WO2014066505A1 - Double knockout (gt/cmah-ko) pigs, organs and tissues - Google Patents

Double knockout (gt/cmah-ko) pigs, organs and tissues Download PDF

Info

Publication number
WO2014066505A1
WO2014066505A1 PCT/US2013/066387 US2013066387W WO2014066505A1 WO 2014066505 A1 WO2014066505 A1 WO 2014066505A1 US 2013066387 W US2013066387 W US 2013066387W WO 2014066505 A1 WO2014066505 A1 WO 2014066505A1
Authority
WO
WIPO (PCT)
Prior art keywords
base pair
cmah
cells
pig
human
Prior art date
Application number
PCT/US2013/066387
Other languages
French (fr)
Inventor
A. Joseph TECTOR
Original Assignee
Indiana University Research & Technology Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Indiana University Research & Technology Corporation filed Critical Indiana University Research & Technology Corporation
Priority to US14/436,963 priority Critical patent/US9888674B2/en
Publication of WO2014066505A1 publication Critical patent/WO2014066505A1/en
Priority to US15/848,870 priority patent/US10667500B2/en
Priority to US16/855,905 priority patent/US11666039B2/en
Priority to US18/101,810 priority patent/US20240196872A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/407Liver; Hepatocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • 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
    • C12N15/873Techniques for producing new embryos, e.g. nuclear transfer, manipulation of totipotent cells or production of chimeric embryos
    • C12N15/877Techniques for producing new mammalian cloned embryos
    • C12N15/8778Swine embryos
    • 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
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0071Oxidoreductases (1.) acting on paired donors with incorporation of molecular oxygen (1.14)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1048Glycosyltransferases (2.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • C12N9/2465Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1) acting on alpha-galactose-glycoside bonds, e.g. alpha-galactosidase (3.2.1.22)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/15Animals comprising multiple alterations of the genome, by transgenesis or homologous recombination, e.g. obtained by cross-breeding
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/108Swine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/02Animal zootechnically ameliorated
    • A01K2267/025Animal producing cells or organs for transplantation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/80Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor

