WO2023083250A1 - Animal non humain génétiquement modifié comportant un cd2 humain ou chimérique - Google Patents

Animal non humain génétiquement modifié comportant un cd2 humain ou chimérique Download PDF

Info

Publication number
WO2023083250A1
WO2023083250A1 PCT/CN2022/131092 CN2022131092W WO2023083250A1 WO 2023083250 A1 WO2023083250 A1 WO 2023083250A1 CN 2022131092 W CN2022131092 W CN 2022131092W WO 2023083250 A1 WO2023083250 A1 WO 2023083250A1
Authority
WO
WIPO (PCT)
Prior art keywords
animal
human
exon
endogenous
sequence
Prior art date
Application number
PCT/CN2022/131092
Other languages
English (en)
Inventor
Ying Zhao
Chong Li
Zhiyuan Shen
Original Assignee
Biocytogen Pharmaceuticals (Beijing) Co., Ltd.
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 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. filed Critical Biocytogen Pharmaceuticals (Beijing) Co., Ltd.
Publication of WO2023083250A1 publication Critical patent/WO2023083250A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70507CD2
    • 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
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • This disclosure relates to genetically modified animal expressing human or chimeric (e.g., humanized) CD2, and methods of use thereof.
  • test results obtained from the use of conventional experimental animals for in vivo pharmacological test may not reflect the real disease state and the interaction at the targeting sites, resulting in that the results in many clinical trials are significantly different from the animal experimental results.
  • the animal model can express human CD2 or chimeric CD2 (e.g., humanized CD2) protein in its body. It can be used in the studies on the function of CD2 gene, and can be used in the screening and evaluation of anti-human CD2 antibodies or drugs targeting CD2.
  • the animal models prepared by the methods described herein can be used in drug screening, pharmacodynamics studies, treatments for immune-related diseases, and cancer therapy for human CD2 target sites; they can also be used to facilitate the development and design of new drugs, and save time and cost.
  • this disclosure provides a powerful tool for studying the function of CD2 protein and a platform for screening cancer drugs.
  • the disclosure is related to a genetically-modified, non-human animal whose genome comprises at least one chromosome comprising a sequence encoding a human or chimeric cluster of differentiation 2 (CD2) .
  • the sequence encoding the human or chimeric CD2 is operably linked to an endogenous regulatory element at the endogenous CD2 gene locus in the at least one chromosome.
  • the sequence encoding a human or chimeric CD2 comprises a sequence encoding an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%identical to human CD2 (NP_001758.2 (SEQ ID NO: 2) ) .
  • the sequence encoding a human or chimeric CD2 comprises a sequence encoding an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%identical to SEQ ID NO: 10. In some embodiments, the sequence encoding a human or chimeric CD2 comprises a sequence encoding an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%identical to amino acids 1-208 of SEQ ID NO: 2.
  • the animal is a mammal, e.g., a monkey, a rodent, a mouse, or a rat. In some embodiments, the animal is a mouse.
  • the animal does not express endogenous CD2 or expresses a decreased level of endogenous CD2 as compared to CD2 expression level in a wild-type animal.
  • the animal has one or more cells expressing human or chimeric CD2.
  • the animal has one or more cells expressing human or chimeric CD2, and the expressed human or chimeric CD2 can interact with a human lymphocyte function-associated antigen 3 (CD58) , activating downstream signaling pathways.
  • the animal has one or more cells expressing human or chimeric CD2, and the expressed human or chimeric CD2 can interact with an endogenous B-lymphocyte activation marker (CD48) , activating downstream signaling pathways.
  • CD48 endogenous B-lymphocyte activation marker
  • the disclosure is related to a genetically-modified, non-human animal
  • the genome of the animal comprises a replacement of a sequence encoding a region of endogenous CD2 with a sequence encoding a corresponding region of human CD2 at an endogenous CD2 gene locus.
  • the sequence encoding the corresponding region of human CD2 is operably linked to an endogenous regulatory element at the endogenous CD2 locus, and one or more cells of the animal expresses a human or chimeric CD2.
