900640

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1991900640

Country of ref document: EP