Definitions

  • the present invention is generally in the field of xenotransplantation and genetic modification to produce transgenic animals, orga ns tissue, or cells suita ble for transplantation into a hu man.
  • transplants from one animal into another animal of the same species are a routine treatment option for many serious conditions, including kidney, heart, lung, liver and other organ disease.
  • transplants from one animal into another animal of the same species are a routine treatment option for many serious conditions, including kidney, heart, lung, liver and other organ disease.
  • there are not enough suita ble organs availa ble for transplant to meet current or expected clinical demands for organ transplants.
  • Xenotransplantation the transplant of organs, tissue or cells from one animal into another animal of a different species, such as the transplantation of a pig organ into a human recipient, has the potential to eliminate the shortage of organs ava ila ble for transplant, potentially helping hundreds of thousands of people worldwide. For instance, suita ble organs for transplant from non-human donors, such as from a pig, could help keep seriously ill patients alive, either permanently or temporarily, until a suita ble human organ is availa ble for transplant. [0007] While many mammalian animals may be suitable candidates for xenotransplantation, much of the current focus is on the pig.
  • pig organs tissue or cells for xenotransplantaion offers many advantages over other non-human mammalian donors. For instance, pigs are easily obtainable, they are inexpensive to breed and maintain, and, most importantly, many pig organs are similar to humans in size, shape and function.
  • xenotransplantation using standard, unmodified pig tissue into a human (or other primate) is accompanied by severe rejection of the transplanted tissue.
  • the rejection may be a hyperacute rejection, an acute rejection or a chronic rejection.
  • the hyperacute response to the pig antibodies present on the transplanted tissue is so strong that the transplant is typically damaged by the human immune system within minutes or hours of transplant into the human recipient.
  • Pig cells express al,3 galactosyltransferase (aGal) and cytidine monophosphate-N- acetylneuraminic acid hydroxylase (CMAH), which are not found in human cells.
  • the aGal enzyme produces the aGal epitope.
  • CMAH converts the sialic acid N-acetylneuraminic acid (Neu5Ac) to N- glycolylneuraminic acid (Neu5Gc). Accordingly, when pig tissue is transplanted into a human, these epitopes elicit an antibody-mediated rejection from the human patient immediately following implantation.
  • the antibodies are present in the patient's blood prior to implantation of the tissue, resulting in the intense and immediate rejection of the implanted tissue.
  • GTKO pig may have eliminated anti-aGal antibodies as a barrier to xenotransplantation
  • studies using GTKO cardiac and renal xenografts in baboons show that the GTKO organs still trigger an immunogenic response, resulting in rejection or damage to the transplanted organ.
  • Baboons transplanted with GTKO kidneys and treated with two different immunosuppressive regimens died within 16 days of surgery. Chen et al. concluded "genetic depletion of Gal antigens does not provide a major benefit in xenograft survival" (Chen et al., 2005, Nature Med. 11(12):1295-1298.
  • Basnet et al examined the cytotoxic response of human serum to CMAH-/- mouse cells. Basnet et al. concluded "the anti-NeuGc Ab-mediated immune response may be significantly involved in graft loss in xenogeneic cell transplantation, but not in organ transplantation" (Basnet et al., 2010, Xenotransplantation, 17(6):440-448).
  • This disclosure relates generally to methods of making porcine organs, tissues or cells for transplantation into a human that do not express aGal and CMAH.
  • the present disclosure provides, in one embodiment, a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)- galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and when tissue from said pig is transplanted into a human, hyperacute rejection is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
  • the specification provides porcine organs, tissue or cells for transplantation into a human having reduced expression of aGal and CMAH on the porcine organs, tissue or cells.
  • the specification provides a method for modifying a porcine organs, tissue or cells for transplantation into a human, the method comprising removing or reducing expression of aGal and CMAH on the porcine organs, tissue or cells.
  • the porcine organs, tissue, or cells may be selected from the group consisting of red blood cells, skin, heart, livers, kidneys, lung, pancreas, thyroid, small bowel, and components thereof.
  • the specification provides a method for making porcine organs, tissue or cells for transplantation into a human, the method comprising reducing expression of aGal and CMAH on the porcine organs, tissue or cells.
  • the porcine organs, tissue, or cells may be selected from the group consisting of red blood cells, skin, heart, liver, kidneys, lung, pancreas, small bowel, and components thereof.
  • the specification provides a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and wherein when tissue from a knockout pig is transplanted into a human, thrombocytopenia is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
  • the specification provides a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and wherein a liver from said pig exhibits reduced uptake of human platelets when said liver is exposed to said human platelets.
  • the specification provides a method of increasing the duration of the period between when a human subject is identified as a subject in need of human liver transplant and when said human liver transplant occurs, said method comprising providing a liver from a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type swine and surgically attaching said liver from said knockout pig to said human su bject in a therapeutically effective manner.
  • the liver from the knockout pig may be internal or external to the human subject, and may be directly or indirectly attached to the human subject.
  • the specification provides a method of preparing organs, tissues, or cells for xenotransplantation into human patients with reduced rejection, the method comprising providing a transgenic pig as a source of transplant material wherein the transplant material is selected from the group consisting of organs, tissues, or cells, and wherein the pig masks or reduces the expression of at least two xenoreactive antigens on the transplant material.
  • At least two xenoreactive antigens may be aGal and Neu5Gc.
  • the specification provides a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein the disruption of said a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; wherein the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and wherein expression of functional a(l,3)-galacto
  • the specification provides a method of improving symptoms of hyperacute rejection in a patient comprising transplanting porcine organs, tissue or cells having reduced expression of aGal and CMAH on the porcine organs, tissue or cells into a human, wherein the symptoms of hyperacute rejection are improved as compared to tissue from a wild-type swine when transplanted into a human.
  • the disclosure provides a cell culture reagent derived from a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein the disruption of said a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; wherein the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and wherein expression of functional
  • a cell culture reagent may be selected from the group of cell culture reagents comprising cell culture media, cell culture serum, a cell culture additive and an isolated cell capable of proliferation.
  • the invention provides a method of producing a glycoprotein of interest comprising the step of incubating an isolated cell capable of expressing the glycoprotein of interest with a cell culture reagent derived from a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein the amount of Neu5Gc or aGal epitopes on the glycoprotein of interest is lower than the amount of Neu5Gc or aGal epitopes on the glycoprotein of interest when an isolated cell capable of expressing said glycoprotein of interest is incubated with a cell culture reagent derived from a wild-type pig.
  • the glycoprotein of interest may be selected from the group comprising an antibody, growth factor, cytokine, hormone and clotting factor.
  • the disruption of the a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence;
  • the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and expression of the functional a(l,3) galactosyltransferase and CMAH in the
  • Figure 1 depicts a schematic of a protocol used to develop the double knock-out CMAH/GGTA1- pigs.
  • Step a) shows the delivery of CMAH ZFNs to liver derived cells.
  • Step b) illustrates identified CMAH mutant cells by screening individual clones.
  • Step c) illustrates the delivery of GGTA1 ZFNs to CMAH KO cells.
  • Step d) illustrates the counterselection of Gal negative cells.
  • Step e) illustrates CMAH and GGTA1 double KO cells.
  • Step f) illustrates somatic cell nuclear transfer (SCNT) resulting in CMAH and GGTAl double KO cells.
  • SCNT somatic cell nuclear transfer
  • Figure 2 provides genotype and phenotype analysis of double-KO fetuses.
  • Panel A presents a photographic image of harvested double-KO fetuses.
  • Panel B provides representative electropherograms of the mutations found in the CMAH gene in the double knockout fetuses. Mutations are underlined while the DNA binding sites of the ZFN are italicized.
  • Panel C provides representative electropherograms of the mutations found in the GGTAl gene in the double knockout fetuses. Mutations are underlined while the DNA binding sites of the ZFN are italicized.
  • Panel D presents data obtained from flow cytometric analysis of red blood cells (RBC) obtained from six month old wild-type piglets (WT), six-month-old GGTA1-KO (GGTA1-KO) piglets, double-KO fetuses and adult humans showing cells stained with an antibody recognizing Neu5GC.
  • Panel E presents data obtained from flow cytometric analysis of red blood cells (RBC) obtained from six month old wild-type piglets (WT), six-month-old GGTA1-KO (GGTA1-KO) piglets, double-KO fetuses and adult humans showing cells stained with fluorescently labeled IB4 lectin to measure the level of the Gal epitopes. Unstained RBC were used as negative controls for IB4 lectin staining and an isotype matched control was used for Neu5Gc staining. Some negative control histograms are difficult to see because of significant overlap with the experimental group.
  • Figure 3A presents a photograph of viable double-KO piglets.
  • Panel B provides sequence information regarding the wild-type (WT) sequences for the CMAH and GGTAl target regions. The alterations that occur in the double-KO piglets in either the CMAH or GGTAl target region are underlined, while the binding sites are italicized.
  • WT wild-type
  • Figure 4 presents results from a series of experiments analyzing carbohydrate expression in genetically modified pigs.
  • Panels A and B provide confocal micrographs of tissues from wild type (WT), single (GGTA1-KO) and CMAH-/-/GGTA1-/- (dou ble-KO) pigs. DAPI staining of nuclei may be visible. Heart, kidney and liver tissues were stained with anti-Neu5Gc antibody in the micrographs of panel A. Limited staining of Neu5Gc occurs in tissues from double-KO pigs. Heart, kidney and liver tissues were stained with IB4 lectin in the micrographs of panel B. Limited IB4 binding occurs in tissues from GGTA1- KO and double-KO pigs.
  • Panel C presents results obtained from flow cytometry analysis of peripheral blood mononuclear cells (PBMC). Traces were obtained from cells labeled with anti-Neu5Gc antibody (left column) and IB4 lectin (right column). Unstained PBMC were the negative controls for IB4 lectin; an isotype negative control was used in the anti-Neu5Gc staining. Negative controls are shown (*) but are difficult to see in some panels because of significant overlap with the experimental group.
  • PBMC peripheral blood mononuclear cells
  • Figure 5 presents results from a series of experiments examining human antibody recognition of PBMC from GGTA1-KO and double-KO pigs.
  • Panels A and B show IgG (A) and IgM (B) histograms of a representative sample (Subject 5) of randomly chosen normal human serum against GGTA1-KO or double-KO cells.
  • Bar graphs C and D show the mean fluorescent intensities (MFI) of IgM) or IgG binding to peripheral blood monocytes (PBMCs). The results of testing ten unique human subjects are shown.
  • Panels E and F illustrate example curves (Subject 3) for antibody-mediated complement-dependent cytotoxicity of normal human serum against GGTA1-KO and double KOPBMCs. Percent cytotoxicity for each serum tested (2.0% final concentration) is shown.
  • Figure 6 shows flow cytometry used to analyze human antibody binding to GT-KO and GT/CMAH-KO porcine fetal fibroblasts in a dose dependent manner.
  • GT/CMAH-KO cells bind less human IgM as compared to GT-KO porcine fetal fibroblasts.
  • Figure 7 provides panels pertaining to the generation of GGTA1/CMAH double knockout porcine LDCs through a C ISP /Cas9 technique. LDCs that survived IB4 lectin counter-selection were expanded and stained with IB4-FITC.
  • Figure 7A provides results of flow cytometry analysis of the a-Gal epitope on LDCs that survived IB4 lectin counter-selection. Also shown are unlabeled LDC and labeled, non- transfected LDC. In the particular example shown, 91.7% of the selected cells were free of the a-Gal epitope.
  • Figure 7B provides an electropherogram of mutations found in Crispr-targeted GGTA1 and CMAH regions. The wild-type sequence of a portion of each targeted region is shown above the sequencing results. The top electropherogram is a portion of the GGTA1 gene; the bottom electropherogram is a portion of the CMAH gene.
  • Figure 8 depicts flow cytometry results of labeled liver derived cells (LDCs) simultaneously transfected with CrispR CMAH and a-Gal targeting plasmids (CRISPR LDC); labeled, non-transfected LDC; and unlabeled LDC.
  • CMAH expression in the cells was evaluated by assessing the anti-Neu5Gc antibody binding to Neu5Gc epitopes on the cell surface.
  • CMAH produces Neu5Gc.
  • Panel B Unselected
  • a small percentage of the bulk transfected cell population shows a CMAH deficiency.
  • FIG. 9 provides panels pertaining to generation of CMAH/aGal DKO fetuses from CMAH- CrispR and aGal-CrispR transfected LDC.
  • Figure 9A provides a photograph of 10 fetuses harvested at day 32 of gestation after SCNT of Crispr-transfected LDC that survived IB4 counterselection and implantation of resulting embryos in recipient sows.
  • DNA sequencing analysis of the GGTA1 and CMAH target regions was performed on each fetus.
  • GGTA1 seven fetuses contained biallelic mutations, one fetus contained a monoallelic mutation, and two fetuses were wild-type.
  • CMAH five fetuses contained biallelic mutations, one fetus contained a monoallelic mutation, and four fetuses were wild-type.
  • Figure 9B provides the results of DNA sequencing analysis of the targeted GGTA1 and CMAH regions of five DKO fetuses with mutations in both alleles of both CMAH and GGTA1. If the two alleles have different mutations in a particular fetus, the sequence of both alleles is shown. If the two alleles of a fetus have the same mutation, the sequence is only shown once. The net change in nucleotide number is indicated to the right of the sequences. The wild-type sequence of the relevant portions of the GGTA1 and CMAH genes is shown above the fetal sequences. Fetal fibroblasts were derived from fetus 7.
  • Panel C provides data from flow cytometry analysis of aGal and Neu5Gc on double knockout (DKO) and wild-type (WT) fetal fibroblasts. Unlabeled DKO and WT cells were also analyzed. The upper trace shows aGal results; the lower trace shows Neu5Gc results.
  • DKO double knockout
  • WT wild-type
  • Figure 10 provides panels of flow cytometry analysis of Neu5Gc on CMAH/GAL double knockout fibroblast cells grown in media supplemented with either fetal bovine serum (FBS) or CMAH/aGAL double knockout derived serum (CMAH/Gal KO).
  • Figure 10A illustrates data from an isotype control.
  • Figure 10B provides Neu5Gc data. Cells grown in the CMAH deficient serum show lower anti-Neu5Gc antibody binding than cells grown in FBS.
  • Figure 11 provides panels of flow cytometry analysis of Neu5Gc on double knockout (CMAH/Gal KO) fibroblasts and single aGal knockout (GGTA KO) renal cells grown with a variety of cell culture reagents.
  • the upper panel provides flow cytometry results obtained from isotype control analysis of a CMAH/aGal DKO cell line (CMAH/Gal KO Fibroblast cell line) and aGall single knockout kidney cells (GGTA KO renal cells).
  • the lower panel provides flow cytometry results of Neu5Gc on a CMAH/aGal DKO cell line (CMAH/Gal KO Fibroblast cell line) grown in culture media supplemented with CMAH/aGal derived serum and a GGTA-1 kidney cell line grown in culture media supplemented with FBS.
  • CMAH/aGal DKO cell line CMAH/Gal KO Fibroblast cell line
  • Figure 12 provides a graph of data obtained from perfusion experiments involving porcine livers perfused with human platelets.
  • the post-perfusion initiation time in minutes is indicated on the x-axis (Time); the percent of human platelets remaining in the perfusion solution is indicated on the y-axis (Percent Platelets Remaining in Perfusion).
  • Figure 13 provides individual curves for antibody-mediated, complement-dependent cytotoxicity of 10 randomly selected baboon serum samples against single (GGTA1-KO) and GGTA1/CMAH double (double-KO) peripheral blood monocytes (PBMCs). Results from a human serum sample against single (GGTA1-KO) and double-KO PBMC are also shown on each graph. Unexpectedly cytotoxicity of the baboon serum to the CMAH/aGAL double knockout PBMC differed widely from cytotoxicity of human serum to CMAH/aGal PBMCs.
  • Cytotoxicity of baboon serum with CMAH/aGal double knockout PBMCs consistently increased as compared to cytotoxicity of baboon serum with aGal single knockout PBMCs.
  • the dashed horizontal line indicates 50% killing.
  • the %CTX for each sample was plotted against the log of each serum dilution and the sigmoid curve was analyzed by non-linear regression.
  • Figure 14 provides histograms showing results of flow cytometry analysis of IgM ( Figure 14) and IgG ( Figure 14B) antibody recognition. Histograms show the results of 5 randomly selected human sera (1, 2, 3, 4 and 5) incu bated with GGTA1-KO pig, double-KO pig, baboon, chimpanzee and human PBMCs. Secondary only control antibody staining is shown in the bottom graph in each column. In four of the five samples shown, IgG antibody recognition for CMAH/GAL DKO PBMC and human PBMC yield almost overlaying traces.
  • the IgM traces for the CMAH/GAL DKO PBMC are shifted closer to the traces for the human PBMC than the baboon, chimpanzee and single knockout (GGTA1-KO) traces.
  • the present invention provides pigs and porcine organs, tissues or cells for transplantation into a human that do not express aGal and CMAH and methods of making the same.
  • the invention provides a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and when tissue from said pig is transplanted into a human, hyperacute rejection is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
  • the present invention provides transgenic animals suitable for use in xenotransplantation and methods of producing mammals suitable for use in xenotransplantation.
  • the present invention describes the production of homozygous double knockout pigs lacking any functional expression of alpha 1,3 galactosyltransferase (aGAL) and CMAH.
  • aGAL alpha 1,3 galactosyltransferase
  • CMAH CMAH
  • SCNT somatic cell nuclear transfer
  • knockout mammal refers to a transgenic mammal wherein a given gene has been altered, removed or disrupted.
  • double knockout we mean a transgenic mammal wherein two genes have been altered, removed or disrupted.
  • the term is intended to include all progeny generations.
  • the founder animal and all Fl, F2, F3, and so on, progeny thereof are included.
  • knockout animals may have one or both copies of the gene sequence of interest disrupted. In the latter case, in which a homozygous disruption is present, the mutation is termed a "null" mutation. In the case where only one copy of the nucleic acid sequence of interest is disrupted, the knockout animal is termed a "heterozygous knockout animal".
  • the knockout animals of the invention are typically homozygous for disruptions of both genes being targeted.
  • chimera refers to a transgenic mammal with a knockout in some of its genome-containing cells.
  • heterozygote or “heterozygotic mammal” refers to a transgenic mammal with a disruption on one of a chromosome pair in all of its genome containing cells.
  • homozygote or “homozygotic mammal” refers to a transgenic mammal with a disruption on both members of a chromosome pair in all of its genome-containing cells.
  • a "non-human mammal" of the invention includes mammals such as rodents, sheep, dogs, ovine such as lamb, bovine such as beef cattle and milk cows, and swine such as pigs and hogs.
  • mammals such as rodents, sheep, dogs, ovine such as lamb, bovine such as beef cattle and milk cows, and swine such as pigs and hogs.
  • porcine a typical non-human animal
  • other mammals can similarly be genetically modified using the methods and compositions of the invention.
  • a “mutation” is a detecta ble change in the genetic material in the animal, which is transmitted to the animal's progeny.
  • a mutation is usually a change in one or more deoxyribonucleotides, such as, for example, adding, deleting, inverting, or substituting nucleotides.
  • pig we mean any pig known to the art, including a wild pig, a domestic pig, mini pigs, a Sus scrofa pig, a Sus scrofa domesticus pig, as well as inbred pigs.
  • the pig can be selected from the group consisting of, for example, Landrace, Hampshire, Duroc, Chinese Meishan, Chester White, Berkshire Goettingen, Landrace/York/Chester White, Yucatan, Bama Xiang Zhu, Wuzhishan, Xi Shuang Banna, and Pietrain pigs.
  • Porcine organs, tissue or cells are organs, tissue or cells from a pig.
  • the present invention provides a transgenic animal lacking any expression of functional aGal and CMAH genes.
  • the animal can be any mammal suitable for xenotransplantation.
  • the animal is a pig.
  • CMAH/aGAL double knockouts "CMAH/aGAL DKO”, “CMAH/aGal”, “CMAH/aGal DKO”, "CMAH 7 ⁇ /GAL.
  • the invention provides organs, tissue and/or cells from animals lacking any expression of functional aGal and CMAH for use as xenografts.
  • the tissues from animals lacking any functional expression of the aGal and CMAH gene can be obtained from a prenatal, neonatal, immature, or fully mature animal, such as a porcine, bovine or ovine.
  • the organ may be used as a temporary or permanent organ replacement for a patient in need of an organ transplant.
  • Any porcine organ can be used, including but not limited to the brain, heart, lungs, eye, stomach, pancreas, kidneys, liver, intestines, uterus, bladder, skin, hair, nails, ears, glands, nose, mouth, lips, spleen, gums, teeth, tongue, salivary glands, tonsils, pharynx, esophagus, large intestine, small intestine, rectum, anus, thyroid gland, thymus gland, bones, cartilage, tendons, ligaments, suprarenal capsule, skeletal muscles, smooth muscles, blood vessels, blood, spinal cord, trachea, ureters, urethra, hypothalamus, pituitary, pylorus, adrenal glands, ovaries, oviducts, vagina, mammary glands, testes, seminal vesicles, penis, lymph, lymph nodes and lymph vessels.
  • the invention provides non-human tissues that are useful for xenotransplantation.
  • the non-human tissue is porcine tissue. Any porcine tissue can be used, including but not limited to epithelium, connective tissue, blood, bone, cartilage, muscle, nerve, adenoid, adipose, areolar, bone, brown adipose, cancellous, muscle, cartilaginous, cavernous, chondroid, chromaffin, dartoic, elastic, epithelial, fatty, fibrohyaline, fibrous, Gamgee, gelatinous, granulation, gut-associated lymphoid, skeletal muscle, Haller's vascular, indifferent, interstitial, investing, islet, lymphatic, lymphoid, mesenchymal, mesonephric, multilocular adipose, mucous connective, myeloid, nasion soft, nephrogenic, nodal, osteoid, osse
  • the invention also provides cells and cell lines from porcine animals that lack expression of functional aGal and CMAH.
  • these cells or cell lines can be used for xenotransplantation.
  • Cells from any porcine tissue or organ can be used, including, but not limited to: epithelial cells, fibroblast cells, neural cells, keratinocytes, hematopoietic cells, melanocytes, chondrocytes, lymphocytes (B and T), macrophages, monocytes, mononuclear cells, cardiac muscle cells, other muscle cells, granulosa cells, cumulus cells, epidermal cells, endothelial cells, Islets of Langerhans cells, pancreatic insulin secreting cells, pancreatic alpha-2 cells, pancreatic beta cells, pancreatic alpha-1 cells, blood cells, blood precursor cells, bone cells, bone precursor cells, neuronal stem cells, primordial stem cells, hepatocytes, keratinocytes, umbilical vein endotheli
  • Nonviable derivatives include tissues stripped of viable cells by enzymatic or chemical treatment these tissue derivatives can be further processed through crosslinking or other chemical treatments prior to use in transplantation.
  • the derivatives include extracellular matrix derived from a variety of tissues, including skin, bone, urinary, bladder or organ submucosal tissues.
  • tendons, joints, and bones stripped of viable tissue to including but not limited to heart valves and other nonviable tissues as medical devices are provided.
  • serum or medium suitable for cell culture and isolated from a knockout pig of the invention are provided.
  • Components of porcine knockout organs, tissues or cells are also provided.
  • Components may also be modified through any means known in the art including but not limited to crosslinking and aldehyde crosslinking. Components may vary depending on the larger organ or tissue from which the component is obtained. Skin components may include but are not limited to stripped skin, collagen, epithelial cells, fibroblasts and dermis. Bone components may include but are not limited to collagen and extracellular matrix. Heart components may include but are not limited to valves and valve tissue.
  • Expression of a gene product is decreased when total expression of the gene product is decreased, a gene product of an altered size is produced or when the gene product exhibits an altered functionality. Thus if a gene expresses a wild-type amount of product but the product has an altered enzymatic activity, altered size, altered cellular localization pattern, altered receptor-ligand binding or other altered activity, expression of that gene product is considered decreased.
  • Expression may be analyzed by any means known in the art including, but not limited to, T-PC , Western blots, Northern blots, microarray analysis, immunoprecipitation, radiological assays, polypeptide purification, spectrophotometric analysis, Coomassie staining of acrylamide gels, ELISAs, 2-D gel electrophoresis, in situ hybridization, chemiluminescence, silver staining, enzymatic assays, ponceau S staining, multiplex T-PC , immunohistochemical assays, radioimmunoassay, colorimetric assays, immunoradiometric assays, positron emission tomography, fluorometric assays, fluorescence activated cell sorter staining of permeablized cells, radioimunnosorbent assays, real-time PCR, hybridization assays, sandwich immunoassays, flow cytometry, SAGE, differential amplification or electronic analysis.
  • T-PC Western
  • Expression may be analyzed directly or indirectly.
  • Indirect expression analysis may include but is not limited to, analyzing levels of a product catalyzed by an enzyme to evaluate expression of the enzyme. See for example, Ausubel et al, eds (2013) Current Protocols in Molecular Biology, Wiley-lnterscience, New York, N.Y. and Coligan et al (2013) Current Protocols in Protein Science, Wiley-lnterscience New York, NY. Gene expression assays for porcine ASGR1 are commercially available (Applied BiosystemsTM, Carlsbad CA).
  • As compared to is intended encompass comparing something to a similar but different thing, such as comparing a data point obtained from an experiment with a knockout pig to a data point obtained from a similar experiment with a wildtype pig.
  • the word "comparing” is intended to encompass examining character, qualities, values, quantities, or ratios in order to discover resemblances or differences between that which is being compared. Comparing may reveal a significant difference in that which is being compared.
  • significant difference is intended a statistically significant difference in results obtained for multiple groups such as the results for material from a knockout pig and material from a wild-type pig.
  • Statistical significance is assessed by a statistical significance test such as but not limited to the student's t-test, Chi-square, one-tailed t-test, two-tailed t-test, ANOVA, Dunett's post hoc test, Fisher's test and z-test.
  • a significant difference between two results may be results with a p ⁇ 0.1, p ⁇ 0.05, p ⁇ 0.04, p ⁇ 0.03, p ⁇ 0.02, p ⁇ 0.01 or greater.
  • isolated is intended to encompass an entity that is physically separated from another entity or group.
  • An isolated cell is physically separated from another group of cells. Examples of a group of cells include, but are not limited to, a developing cell mass, a cell culture, a cell line, a tissue, and an animal.
  • isolated is intended to encompass physically separating an entity from another entity or group. Examples include physically separating a cell from other cells, physically separating a cell component from the remainder of the cell and physically separating tissue or organ from an animal.
  • An isolated cell or cell component is separated by 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, up to 100% of other naturally occurring cells or cell components.
  • Methods for isolating one or more cells from another group of cells are known in the art. See for example Freshney (ED) Culture of Animal Cells: a manual of basic techniques (3 Ed.) 1994, Wiley-Liss; Spector et al (Eds)(1998) Cells: a Laboratory Manual (vol.1) Cold Spring Harbor Laboratory Press and Darling et al (1994) Animal Cells: culture and media John Wiley & Sons.
  • Methods of isolating a tissue or an organ from an animal are known in the art and vary depending on the tissue or organ to be isolated and the desired method of transplanting the tissue or organ.