  • the animal does not express endogenous CD2 or expresses a decreased level of endogenous CD2 as compared to CD2 expression level in a wild-type animal.
  • the replaced sequence encodes all or a portion of the extracellular region of CD2, optionally including the signal peptide.
  • the animal has one or more cells expressing a chimeric CD2 having a signal peptide, an extracellular region, a transmembrane region, and a cytoplasmic region, in some embodiments, the extracellular region comprises a sequence that is at least 50%, 60%, 70%, 80%, 90%, 95%, or 99%identical to the extracellular region of human CD2 (NP_001758.2 (SEQ ID NO: 2) ) .
  • the extracellular region of the chimeric CD2 has a sequence that has at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 181, 182, 183, 184, or 185 contiguous amino acids that are identical to a contiguous sequence present in the extracellular region of human CD2 (e.g., amino acids 25-208 of SEQ ID NO: 2) .
  • the signal peptide of the chimeric CD2 has a sequence that is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 contiguous amino acids that are identical to a contiguous sequence present in the signal peptide of human CD2 (e.g., amino acids 1-24 of SEQ ID NO: 2) .
  • the sequence encoding a region of endogenous CD2 comprises exon 1, exon 2, exon 3, and/or exon 4, or a part thereof, of the endogenous CD2 gene.
  • the animal is a mouse.
  • the animal is heterozygous with respect to the replacement at the endogenous CD2 gene locus.
  • the animal is homozygous with respect to the replacement at the endogenous CD2 gene locus.
  • the disclosure is related to a method for making a genetically-modified, non-human animal, comprising: replacing in at least one cell of the animal, at an endogenous CD2 gene locus, a sequence encoding a region of endogenous CD2 with a sequence encoding a corresponding region of human CD2.
  • the sequence encoding the corresponding region of human CD2 comprises exon 1, exon 2, exon 3, and/or exon 4, or a part thereof, of a human CD2 gene.
  • the sequence encoding the corresponding region of human CD2 comprises a portion of exon 1, exon 2, exon 3, and a portion of exon 4, of a human CD2 gene.
  • the sequence encoding the corresponding region of human CD2 encodes amino acids 1-208 of SEQ ID NO: 2.
  • the region comprises all or a portion of the extracellular region, optionally the signal peptide, of CD2.
  • the sequence encoding a region of endogenous CD2 comprises exon 1, exon 2, exon 3, and/or exon 4, or a part thereof, of the endogenous CD2 gene.
  • the animal is a mouse, and the sequence encoding a region of endogenous CD2 comprises a portion of exon 1, exon 2, exon 3, and a portion of exon 4 of the endogenous CD2 gene.
  • the disclosure is related to a non-human animal comprising at least one cell comprising a nucleotide sequence encoding a humanized CD2 polypeptide
  • the humanized CD2 polypeptide comprises at least 50 contiguous amino acid residues that are identical to the corresponding contiguous amino acid sequence of a human CD2, in some embodiments, the animal expresses the humanized CD2 polypeptide.
  • the humanized CD2 polypeptide has at least 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 181, 182, 183, 184, or 185 contiguous amino acid residues that are identical to the corresponding contiguous amino acid sequence of human CD2 extracellular region (e.g., amino acids 25-208 of SEQ ID NO: 2) .
  • the humanized CD2 polypeptide has at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 contiguous amino acid residues that are identical to the corresponding contiguous amino acid sequence of human CD2 signal peptide (e.g., amino acids 1-24 of SEQ ID NO: 2) .
  • the humanized CD2 polypeptide comprises a sequence that is at least 90%, 95%, or 99%identical to amino acids 1-208 of SEQ ID NO: 2.
  • the nucleotide sequence is operably linked to an endogenous CD2 regulatory element of the animal.
  • the chimeric CD2 polypeptide comprises an endogenous CD2 transmembrane region and/or an endogenous CD2 cytoplasmic region.
  • the nucleotide sequence is integrated to an endogenous CD2 gene locus of the animal.
  • the humanized CD2 polypeptide has at least one mouse CD2 activity and/or at least one human CD2 activity.