  • a "skin related product” encompasses products isolated from skin and products intended for use with skin. Skin related products isolated from skin or other tissues may be modified before use with skin. Skin related products include but are not limited to replacement dressings, burn coverings, dermal products, replacement dermis, dermal fibroblasts,collagen, chondroitin, connective tissue, keratinocytes, cell-free xenodermis,cell-free pig dermis,composite skin su bstitutes and epidermis and temporary wound coverings. See for example Matou-Kovd et al (1994) Ann Med Burn Club 7:143, herein incorporated by reference in its entirety.
  • Xenotransplantation encompasses any procedure that involves the transplantation, implantation or infusion of cells, tissues or organs into a recipient subject from a different species. Xenotransplantation in which the recipient is a human is particularly envisioned. Thus xenotransplantation includes but is not limited to vascularized xenotransplant, partially vascularized xenotransplant, unvascularized xenotransplant, xenodressings, xenobandages, and xenostructures.
  • the invention provides a method of improving a hyperacute rejection related symptom in a patient comprising transplanting porcine organs, tissue or cells having reduced expression of aGal and Neu5Gc on the porcine organs, tissue or cells into a human, wherein the hyperacute rejection related symptoms are improved as compared to when tissue from a wild-type swine is transplanted into a human.
  • a hyperacute rejection related symptom may encompass a decrease, lessening, or diminishing of an undesirable symptom.
  • hyperacute rejection we mean rejection of the transplanted material or tissue occurring or beginning within the first 24 hours post-transplant involving one or more mechanisms of rejection. Hyperacute rejection encompasses but is not limited to “acute humoral rejection” and “antibody mediated rejection”.
  • Hyperacute rejection related symptom is intended to encompass any symptom known to the field as related to or caused by hyperacute rejection. It is recognized that hyperacute rejection related symptoms may vary depending upon the type of organ, tissue or cell that was transplanted.
  • Hyperacute rejection related symptoms may include, but are not limited to, thrombotic occlusion, hemorrhage of the graft vasculature, neutrophil influx, ischemia, mottling, cyanosis, edema, organ failure, reduced organ function, necrosis, glomerular capillary thrombosis, lack of function, hemolysis, fever, clotting, decreased bile production, asthenia, hypotension, oliguria, coagulopathy, elevated serum aminotransferase levels, elevated alkaline phosphatase levels, jaundice, lethargy, acidosis and hyperbilirubenemia and thrombocytopenia.
  • Thrombocytopenia is a quantity of platelets below the normal range of 140,000 to 440,000/ ⁇ .
  • Thrombocytopenia related symptoms include, but are not limited to, internal hemorrhage, intracranial bleeding, hematuria, hematemesis, bleeding gums, abdominal distension, melena, prolonged menstruation, epistaxis, ecchymosis, petechiae or purpura. Uptake of human platelets by pig livers contributes to the development of thrombocytopenia in xenograft recipients.
  • Platelets also known as thrombocytes, are enucleate fragments of megakaryocytes involved in blood coagulation, hemostasis and blood thrombus formation. Human platelets are routinely isolated through a variety of methods including, but not limited to, platelet apheresis, plateletpheresis and ultracentrifugation.
  • platelet uptake is intended to encompass the incorporation of a platelet into a liver or liver cell. While not being limited by mechanism, such uptake may occur through a phagocytic process. Platelet uptake may be monitored by any platelet uptake monitoring assay known in the art. Platelet uptake monitoring assays include, but are not limited to immunological methods, western blots, immunoblotting, microscopy, confocal microscopy, transmission electron microscopy and phagosome isolation. It is recognized that the appropriate platelet uptake monitoring assay may depend upon the type of label used.
  • Platelet uptake may be measured as a percentage of total platelets absorbed, percentage of total platelets not absorbed, a ratio of absorbed to unabsorbed platelets, percentage of cells absorbing at least one platelet, percentage of cells not absorbing a platelet, or number of platelets absorbed per cell. It is recognized that platelet uptake by more than one cell type may contribute to the total platelet uptake of the liver.
  • Total platelet uptake by an animal liver may include platelet uptake by liver sinusoidal endothelial cells, platelet uptake by Kuppffer cells, platelet uptake by LSECs and Kupffer cells and platelet uptake by additional cell types. It is recognized that platelet uptake by different cell types may contribute similar or disparate fractions of the total platelet uptake by a liver.
  • an alteration, inhibition, reduction, decrease, or lowering of platelet uptake by a liver comprises an alteration, inhibition, reduction, decrease, or lowering of platelet uptake by one or more liver cell types.
  • the word "providing” is intended to encompass preparing, procuring, getting ready, making ready, supplying or furnishing. It is recognized that methods of providing a cell may differ from methods of providing a subject and that methods of providing a liver may differ from methods of providing a pig.
  • a cell culture reagent derived from a transgenic pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes is provided.
  • Cell culture reagents are reagents utilized for tissue culture, in vitro tissue culture, microfluidic tissue culture, cell culture or other means of growing isolated cells or cell lines.
  • Cell culture reagents may include but are not limited to cell culture media, cell culture serum, a cell culture additive, a feeder cell, and an isolated cell capable of proliferation.
  • an isolated cell capable of proliferation is intended a cell isolated or partially isolated from other cell types or other cells wherein the cell is capable of proliferating, dividing, or multiplying into at least one additional clonal cell.
  • Cells grown in culture may synthesize or meta bolically incorporate antigenic epitopes into glycoproteins secreted by the cultured cell.
  • the antigenic epitopes may result in increased binding by human antibodies and decreased efficacy of the glycoprotein. See Ghaderi et al, 2010 Nature Biotechnology 28(8):863-867, herein incorporated by reference in its entirety.
  • Growing the producing cell in a cell culture reagent with less aGal or Neu5Gc may reduce the amount of aGal, Neu5Gc, or both aGal and Neu5Gc epitopes on the glycoprotein of interest.
  • Glycoproteins of interest may include any glycoprotein, particularly glycoproteins intended for use in human su bjects such as but not limited to, an antibody, growth factor, cytokine, hormone, or clotting factor.
  • xenoantigens aGal and Neu5Gc were eliminated in transgenic pigs by genetic modification.
  • Double knockout pigs (GT/CMAH-KO) were produced within 5-10 months or less.
  • tissues are provided in which both the aGal and CMAH genes are rendered inactive, such that the resultant aGal and CMAH products can no longer generate alpha 1,3-galactosyl epitopes or Neu5Gc on the cell surface.
  • the aGal and CMAH genes can be inactivated in such a way that no transcription of the gene occurs.
  • the present invention provides a method for producing viable pigs lacking any functional expression of aGal and CMAH.
  • the pigs are produced as described below. Methods of making transgenic pigs, and the challenges thereto, are discussed in Galli et al. 2010 Xenotransplantation, 17(6) p. 397-410, incorporated by reference herein for all purposes. The methods and cell cultures of the invention are further detailed below. EXAMPLES
  • a pair of ZFN were designed to bind and cleave the sequence of porcine CMAH exon 9 (SEQ ID NO: 1-AAACTCCTGAACTACAAGGCTCGGCTGGTGAAGGA) beginning at position 1,341 of Ensemble transcript ENSSSCT00000001195.
  • Another pair of ZFN were designed to bind and cleave the region of GGTA1 exon 8 (SEQ ID NO: 2-GTCATCTTTTACATCATGGTGGATGATATCTCCAGGATGCC) beginning at position 1165 of Ensemble transcript ENSSSCT00000006069.
  • the ZFN activities were validated in yeast (Sigma-Aldrich, St. Louis, MO). Additional detail regarding the ZFN of the present invention can be found in Li et al., Journal of Surgical Research, (2012)E1-E7 Epub 2012 July 3, which is incorporated by reference herein for all purposes.
  • LDC Porcine adult liver derived cells
  • LDC were harvested by trypsin digestion, washed with calcium and magnesium free Dulbecco's phosphate buffered saline (DPBS) (Invitrogen) and centrifuged. 10 s cells were suspended in 120 ⁇ of resuspension buffer (Invitrogen) containing 2 ⁇ gs of each CMAH ZFN plasmid. pEGFP-N 1 (Clonetech, Mountain View, CA) was used as a control to measure transfection efficiency as well as ZFN activity. Cells were electroporated at 1300 V, 30 msec for 1 pulse.
  • DPBS calcium and magnesium free Dulbecco's phosphate buffered saline
  • ZFN-induced mutation was detected using the Surveyor Mutation Detection kit (Transgenomic, Omaha, NE). At day 5 post-transfection, genomic DNA from ZFN-treated and control plasmid pEGFP-Nl treated cells was extracted and PCR was performed using primers ZFN-CMAH-F (SEQ ID NO: 3-5' GGACCTGCTTTATCTTGCTCGC 3'), ZFN-CMAH-R (SEQ ID NO: 4-5' CC ATACTTGTCTG CTGGGTGGG 3').
  • PCR product was denatured and annealed using the following program on a MyCycler (Bio-Rad): 95°C, 10 minutes; 95°C to 85°C, -2°C /second; 85°C to 25°C, -0.1°C /second. 200-400 ng of PCR product was digested with 1 ⁇ of Nuclease S and 1 ⁇ of enhancer (Transgenomic, Omaha, NE) at 42°C for 40 minutes. The product was separated on a 10% polyacrylamide gel and stained with SYBR Safe to assess ZFN-induced mutations.
  • CMAH-S1 SEQ ID NO: 5-5' CCAAACCCTGTCATTCCAG 3' was used to sequence the ZFN targeted CMAH region. A clone with an identical mutation in both CMAH gene copies was identified.
  • CMAH deficient LDC were treated with zinc finger plasmids targeting porcine 1,3 aGal.
  • LDC were transfected with Crispr CMAH sgRNA expression plasmids and GGTAl sgRNA expression plasmids and grown for 48 hours as described elsewhere herein. The cells were harvested, washed and counted.
  • the IB4 lectin is specific for the Gal epitope; cells that did not bind the Dynabeads remained in the supernatant.
  • CMAH cells that did not bind IB4 and the Dynabeads were identified as Gal-/- cells.
  • Crispr CMAH and GGTAl treated cells that did not bind IB4 and the Dynabeads were utilized in SCNT as described below herein.
  • TALEN constructs are designed to bind and cleave the sequence of porcine CMAH at a suitable site.
  • Additional Talen constructs are designed to bind and cleave GGTAl at a suitable site.
  • Somatic Cell Nuclear Transfer was performed using in vitro matured oocytes (DeSoto Biosciences Inc., St. Seymour TN and Minitube of America, Mount Horeb, Wl). Cumulus cells were removed from the oocytes by pipetting in 0.1% hyaluronidase. Oocytes with normal morphology and a visible polar body were selected and incubated in manipulation media (calcium-free NCSU-23 with 5% fetal bovine serum (FBS) containing 5 ⁇ g/ml bisbenzimide and 7.5 ⁇ g/mL cytochalasin B for 15 minutes.
  • manipulation media calcium-free NCSU-23 with 5% fetal bovine serum (FBS) containing 5 ⁇ g/ml bisbenzimide and 7.5 ⁇ g/mL cytochalasin B for 15 minutes.
  • oocytes were enucleated by removing the first polar body and metaphase II plate.
  • Single cells of CMAH deficient LDC that survived IB4 counterselection were injected into each enucleated oocyte.
  • Crispr transfected cells that survived IB4 counterselection were injected into each enucleated oocyte.
  • Electrical fusion was induced with a BTX cell electroporator (Harvard Apparatus, Holliston, MA).
  • Enucleated oocytes injected with a cell (couples) were exposed to two DC pulses of 140 V for 50 ⁇ in 280 mM mannitol, 0.001 mM CaCI 2 and 0.05 mM MgCI 2 .
  • reconstructed oocytes were activated by two DC pulses of 120 V for 60 ⁇ in 280 mM mannitol, 0. 1 mM CaCI 2 and 0.05 mM MgCI 2 .
  • oocytes were placed in NCSU-23 medium with 0.4% bovine serum albumin (BSA) and incubated at 38.5°C, 5% C0 2 in a humidified atmosphere for less than one hour.
  • BSA bovine serum albumin
  • oocytes were transferred into a recipient pig.
  • Recipient pigs were synchronized occidental pigs on their first day of estrus. Pregnancies were verified by ultrasound at day 25 or day 26 after embryo transfer.
  • Results of SCNT using double knockout cells are summarized in Table 1.
  • Results of SCNT using Crispr-transfected IB4 counterselected cells are summarized in Table 2.
  • Table 1 Results of somatic cell nuclear transfer using double-KO cells.
  • Dou ble-KO cells (cl22) were used as donor cells in SCNT and transferred into 2 recipients, 01 and 02. Fetal fibroblasts from fetus num ber 1 of recipient 02 were re-cloned and transferred to animals 03, 04, 05, and 06. 477 embryos were delivered to four gilts, resulting in one pregnancy and the birth of 5 piglets. The four via ble piglets appeared healthy as they were nursing since birth and needed no feeding support.
  • SCNT of the selected cells deficient for CMAH and GGTAl resulted in a pregnancy that was terminated at day 30 of gestation.
  • Five normal fetuses and one resorbed fetus were harvested, photogra phed and shown in Figure 1A.
  • Red blood cells from the dou ble knockout fetuses were analyzed by flow cytometry as described below herein.
  • Fibroblasts were grown from fetus number one. The fibroblasts were utilized in SCNT to generate 477 em bryos which were transferred into four recipient gilts. The SCNT resulted in one pregnancy and the birth of four live piglets and one still born piglet. The four via ble piglets needed no feeding support and appeared healthy. Three of the aGal/CMAH dou ble knockout pigs are shown in Figure 3A. The above described disruptions of the CMAH and GGTAl genes were validated in the four via ble pigs from the second pregnancy as described below herein. Results of somatic cell nuclear transfer using dou ble-KO cells are su mmarized in Ta ble 1 a bove.
  • CMAH complementary metal-oxide-semiconductor
  • five fetuses were biallelic mutations, one fetus was a monoallelic mutation and four fetuses were wild-type.
  • two homozygous mutations were found in fetus 7 at the GGTA1 locus and in fetus 2 at the CMAH locus.
  • the remaining mutations were heterozygous biallelic mutations. Results are summarized in Table 2 below and sequence data are presented in Figure 9B.
  • Fetal fibroblasts were harvested from one dou ble knockout fetus. The fetal fibroblasts were utilized in SCNT to generate embryos which were transferred into recipient sows. SCNT of the double knockout fetal fibroblasts resulted in at least one pregnancy.
  • Genomic DNA from double knockout Zn finger cloned fetuses and the four viable piglets was extracted using DNeasy Blood & Tissue Kit (Qiagen, Valencia, CA). PCR amplification of the CMAH region was performed as described above herein. [0095] Primer CMAH-S1 (SEQ ID NO: 5-5' CCAAACCCTGTCATTCCAG 3') and GGTA1-F primer (SEQ ID NO: 6-5' CTAGAAATCCCAGAGGTTAC 3') were used to sequence the targeted CMAH region and GGTAl region, respectively.
  • the GGTAl region was amplified by PCR using the GGTA1-F primer (SEQ ID NO: 6 (5' CTAGAAATCCCAGAGGTTAC 3')) and GGTA1-R primer (SEQ ID NO:7 (5'TCCTTGTCCTGGAGGATTCC3')).
  • Pwo Master Roche, Indianapolis IN
  • PCR conditions were as follows: 94°C, 2 min; 94°C, 15 s, 57°C, 30 s, and 72°C, 30s for 40 cycles; and a final extension step of 72°C for 5 min.
  • a total of 200-400 ng of PCR product was denatured and annealed using the following program on a Mycycler (Bio-Rad): 95°C, 10 min; 95° to 85°C, -2°C/s; 85°C to 25°C, -0.1°C/s.
  • One microliter of enhancer and 1 ⁇ of Nuclease S was added to each reaction and incubated at 42°C for 40 minutes. The product was separated on a 10% polyacrylamide gel and stained with SYBR Safe (Invitrogen USA Eugene OR).
  • Results from an exemplary DNA sequence analysis are shown in Figure 2B.
  • DNA sequence analysis confirmed homozygous alterations of the GGTAl and CMAH genes in at least one fetus.
  • the CMAH gene sequence is altered by a four base pair insertion.
  • the inserted sequence is "GGAA”.
  • the GGTAl gene sequence is altered by a 3 base pair deletion adjacent to a G to A substitution.
  • the sequences of the targeted CMAH and GGTAl regions of the genomic DNA of four viable piglets are shown in Figure 3B.
  • Genomic DNA from IB4 counter-selected cells and Crispr cloned fetuses was extracted using GenElute Mammalian Genomic DNA Miniprep Kit (Sigma Aldrich, St. Louis MO).
  • the GGTAl region was amplified by PCR using the GGTA1-F seq primer (SEQ ID NO: 8 (5' CCTTAGTATCCTTCCC AACCC AG AC-3' ) ) and GGTA1-R seq primer (SEQ ID NO: 9 (5' GCTTTCTTTACGGTGTCAGTGAATCC-3')).
  • the CMAH region was amplified by PCR using the CMAH-F seq primer (SEQ ID NO: 10 (5'- CTTGGAGGTGATTTGAGTTGGG-3')) and the CMAH-R seq primer (SEQ ID NO: 11 (5'- CATTTTCTTCGGAGTTGAGGGC-3')).
  • GGTAl Pwo Superyield DNA polymerase (Roche, Indianapolis IN) was used and PCR conditions for GGTAl were as follows: 94°C, 2 min; 94°C, 15 s, 54°C, 30 s, and 72°C, 45s for 15 cycles; 94°C, 15 s, 54°C, 30 s, and 72°C, 45s + 5 seconds/cycle for 25 cycles; and a final extension step of 72°C for 5 min.
  • CMAH CMAH-derived DNA sequencer
  • GenElute Gel Extraction kit Sigma-Aldrich, St. Louis MO
  • Sanger method DNA Sequencing Core Facility, Indiana University School of Medicine
  • CMAH-F seq (SEQ ID NO: 10) and GGTA1-F seq primer (SEQ ID NO: 8) were used to sequence the targeted CMAH region and GGTAl region, respectively.
  • both GGTAl alleles have an eight nucleotide insertion that substitute for a five nucleotide sequence.
  • one CMAH allele has a double base pair deletion (deletes TG), the other allele has a single nucleotide insertion (inserts T).
  • both CMAH alleles have an 8 base pair deletion.
  • one CMAH allele has a 5 nucleotide deletion, the other has a 3 nucleotide deletion.
  • one CMAH allele has a single base pair deletion (T), the other has single base pair deletion and a colocalized two base pair insertion.
  • CMAH allele has a 5 base pair deletion, the other has a 20 base pair deletion.
  • Sequences of the targeted CMAH and GGTAl regions of the genomic DNA of five fetuses are shown in Figure 9B.
  • Fetal livers were removed from double knockout fetuses and incubated in RPM I1640 for
  • RBCs were collected from cells released into the media after incubation of the fetal livers. RBCs were also obtained from adult human donors, six month old wild-type pigs and GGTA- knockout pigs (GGTA-KO pigs (fetal or 6 month old)). Porcine and human peripheral blood monocytes (PBMCs) were prepared using Ficoll-Paque Plus from whole blood collected in anticoagulant citrate dextrose (ACD).
  • ACD anticoagulant citrate dextrose
  • Neu5Gc antibody (Sialix, Vista CA). A negative control antibody for comparison with anti-Neu5Gc antibody was also used (Sialix, Vista CA). Cells were incubated with IB4 lectin for 20 minutes at 4°C. The cells were washed with blocking agent (Sialix, Vista, Ca) diluted in HBSS. Gal epitopes bind IB4 lectin. Cells were then stained with anti-Neu5Gc antibody for one hour at 4°C followed by Donkey anti-chicken DyLight 649 (Jackson ImmunoResearch Laboratories Inc, West Grove PA) for 40-60 minutes at 4°C. Cells stained with anti-Neu5Gc antibody were washed before and after secondary antibody with Sialix blocking agent diluted in PBS. In various experiments unstained BC or PBMC were used as negative controls for IB4 lectin staining. An Accuri C6 flow cytometer and CFlow Software (Accuri, Ann Arbor, Ml) were used for analysis.
  • One of the four double knockout (Double-KO, DKO, CMAH-/aGAL-) piglets with mutations generated by Zn-finger targeting was euthanized.
  • Liver, heart and kidney tissues were obtained from the double-ko pig.
  • Liver, heart and kidney tissues were also obtained from wild-type (WT) and GGTA1 knockout (GGTA-ko) pigs. Frozen sections of each tissue were prepared. Mounted tissues were blocked in Odyssey blocking buffer (Ll-Cor Biosciences, Lincoln NE) in HBSS for one hour. The slides were then fixed in 4% paraformaldehyde for 10 minutes.
  • Tissues were stained with IB4 lectin Alexa Fluor 647 (Invitrogen, Grand Island NY) to visualize the presence of the Gal epitope.
  • IB4 lectin Alexa Fluor 647 Invitrogen, Grand Island NY
  • tissues were stained with a chicken anti-Neu5Gc antibody or with a control antibody (Sialix, Vista, CA) for one hour.
  • Tissues were washed three times with HBSS.
  • Donkey anti- chicken DyLight 649 Jackson ImmunoResearch Laboratories Inc., West Grove PA
  • secondary antibody was incubated with the tissue for approximately one hour.
  • Tissues were washed three times with 0.1% HBSS Tween.
  • DAPI stain Invitrogen, Grand Island NY was added to all the slides for 1 minute followed by two 0.1% HBSS Tween washes. Tissues were mounted in ProLong Gold (Invitrogen, Grand Island NY). Confocal microscopy was performed using an Olympus FV1000.
  • Confocal microscopy indicated the presence of a-Gal and Neu5Gc in liver, heart and kidney tissues obtained from a wild-type pig.
  • GGTA1-KO pigs displayed only Neu5Gc in liver, heart and kidney tissues.
  • Confocal microscopy indicated the absence of a-Gal and Neu5Gc in liver, heart and kidney in tissues obtained from a double knockout piglet (exemplary fields are shown in Figure 4A and 4B).
  • Example 11 Crossmatch of Human Sera with GGTA1-KO and double-KO PBMCs.
  • Porcine whole blood from GGTA1-KO and dou ble-KO pigs was collected in ACD. Porcine
  • PBMCs were prepared from the whole blood using Ficoll-Paque Plus. Cell viability was assessed microscopically with Trypan Blue. Sera were obtained from ten healthy human volunteers. Twenty-five percent heat-inactivated serum was prepared. Approximately 2 X 10 6 /ml GGTA-KO and double-KO PBMCs were incubated with each human serum sample for 2 hours at 4°C. After incubation of the serum and PBMCs, the PBMCs were washed three times in 0.5% PBS Sialix Blocking agent.
  • PBMCs were stained with DyLight 649-conjugated Donkey anti-human IgM or DyLight 488 Donkey anti-human IgG (Jackson Immunoresearch Laboratories Inc, West Grove PA) for 1 hour at 4°C.
  • PBMCs were washed three times using 0.5% PBS Sialix blocking agent.
  • Analyses were performed using an Accuri C6 flow cytometer and BD CFlow Plus Software (Accuri, Ann Arbor Ml). Overlays were produced using Kaluza version 1.2 software from Beckman Coulter (Brea, CA).
  • a representative histogram is shown in Figure 5A. MFI's obtained from flow cytometry crossmatch analysis of 10 unique human subjects are also shown in Figure 5A.
  • MFI Mean fluorescent Intensities
  • Antibody-mediated complement dependent cytotoxicity assays are known in the art.
  • a modified method of Diaz et al (Diaz et al, 2004, Transplant Immunology 13(4):313-317) was performed.
  • Human serum was obtained from ten healthy volunteers ( Figures 5 and 6). Twenty-five percent heat- inactivated human serum was prepared. The heat-inactivated human sera were serially diluted and 100 ⁇ of each concentration were placed in a 96 well v-bottom assay plate. Sera were mixed with a 100 ⁇ aliquot of PBMC obtained from either GGTA1-KO or double-KO pigs.
  • the final concentration of PBMC in each well was 5 X 10 6 /ml; in some experiments 1 x 10 6 /ml PBMC were used.
  • the serum concentrations varied from 50%, 17%, 6%, 2%, 0.6%, 0.2% and 0.07%.
  • the mixtures were incubated for 30 minutes at 4°C. After 30 minutes, the plates were centrifuged for 4 minutes at 400 x g. The plates were decanted and washed with HBSS. Rabbit complement (150 ⁇ of a 1:15 dilution) was added to each well and incubated for 30 minutes at 37°C.
  • PBMC peripheral blood mononuclear cells
  • %CTX (%CTX eX p-%CTX S pont)/(100-%CTX S p O nt) x 100, where %CTX exp is the percentage of dead cells under the experimental condition.
  • Antibody-mediated, complement dependent cytotoxicity data was also analyzed as above.
  • the log of each serum dilution was plotted against the %CTX for each sample and the sigmoid curve was analyzed by non-linear regression to estimate the serum LD50.
  • the %CTX of GGTAl-KO and double-KO PBMCs at each serum dilution for each sample was analyzed using the Wilcoxon matched pairs signed rank test.
  • Example 14 Ex vivo Perfusion of Human Platelet through a Double KO Liver.
  • a double knockout CMAH/aGal pig is anesthetized and intubated. A midline abdominal incision is made. The liver is removed and placed in a perfusion device under normothermic conditions. Humidity, temperature and air flow are maintained in the perfusion device. Human platelets obtained from healthy volunteer subjects are circulated through the double knockout liver. Platelet levels in the pre-perfusion and post-perfusion samples are evaluated. Pre and post-perfusion evaluation of the pig liver is performed.
  • Example 15 Evaluation of Response to a Double Knockout (ctGal and CMAH) Xenograft.
  • a porcine liver obtained from double knockout (aGal-/CMAH-) pigs is surgically transplanted into a recently deceased human cadaver using the piggyback method. After the surgery, biological samples are obtained from the human cadaver. Clinical indicia of graft rejection are monitored.
  • Example 16 Evaluation of a Response to a Double Knockout (ctGal and CMAH) Xenograft.
  • Porcine kidneys are obtained from double knockout (GGTA1/CMAH double-KO) pigs. A highly sensitized human subject is administered compounds to manage preexisting and de novo donor- specific antibodies. Porcine double-KO kidneys are surgically transplanted into the subject. Clinical indicia of graft rejection are monitored.
  • Targeting sites for the CMAH and GGTA1 genes were identified. Forward and reverse oligonucleotides for each targeting site were obtained.
  • the GGTA1 forward sgRNA sequence was CACCGAGAAAATAATGAATGTCAA (SEQ ID NO: 12); the GGTA1 reverse sgRNA sequence was AAACTTGACATTCATTATTTTCTC (SEQ ID NO: 13).
  • the CMAH forward sgRNA sequence was CACCGAGTAAGGTACGTGATCTGT (SEQ ID NO: 14); the CMAH reverse sgRNA sequence was AAACACAGATCACGTACCTTACT (SEQ ID NO: 15).
  • the oligonucleotide pairs were annealed together to generate short double strand DNA fragments with Bbsl compatible overhangs.
  • the pX330 bicistronic expression vector expressing Cas9 and sgRNA of interest was linearized with Bbsl.
  • pX330 expresses a mammalian codon optimized Cas9.
  • the linearized pX330 expression vector and the double strand DNA fragments for each sg NA (GGTA1 and CMAH) were incubated with ligase.
  • LDCs Liver derived cells
  • LDC were cotransfected with pX330- CMAHsgRNA and pX300-GGTAlsgRNA expression plasmids using the Neon transfection system (Invitrogen) according to the manufacturer's instructions. Forty-eight hours after transfection, cells were harvested and used for isolectin B4 counter-selection.
  • IB4 Isolectin B4 counter-selected LDC, cloned fetal fibroblasts from DKO fetuses, wild- type LDC and wild-type fetal fibroblasts were stained with IB4 conjugated with FITC (Enzo Life Science, Farmingdale NY) to assess the aGal epitope level.
  • IB4 counter-selected LDC cloned fetal fibroblasts from DKO fetuses, wild-type LDC and wild-type fetal fibroblasts were stained with antiNeu5Gc antibody (Sialix, Vista CA) followed by donkey anti-chicken DyLight 649 (Jackson ImmunoResearch Laboratories Inc., West Grove PA).
  • a negative control antibody for comparison with anti-Neu5Gc antibody was also used (Sialix, Vista CA).
  • An Accuri C6 flow cytometer (Accuri, Ann Arbor Ml) and FlowJo software (Tree Star, Inc. Ashland OR) were used for analysis.
  • Representative flow cytometry results of aGal from IB4 counter selected cells are shown in Figure 7A.
  • Representative flow cytometry results of Neu5Gc on IB4 counter selected cells are shown in Figure 8A.
  • Representative flow cytometry results of the aGal and Neu5Gc on fetal fibroblasts derived from fetus 7 are shown in Figure 9C.
  • Example 20 Neu5Gc Levels in Cells Cultured with a DKO (aGal/CMAH) Piq Cell Culture Reagent.
  • Fibroblasts were cultured from a CMAH/aGal Double knockout pig. Because the DKO fibroblasts lack CMAH, they do not produce Neu5Gc. DKO serum was isolated from the blood of CMAH/aGAL pigs. The DKO fibroblasts were cultured in cell culture media supplemented with either DKO serum or bovine serum (FBS) for six weeks. After six weeks, cells were evaluated by flow cytometry with anti-Neu5Gc antibodies. Results from one such experiment are presented in Figure 10.
  • Example 21 Concordant Analysis of DKO Pig, Baboon and Chimpanzee Material.
  • Antibody-mediated complement dependent cytotoxicity assays are known in the art.
  • a modified method of Diaz et al (Diaz et al, 2004, Transplant Immunology 13(4):313-317) was performed. Serum samples were obtained from 10 randomly selected baboons. Twenty-five percent heat- inactivated baboon serum was prepared. The heat-inactivated baboon sera were serially diluted and 100 ⁇ of each concentration were placed in a 96 well v-bottom assay plate. Sera were mixed with a 100 ⁇ aliquot of PBMC obtained from either GGTA1-KO or double-KO (GGTA1/CMAH DKO) pigs. The final concentration of PBMC in each well was 5 X 10 6 /ml; in some experiments 1 x 1010 6 /ml PBMC were used.
  • Serum was obtained from five randomly selected human donors. Twenty-five percent heat-inactivated serum was prepared. Human, baboon and chimpanzee PBMCs were obtained. PBMCs were obtained from single aGal knockout pigs (GGTA1-KO) and double CMAH/aGal (double-KO) pigs. Approximately 2 X 10 6 /ml PBMCs were incubated with each human serum sample for 2 hours at 4°C. After incu bation of the serum and PBMCs, the PBMCs were washed three times in 0.5% PBS Sialix Blocking agent.
  • PBMCs were stained with DyLight 649-conjugated Donkey anti-human IgM or DyLight 488 Donkey anti-human IgG (Jackson Immunoresearch Laboratories Inc, West Grove PA) for 1 hour at 4°C.
  • PBMCs were washed three times using 0.5% PBS Sialix blocking agent.
  • Analyses were performed using an Accuri C6 flow cytometer and BD CFlow Plus Software (Accuri, Ann Arbor Ml). Overlays were produced using Kaluza version 1.2 software from Beckman Coulter (Brea, CA). Secondary only control antibody staining was also evaluated. Histograms from one such experiment are presented in Figure 14.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Developmental Biology & Embryology (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Environmental Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Physiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Nutrition Science (AREA)
  • Mycology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention provides double knockout transgenic pigs (GT/CMAH-KO pigs) lacking expression of any functional aGAL and CMAH. Double knockout GT/CMAH-KO transgenic organs, tissues and cells are also provided. Methods of making and using the GT/CMAH-KO pigs and tissue are also provided.

Description

DOUBLE KNOCKOUT (GT/CMAH-KO) PIGS, ORGANS AND TISSUES
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to, and the benefit of, U.S. Provisional Patent Application No:61/717,845, filed October 24, 2012, and U.S. Patent Application 13/804,365, filed March 14, 2013, both of which are incorporated herein by reference in their entirety for all purposes.
INCORPORATION OF SEQUENCE LISTING
[0002] The sequence listing in text format su bmitted herewith is herein incorporated by reference in its entirety for all purposes.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0003] Not applica ble.
FIELD OF THE INVENTION
[0004] The present invention is generally in the field of xenotransplantation and genetic modification to produce transgenic animals, orga ns tissue, or cells suita ble for transplantation into a hu man.
BACKGROUND
[0005] It is well known that transplants from one animal into another animal of the same species, such as human to human, are a routine treatment option for many serious conditions, including kidney, heart, lung, liver and other organ disease. However, it is also well known that there are not enough suita ble organs availa ble for transplant to meet current or expected clinical demands for organ transplants.
[0006] Xenotransplantation, the transplant of organs, tissue or cells from one animal into another animal of a different species, such as the transplantation of a pig organ into a human recipient, has the potential to eliminate the shortage of organs ava ila ble for transplant, potentially helping hundreds of thousands of people worldwide. For instance, suita ble organs for transplant from non-human donors, such as from a pig, could help keep seriously ill patients alive, either permanently or temporarily, until a suita ble human organ is availa ble for transplant. [0007] While many mammalian animals may be suitable candidates for xenotransplantation, much of the current focus is on the pig. Using pig organs, tissue or cells for xenotransplantaion offers many advantages over other non-human mammalian donors. For instance, pigs are easily obtainable, they are inexpensive to breed and maintain, and, most importantly, many pig organs are similar to humans in size, shape and function.
[0008] However, xenotransplantation using standard, unmodified pig tissue into a human (or other primate) is accompanied by severe rejection of the transplanted tissue. The rejection may be a hyperacute rejection, an acute rejection or a chronic rejection. The hyperacute response to the pig antibodies present on the transplanted tissue is so strong that the transplant is typically damaged by the human immune system within minutes or hours of transplant into the human recipient.
[0009] Pig cells express al,3 galactosyltransferase (aGal) and cytidine monophosphate-N- acetylneuraminic acid hydroxylase (CMAH), which are not found in human cells. The aGal enzyme produces the aGal epitope. CMAH converts the sialic acid N-acetylneuraminic acid (Neu5Ac) to N- glycolylneuraminic acid (Neu5Gc). Accordingly, when pig tissue is transplanted into a human, these epitopes elicit an antibody-mediated rejection from the human patient immediately following implantation. The antibodies are present in the patient's blood prior to implantation of the tissue, resulting in the intense and immediate rejection of the implanted tissue.
[0010] Many strategies have been employed to address the rejection caused by aGal and CMAH, including removing the genes encoding aGal or CMAH to prevent expression of the enzymes, modifying the genes encoding aGal or CMAH to reduce or limit expression of the enzymes or otherwise limiting the ability of the enzymes to trigger a rejection response. For instance, U.S Patent 7,547,816 to Day et al. describes a knockout pig with decreased expression of aGal as compared to wild-type pigs. U.S. Patents 7, 166,278 and 8,034,330 to Zhu et al. describe methods for making porcine organs for transplantation that are less likely to be subject to hyperacute rejection. However, progress in this field is critically dependent upon the development of the genetically modified pigs.
[0011] Unfortunately, developing homozygous knockout pigs is a slow process, requiring as long as three years using homologous recombination in fetal fibroblasts followed by somatic cell nuclear transfer (SCNT), and then breeding of heterozygous knockout animals. The development of new knockout pigs for xenotransplantation has been hampered by the lack of pluripotent stem cells, relying instead on the fetal fibroblast as the cell upon which genetic engineering was carried out. For instance, the production of the first live pigs lacking any functional expression of aGal (GTKO) was first reported in 2003. U.S. Patent 7,795,493 to Phelps et al. describes a method for the production of a pig that lacks any expression of functional aGal.
[0012] Unfortunately, while the GTKO pig may have eliminated anti-aGal antibodies as a barrier to xenotransplantation, studies using GTKO cardiac and renal xenografts in baboons show that the GTKO organs still trigger an immunogenic response, resulting in rejection or damage to the transplanted organ. Baboons transplanted with GTKO kidneys and treated with two different immunosuppressive regimens died within 16 days of surgery. Chen et al. concluded "genetic depletion of Gal antigens does not provide a major benefit in xenograft survival" (Chen et al., 2005, Nature Med. 11(12):1295-1298. Basnet et al examined the cytotoxic response of human serum to CMAH-/- mouse cells. Basnet et al. concluded "the anti-NeuGc Ab-mediated immune response may be significantly involved in graft loss in xenogeneic cell transplantation, but not in organ transplantation" (Basnet et al., 2010, Xenotransplantation, 17(6):440-448).
[0013] Thus, there is a need in the art for an improved, simple, replica ble, efficient and standardized method of producing double knockout (aGAL and CMAH) pigs having no aGAL and CMAH expression (GT/CMAH-KO) as a source of human transplant material for organs, tissue and cells for human transplant recipients.
BRIEF SUMMARY
[0014] This disclosure relates generally to methods of making porcine organs, tissues or cells for transplantation into a human that do not express aGal and CMAH.
[0015] The present disclosure provides, in one embodiment, a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)- galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and when tissue from said pig is transplanted into a human, hyperacute rejection is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
[0016] In another embodiment, the specification provides porcine organs, tissue or cells for transplantation into a human having reduced expression of aGal and CMAH on the porcine organs, tissue or cells.
[0017] In another embodiment, the specification provides a method for modifying a porcine organs, tissue or cells for transplantation into a human, the method comprising removing or reducing expression of aGal and CMAH on the porcine organs, tissue or cells. The porcine organs, tissue, or cells may be selected from the group consisting of red blood cells, skin, heart, livers, kidneys, lung, pancreas, thyroid, small bowel, and components thereof.
[0018] In another embodiment, the specification provides a method for making porcine organs, tissue or cells for transplantation into a human, the method comprising reducing expression of aGal and CMAH on the porcine organs, tissue or cells. The porcine organs, tissue, or cells may be selected from the group consisting of red blood cells, skin, heart, liver, kidneys, lung, pancreas, small bowel, and components thereof.
[0019] In another embodiment, the specification provides a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and wherein when tissue from a knockout pig is transplanted into a human, thrombocytopenia is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
[0020] In another embodiment, the specification provides a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and wherein a liver from said pig exhibits reduced uptake of human platelets when said liver is exposed to said human platelets.
[0021] In another embodiment, the specification provides a method of increasing the duration of the period between when a human subject is identified as a subject in need of human liver transplant and when said human liver transplant occurs, said method comprising providing a liver from a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type swine and surgically attaching said liver from said knockout pig to said human su bject in a therapeutically effective manner. The liver from the knockout pig may be internal or external to the human subject, and may be directly or indirectly attached to the human subject.
[0022] In another embodiment, the specification provides a method of preparing organs, tissues, or cells for xenotransplantation into human patients with reduced rejection, the method comprising providing a transgenic pig as a source of transplant material wherein the transplant material is selected from the group consisting of organs, tissues, or cells, and wherein the pig masks or reduces the expression of at least two xenoreactive antigens on the transplant material. At least two xenoreactive antigens may be aGal and Neu5Gc. [0023] In another embodiment, the specification provides a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein the disruption of said a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; wherein the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and wherein expression of functional a(l,3)-galactosyltransferase and CMAH in said knockout pig is decreased as compared to a wild-type pig and when tissue from said knockout pig is transplanted into a human, a hyperacute rejection related symptom is improved as compared to when tissue from a wild-type pig is transplanted into a human.
[0024] In another embodiment, the specification provides a method of improving symptoms of hyperacute rejection in a patient comprising transplanting porcine organs, tissue or cells having reduced expression of aGal and CMAH on the porcine organs, tissue or cells into a human, wherein the symptoms of hyperacute rejection are improved as compared to tissue from a wild-type swine when transplanted into a human.
[0025] In another embodiment, the disclosure provides a cell culture reagent derived from a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein the disruption of said a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; wherein the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and wherein expression of functional a(l,3)-galactosyltransferase and CMAH in said knockout pig is decreased as compared to a wild-type pig. A cell culture reagent may be selected from the group of cell culture reagents comprising cell culture media, cell culture serum, a cell culture additive and an isolated cell capable of proliferation. [0026] In a further embodiment, the invention provides a method of producing a glycoprotein of interest comprising the step of incubating an isolated cell capable of expressing the glycoprotein of interest with a cell culture reagent derived from a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein the amount of Neu5Gc or aGal epitopes on the glycoprotein of interest is lower than the amount of Neu5Gc or aGal epitopes on the glycoprotein of interest when an isolated cell capable of expressing said glycoprotein of interest is incubated with a cell culture reagent derived from a wild-type pig. The glycoprotein of interest may be selected from the group comprising an antibody, growth factor, cytokine, hormone and clotting factor. In an aspect of the disclosure, the disruption of the a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and expression of the functional a(l,3) galactosyltransferase and CMAH in the knockout pig from which the cell culture reagent is derived is decreased as compared to a wild-type pig.
[0027] While multiple embodiments are disclosed, still other embodiments of the present invention will become apparent to those skilled in the art from the following detailed description. As will be apparent, the invention is capable of modifications in various obvious aspects, all without departing from the spirit and scope of the present invention. Accordingly, the detailed descriptions are to be regarded as illustrative in nature and not restrictive.
BRIEF DESCRIPTION OF THE DRAWINGS
[0028] The disclosure will be better understood and features, aspects and advantages other than those set forth above will become apparent when consideration is given to the following detailed description thereof. Such detailed description makes reference to the following drawings.
[0029] Figure 1 depicts a schematic of a protocol used to develop the double knock-out CMAH/GGTA1- pigs. Step a) shows the delivery of CMAH ZFNs to liver derived cells. Step b) illustrates identified CMAH mutant cells by screening individual clones. Step c) illustrates the delivery of GGTA1 ZFNs to CMAH KO cells. Step d) illustrates the counterselection of Gal negative cells. Step e) illustrates CMAH and GGTA1 double KO cells. Step f) illustrates somatic cell nuclear transfer (SCNT) resulting in CMAH and GGTAl double KO cells.
[0030] Figure 2 provides genotype and phenotype analysis of double-KO fetuses. Panel A presents a photographic image of harvested double-KO fetuses. Panel B provides representative electropherograms of the mutations found in the CMAH gene in the double knockout fetuses. Mutations are underlined while the DNA binding sites of the ZFN are italicized. Panel C provides representative electropherograms of the mutations found in the GGTAl gene in the double knockout fetuses. Mutations are underlined while the DNA binding sites of the ZFN are italicized. Panel D presents data obtained from flow cytometric analysis of red blood cells (RBC) obtained from six month old wild-type piglets (WT), six-month-old GGTA1-KO (GGTA1-KO) piglets, double-KO fetuses and adult humans showing cells stained with an antibody recognizing Neu5GC. Panel E presents data obtained from flow cytometric analysis of red blood cells (RBC) obtained from six month old wild-type piglets (WT), six-month-old GGTA1-KO (GGTA1-KO) piglets, double-KO fetuses and adult humans showing cells stained with fluorescently labeled IB4 lectin to measure the level of the Gal epitopes. Unstained RBC were used as negative controls for IB4 lectin staining and an isotype matched control was used for Neu5Gc staining. Some negative control histograms are difficult to see because of significant overlap with the experimental group.
[0031] Figure 3A presents a photograph of viable double-KO piglets. Panel B provides sequence information regarding the wild-type (WT) sequences for the CMAH and GGTAl target regions. The alterations that occur in the double-KO piglets in either the CMAH or GGTAl target region are underlined, while the binding sites are italicized.
[0032] Figure 4 presents results from a series of experiments analyzing carbohydrate expression in genetically modified pigs. Panels A and B provide confocal micrographs of tissues from wild type (WT), single (GGTA1-KO) and CMAH-/-/GGTA1-/- (dou ble-KO) pigs. DAPI staining of nuclei may be visible. Heart, kidney and liver tissues were stained with anti-Neu5Gc antibody in the micrographs of panel A. Limited staining of Neu5Gc occurs in tissues from double-KO pigs. Heart, kidney and liver tissues were stained with IB4 lectin in the micrographs of panel B. Limited IB4 binding occurs in tissues from GGTA1- KO and double-KO pigs. Panel C presents results obtained from flow cytometry analysis of peripheral blood mononuclear cells (PBMC). Traces were obtained from cells labeled with anti-Neu5Gc antibody (left column) and IB4 lectin (right column). Unstained PBMC were the negative controls for IB4 lectin; an isotype negative control was used in the anti-Neu5Gc staining. Negative controls are shown (*) but are difficult to see in some panels because of significant overlap with the experimental group.
[0033] Figure 5 presents results from a series of experiments examining human antibody recognition of PBMC from GGTA1-KO and double-KO pigs. Panels A and B show IgG (A) and IgM (B) histograms of a representative sample (Subject 5) of randomly chosen normal human serum against GGTA1-KO or double-KO cells. Bar graphs C and D show the mean fluorescent intensities (MFI) of IgM) or IgG binding to peripheral blood monocytes (PBMCs). The results of testing ten unique human subjects are shown. Panels E and F illustrate example curves (Subject 3) for antibody-mediated complement-dependent cytotoxicity of normal human serum against GGTA1-KO and double KOPBMCs. Percent cytotoxicity for each serum tested (2.0% final concentration) is shown.
[0034] Figure 6 shows flow cytometry used to analyze human antibody binding to GT-KO and GT/CMAH-KO porcine fetal fibroblasts in a dose dependent manner. GT/CMAH-KO cells bind less human IgM as compared to GT-KO porcine fetal fibroblasts.
[0035] Figure 7 provides panels pertaining to the generation of GGTA1/CMAH double knockout porcine LDCs through a C ISP /Cas9 technique. LDCs that survived IB4 lectin counter-selection were expanded and stained with IB4-FITC. Figure 7A provides results of flow cytometry analysis of the a-Gal epitope on LDCs that survived IB4 lectin counter-selection. Also shown are unlabeled LDC and labeled, non- transfected LDC. In the particular example shown, 91.7% of the selected cells were free of the a-Gal epitope. Figure 7B provides an electropherogram of mutations found in Crispr-targeted GGTA1 and CMAH regions. The wild-type sequence of a portion of each targeted region is shown above the sequencing results. The top electropherogram is a portion of the GGTA1 gene; the bottom electropherogram is a portion of the CMAH gene.
[0036] Figure 8 depicts flow cytometry results of labeled liver derived cells (LDCs) simultaneously transfected with CrispR CMAH and a-Gal targeting plasmids (CRISPR LDC); labeled, non-transfected LDC; and unlabeled LDC. CMAH expression in the cells was evaluated by assessing the anti-Neu5Gc antibody binding to Neu5Gc epitopes on the cell surface. CMAH produces Neu5Gc. As shown in Panel B (Unselected) a small percentage of the bulk transfected cell population shows a CMAH deficiency. As shown in Panel A, after IB4 lectin counter-selection of the bulk cell population, a significantly higher percentage of the IB4 nonbinding cells (IB4 lectin selected) were CMAH deficient cells. In one experiment, 41.6% of the IB4 counter-selected cells were CMAH deficient while 6.7% of unselected bulk transfected cells were CMAH deficient. [0037] Figure 9 provides panels pertaining to generation of CMAH/aGal DKO fetuses from CMAH- CrispR and aGal-CrispR transfected LDC. Figure 9A provides a photograph of 10 fetuses harvested at day 32 of gestation after SCNT of Crispr-transfected LDC that survived IB4 counterselection and implantation of resulting embryos in recipient sows. DNA sequencing analysis of the GGTA1 and CMAH target regions was performed on each fetus. For GGTA1, seven fetuses contained biallelic mutations, one fetus contained a monoallelic mutation, and two fetuses were wild-type. For CMAH, five fetuses contained biallelic mutations, one fetus contained a monoallelic mutation, and four fetuses were wild-type. Figure 9B provides the results of DNA sequencing analysis of the targeted GGTA1 and CMAH regions of five DKO fetuses with mutations in both alleles of both CMAH and GGTA1. If the two alleles have different mutations in a particular fetus, the sequence of both alleles is shown. If the two alleles of a fetus have the same mutation, the sequence is only shown once. The net change in nucleotide number is indicated to the right of the sequences. The wild-type sequence of the relevant portions of the GGTA1 and CMAH genes is shown above the fetal sequences. Fetal fibroblasts were derived from fetus 7. Panel C provides data from flow cytometry analysis of aGal and Neu5Gc on double knockout (DKO) and wild-type (WT) fetal fibroblasts. Unlabeled DKO and WT cells were also analyzed. The upper trace shows aGal results; the lower trace shows Neu5Gc results.
[0038] Figure 10 provides panels of flow cytometry analysis of Neu5Gc on CMAH/GAL double knockout fibroblast cells grown in media supplemented with either fetal bovine serum (FBS) or CMAH/aGAL double knockout derived serum (CMAH/Gal KO). Figure 10A illustrates data from an isotype control. Figure 10B provides Neu5Gc data. Cells grown in the CMAH deficient serum show lower anti-Neu5Gc antibody binding than cells grown in FBS.
[0039] Figure 11 provides panels of flow cytometry analysis of Neu5Gc on double knockout (CMAH/Gal KO) fibroblasts and single aGal knockout (GGTA KO) renal cells grown with a variety of cell culture reagents. The upper panel provides flow cytometry results obtained from isotype control analysis of a CMAH/aGal DKO cell line (CMAH/Gal KO Fibroblast cell line) and aGall single knockout kidney cells (GGTA KO renal cells). The lower panel provides flow cytometry results of Neu5Gc on a CMAH/aGal DKO cell line (CMAH/Gal KO Fibroblast cell line) grown in culture media supplemented with CMAH/aGal derived serum and a GGTA-1 kidney cell line grown in culture media supplemented with FBS.
[0040] Figure 12 provides a graph of data obtained from perfusion experiments involving porcine livers perfused with human platelets. The post-perfusion initiation time in minutes is indicated on the x-axis (Time); the percent of human platelets remaining in the perfusion solution is indicated on the y-axis (Percent Platelets Remaining in Perfusion). Data obtained from experiments with single aGal knockout pig livers (GGTA17-, n=2) and CMAH/aGal DKO livers (CMAH7~/GGTA17 ~ , n=3) are provided.
[0041] Figure 13 provides individual curves for antibody-mediated, complement-dependent cytotoxicity of 10 randomly selected baboon serum samples against single (GGTA1-KO) and GGTA1/CMAH double (double-KO) peripheral blood monocytes (PBMCs). Results from a human serum sample against single (GGTA1-KO) and double-KO PBMC are also shown on each graph. Unexpectedly cytotoxicity of the baboon serum to the CMAH/aGAL double knockout PBMC differed widely from cytotoxicity of human serum to CMAH/aGal PBMCs. Cytotoxicity of baboon serum with CMAH/aGal double knockout PBMCs consistently increased as compared to cytotoxicity of baboon serum with aGal single knockout PBMCs. The dashed horizontal line indicates 50% killing. The %CTX for each sample was plotted against the log of each serum dilution and the sigmoid curve was analyzed by non-linear regression.
[0042] Figure 14 provides histograms showing results of flow cytometry analysis of IgM (Figure 14) and IgG (Figure 14B) antibody recognition. Histograms show the results of 5 randomly selected human sera (1, 2, 3, 4 and 5) incu bated with GGTA1-KO pig, double-KO pig, baboon, chimpanzee and human PBMCs. Secondary only control antibody staining is shown in the bottom graph in each column. In four of the five samples shown, IgG antibody recognition for CMAH/GAL DKO PBMC and human PBMC yield almost overlaying traces. In the same four samples shown, the IgM traces for the CMAH/GAL DKO PBMC are shifted closer to the traces for the human PBMC than the baboon, chimpanzee and single knockout (GGTA1-KO) traces.
DETAILED DESCRIPTION OF THE INVENTION
[0043] The present invention provides pigs and porcine organs, tissues or cells for transplantation into a human that do not express aGal and CMAH and methods of making the same. In one embodiment, the invention provides a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and when tissue from said pig is transplanted into a human, hyperacute rejection is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
I. In General. [0044] In the specification and in the claims, the terms "including" and "comprising" are open-ended terms and should be interpreted to mean "including, but not limited to. . . . " These terms encompass the more restrictive terms "consisting essentially of" and "consisting of."
[0045] As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. As well, the terms "a" (or "an"), "one or more" and "at least one" can be used interchangeably herein. It is also to be noted that the terms "comprising", "including", "characterized by" and "having" can be used interchangeably.
[0046] Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications and patents specifically mentioned herein are incorporated by reference in their entirety for all purposes including describing and disclosing the chemicals, instruments, statistical analyses and methodologies which are reported in the publications which might be used in connection with the invention. All references cited in this specification are to be taken as indicative of the level of skill in the art. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
II. The Invention.
[0047] The present invention provides transgenic animals suitable for use in xenotransplantation and methods of producing mammals suitable for use in xenotransplantation. Specifically, the present invention describes the production of homozygous double knockout pigs lacking any functional expression of alpha 1,3 galactosyltransferase (aGAL) and CMAH. In one embodiment, the production of pigs with copies of the aGal and CMAH genes knocked out prior to somatic cell nuclear transfer (SCNT) is described. The time to create a two gene homozygous knockout took less than 10 months, significantly reducing the time required to create new pigs to speed up the progress of xenotransplantation research.
[0048] The term "knockout mammal" refers to a transgenic mammal wherein a given gene has been altered, removed or disrupted. By a "double knockout" we mean a transgenic mammal wherein two genes have been altered, removed or disrupted. It is to be emphasized that the term is intended to include all progeny generations. Thus, the founder animal and all Fl, F2, F3, and so on, progeny thereof are included. In principle, knockout animals may have one or both copies of the gene sequence of interest disrupted. In the latter case, in which a homozygous disruption is present, the mutation is termed a "null" mutation. In the case where only one copy of the nucleic acid sequence of interest is disrupted, the knockout animal is termed a "heterozygous knockout animal". The knockout animals of the invention are typically homozygous for disruptions of both genes being targeted.