  • the disclosure is related to a method of making a genetically-modified animal cell that expresses a chimeric CD2, the method comprising: replacing at an endogenous CD2 gene locus, a nucleotide sequence encoding a region of endogenous CD2 with a nucleotide sequence encoding a corresponding region of human CD2, thereby generating a genetically-modified animal cell that includes a nucleotide sequence that encodes the chimeric CD2, in some embodiments, the animal cell expresses the chimeric CD2. In some embodiments, the animal is a mouse.
  • the chimeric CD2 comprises a human or humanized CD2 extracellular region; and a transmembrane and/or a cytoplasmic region of mouse CD2. In some embodiments, the chimeric CD2 further comprises a human or humanized CD2 signal peptide. In some embodiments, the nucleotide sequence encoding the chimeric CD2 is operably linked to an endogenous CD2 regulatory region, e.g., promoter.
  • the animal further comprises a sequence encoding an additional human or chimeric protein.
  • the additional human or chimeric protein is CD3e molecule, epsilon (CD3E) , CD28, CD226, IL2, IL4, IL6, programmed cell death protein 1 (PD-1) , tumor necrosis factor receptor superfamily member 9 (4-1BB) , CD40, programmed cell death ligand 1 (PD-L1) , lymphocyte-activation gene 3 (LAG3) , tumor necrosis factor alpha (TNF- ⁇ ) and/or T cell immunoreceptor with Ig and ITIM domains (TIGIT) .
  • CD3e molecule CD3e molecule, epsilon (CD3E) , CD28, CD226, IL2, IL4, IL6, programmed cell death protein 1 (PD-1) , tumor necrosis factor receptor superfamily member 9 (4-1BB) , CD40, programmed cell death ligand 1 (PD-L1) , lymphocyte-activation gene 3
  • the disclosure is related to a method of determining effectiveness of a therapeutic agent for the treatment of cancer, comprising: a) administering the therapeutic agent to the animal as described herein, in some embodiments, the animal has a tumor; and b) determining inhibitory effects of the therapeutic agent to the tumor.
  • the therapeutic agent is an anti-CD2 antibody.
  • the tumor comprises one or more cancer cells that are injected into the animal.
  • determining inhibitory effects of the anti-CD2 antibody to the tumor involves measuring the tumor volume in the animal.
  • the cancer is colorectal, endometrial, or ovarian cancer.
  • the disclosure is related to a method of determining effectiveness of an anti-CD2 antibody and an additional therapeutic agent for the treatment of cancer, comprising a) administering the anti-CD2 antibody and the additional therapeutic agent to the animal as described herein, in some embodiments, the animal has a tumor; and b) determining inhibitory effects on the tumor.
  • the animal further comprises a sequence encoding a human or chimeric PD-1, a human or chimeric PD-L1, and/or a human or chimeric CTLA4.
  • the additional therapeutic agent is an anti-PD-1 antibody, an anti-PD-L1 antibody, or an anti-CTLA4 antibody.
  • the tumor comprises one or more tumor cells that express PD-L1.
  • the tumor comprises one or more cancer cells that are injected into the animal.
  • determining inhibitory effects of the treatment involves measuring the tumor volume in the animal.
  • the animal has colorectal, endometrial, or ovarian cancer.
  • the disclosure is related to a method of determining effectiveness of a therapeutic agent for treatment an immune disorder, comprising: a) administering the therapeutic agent to the animal as described herein, in some embodiments, the animal has the immune disorder; and b) determining effects of the therapeutic agent to the immune disorder.
  • the disclosure is related to a method of determining effectiveness of a therapeutic agent for reducing an inflammation, comprising: a) administering the therapeutic agent to the animal as described herein, in some embodiments, the animal has the inflammation; and b) determining effects of the therapeutic agent to the inflammation.
  • the disclosure is related to a method of determining effectiveness of a therapeutic agent for treating an autoimmune disease, comprising: a) administering the therapeutic agent to the animal as described herein, in some embodiments, the animal has the autoimmune disease; and b) determining effects of the therapeutic agent to the autoimmune disease.
  • the autoimmune disease is graft versus host disease (GVHD) or psoriasis.