[0049] The term "chimera," "mosaic" or "chimeric mammal" refers to a transgenic mammal with a knockout in some of its genome-containing cells.
[0050] The term "heterozygote" or "heterozygotic mammal" refers to a transgenic mammal with a disruption on one of a chromosome pair in all of its genome containing cells.
[0051] The term "homozygote" or "homozygotic mammal" refers to a transgenic mammal with a disruption on both members of a chromosome pair in all of its genome-containing cells.
[0052] A "non-human mammal" of the invention includes mammals such as rodents, sheep, dogs, ovine such as lamb, bovine such as beef cattle and milk cows, and swine such as pigs and hogs. Although the invention uses a typical non-human animal (e.g., porcine), other mammals can similarly be genetically modified using the methods and compositions of the invention.
[0053] A "mutation" is a detecta ble change in the genetic material in the animal, which is transmitted to the animal's progeny. A mutation is usually a change in one or more deoxyribonucleotides, such as, for example, adding, deleting, inverting, or substituting nucleotides.
[0054] By "pig" we mean any pig known to the art, including a wild pig, a domestic pig, mini pigs, a Sus scrofa pig, a Sus scrofa domesticus pig, as well as inbred pigs. Without limitation, the pig can be selected from the group consisting of, for example, Landrace, Yorkshire, Hampshire, Duroc, Chinese Meishan, Chester White, Berkshire Goettingen, Landrace/York/Chester White, Yucatan, Bama Xiang Zhu, Wuzhishan, Xi Shuang Banna, and Pietrain pigs. Porcine organs, tissue or cells are organs, tissue or cells from a pig.
[0055] Transgenic Animals. The present invention provides a transgenic animal lacking any expression of functional aGal and CMAH genes. The animal can be any mammal suitable for xenotransplantation. In a specific embodiment, the animal is a pig. "CMAH/aGAL double knockouts", "CMAH/aGAL DKO", "CMAH/aGal", "CMAH/aGal DKO", "CMAH7~/GAL.7~", "aGal/CMAH DKOs", "aGAL/CMAH double knockouts", "GGTA1/CMAH DKO", "GT1/CMAH DKO", "GGTA17~/CMAH7~", "GGT17~/CMAH7", "CMAH/GGTA DKO", "GT/CMAH-KO", "GGTA1/CMAH KO", "DKO (aGal/CMAH)", "DKO (aGAL & CMAH)", "CMAH-/aGal-", "aGal-/CMAH-", "CMAH-/aGAL-" and variants thereof refer to animals, cells, or tissues that lack expression of functional alpha 1,3 galactosyltransferase and cytidine monophosphate-N- acetylneuraminic acid hydroxylase. [0056] Transgenic Material. In another embodiment, the invention provides organs, tissue and/or cells from animals lacking any expression of functional aGal and CMAH for use as xenografts. The tissues from animals lacking any functional expression of the aGal and CMAH gene can be obtained from a prenatal, neonatal, immature, or fully mature animal, such as a porcine, bovine or ovine. The organ may be used as a temporary or permanent organ replacement for a patient in need of an organ transplant. Any porcine organ can be used, including but not limited to the brain, heart, lungs, eye, stomach, pancreas, kidneys, liver, intestines, uterus, bladder, skin, hair, nails, ears, glands, nose, mouth, lips, spleen, gums, teeth, tongue, salivary glands, tonsils, pharynx, esophagus, large intestine, small intestine, rectum, anus, thyroid gland, thymus gland, bones, cartilage, tendons, ligaments, suprarenal capsule, skeletal muscles, smooth muscles, blood vessels, blood, spinal cord, trachea, ureters, urethra, hypothalamus, pituitary, pylorus, adrenal glands, ovaries, oviducts, vagina, mammary glands, testes, seminal vesicles, penis, lymph, lymph nodes and lymph vessels.
[0057] In another embodiment, the invention provides non-human tissues that are useful for xenotransplantation. In one embodiment, the non-human tissue is porcine tissue. Any porcine tissue can be used, including but not limited to epithelium, connective tissue, blood, bone, cartilage, muscle, nerve, adenoid, adipose, areolar, bone, brown adipose, cancellous, muscle, cartilaginous, cavernous, chondroid, chromaffin, dartoic, elastic, epithelial, fatty, fibrohyaline, fibrous, Gamgee, gelatinous, granulation, gut-associated lymphoid, skeletal muscle, Haller's vascular, indifferent, interstitial, investing, islet, lymphatic, lymphoid, mesenchymal, mesonephric, multilocular adipose, mucous connective, myeloid, nasion soft, nephrogenic, nodal, osteoid, osseous, osteogenic, retiform, periapical, reticular, rubber, smooth muscle, hard hemopoietic, and subcutaneous tissue.
[0058] The invention also provides cells and cell lines from porcine animals that lack expression of functional aGal and CMAH. In one embodiment, these cells or cell lines can be used for xenotransplantation. Cells from any porcine tissue or organ can be used, including, but not limited to: epithelial cells, fibroblast cells, neural cells, keratinocytes, hematopoietic cells, melanocytes, chondrocytes, lymphocytes (B and T), macrophages, monocytes, mononuclear cells, cardiac muscle cells, other muscle cells, granulosa cells, cumulus cells, epidermal cells, endothelial cells, Islets of Langerhans cells, pancreatic insulin secreting cells, pancreatic alpha-2 cells, pancreatic beta cells, pancreatic alpha-1 cells, blood cells, blood precursor cells, bone cells, bone precursor cells, neuronal stem cells, primordial stem cells, hepatocytes, keratinocytes, umbilical vein endothelial cells, aortic endothelial cells, microvascular endothelial cells, fibroblasts, liver stellate cells, aortic smooth muscle cells, cardiac myocytes, neurons, Kupffer cells, smooth muscle cells, Schwann cells, and epithelial cells, erythrocytes, platelets, neutrophils, lymphocytes, monocytes, eosinophils, basophils, adipocytes, chondrocytes, pancreatic islet cells, thyroid cells, parathyroid cells, parotid cells, tumor cells, glial cells, astrocytes, red blood cells, white blood cells, macrophages, epithelial cells, somatic cells, pituitary cells, adrenal cells, hair cells, bladder cells, kidney cells, retinal cells, rod cells, cone cells, heart cells, pacemaker cells, spleen cells, antigen presenting cells, memory cells, T cells, B cells, plasma cells, muscle cells, ovarian cells, uterine cells, prostate cells, vaginal epithelial cells, sperm cells, testicular cells, germ cells, egg cells, leydig cells, peritubular cells, Sertoli cells, lutein cells, cervical cells, endometrial cells, mammary cells, follicle cells, mucous cells, ciliated cells, nonkeratinized epithelial cells, keratinized epithelial cells, lung cells, goblet cells, columnar epithelial cells, dopamiergic cells, squamous epithelial cells, osteocytes, osteoblasts, osteoclasts, dopaminergic cells, embryonic stem cells, fibroblasts and fetal fibroblasts.
[0059] Nonviable derivatives include tissues stripped of viable cells by enzymatic or chemical treatment these tissue derivatives can be further processed through crosslinking or other chemical treatments prior to use in transplantation. In a preferred embodiment, the derivatives include extracellular matrix derived from a variety of tissues, including skin, bone, urinary, bladder or organ submucosal tissues. In addition, tendons, joints, and bones stripped of viable tissue to including but not limited to heart valves and other nonviable tissues as medical devices are provided. In an embodiment, serum or medium suitable for cell culture and isolated from a knockout pig of the invention are provided. Components of porcine knockout organs, tissues or cells are also provided. Components may also be modified through any means known in the art including but not limited to crosslinking and aldehyde crosslinking. Components may vary depending on the larger organ or tissue from which the component is obtained. Skin components may include but are not limited to stripped skin, collagen, epithelial cells, fibroblasts and dermis. Bone components may include but are not limited to collagen and extracellular matrix. Heart components may include but are not limited to valves and valve tissue.
[0060] Expression of a gene product is decreased when total expression of the gene product is decreased, a gene product of an altered size is produced or when the gene product exhibits an altered functionality. Thus if a gene expresses a wild-type amount of product but the product has an altered enzymatic activity, altered size, altered cellular localization pattern, altered receptor-ligand binding or other altered activity, expression of that gene product is considered decreased. Expression may be analyzed by any means known in the art including, but not limited to, T-PC , Western blots, Northern blots, microarray analysis, immunoprecipitation, radiological assays, polypeptide purification, spectrophotometric analysis, Coomassie staining of acrylamide gels, ELISAs, 2-D gel electrophoresis, in situ hybridization, chemiluminescence, silver staining, enzymatic assays, ponceau S staining, multiplex T-PC , immunohistochemical assays, radioimmunoassay, colorimetric assays, immunoradiometric assays, positron emission tomography, fluorometric assays, fluorescence activated cell sorter staining of permeablized cells, radioimunnosorbent assays, real-time PCR, hybridization assays, sandwich immunoassays, flow cytometry, SAGE, differential amplification or electronic analysis. Expression may be analyzed directly or indirectly. Indirect expression analysis may include but is not limited to, analyzing levels of a product catalyzed by an enzyme to evaluate expression of the enzyme. See for example, Ausubel et al, eds (2013) Current Protocols in Molecular Biology, Wiley-lnterscience, New York, N.Y. and Coligan et al (2013) Current Protocols in Protein Science, Wiley-lnterscience New York, NY. Gene expression assays for porcine ASGR1 are commercially available (Applied Biosystems™, Carlsbad CA).
[0061] "As compared to" is intended encompass comparing something to a similar but different thing, such as comparing a data point obtained from an experiment with a knockout pig to a data point obtained from a similar experiment with a wildtype pig. The word "comparing" is intended to encompass examining character, qualities, values, quantities, or ratios in order to discover resemblances or differences between that which is being compared. Comparing may reveal a significant difference in that which is being compared. By "significant difference" is intended a statistically significant difference in results obtained for multiple groups such as the results for material from a knockout pig and material from a wild-type pig. Generally statistical significance is assessed by a statistical significance test such as but not limited to the student's t-test, Chi-square, one-tailed t-test, two-tailed t-test, ANOVA, Dunett's post hoc test, Fisher's test and z-test. A significant difference between two results may be results with a p<0.1, p<0.05, p<0.04, p<0.03, p<0.02, p<0.01 or greater.
[0062] The word "isolated" is intended to encompass an entity that is physically separated from another entity or group. An isolated cell is physically separated from another group of cells. Examples of a group of cells include, but are not limited to, a developing cell mass, a cell culture, a cell line, a tissue, and an animal. The word "isolating" is intended to encompass physically separating an entity from another entity or group. Examples include physically separating a cell from other cells, physically separating a cell component from the remainder of the cell and physically separating tissue or organ from an animal. An isolated cell or cell component is separated by 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, up to 100% of other naturally occurring cells or cell components. Methods for isolating one or more cells from another group of cells are known in the art. See for example Freshney (ED) Culture of Animal Cells: a manual of basic techniques (3 Ed.) 1994, Wiley-Liss; Spector et al (Eds)(1998) Cells: a Laboratory Manual (vol.1) Cold Spring Harbor Laboratory Press and Darling et al (1994) Animal Cells: culture and media John Wiley & Sons. Methods of isolating a tissue or an organ from an animal are known in the art and vary depending on the tissue or organ to be isolated and the desired method of transplanting the tissue or organ.
[0063] A "skin related product" encompasses products isolated from skin and products intended for use with skin. Skin related products isolated from skin or other tissues may be modified before use with skin. Skin related products include but are not limited to replacement dressings, burn coverings, dermal products, replacement dermis, dermal fibroblasts,collagen, chondroitin, connective tissue, keratinocytes, cell-free xenodermis,cell-free pig dermis,composite skin su bstitutes and epidermis and temporary wound coverings. See for example Matou-Kovd et al (1994) Ann Med Burn Club 7:143, herein incorporated by reference in its entirety.
[0064] "Xenotransplantation" encompasses any procedure that involves the transplantation, implantation or infusion of cells, tissues or organs into a recipient subject from a different species. Xenotransplantation in which the recipient is a human is particularly envisioned. Thus xenotransplantation includes but is not limited to vascularized xenotransplant, partially vascularized xenotransplant, unvascularized xenotransplant, xenodressings, xenobandages, and xenostructures.
[0065] In another embodiment, the invention provides a method of improving a hyperacute rejection related symptom in a patient comprising transplanting porcine organs, tissue or cells having reduced expression of aGal and Neu5Gc on the porcine organs, tissue or cells into a human, wherein the hyperacute rejection related symptoms are improved as compared to when tissue from a wild-type swine is transplanted into a human. By "improving", "bettering", "ameliorating", "enhancing", and "helping" is intended advancing or making progress in what is desirable. It is also envisioned that improving a hyperacute rejection related symptom may encompass a decrease, lessening, or diminishing of an undesirable symptom. By "hyperacute rejection" we mean rejection of the transplanted material or tissue occurring or beginning within the first 24 hours post-transplant involving one or more mechanisms of rejection. Hyperacute rejection encompasses but is not limited to "acute humoral rejection" and "antibody mediated rejection".
[0066] "Hyperacute rejection related symptom" is intended to encompass any symptom known to the field as related to or caused by hyperacute rejection. It is recognized that hyperacute rejection related symptoms may vary depending upon the type of organ, tissue or cell that was transplanted. Hyperacute rejection related symptoms may include, but are not limited to, thrombotic occlusion, hemorrhage of the graft vasculature, neutrophil influx, ischemia, mottling, cyanosis, edema, organ failure, reduced organ function, necrosis, glomerular capillary thrombosis, lack of function, hemolysis, fever, clotting, decreased bile production, asthenia, hypotension, oliguria, coagulopathy, elevated serum aminotransferase levels, elevated alkaline phosphatase levels, jaundice, lethargy, acidosis and hyperbilirubenemia and thrombocytopenia.
[0067] Thrombocytopenia is a quantity of platelets below the normal range of 140,000 to 440,000/μΙ. Thrombocytopenia related symptoms include, but are not limited to, internal hemorrhage, intracranial bleeding, hematuria, hematemesis, bleeding gums, abdominal distension, melena, prolonged menstruation, epistaxis, ecchymosis, petechiae or purpura. Uptake of human platelets by pig livers contributes to the development of thrombocytopenia in xenograft recipients.
[0068] Platelets, also known as thrombocytes, are enucleate fragments of megakaryocytes involved in blood coagulation, hemostasis and blood thrombus formation. Human platelets are routinely isolated through a variety of methods including, but not limited to, platelet apheresis, plateletpheresis and ultracentrifugation.
[0069] The phrase "platelet uptake" is intended to encompass the incorporation of a platelet into a liver or liver cell. While not being limited by mechanism, such uptake may occur through a phagocytic process. Platelet uptake may be monitored by any platelet uptake monitoring assay known in the art. Platelet uptake monitoring assays include, but are not limited to immunological methods, western blots, immunoblotting, microscopy, confocal microscopy, transmission electron microscopy and phagosome isolation. It is recognized that the appropriate platelet uptake monitoring assay may depend upon the type of label used. Platelet uptake may be measured as a percentage of total platelets absorbed, percentage of total platelets not absorbed, a ratio of absorbed to unabsorbed platelets, percentage of cells absorbing at least one platelet, percentage of cells not absorbing a platelet, or number of platelets absorbed per cell. It is recognized that platelet uptake by more than one cell type may contribute to the total platelet uptake of the liver. Total platelet uptake by an animal liver may include platelet uptake by liver sinusoidal endothelial cells, platelet uptake by Kuppffer cells, platelet uptake by LSECs and Kupffer cells and platelet uptake by additional cell types. It is recognized that platelet uptake by different cell types may contribute similar or disparate fractions of the total platelet uptake by a liver. Thus an alteration, inhibition, reduction, decrease, or lowering of platelet uptake by a liver comprises an alteration, inhibition, reduction, decrease, or lowering of platelet uptake by one or more liver cell types. [0070] The word "providing" is intended to encompass preparing, procuring, getting ready, making ready, supplying or furnishing. It is recognized that methods of providing a cell may differ from methods of providing a subject and that methods of providing a liver may differ from methods of providing a pig.
[0071] In an embodiment, a cell culture reagent derived from a transgenic pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes is provided. Cell culture reagents are reagents utilized for tissue culture, in vitro tissue culture, microfluidic tissue culture, cell culture or other means of growing isolated cells or cell lines. Cell culture reagents may include but are not limited to cell culture media, cell culture serum, a cell culture additive, a feeder cell, and an isolated cell capable of proliferation. By an "isolated cell capable of proliferation" is intended a cell isolated or partially isolated from other cell types or other cells wherein the cell is capable of proliferating, dividing, or multiplying into at least one additional clonal cell.
[0072] Cells grown in culture may synthesize or meta bolically incorporate antigenic epitopes into glycoproteins secreted by the cultured cell. The antigenic epitopes may result in increased binding by human antibodies and decreased efficacy of the glycoprotein. See Ghaderi et al, 2010 Nature Biotechnology 28(8):863-867, herein incorporated by reference in its entirety. Growing the producing cell in a cell culture reagent with less aGal or Neu5Gc may reduce the amount of aGal, Neu5Gc, or both aGal and Neu5Gc epitopes on the glycoprotein of interest. Glycoproteins of interest may include any glycoprotein, particularly glycoproteins intended for use in human su bjects such as but not limited to, an antibody, growth factor, cytokine, hormone, or clotting factor.
[0073] In summary, xenoantigens aGal and Neu5Gc were eliminated in transgenic pigs by genetic modification. Double knockout pigs (GT/CMAH-KO) were produced within 5-10 months or less.
[0074] In embodiments of the present invention, tissues are provided in which both the aGal and CMAH genes are rendered inactive, such that the resultant aGal and CMAH products can no longer generate alpha 1,3-galactosyl epitopes or Neu5Gc on the cell surface. In an alternative embodiment, the aGal and CMAH genes can be inactivated in such a way that no transcription of the gene occurs.
[0075] In yet another aspect, the present invention provides a method for producing viable pigs lacking any functional expression of aGal and CMAH. In one embodiment, the pigs are produced as described below. Methods of making transgenic pigs, and the challenges thereto, are discussed in Galli et al. 2010 Xenotransplantation, 17(6) p. 397-410, incorporated by reference herein for all purposes. The methods and cell cultures of the invention are further detailed below. EXAMPLES
[0076] The following Examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and the following examples and fall within the scope of the appended claims.
Example 1. Design of Zinc Finger Nucleases (ZFN).
[0077] A pair of ZFN were designed to bind and cleave the sequence of porcine CMAH exon 9 (SEQ ID NO: 1-AAACTCCTGAACTACAAGGCTCGGCTGGTGAAGGA) beginning at position 1,341 of Ensemble transcript ENSSSCT00000001195. Another pair of ZFN were designed to bind and cleave the region of GGTA1 exon 8 (SEQ ID NO: 2-GTCATCTTTTACATCATGGTGGATGATATCTCCAGGATGCC) beginning at position 1165 of Ensemble transcript ENSSSCT00000006069. The ZFN activities were validated in yeast (Sigma-Aldrich, St. Louis, MO). Additional detail regarding the ZFN of the present invention can be found in Li et al., Journal of Surgical Research, (2012)E1-E7 Epub 2012 July 3, which is incorporated by reference herein for all purposes.
Example 2. Cell Culture and Transfection of Porcine Liver Derived Cells.
[0078] Porcine adult liver derived cells (LDC) were isolated with modifications from the method described in Li et al, 2010 Cell Reprogram. 12:599. Isolated LDC were cultured in a combination stem cell media (SCM) (a-MEM :EGM-MV 3:1) (Invitrogen/Lonza, Switzerland) supplemented with 10% fetal bovine serum (FBS) (Hyclone, Logan, UT), 10% horse serum (Invitrogen, Carlsbad, CA), 12mM HEPES, and 1% Pen/Strep (Invitrogen) as described in Li et al. 2012 JSR Epub 2012 July 3. A commercial porcine strain (Landrace/York/Chester White) was used as the source of the LDC. Neon transfection system was used according to the manufacturer's instruction (Invitrogen).
[0079] Briefly, LDC were harvested by trypsin digestion, washed with calcium and magnesium free Dulbecco's phosphate buffered saline (DPBS) (Invitrogen) and centrifuged. 10s cells were suspended in 120 μΙ of resuspension buffer (Invitrogen) containing 2 μgs of each CMAH ZFN plasmid. pEGFP-N 1 (Clonetech, Mountain View, CA) was used as a control to measure transfection efficiency as well as ZFN activity. Cells were electroporated at 1300 V, 30 msec for 1 pulse.
[0080] Then cells were transferred in SCM without antibiotics and plated onto collagen I coated plates. Cells were cultured with 5% C02 and 10% 02 at 30°C for 3 days and 37°C for 2 days. Example 3. Surveyor Mutation Detection Assay (CEL I assay).
[0081] ZFN-induced mutation was detected using the Surveyor Mutation Detection kit (Transgenomic, Omaha, NE). At day 5 post-transfection, genomic DNA from ZFN-treated and control plasmid pEGFP-Nl treated cells was extracted and PCR was performed using primers ZFN-CMAH-F (SEQ ID NO: 3-5' GGACCTGCTTTATCTTGCTCGC 3'), ZFN-CMAH-R (SEQ ID NO: 4-5' CC ATACTTGTCTG CTGGGTGGG 3'). Pwo SuperYield DNA Polymerase, dNTPack (Roche, Indianapolis, IN) was used and the PCR conditions were as follows: 94°C, 2 minutes; 94°C, 15 seconds, 55°C, 30 seconds and 68°C, 50 seconds for 15 cycles; 94°C, 15 seconds, 5 5°C, 30 seconds and 68°C, 50 seconds with additional 5 seconds for each cycle, for 25 cycles and a final extension step of 68°C for 5 minutes.
[0082] PCR product was denatured and annealed using the following program on a MyCycler (Bio-Rad): 95°C, 10 minutes; 95°C to 85°C, -2°C /second; 85°C to 25°C, -0.1°C /second. 200-400 ng of PCR product was digested with 1 μΙ of Nuclease S and 1 μΙ of enhancer (Transgenomic, Omaha, NE) at 42°C for 40 minutes. The product was separated on a 10% polyacrylamide gel and stained with SYBR Safe to assess ZFN-induced mutations.
Example 4. Screening CMAH Mutant Cells.
[0083] ZFN-treated cells were plated at 1 cell/well in ten 96-well plates coated with collagen I (BD, Franklin Lakes New Jersey). After 14 days, single cell clones became evident. Cells were expanded to 48- well plate. Some cells were harvested for mutation screening. PCR was performed as described above herein. PCR products were resolved on a 0.8% agarose gel and purified by QIAquick Gel Extraction Kit (Qiagen). Primer CMAH-S1 (SEQ ID NO: 5-5' CCAAACCCTGTCATTCCAG 3') was used to sequence the ZFN targeted CMAH region. A clone with an identical mutation in both CMAH gene copies was identified.
[0084] Disruption of GGTA 1 gene on CMAH mutant cells and counter selection of a-Gal- cells 10s of CMAH mutant cells were transfected with 2 μgs of each GGTAl ZFN plasmid as described below herein. aGal- cells were isolated by the counter selection method described below herein.
Example 5. Counter Selection of aGal- Cells.
[0085] CMAH deficient LDC were treated with zinc finger plasmids targeting porcine 1,3 aGal. Alternatively LDC were transfected with Crispr CMAH sgRNA expression plasmids and GGTAl sgRNA expression plasmids and grown for 48 hours as described elsewhere herein. The cells were harvested, washed and counted. A total of 2.3 x 10s cells were incubated with 5 μg of biotin-conjugated Griffonia simplicifolia isolectin IB4 (IB4 lectin from Enzo Life Science, Farmingdale NY) in 600 μΙ of PBS supplemented with 0.1% BSA and 2 mM ethylenediaminetetracetic acid (pH 7.4) on ice for 30 minutes. After 30 minutes, the cells were washed once and incubated with 50 μΙ Dynabeads Biotin Binder (Invitrogen) at 4°C with rotation for 30 minutes. A magnet was applied to deplete Dynabeads-bound a- Gal+ cells. The IB4 lectin is specific for the Gal epitope; cells that did not bind the Dynabeads remained in the supernatant. CMAH cells that did not bind IB4 and the Dynabeads were identified as Gal-/- cells. Crispr CMAH and GGTAl treated cells that did not bind IB4 and the Dynabeads were utilized in SCNT as described below herein.
Example 6. TALEN Constructs.
[0086] TALEN constructs are designed to bind and cleave the sequence of porcine CMAH at a suitable site. Additional Talen constructs are designed to bind and cleave GGTAl at a suitable site.
Example 7. Production of Double Knockout (otGal and CMAH) Pigs.