  • the disclosure is related to a method of determining toxicity of a therapeutic agent comprising: a) administering the therapeutic agent to the animal as described herein; and b) determining effects of the therapeutic agent to the animal.
  • the therapeutic agent is an anti-CD2 antibody.
  • determining effects of the therapeutic agent to the animal involves measuring the body weight, red blood cell count, hematocrit, and/or hemoglobin of the animal.
  • the disclosure is related to a protein comprising an amino acid sequence
  • the amino acid sequence is one of the following: (a) an amino acid sequence set forth in SEQ ID NO: 1, 2, or 10; (b) an amino acid sequence that is at least 90%identical to SEQ ID NO: 1, 2, or 10; (c) an amino acid sequence that is at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to SEQ ID NO: 1, 2, or 10; (d) an amino acid sequence that is different from the amino acid sequence set forth in SEQ ID NO: 1, 2, or 10 by no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid; and (e) an amino acid sequence that comprises a substitution, a deletion and /or insertion of one, two, three, four, five or more amino acids to the amino acid sequence set forth in SEQ ID NO: 1, 2, or 10.
  • the disclosure is related to a nucleic acid comprising a nucleotide sequence
  • the nucleotide sequence is one of the following: (a) a sequence that encodes the protein as described herein; (b) SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14; (c) a sequence that is at least 90%identical to SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14; and (d) a sequence that is at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14.
  • the disclosure is related to a cell comprising the protein and/or the nucleic acid as described herein. In one aspect, the disclosure is related to an animal comprising the protein and/or the nucleic acid as described herein.
  • the disclosure further relates to a CD2 genomic DNA sequence of a humanized mouse, a DNA sequence obtained by a reverse transcription of the mRNA obtained by transcription thereof is consistent with or complementary to the DNA sequence; a construct expressing the amino acid sequence thereof; a cell comprising the construct thereof; a tissue comprising the cell thereof.
  • the disclosure further relates to the use of the non-human mammal or an offspring thereof, or the tumor bearing non-human mammal, the animal model generated through the method as described herein in the development of a product related to an immunization processes of human cells, the manufacture of a human antibody, or the model system for a research in pharmacology, immunology, microbiology and medicine.
  • the disclosure also relates to the use of the non-human mammal or an offspring thereof, or the tumor bearing non-human mammal, the animal model generated through the method as described herein in the production and utilization of an animal experimental disease model of an immunization processes involving human cells, the study on a pathogen, or the development of a new diagnostic strategy and /or a therapeutic strategy.
  • the disclosure further relates to the use of the non-human mammal or an offspring thereof, or the tumor bearing non-human mammal, the animal model generated through the methods as described herein, in the screening, verifying, evaluating or studying the CD2 gene function, human CD2 antibodies, the drugs or efficacies for human CD2 targeting sites, and the drugs for immune-related diseases and antitumor drugs.
  • FIG. 1 is a schematic diagram showing mouse and human CD2 gene loci.
  • FIG. 2 is a schematic diagram showing humanized CD2 gene locus.
  • FIG. 3 is a schematic diagram showing a CD2 gene targeting strategy.
  • FIG. 4 is a schematic diagram showing the Frt recombination process in CD2 gene humanized mice.
  • FIG. 5 is a schematic diagram showing a CD2 gene targeting strategy.
  • FIGS. 6A-6B show mouse tail PCR identification results of F1 generation mice by primers F1-F and F1-R.
  • M is a marker.
  • WT is a wild-type control.
  • H 2 O is a water control.
  • FIG. 7 shows Southern Blot results of cells after recombination using the 5’ Probe and A Probe (3’) .
  • WT is a wild-type control.
  • FIGS. 8A-8C show RT-PCR detection results of mouse CD2 mRNA (by primers PCR-F1/PCR-R1) , humanized CD2 mRNA (by primers PCR-F2/PCR-R2) , and GAPDH mRNA (by primers GAPDH-F/GAPDH-R) , respectively, in the spleen tissues of a wild-type C57BL/6 mouse (+/+) and a CD2 gene humanized homozygous mouse (H/H) .
  • H 2 O is a water control.