[0087] Somatic Cell Nuclear Transfer (SCNT) was performed using in vitro matured oocytes (DeSoto Biosciences Inc., St. Seymour TN and Minitube of America, Mount Horeb, Wl). Cumulus cells were removed from the oocytes by pipetting in 0.1% hyaluronidase. Oocytes with normal morphology and a visible polar body were selected and incubated in manipulation media (calcium-free NCSU-23 with 5% fetal bovine serum (FBS) containing 5 μg/ml bisbenzimide and 7.5 μg/mL cytochalasin B for 15 minutes. Following this incubation period, oocytes were enucleated by removing the first polar body and metaphase II plate. Single cells of CMAH deficient LDC that survived IB4 counterselection were injected into each enucleated oocyte. Alternatively Crispr transfected cells that survived IB4 counterselection were injected into each enucleated oocyte. Electrical fusion was induced with a BTX cell electroporator (Harvard Apparatus, Holliston, MA). Enucleated oocytes injected with a cell (couples) were exposed to two DC pulses of 140 V for 50 μ≤ in 280 mM mannitol, 0.001 mM CaCI2 and 0.05 mM MgCI2. After one hour, reconstructed oocytes were activated by two DC pulses of 120 V for 60 μ≤ in 280 mM mannitol, 0. 1 mM CaCI2 and 0.05 mM MgCI2. After activation, oocytes were placed in NCSU-23 medium with 0.4% bovine serum albumin (BSA) and incubated at 38.5°C, 5% C02 in a humidified atmosphere for less than one hour. Within an hour after activation, oocytes were transferred into a recipient pig. Recipient pigs were synchronized occidental pigs on their first day of estrus. Pregnancies were verified by ultrasound at day 25 or day 26 after embryo transfer. Results of SCNT using double knockout cells are summarized in Table 1. Results of SCNT using Crispr-transfected IB4 counterselected cells are summarized in Table 2. Table 1: Results of somatic cell nuclear transfer using double-KO cells.
Figure imgf000024_0001
[0088] Dou ble-KO cells (cl22) were used as donor cells in SCNT and transferred into 2 recipients, 01 and 02. Fetal fibroblasts from fetus num ber 1 of recipient 02 were re-cloned and transferred to animals 03, 04, 05, and 06. 477 embryos were delivered to four gilts, resulting in one pregnancy and the birth of 5 piglets. The four via ble piglets appeared healthy as they were nursing since birth and needed no feeding support.
[0089] In at least one experiment, SCNT of the selected cells deficient for CMAH and GGTAl resulted in a pregnancy that was terminated at day 30 of gestation. Five normal fetuses and one resorbed fetus were harvested, photogra phed and shown in Figure 1A. Red blood cells from the dou ble knockout fetuses were analyzed by flow cytometry as described below herein.
[0090] Fibroblasts were grown from fetus number one. The fibroblasts were utilized in SCNT to generate 477 em bryos which were transferred into four recipient gilts. The SCNT resulted in one pregnancy and the birth of four live piglets and one still born piglet. The four via ble piglets needed no feeding support and appeared healthy. Three of the aGal/CMAH dou ble knockout pigs are shown in Figure 3A. The above described disruptions of the CMAH and GGTAl genes were validated in the four via ble pigs from the second pregnancy as described below herein. Results of somatic cell nuclear transfer using dou ble-KO cells are su mmarized in Ta ble 1 a bove.
[0091] Crispr-transfected I B4 counterselected cells were used as donor cells in SCNT and transferred into 2 recipients, Juneau and Honda. Eighty-eight em bryos were transferred into Ju neau; pregnancy did not occur in Juneau. Over 100 em bryos were transferred into Honda; pregnancy occurred. Ten healthy fetuses were collected at day 32 of gestation (Figure 9A). DNA sequencing results showed that five out of the ten fetuses were GGTA1 and CMAH double knockouts. For GGTA1, seven fetuses were biallelic mutations, one fetus was a monoallelic mutation and two fetuses were wild-type. For CMAH, five fetuses were biallelic mutations, one fetus was a monoallelic mutation and four fetuses were wild-type. Among the five double knockout fetuses, two homozygous mutations were found in fetus 7 at the GGTA1 locus and in fetus 2 at the CMAH locus. The remaining mutations were heterozygous biallelic mutations. Results are summarized in Table 2 below and sequence data are presented in Figure 9B. Fetal fibroblasts were harvested from one dou ble knockout fetus. The fetal fibroblasts were utilized in SCNT to generate embryos which were transferred into recipient sows. SCNT of the double knockout fetal fibroblasts resulted in at least one pregnancy.
[0092] Table 2 Results of Somatic Cell Nuclear Transfer Using Crispr-Transfected Cells
Figure imgf000025_0002
Figure imgf000025_0001
[0093] Somatic Cell Nuclear Transfer. All the animals used in this study were approved by Institutional Biosafety Committee (IBC) and Institutional Animal Care and Use Committee (IACUC). In Table 2, * indicates pregnant animals/total recipients and ** indicates fetuses/embryos transferred.
Example 8. DNA Sequencing Analysis of Zn-finger Targeted CMAH and GGTA1 Regions.
[0094] Genomic DNA from double knockout Zn finger cloned fetuses and the four viable piglets was extracted using DNeasy Blood & Tissue Kit (Qiagen, Valencia, CA). PCR amplification of the CMAH region was performed as described above herein. [0095] Primer CMAH-S1 (SEQ ID NO: 5-5' CCAAACCCTGTCATTCCAG 3') and GGTA1-F primer (SEQ ID NO: 6-5' CTAGAAATCCCAGAGGTTAC 3') were used to sequence the targeted CMAH region and GGTAl region, respectively.
[0096] The GGTAl region was amplified by PCR using the GGTA1-F primer (SEQ ID NO: 6 (5' CTAGAAATCCCAGAGGTTAC 3')) and GGTA1-R primer (SEQ ID NO:7 (5'TCCTTGTCCTGGAGGATTCC3')). Pwo Master (Roche, Indianapolis IN) was used and PCR conditions were as follows: 94°C, 2 min; 94°C, 15 s, 57°C, 30 s, and 72°C, 30s for 40 cycles; and a final extension step of 72°C for 5 min. A total of 200-400 ng of PCR product was denatured and annealed using the following program on a Mycycler (Bio-Rad): 95°C, 10 min; 95° to 85°C, -2°C/s; 85°C to 25°C, -0.1°C/s. One microliter of enhancer and 1 μΙ of Nuclease S (Transgenomic Omaha NE) was added to each reaction and incubated at 42°C for 40 minutes. The product was separated on a 10% polyacrylamide gel and stained with SYBR Safe (Invitrogen USA Eugene OR).
[0097] Results from an exemplary DNA sequence analysis are shown in Figure 2B. DNA sequence analysis confirmed homozygous alterations of the GGTAl and CMAH genes in at least one fetus. The CMAH gene sequence is altered by a four base pair insertion. The inserted sequence is "GGAA". The GGTAl gene sequence is altered by a 3 base pair deletion adjacent to a G to A substitution. The sequences of the targeted CMAH and GGTAl regions of the genomic DNA of four viable piglets are shown in Figure 3B.
[0098] Genomic DNA from IB4 counter-selected cells and Crispr cloned fetuses was extracted using GenElute Mammalian Genomic DNA Miniprep Kit (Sigma Aldrich, St. Louis MO).
[0099] The GGTAl region was amplified by PCR using the GGTA1-F seq primer (SEQ ID NO: 8 (5' CCTTAGTATCCTTCCC AACCC AG AC-3' ) ) and GGTA1-R seq primer (SEQ ID NO: 9 (5' GCTTTCTTTACGGTGTCAGTGAATCC-3')). The CMAH region was amplified by PCR using the CMAH-F seq primer (SEQ ID NO: 10 (5'- CTTGGAGGTGATTTGAGTTGGG-3')) and the CMAH-R seq primer (SEQ ID NO: 11 (5'- CATTTTCTTCGGAGTTGAGGGC-3')). Pwo Superyield DNA polymerase (Roche, Indianapolis IN) was used and PCR conditions for GGTAl were as follows: 94°C, 2 min; 94°C, 15 s, 54°C, 30 s, and 72°C, 45s for 15 cycles; 94°C, 15 s, 54°C, 30 s, and 72°C, 45s + 5 seconds/cycle for 25 cycles; and a final extension step of 72°C for 5 min. The PCR conditions for CMAH were as follows: 94°C, 2 min; 94°C, 15 s, 56°C, 30 s, and 72°C, 45s for 15 cycles; 94°C, 15 s, 56°C, 30 s, and 72°C, 45s + 5 seconds/cycle for 25 cycles; and a final extension step of 72°C for 5 min. The products were separated on a 1% agarose gel, purified by the GenElute Gel Extraction kit (Sigma-Aldrich, St. Louis MO) and sequenced by the Sanger method (DNA Sequencing Core Facility, Indiana University School of Medicine).
[00100] Primer CMAH-F seq (SEQ ID NO: 10) and GGTA1-F seq primer (SEQ ID NO: 8) were used to sequence the targeted CMAH region and GGTAl region, respectively.
[00101] Results from exemplary DNA sequence analyses are shown in Figure 9B. DNA sequence analysis confirmed bia I lei ic alterations of the GGTAl and CMAH genes in five fetuses. In fetus 1 and 5, one GGTAl allele has a single base pair deletion (deletes a C), the other allele has a single nucleotide insertion (T). In fetus 2, one GGTAl allele has a six base pair deletion, the other allele has a two nucleotide insertion (AT). In fetus 6, one GGTAl allele has a ten base pair deletion, the other allele has a seven base pair deletion. In fetus 7, both GGTAl alleles have an eight nucleotide insertion that substitute for a five nucleotide sequence. In fetus 1, one CMAH allele has a double base pair deletion (deletes TG), the other allele has a single nucleotide insertion (inserts T). In fetus 2, both CMAH alleles have an 8 base pair deletion. In fetus 5, one CMAH allele has a 5 nucleotide deletion, the other has a 3 nucleotide deletion. In fetus 6, one CMAH allele has a single base pair deletion (T), the other has single base pair deletion and a colocalized two base pair insertion. In fetus 7, one CMAH allele has a 5 base pair deletion, the other has a 20 base pair deletion. Sequences of the targeted CMAH and GGTAl regions of the genomic DNA of five fetuses are shown in Figure 9B.
Example 9. Flow Cytometry of Red Blood Cells (RBCs).
[00102] Fetal livers were removed from double knockout fetuses and incubated in RPM I1640 for
24 hours at 37°C. RBCs were collected from cells released into the media after incubation of the fetal livers. RBCs were also obtained from adult human donors, six month old wild-type pigs and GGTA- knockout pigs (GGTA-KO pigs (fetal or 6 month old)). Porcine and human peripheral blood monocytes (PBMCs) were prepared using Ficoll-Paque Plus from whole blood collected in anticoagulant citrate dextrose (ACD).
[00103] Cells were stained with IB4 lectin Alexa Fluor 647 (Invitrogen, Grand Island, NY) and anti-
Neu5Gc antibody (Sialix, Vista CA). A negative control antibody for comparison with anti-Neu5Gc antibody was also used (Sialix, Vista CA). Cells were incubated with IB4 lectin for 20 minutes at 4°C. The cells were washed with blocking agent (Sialix, Vista, Ca) diluted in HBSS. Gal epitopes bind IB4 lectin. Cells were then stained with anti-Neu5Gc antibody for one hour at 4°C followed by Donkey anti-chicken DyLight 649 (Jackson ImmunoResearch Laboratories Inc, West Grove PA) for 40-60 minutes at 4°C. Cells stained with anti-Neu5Gc antibody were washed before and after secondary antibody with Sialix blocking agent diluted in PBS. In various experiments unstained BC or PBMC were used as negative controls for IB4 lectin staining. An Accuri C6 flow cytometer and CFlow Software (Accuri, Ann Arbor, Ml) were used for analysis.
[00104] FACS analysis validated the absence of functional GGTA1 and CMAH genes eliminated the a-Gal and Neu5Gc modifications on the RBCs isolated from the cloned fetuses. Results obtained from one such experiment are shown in Figure 2C.
Example 10. Confocal microscopy analysis.
[00105] One of the four double knockout (Double-KO, DKO, CMAH-/aGAL-) piglets with mutations generated by Zn-finger targeting was euthanized. Liver, heart and kidney tissues were obtained from the double-ko pig. Liver, heart and kidney tissues were also obtained from wild-type (WT) and GGTA1 knockout (GGTA-ko) pigs. Frozen sections of each tissue were prepared. Mounted tissues were blocked in Odyssey blocking buffer (Ll-Cor Biosciences, Lincoln NE) in HBSS for one hour. The slides were then fixed in 4% paraformaldehyde for 10 minutes. Tissues were stained with IB4 lectin Alexa Fluor 647 (Invitrogen, Grand Island NY) to visualize the presence of the Gal epitope. To visualize the Neu5Gc epitope, tissues were stained with a chicken anti-Neu5Gc antibody or with a control antibody (Sialix, Vista, CA) for one hour. Tissues were washed three times with HBSS. Donkey anti- chicken DyLight 649 (Jackson ImmunoResearch Laboratories Inc., West Grove PA) secondary antibody was incubated with the tissue for approximately one hour. Tissues were washed three times with 0.1% HBSS Tween. To stain the nucleus, DAPI stain (Invitrogen, Grand Island NY) was added to all the slides for 1 minute followed by two 0.1% HBSS Tween washes. Tissues were mounted in ProLong Gold (Invitrogen, Grand Island NY). Confocal microscopy was performed using an Olympus FV1000.
[00106] Confocal microscopy indicated the presence of a-Gal and Neu5Gc in liver, heart and kidney tissues obtained from a wild-type pig. GGTA1-KO pigs displayed only Neu5Gc in liver, heart and kidney tissues. Confocal microscopy indicated the absence of a-Gal and Neu5Gc in liver, heart and kidney in tissues obtained from a double knockout piglet (exemplary fields are shown in Figure 4A and 4B). Example 11. Crossmatch of Human Sera with GGTA1-KO and double-KO PBMCs.
[00107] Porcine whole blood from GGTA1-KO and dou ble-KO pigs was collected in ACD. Porcine
PBMCs were prepared from the whole blood using Ficoll-Paque Plus. Cell viability was assessed microscopically with Trypan Blue. Sera were obtained from ten healthy human volunteers. Twenty-five percent heat-inactivated serum was prepared. Approximately 2 X 106/ml GGTA-KO and double-KO PBMCs were incubated with each human serum sample for 2 hours at 4°C. After incubation of the serum and PBMCs, the PBMCs were washed three times in 0.5% PBS Sialix Blocking agent. PBMCs were stained with DyLight 649-conjugated Donkey anti-human IgM or DyLight 488 Donkey anti-human IgG (Jackson Immunoresearch Laboratories Inc, West Grove PA) for 1 hour at 4°C. PBMCs were washed three times using 0.5% PBS Sialix blocking agent. Analyses were performed using an Accuri C6 flow cytometer and BD CFlow Plus Software (Accuri, Ann Arbor Ml). Overlays were produced using Kaluza version 1.2 software from Beckman Coulter (Brea, CA). A representative histogram is shown in Figure 5A. MFI's obtained from flow cytometry crossmatch analysis of 10 unique human subjects are also shown in Figure 5A.
[00108] Lower Mean fluorescent Intensities (MFI) of double-KO PBMCs indicate less IgM bound to double-KO cells than to GGTA1-KO cells in 9 of 10 samples. In an experiment, the median MFI observed for human IgM binding to double-KO and GGTA-KO PBMCs were 4,411 and 15,059 respectively (n=10, p=0.0039). Lower MFI of double-KO PBMCs indicate less IgG bound to double-KO cells than to GGTA1-KO cells in 10 of 10 samples. In an experiment, the median MFI observed for human IgG binding to double-KO and GGTA-KO PBMC were 714 and 2306 respectively (n=10, p=0.002). (Figure 5A).
Example 12. Antibody-mediated Complement-Dependent Cytotoxicity.
[00109] Antibody-mediated complement dependent cytotoxicity assays are known in the art. A modified method of Diaz et al (Diaz et al, 2004, Transplant Immunology 13(4):313-317) was performed. Human serum was obtained from ten healthy volunteers (Figures 5 and 6). Twenty-five percent heat- inactivated human serum was prepared. The heat-inactivated human sera were serially diluted and 100 μΙ of each concentration were placed in a 96 well v-bottom assay plate. Sera were mixed with a 100 μΙ aliquot of PBMC obtained from either GGTA1-KO or double-KO pigs. The final concentration of PBMC in each well was 5 X 106/ml; in some experiments 1 x 106/ml PBMC were used. The serum concentrations varied from 50%, 17%, 6%, 2%, 0.6%, 0.2% and 0.07%. The mixtures were incubated for 30 minutes at 4°C. After 30 minutes, the plates were centrifuged for 4 minutes at 400 x g. The plates were decanted and washed with HBSS. Rabbit complement (150 μΙ of a 1:15 dilution) was added to each well and incubated for 30 minutes at 37°C. PBMC were labeled with a fluorescein diacetate (FDA) stock solution, prepared fresh daily in HBSS (1 μ§/ιηΙ-) from a 1 mg/ml stock solution in acetone and with propidium iodide (PI), prepared at 50 μ§/ιηΙ in phosphate buffered saline (PBS). After incubation in complement, the samples were transferred by pipette to tubes containing 250 μΙ of HBSS and 10 μΙ of FDA/PI for analysis using an Accuri C6 flow cytometer.
[00110] The percentage of dead cells (PI+/FDA-), damaged cells (PI+/FDA+) and live cells (Pl-
/FDA+) was determined. Double negative events (PI-/FDA-) were excluded from calculations. The percentage of cytotoxicity in cells not exposed to serum was considered spontaneous killing (% CTXspont). Values for cytotoxicity (%CTX) are shown after correction for spontaneous cytotoxicity (%CTXspont), using the following formula: %CTX = (%CTXeXp-%CTXSpont)/(100-%CTXSpOnt) x 100, where %CTXexp is the percentage of dead cells under the experimental condition.
[00111] Data from a series of antibody-mediated complement dependent cytotoxicity assays are shown in Figure 5B. At 2% concentration, most of the subjects' sera lysed greater than 90% of GGTAl- KO PBMCs but less than 50% of the double-KO PBMCs. The median percent cytotoxicity of GGTAl-KO and double-KO PBMCs in this series was 98% and 29% respectively, at 2% human serum (n=10, p=0.002). The data shown indicate these human sera were less cytotoxic to double-KO cells than to GGTAl-KO cells. While not being bound by mechanism, the data suggest in the absence of the Gal epitope, a significant portion of IgM and IgG binding is to Neu5Gc.
Example 13. Statistical Analysis.
[00112] Flow cytometric crossmatch results were reported as medians of the M FI values for each human serum crossmatched with GGTAl-KO and double-KO PBMCs for both IgM and IgG. The Wilcoxon matched pairs signed rank test was used to analyze the data using Prism 5 for Windows (GraphPad Software Inc, La Jolla Ca).
[00113] Antibody-mediated, complement dependent cytotoxicity data was also analyzed as above. The log of each serum dilution was plotted against the %CTX for each sample and the sigmoid curve was analyzed by non-linear regression to estimate the serum LD50. The %CTX of GGTAl-KO and double-KO PBMCs at each serum dilution for each sample was analyzed using the Wilcoxon matched pairs signed rank test. Example 14. Ex vivo Perfusion of Human Platelet through a Double KO Liver.
[00114] A double knockout CMAH/aGal pig is anesthetized and intubated. A midline abdominal incision is made. The liver is removed and placed in a perfusion device under normothermic conditions. Humidity, temperature and air flow are maintained in the perfusion device. Human platelets obtained from healthy volunteer subjects are circulated through the double knockout liver. Platelet levels in the pre-perfusion and post-perfusion samples are evaluated. Pre and post-perfusion evaluation of the pig liver is performed.
Example 15. Evaluation of Response to a Double Knockout (ctGal and CMAH) Xenograft.
[00115] A porcine liver obtained from double knockout (aGal-/CMAH-) pigs is surgically transplanted into a recently deceased human cadaver using the piggyback method. After the surgery, biological samples are obtained from the human cadaver. Clinical indicia of graft rejection are monitored.
Example 16. Evaluation of a Response to a Double Knockout (ctGal and CMAH) Xenograft.
[00116] Porcine kidneys are obtained from double knockout (GGTA1/CMAH double-KO) pigs. A highly sensitized human subject is administered compounds to manage preexisting and de novo donor- specific antibodies. Porcine double-KO kidneys are surgically transplanted into the subject. Clinical indicia of graft rejection are monitored.
Example 17. Construction of single guide RNAs (sgRNAs) Expression Vector
[00117] Targeting sites for the CMAH and GGTA1 genes were identified. Forward and reverse oligonucleotides for each targeting site were obtained. The GGTA1 forward sgRNA sequence was CACCGAGAAAATAATGAATGTCAA (SEQ ID NO: 12); the GGTA1 reverse sgRNA sequence was AAACTTGACATTCATTATTTTCTC (SEQ ID NO: 13). The CMAH forward sgRNA sequence was CACCGAGTAAGGTACGTGATCTGT (SEQ ID NO: 14); the CMAH reverse sgRNA sequence was AAACACAGATCACGTACCTTACT (SEQ ID NO: 15). The oligonucleotide pairs were annealed together to generate short double strand DNA fragments with Bbsl compatible overhangs. The pX330 bicistronic expression vector expressing Cas9 and sgRNA of interest (Addgene, www.addgene.org/crispr-cas) was linearized with Bbsl. pX330 expresses a mammalian codon optimized Cas9. The linearized pX330 expression vector and the double strand DNA fragments for each sg NA (GGTA1 and CMAH) were incubated with ligase.
[00118] Liver derived cells (LDCs) were cultured as described in Li et al (2013) J. Surg Res
181:e39-45, herein incorporated by reference in its entirety. LDC were cotransfected with pX330- CMAHsgRNA and pX300-GGTAlsgRNA expression plasmids using the Neon transfection system (Invitrogen) according to the manufacturer's instructions. Forty-eight hours after transfection, cells were harvested and used for isolectin B4 counter-selection.
Example 18. Flow cytometry analysis
[00119] Isolectin B4 (IB4) counter-selected LDC, cloned fetal fibroblasts from DKO fetuses, wild- type LDC and wild-type fetal fibroblasts were stained with IB4 conjugated with FITC (Enzo Life Science, Farmingdale NY) to assess the aGal epitope level. To evaluate the Neu5Gc level, IB4 counter-selected LDC cloned fetal fibroblasts from DKO fetuses, wild-type LDC and wild-type fetal fibroblasts were stained with antiNeu5Gc antibody (Sialix, Vista CA) followed by donkey anti-chicken DyLight 649 (Jackson ImmunoResearch Laboratories Inc., West Grove PA). A negative control antibody for comparison with anti-Neu5Gc antibody was also used (Sialix, Vista CA). An Accuri C6 flow cytometer (Accuri, Ann Arbor Ml) and FlowJo software (Tree Star, Inc. Ashland OR) were used for analysis. Representative flow cytometry results of aGal from IB4 counter selected cells are shown in Figure 7A. Representative flow cytometry results of Neu5Gc on IB4 counter selected cells are shown in Figure 8A. Representative flow cytometry results of the aGal and Neu5Gc on fetal fibroblasts derived from fetus 7 are shown in Figure 9C.
Example 19. Ex vivo Perfusion of Human Platelets through a Double KO Liver
[00120] Double knockout CMAH/aGAL and single aGal knockout pigs were anesthetized and intubated. A midline abdominal incision was made in each pig. Livers were removed and placed in a perfusion device under normothermic conditions. Humidity, temperature and air flow were maintained in the perfusion device. Human platelets obtained from healthy volunteer subjects were circulated through the double knockout liver. Platelet levels in the pre-perfusion and post-perfusion samples were evaluated after circulation through three double knockout (CMAH ^/GGTAl7 ) livers. Pre and post- perfusion evaluation of the pig livers were performed. GGTA17" livers (n=2) were obtained, and the livers were perfused under similar conditions. Data from one experimental series are summarized in Figure 12.
Example 20. Neu5Gc Levels in Cells Cultured with a DKO (aGal/CMAH) Piq Cell Culture Reagent.
[00121] Fibroblasts were cultured from a CMAH/aGal Double knockout pig. Because the DKO fibroblasts lack CMAH, they do not produce Neu5Gc. DKO serum was isolated from the blood of CMAH/aGAL pigs. The DKO fibroblasts were cultured in cell culture media supplemented with either DKO serum or bovine serum (FBS) for six weeks. After six weeks, cells were evaluated by flow cytometry with anti-Neu5Gc antibodies. Results from one such experiment are presented in Figure 10.
Example 21. Concordant Analysis of DKO Pig, Baboon and Chimpanzee Material.
[00122] Antibody-mediated complement dependent cytotoxicity assays are known in the art. A modified method of Diaz et al (Diaz et al, 2004, Transplant Immunology 13(4):313-317) was performed. Serum samples were obtained from 10 randomly selected baboons. Twenty-five percent heat- inactivated baboon serum was prepared. The heat-inactivated baboon sera were serially diluted and 100 μΙ of each concentration were placed in a 96 well v-bottom assay plate. Sera were mixed with a 100 μΙ aliquot of PBMC obtained from either GGTA1-KO or double-KO (GGTA1/CMAH DKO) pigs. The final concentration of PBMC in each well was 5 X 106/ml; in some experiments 1 x 10106/ml PBMC were used.
[00123] The mixtures were incubated for 30 minutes at 4°C. After 30 minutes, the plates were centrifuged for 4 minutes at 400 x g. The plates were decanted and washed with HBSS. Rabbit complement (150 μΙ of a 1:15 dilution) was added to each well and incubated for 30 minutes at 37°C. PBMC were labeled with a fluorescein diacetate (FDA) stock solution, prepared fresh daily in HBSS (1 μg/mL) from a 1 mg/ml stock solution in acetone and with propidium iodide (PI), prepared at 50 μg/ml in phosphate buffered saline (PBS). After incubation in complement, the samples were transferred by pipette to tu bes containing 250 μΙ of HBSS and 10 μΙ of FDA/PI for analysis using an Accuri C6 flow cytometer.
[00124] The %CTX for each sample was plotted against the log of each serum dilution and the sigmoid curve was analyzed by non-linear regression. Data from one such set of experiments are presented in Figure 13.
[00125] Serum was obtained from five randomly selected human donors. Twenty-five percent heat-inactivated serum was prepared. Human, baboon and chimpanzee PBMCs were obtained. PBMCs were obtained from single aGal knockout pigs (GGTA1-KO) and double CMAH/aGal (double-KO) pigs. Approximately 2 X 106/ml PBMCs were incubated with each human serum sample for 2 hours at 4°C. After incu bation of the serum and PBMCs, the PBMCs were washed three times in 0.5% PBS Sialix Blocking agent. PBMCs were stained with DyLight 649-conjugated Donkey anti-human IgM or DyLight 488 Donkey anti-human IgG (Jackson Immunoresearch Laboratories Inc, West Grove PA) for 1 hour at 4°C. PBMCs were washed three times using 0.5% PBS Sialix blocking agent. Analyses were performed using an Accuri C6 flow cytometer and BD CFlow Plus Software (Accuri, Ann Arbor Ml). Overlays were produced using Kaluza version 1.2 software from Beckman Coulter (Brea, CA). Secondary only control antibody staining was also evaluated. Histograms from one such experiment are presented in Figure 14.
[00126] The invention is not limited to the embodiments set forth herein for illustration, but includes everything that is within the scope of the claims. Furthermore, all references cited herein are hereby incorporated by reference in their entirety and for all purposes as if fully set forth herein.

Claims

CLAIMS We claim:
1. A knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and when tissue from said pig is transplanted into a human, hyperacute rejection is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
2. Porcine organs, tissue or cells for transplantation into a human having reduced expression of aGal and Neu5Gc on the porcine organs, tissue or cells.
3. A method for modifying a porcine organs, tissue or cells for transplantation into a human, the method comprising removing or reducing expression of aGal and Neu5Gc on the porcine organs, tissue or cells.
4. The method of claim 3 wherein the porcine organs, tissue, or cells are selected from the group consisting of skin, heart, liver, kidneys, lung, pancreas, thyroid, small bowel, and components thereof.
5. A method for making porcine organs, tissue or cells for transplantation into a human, the method comprising reducing expression of aGal and CMAH on the porcine organs, tissue or cells.
6. The method of claim 5 wherein said porcine organs, tissue, or cells are selected from the group consisting of skin, heart, liver, kidneys, lung, pancreas, small bowel, and components thereof.
7. A knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and wherein when tissue from said knockout pig is transplanted into a human, thrombocytopenia is decreased as compared to when tissue from a wild-type pig is transplanted into a human.
8. A knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig and wherein a liver from said pig exhibits reduced uptake of human platelets when said liver is exposed to said human platelets.
9. A method of increasing the duration of the period between when a human subject is identified as a subject in need of human liver transplant and when said human liver transplant occurs, said method comprising providing a liver from a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in the knockout pig is decreased as compared to a wild-type pig, and surgically attaching said liver from said knockout pig to said human subject in a therapeutically effective manner.
10. The method of claim 9, wherein said liver from said knockout pig is internal to said human subject.
11. The method of claim 9, wherein said liver from said knockout pig is external to said human subject.
12. The method of claim 9, wherein said liver is directly or indirectly attached to said subject.
13. A method of preparing organs, tissues, or cells for xenotransplantation into human patients with reduced rejection, the method comprising providing a transgenic pig as a source of transplant material wherein the transplant material is selected from the group consisting of organs, tissues, or cells, and wherein the pig masks or reduces the expression of at least two xenoreactive antigens on the transplant material.
14. The method of claim 13, wherein the at least two xenoreactive antigens are aGal and Neu5Gc.
15. A knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein the disruption of said a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; wherein the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and wherein expression of functional a(l,3)-galactosyltransferase and CMAH in said knockout pig is decreased as compared to a wild-type pig and when tissue from said knockout pig is transplanted into a human, a hyperacute rejection related symptom is improved as compared to when tissue from a wild- type pig is transplanted into a human.
16. A method of improving a hyperacute rejection related symptom in a patient comprising transplanting porcine organs, tissue or cells having reduced expression of aGal and CMAH into a human, wherein said hyperacute rejection related symptom is improved as compared to when tissue from a wild-type swine is transplanted into a human.
17. A cell culture reagent derived from a knockout pig comprising disrupted a(l,3)- galactosyltransferase and CMAH genes, wherein expression of functional a(l,3)-galactosyltransferase and CMAH in said knockout pig is decreased as compared to a wild-type pig, wherein said cell culture reagent is selected from the group comprising cell culture media, cell culture serum, cell culture additive and an isolated cell capable of proliferation.
18. A method of producing a glycoprotein of interest, said method comprising the step of incubating an isolated cell capable of expressing said glycoprotein of interest with a cell culture reagent derived from a knockout pig comprising disrupted a(l,3)-galactosyltransferase and CMAH genes, wherein the amount of Neu5Gc or alphaGal epitopes on said glycoprotein of interest is lower than the amount of Neu5Gc or alphaGal epitopes on said glycoprotein of interest when said isolated cell capable of expressing said glycoprotein of interest is incu bated with a cell culture reagent derived from a wild-type Pig-
19. The method of claim 18 wherein said glycoprotein of interest is a glycoprotein selected from the group comprising an antibody, growth factor, cytokine, hormone and clotting factor.
20. The method of claim 18, wherein the disruption of said a(l,3)-galactosyltransferase gene is selected from the group of disruptions comprising a 3 base pair deletion adjacent to a G to A substitution, a single base pair deletion, a single base pair insertion, a six base pair deletion, a two base pair insertion, a ten base pair deletion, a seven base pair deletion, and an eight base pair substitution for a five base pair sequence; wherein the disruption of said CMAH gene is selected from the group of disruptions comprising a four base pair insertion, a two base pair deletion, a single base pair insertion, an eight base pair deletion, a five base pair deletion, a three base pair deletion, a two base pair substitution for a single base pair, and a twenty base pair deletion; and wherein expression of functional a(l,3)-galactosyltransferase and CMAH in said knockout pig is decreased as compared to a wild-type pig.
PCT/US2013/066387 2012-10-24 2013-10-23 Double knockout (gt/cmah-ko) pigs, organs and tissues WO2014066505A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/436,963 US9888674B2 (en) 2012-10-24 2013-10-23 Double knockout (GT/CMAH-KO) pigs, organs and tissues
US15/848,870 US10667500B2 (en) 2012-10-24 2017-12-20 Double knockout (GT/CMAH-KO) pigs, organs and tissues
US16/855,905 US11666039B2 (en) 2012-10-24 2020-04-22 Double knockout (GT/CMAH-KO) pigs, organs and tissues
US18/101,810 US20240196872A1 (en) 2012-10-24 2023-01-26 Double knockout (gt/cmah-ko) pigs, organs and tissues

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261717845P 2012-10-24 2012-10-24
US61/717,845 2012-10-24
US13/804,365 2013-03-14
US13/804,365 US20140115728A1 (en) 2012-10-24 2013-03-14 Double knockout (gt/cmah-ko) pigs, organs and tissues

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/804,365 Continuation US20140115728A1 (en) 2012-10-24 2013-03-14 Double knockout (gt/cmah-ko) pigs, organs and tissues

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/436,963 A-371-Of-International US9888674B2 (en) 2012-10-24 2013-10-23 Double knockout (GT/CMAH-KO) pigs, organs and tissues
US15/848,870 Division US10667500B2 (en) 2012-10-24 2017-12-20 Double knockout (GT/CMAH-KO) pigs, organs and tissues

Publications (1)

Publication Number Publication Date
WO2014066505A1 true WO2014066505A1 (en) 2014-05-01

Family

ID=50486651

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/066387 WO2014066505A1 (en) 2012-10-24 2013-10-23 Double knockout (gt/cmah-ko) pigs, organs and tissues

Country Status (2)

Country Link
US (5) US20140115728A1 (en)
WO (1) WO2014066505A1 (en)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
WO2016059161A1 (en) * 2014-10-15 2016-04-21 Xenothera Composition with reduced immunogenicity
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
WO2016065046A1 (en) 2014-10-22 2016-04-28 Indiana University Research & Technology Corporation Triple transgenic pigs suitable for xenograft
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9888673B2 (en) 2014-12-10 2018-02-13 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
JP2018526015A (en) * 2015-09-09 2018-09-13 レビビコア, インコーポレイテッド Multi-transgenic pigs for xenotransplantation
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2020041384A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. 3-phenyl-2-cyano-azetidine derivatives, inhibitors of rna-guided nuclease activity
WO2020041387A1 (en) 2018-08-20 2020-02-27 The Brigham And Women's Hospital, Inc. Degradation domain modifications for spatio-temporal control of rna-guided nucleases
WO2020068304A2 (en) 2018-08-20 2020-04-02 The Broad Institute, Inc. Inhibitors of rna-guided nuclease target binding and uses thereof
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
WO2020236967A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Random crispr-cas deletion mutant
WO2021041922A1 (en) 2019-08-30 2021-03-04 The Broad Institute, Inc. Crispr-associated mu transposase systems
WO2021050974A1 (en) 2019-09-12 2021-03-18 The Broad Institute, Inc. Engineered adeno-associated virus capsids
US11180751B2 (en) 2015-06-18 2021-11-23 The Broad Institute, Inc. CRISPR enzymes and systems
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2022087089A1 (en) 2020-10-21 2022-04-28 Tissue Testing Technologies Llc Minimizing immunogenicity of decellularized tissues
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11479752B2 (en) 2017-10-20 2022-10-25 Indiana University Research And Technology Corporation Scaffold-free 3D bioprinting of porcine cells
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140115728A1 (en) 2012-10-24 2014-04-24 A. Joseph Tector Double knockout (gt/cmah-ko) pigs, organs and tissues
BR112017018932A2 (en) 2015-03-03 2018-07-31 Univ Minnesota etv2 and uses thereof
EP3313176A4 (en) * 2015-06-26 2019-06-26 Indiana University Research&technology Corporation Transgenic pigs with genetic modifications of sla
RU2018103093A (en) 2015-06-30 2019-07-31 Реджентс Оф Зэ Юниверсити Оф Миннесота HUMANIZED skeletal muscle
JP2018523999A (en) 2015-06-30 2018-08-30 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Humanized myocardium
EP3468356A4 (en) * 2016-06-14 2020-02-26 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
KR102363891B1 (en) * 2017-04-28 2022-02-17 서울대학교산학협력단 Transgenic pigs expressing both shTNFRI-Fc and HA-HO-1 without Gal epitope and N-glycolylneuraminic acid epitope and the Use of thereof
AU2019356014A1 (en) 2018-10-05 2021-05-20 Alexis Bio, Inc. Xenotransplantation products and methods
US10883084B2 (en) 2018-10-05 2021-01-05 Xenotherapeutics, Inc. Personalized cells, tissues, and organs for transplantation from a humanized, bespoke, designated-pathogen free, (non-human) donor and methods and products relating to same
EP3863400A4 (en) * 2018-10-10 2022-07-13 Revivicor Inc. Compositions and methods for preventing allergies
CN113425906A (en) * 2020-03-23 2021-09-24 成都中科奥格生物科技有限公司 Cartilage material and preparation method and application thereof
US20230203176A1 (en) 2021-09-17 2023-06-29 Novartis Ag Methods For Prevention Of Graft Rejection In Xenotransplantation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002088351A1 (en) * 2001-04-30 2002-11-07 Rbc Biotechnology, Inc. Modified organs and cells for xenotransplantation
WO2004028243A2 (en) * 2002-08-21 2004-04-08 Revivicor, Inc. Porcine animals lacking any expression of functional alpha 1,3 galactosyltransferase
US7547816B2 (en) 2001-12-21 2009-06-16 The Curators Of The University Of Missouri α(1,3)-galactosyltransferase knockout swine, tissues and organs

Family Cites Families (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5352240A (en) 1989-05-31 1994-10-04 Promedica International, Inc. Human heart valve replacement with porcine pulmonary valve
US5705732A (en) 1989-06-12 1998-01-06 Oklahoma Medical Research Foundation Universal donor cells
WO1991016449A1 (en) 1990-04-16 1991-10-31 The Trustees Of The University Of Pennsylvania Saccharide compositions, methods and apparatus for their synthesis
US5846715A (en) 1990-05-11 1998-12-08 The Austin Research Institute CD46 variants
WO1994021799A1 (en) 1993-03-16 1994-09-29 Austin Research Institute USE OF PORCINE GAL α(1,3) GALACTOSYL TRANSFERASE IN XENOGRAFT THERAPIES
US6331658B1 (en) 1993-04-20 2001-12-18 Integris Baptist Medical Center, Inc. Genetically engineered mammals for use as organ donors
US5560911A (en) 1993-10-12 1996-10-01 Oklahoma Medical Research Foundation Method of inhibiting acute complement mediated cytotoxicity with anti-idiotypic antibodies
US5849991A (en) 1994-01-27 1998-12-15 Bresatch Limited Mice homozygous for an inactivated α 1,3-galactosyl transferase gene
WO1995024494A1 (en) 1994-03-09 1995-09-14 Abbott Laboratories Humanized milk
US6204431B1 (en) 1994-03-09 2001-03-20 Abbott Laboratories Transgenic non-human mammals expressing heterologous glycosyltransferase DNA sequences produce oligosaccharides and glycoproteins in their milk
US6204053B1 (en) 1994-11-08 2001-03-20 Diacrin, Inc. Porcine cortical cells and their use in treatment of neurological deficits due to neurodegenerative diseases
US6294383B1 (en) 1994-11-08 2001-09-25 The Mclean Hospital Corporation Porcine neural cells and their use in treatment of neurological deficits due to neurodegenerative diseases
US5922577A (en) 1995-04-11 1999-07-13 Cytel Corporation Enzymatic synthesis of glycosidic linkages
US5728554A (en) 1995-04-11 1998-03-17 Cytel Corporation Enzymatic synthesis of glycosidic linkages
GB9517780D0 (en) 1995-08-31 1995-11-01 Roslin Inst Edinburgh Biological manipulation
US6166288A (en) 1995-09-27 2000-12-26 Nextran Inc. Method of producing transgenic animals for xenotransplantation expressing both an enzyme masking or reducing the level of the gal epitope and a complement inhibitor
WO1998005768A1 (en) 1996-08-02 1998-02-12 The Austin Research Institute Improved nucleic acids encoding a chimeric glycosyltransferase
AUPO182396A0 (en) 1996-08-23 1996-09-12 Austin Research Institute, The Improved nucleic acids for reducing carbohydrate epitopes
US6423316B1 (en) 1997-03-26 2002-07-23 Imperial College Innovative Limited Anticoagulant fusion protein anchored to cell membrane
GB9809280D0 (en) 1998-04-30 1998-07-01 Rpms Technology Ltd Immunosupression
WO2000004191A1 (en) 1998-07-17 2000-01-27 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods and devices for detection of xenogeneic graft persistence and infectious agents
US6444205B2 (en) 1998-09-30 2002-09-03 Diacrin, Inc. Transplantation of neural cells for the treatment of chronic pain or spasticity
GB2365445A (en) 1998-10-12 2002-02-20 Geron Bio Med Ltd Porcine occcytes with improved developmental competence
US6258998B1 (en) 1998-11-24 2001-07-10 Infigen, Inc. Method of cloning porcine animals
US7531715B1 (en) 1999-01-13 2009-05-12 Ppl Therapeutics (Scotland) Double nuclear transfer method and results thereof
US20020012660A1 (en) 1999-03-04 2002-01-31 Alan Colman Method of preparing a somatic cells for nuclear transfer
EP1158999A2 (en) 1999-03-05 2001-12-05 Diacrin, Inc. Methods for improving graft acceptance in a recipient by administration of a cytokine profile altering agent
US6245890B1 (en) 1999-03-26 2001-06-12 New York Blood Center, Inc. Porcine protein and uses thereof
WO2001030992A2 (en) 1999-10-22 2001-05-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education α1-3 GALACTOSYLTRANSFERASE GENE AND PROMOTER
CA2925231A1 (en) 1999-10-26 2001-05-03 Stichting Dienst Landbouwkundig Onderzoek Mammalian-type glycosylation in transgenic plants expressing galactosyltransferase and sialyl transferase
US20020081654A1 (en) 2000-04-07 2002-06-27 Sandrin Mauro Sergio Targeting hydrolase enzymes
WO2002015681A1 (en) 2000-08-25 2002-02-28 Nippon Meat Packers, Inc. Transgenic mammals
MXPA03008209A (en) 2001-03-12 2004-11-12 Progenetics Llc Production of high levels of transgenic factor viii with engineered stability, and its therapeutic uses.
JP2005510202A (en) 2001-03-28 2005-04-21 ネクストラン インコーポレイテッド Elimination of endogenous porcine retroviruses
US7214660B2 (en) 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20030124023A1 (en) 2001-12-21 2003-07-03 Wilson Burgess Method of sterilizing heart valves
AU2003265415A1 (en) 2002-08-14 2004-03-03 Immerge Biotherapeutics, Inc. Alpha(1,3)-GALACTOSYLTRANSFERASE NULL CELLS, METHODS OF SELECTING AND Alpha(1,3)-GALACTOSYLTRANSFERASE NULL SWINE PRODUCED THEREFROM
US8106251B2 (en) 2002-08-21 2012-01-31 Revivicor, Inc. Tissue products derived from porcine animals lacking any expression of functional alpha 1,3 galactosyltransferase
US7626075B2 (en) 2002-09-19 2009-12-01 Ximerex, Inc. Growth of foreign cells in fetal animals facilitated by conditional and selective destruction of native host cells
CA2528500A1 (en) 2003-06-06 2004-12-16 University Of Pittsburgh Porcine cmp-n-acetylneuraminic acid hydroxylase gene
WO2005010485A2 (en) 2003-07-15 2005-02-03 The Regents Of The University Of California Methods for detecting and analyzing n-glycolylneuraminic acid (neu5gc) in biological materials
CA2533259C (en) 2003-07-21 2014-01-28 Lifecell Corporation Acellular tissue matrices made from galactose .alpha.-1,3-galactose-deficient tissue
EP1685148A2 (en) 2003-11-05 2006-08-02 University Of Pittsburgh Of The Commonwealth System Of Higher Education PORCINE ISOGLOBOSIDE 3 SYNTHASE PROTEIN, cDNA, GENOMIC ORGANIZATION, AND REGULATORY REGION
US20090043383A1 (en) 2004-03-29 2009-02-12 Mayo Foundation For Medical Education And Research Genetically modified heart valve xenografts
AU2005243169A1 (en) 2004-05-07 2005-11-24 University Of Pittsburgh Of The Commonwealth System Of Higher Education Porcine Forssman synthetase protein, cDNA, genomic organization, and regulatory region
US7582741B2 (en) 2004-07-26 2009-09-01 University Of Massachusetts Conditional disruption of dicer1 in cell lines and non-human mammals
NZ539491A (en) 2005-04-15 2008-04-30 Living Cell Products Pty Ltd Swine population and uses thereof
US20080166805A1 (en) 2005-06-08 2008-07-10 The Regents Of The University Of California Culturing human cells and tissues in an N-glycolylneuraminic acid-free environment
US20070033666A1 (en) 2005-06-24 2007-02-08 Harris Reuben S Using cytosine deaminases to diminish retroelement transfer from pigs to humans
PT1904636E (en) 2005-07-12 2010-09-14 Greenovation Biotech Gmbh Improvements in or relating to protein production
CA2617930A1 (en) 2005-08-09 2007-03-29 Revivicor, Inc. Transgenic ungulates expressing ctla4-ig and uses thereof
GB0606231D0 (en) 2006-03-29 2006-05-10 Univ Leeds Improvements relating to decellurisation of tissue matrices for bladder implantation
US20100105140A1 (en) 2008-07-16 2010-04-29 Fahrenkrug Scott C Plaice dna transposon system
WO2010008562A2 (en) 2008-07-16 2010-01-21 Recombinetics Methods and materials for producing transgenic animals
US20120045816A1 (en) 2008-09-09 2012-02-23 Sialix, Inc. Novel Glycosylated Polypeptides
CA2736488A1 (en) 2008-09-09 2010-03-18 The Regents Of The University Of California Elimination of a contaminating non-human sialic acid by metabolic competition
US8709400B2 (en) 2009-07-27 2014-04-29 Washington University Inducement of organogenetic tolerance for pancreatic xenotransplant
ES2550202T3 (en) 2009-08-03 2015-11-05 Recombinetics, Inc. Methods and compositions for targeted gene modification
KR20120050485A (en) 2009-08-14 2012-05-18 레비비코르 인코포레이션 Multi-transgenic pigs for diabetes treatment
WO2011139488A2 (en) 2010-05-06 2011-11-10 Mayo Foundation For Medical Education And Research Methods and materials for reducing cardiac xenograft rejection
US8945868B2 (en) 2010-07-21 2015-02-03 Sangamo Biosciences, Inc. Methods and compositions for modification of a HLA locus
JP2014520533A (en) 2011-06-30 2014-08-25 シグマ−アルドリッチ・カンパニー・リミテッド・ライアビリティ・カンパニー Cells deficient in CMP-N-acetylneuraminic acid hydroxylase and / or glycoprotein alpha-1,3-galactosyltransferase
WO2013019625A2 (en) 2011-07-29 2013-02-07 The University Of Wyoming Methods of using a bacterial glcnac-6-p 2'-epimerase to promote sialylation of glycoconjugates
WO2013151649A1 (en) 2012-04-04 2013-10-10 Sialix Inc Glycan-interacting compounds
WO2013169929A1 (en) 2012-05-08 2013-11-14 The General Hospital Corporation Reducing immunogenicity of xenogeneic transplant tissues
US20140115728A1 (en) 2012-10-24 2014-04-24 A. Joseph Tector Double knockout (gt/cmah-ko) pigs, organs and tissues
CN107106607A (en) 2014-10-22 2017-08-29 印第安纳大学研究与技术公司 Suitable for triple transgene pigs of heterograft

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002088351A1 (en) * 2001-04-30 2002-11-07 Rbc Biotechnology, Inc. Modified organs and cells for xenotransplantation
US7166278B2 (en) 2001-04-30 2007-01-23 Rbc Biotechnology, Inc. Modified organs and cells for xenotransplantation
US8034330B2 (en) 2001-04-30 2011-10-11 Rbc Biotechnology, Inc. Modified organs and cells for xenotransplantation
US7547816B2 (en) 2001-12-21 2009-06-16 The Curators Of The University Of Missouri α(1,3)-galactosyltransferase knockout swine, tissues and organs
WO2004028243A2 (en) * 2002-08-21 2004-04-08 Revivicor, Inc. Porcine animals lacking any expression of functional alpha 1,3 galactosyltransferase
US7795493B2 (en) 2002-08-21 2010-09-14 Revivicor, Inc. Porcine animals lacking any expression of functional alpha 1, 3 galactosyltransferase

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
"Cells: a Laboratory Manual", vol. 1, 1998, COLD SPRING HARBOR LABORATORY PRESS
"Current Protocols in Molecular Biology", 2013, WILEY-INTERSCIENCE
ANDREW J. LUTZ ET AL: "Double knockout pigs deficient in N-glycolylneuraminic acid and Galactose [alpha]-1,3-Galactose reduce the humoral barrier to xenotransplantation", XENOTRANSPLANTATION, vol. 20, no. 1, 5 January 2013 (2013-01-05), pages 27 - 35, XP055069928, ISSN: 0908-665X, DOI: 10.1111/xen.12019 *
BASNET ET AL., XENOTRANSPLANTATION, vol. 17, no. 6, 2010, pages 440 - 448
BASNET NABIN B ET AL: "Deficiency of N-glycolylneuraminic acid and Gal alpha 1-3Gal beta 1-4GlcNAc epitopes in xenogeneic cells attenuates cytotoxicity of human natural antibodies", XENOTRANSPLANTATION, vol. 17, no. 6, November 2010 (2010-11-01), pages 440 - 448, XP002717542, ISSN: 0908-665X *
CESARE GALLI ET AL: "Genetic engineering including superseding microinjection: new ways to make GM pigs", XENOTRANSPLANTATION, vol. 17, no. 6, 1 November 2010 (2010-11-01), pages 397 - 410, XP055007654, ISSN: 0908-665X, DOI: 10.1111/j.1399-3089.2010.00590.x *
CHEN ET AL., NATURE MED., vol. 11, no. 12, 2005, pages 1295 - 1298
CHEN GANG ET AL: "Acute rejection is associated with antibodies to non-Gal antigens in baboons using Gal-knockout pig kidneys", NATURE MEDICINE, vol. 11, no. 12, December 2005 (2005-12-01), pages 1295 - 1298, XP002717545, ISSN: 1078-8956 *
COLIGAN ET AL.: "Current Protocols in Protein Science", 2013, WILEY-INTERSCIENCE
DARLING ET AL.: "Animal Cells: culture and media", 1994, JOHN WILEY & SONS
DIAZ ET AL., TRANSPLANT IMMUNOLOGY, vol. 13, no. 4, 2004, pages 313 - 317
FRESHNEY: "ED) Culture of Animal Cells: a manual of basic techniques", WILEY-LISS
GALLI ET AL., XENOTRANSPLANTATION, vol. 17, no. 6, 2010, pages 397 - 410
GHADERI, NATURE BIOTECHNOLOGY, vol. 28, no. 8, 2010, pages 863 - 867
LI ET AL., CELL REPROGRAM., vol. 12, 2010, pages 599
LI ET AL., J. SURG RES, vol. 181, 2013, pages E39 - 45
LI ET AL., JOURNAL OF SURGICAL RESEARCH, 3 July 2012 (2012-07-03), pages E1 - E7
LI ET AL., JSR, 3 July 2012 (2012-07-03)
LI PING ET AL: "Biallelic knockout of the alpha-1,3 galactosyltransferase gene in porcine liver-derived cells using zinc finger nucleases", JOURNAL OF SURGICAL RESEARCH, vol. 181, no. 1, 3 July 2012 (2012-07-03), pages E39 - E45, XP002717543 *
MATOU-KOVD, ANN MED BURN CLUB, vol. 7, 1994, pages 143
PARK JONG-YI ET AL: "Alpha 1,3-galactosyltransferase deficiency in pigs increases sialyltransferase activities that potentially raise non-gal xenoantigenicity.", JOURNAL OF BIOMEDICINE & BIOTECHNOLOGY, vol. 2011, 560850, 2011, pages 8PP, XP002717544, ISSN: 1110-7251 *

Cited By (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US12006520B2 (en) 2011-07-22 2024-06-11 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US10954548B2 (en) 2013-08-09 2021-03-23 President And Fellows Of Harvard College Nuclease profiling system
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US10227581B2 (en) 2013-08-22 2019-03-12 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US10682410B2 (en) 2013-09-06 2020-06-16 President And Fellows Of Harvard College Delivery system for functional nucleases
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US10912833B2 (en) 2013-09-06 2021-02-09 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10640788B2 (en) 2013-11-07 2020-05-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAs
US11390887B2 (en) 2013-11-07 2022-07-19 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US11124782B2 (en) 2013-12-12 2021-09-21 President And Fellows Of Harvard College Cas variants for gene editing
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10704062B2 (en) 2014-07-30 2020-07-07 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
EP3613764A1 (en) * 2014-10-15 2020-02-26 Xenothera Composition with reduced immunogenicity
WO2016059161A1 (en) * 2014-10-15 2016-04-21 Xenothera Composition with reduced immunogenicity
US11725044B2 (en) 2014-10-15 2023-08-15 Xenothera Method for producing polyclonal antibodies with improved complement-dependent cytotoxicity
CN107001449A (en) * 2014-10-15 2017-08-01 芝诺锡拉公司 The composition of immunogenicity with reduction
EP3220925A4 (en) * 2014-10-22 2018-07-18 Indiana University Research&technology Corporation Triple transgenic pigs suitable for xenograft
EP4129308A1 (en) * 2014-10-22 2023-02-08 Indiana University Research & Technology Corporation Triple transgenic pigs suitable for xenograft
WO2016065046A1 (en) 2014-10-22 2016-04-28 Indiana University Research & Technology Corporation Triple transgenic pigs suitable for xenograft
US11234418B2 (en) 2014-12-10 2022-02-01 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US9888673B2 (en) 2014-12-10 2018-02-13 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US10278372B2 (en) 2014-12-10 2019-05-07 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US10993419B2 (en) 2014-12-10 2021-05-04 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US11180751B2 (en) 2015-06-18 2021-11-23 The Broad Institute, Inc. CRISPR enzymes and systems
JP2018526015A (en) * 2015-09-09 2018-09-13 レビビコア, インコーポレイテッド Multi-transgenic pigs for xenotransplantation
JP2021101741A (en) * 2015-09-09 2021-07-15 レビビコア, インコーポレイテッド Multi-transgenic pig for xenotransplantation
EP3347458A4 (en) * 2015-09-09 2019-08-07 Revivicor Inc. Multi-transgenic pig for xenotransplantation
JP7050665B2 (en) 2015-09-09 2022-04-08 レビビコア, インコーポレイテッド Multi-transgenic pigs for xenotransplantation
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11999947B2 (en) 2016-08-03 2024-06-04 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11479752B2 (en) 2017-10-20 2022-10-25 Indiana University Research And Technology Corporation Scaffold-free 3D bioprinting of porcine cells
WO2020041387A1 (en) 2018-08-20 2020-02-27 The Brigham And Women's Hospital, Inc. Degradation domain modifications for spatio-temporal control of rna-guided nucleases
WO2020041384A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. 3-phenyl-2-cyano-azetidine derivatives, inhibitors of rna-guided nuclease activity
WO2020068304A2 (en) 2018-08-20 2020-04-02 The Broad Institute, Inc. Inhibitors of rna-guided nuclease target binding and uses thereof
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
WO2020236967A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Random crispr-cas deletion mutant
WO2021041922A1 (en) 2019-08-30 2021-03-04 The Broad Institute, Inc. Crispr-associated mu transposase systems
WO2021050974A1 (en) 2019-09-12 2021-03-18 The Broad Institute, Inc. Engineered adeno-associated virus capsids
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2022087089A1 (en) 2020-10-21 2022-04-28 Tissue Testing Technologies Llc Minimizing immunogenicity of decellularized tissues

Also Published As

Publication number Publication date
US20140115728A1 (en) 2014-04-24
US10667500B2 (en) 2020-06-02
US11666039B2 (en) 2023-06-06
US20240196872A1 (en) 2024-06-20
US20210076647A1 (en) 2021-03-18
US9888674B2 (en) 2018-02-13
US20150264900A1 (en) 2015-09-24
US20180153146A1 (en) 2018-06-07

Similar Documents

Publication Publication Date Title
US11666039B2 (en) Double knockout (GT/CMAH-KO) pigs, organs and tissues
AU2017285224B2 (en) Genetically modified cells, tissues, and organs for treating disease
EP3220925B1 (en) Triple transgenic pigs suitable for xenograft
JP2021126129A (en) Genetically modified cells, tissues and organs for treating diseases
JP5329502B2 (en) Pig animals lacking any expression of functional alpha-1,3-galactosyltransferase
US20180184630A1 (en) Transgenic pigs with genetic modifications of sla
US20170251646A1 (en) Transgenic pigs lacking one or more cellular transport genes
Joanna et al. The production of UL16-binding protein 1 targeted pigs using CRISPR technology
US9420770B2 (en) Methods of modulating thrombocytopenia and modified transgenic pigs
JPWO2015155904A1 (en) Gene knockout pig
Hossein et al. Cloning missy: obtaining multiple offspring of a specific canine genotype by somatic cell nuclear transfer
JP5771240B2 (en) Immunodeficient pig
LE ANH Studies on genome editing techniques using CRISPR/Cas9 in porcine embryos
WO2023076897A1 (en) Viable galactosyltransferase knock-out sheep and related methods
Ishikawa et al. Insertion of transgene in sex chromosomes in generating transgenic mice

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13786121

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14436963

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13786121

Country of ref document: EP

Kind code of ref document: A1