  • FIGS. 9A-9D show mouse tail PCR identification results of F1 generation CD2 gene knockout mice.
  • M is a marker.
  • WT is a wild-type control.
  • H 2 O is a water control.
  • FIG. 10 shows the alignment between human CD2 amino acid sequence (NP_001758.2; SEQ ID NO: 2) and mouse CD2 amino acid sequence (NP_038514.1; SEQ ID NO: 1) .
  • FIG. 11 shows the alignment between human CD2 amino acid sequence (NP_001758.2; SEQ ID NO: 2) and rat CD2 amino acid sequence (NP_036962.2; SEQ ID NO: 44) .
  • This disclosure relates to transgenic non-human animal with human or chimeric (e.g., humanized) CD2, and methods of use thereof.
  • CD2 is a transmembrane glycoprotein of the immunoglobulin superfamily expressed on the surface of T cells, NK cells, thymocytes and dendritic cells.
  • the corresponding binding partner of CD2 is lymphocyte-associated antigen 3 (LFA3; also known as CD58) which is expressed on the surface of B cells, T cells, monocytes, granulocytes, thymic epithelial cells.
  • LFA3 lymphocyte-associated antigen 3
  • CD2 also binds to CD48, albeit with a relatively lower affinity.
  • Experimental animal models are an indispensable research tool for studying the effects of these antibodies (e.g., anti-CD2 antibodies) .
  • Common experimental animals include mice, rats, guinea pigs, hamsters, rabbits, dogs, monkeys, pigs, fish and so on.
  • human and animal genes and protein sequences there are many differences between human and animal genes and protein sequences, and many human proteins cannot bind to the animal’s homologous proteins to produce biological activity, leading to that the results of many clinical trials do not match the results obtained from animal experiments.
  • a large number of clinical studies are in urgent need of better animal models.
  • the use of human cells or genes to replace or substitute an animal’s endogenous similar cells or genes to establish a biological system or disease model closer to human, and establish the humanized experimental animal models (humanized animal model) has provided an important tool for new clinical approaches or means.
  • the genetically engineered animal model that is, the use of genetic manipulation techniques, the use of human normal or mutant genes to replace animal homologous genes, can be used to establish the genetically modified animal models that are closer to human gene systems.
  • the humanized animal models have various important applications. For example, due to the presence of human or humanized genes, the animals can express or express in part of the proteins with human functions, so as to greatly reduce the differences in clinical trials between humans and animals, and provide the possibility of drug screening at animal levels.
  • CD2 Cluster of differentiation 2 is a cell adhesion molecule found on the surface of T cells and natural killer (NK) cells. It has also been called T-cell surface antigen T11/Leu-5, LFA-2, LFA-3 receptor, erythrocyte receptor and rosette receptor. It interacts with other adhesion molecules, such as lymphocyte function-associated antigen-3 (LFA-3/CD58) in humans, or CD48 in rodents, which are expressed on the surfaces of other cells. In addition to its adhesive properties, CD2 also acts as a co-stimulatory molecule on T and NK cells.
  • NK natural killer
  • Human CD2 is a transmembrane protein in T cells that consists of 327 amino acids (351 amino acids including signal sequence) and a protein weight of about 40 kDa. Fully-glycosylated CD2 can weigh up to 50 kDa.
  • the extracellular domain (ECD) of CD2 includes two Ig domains, and is linked to a disordered proline-rich cytoplasmic tail via a transmembrane helix.
  • the membrane-proximal domain of the CD2 ECD connects the adhesion (membrane-distal) domain to the cell membrane and may be involved in CD2 clustering following T cell activation.
  • the membrane-distal domain is responsible for CD58 binding.
  • CD2 can bind to its ligand CD58 on APC in humans and on CD48 in rodents. Protein-protein interaction between CD2 and CD58 helps to enhance T cell-APC adhesion and thus promotes T-cell activation. In addition to its adhesion function, CD2 has also been suggested to be involved in signaling. Blocking of the CD2-CD58 interaction leads to immunosuppression in both model systems and humans, indicating its importance for the cellular immune response.
  • Alefacept a recombinant human CD58-Ig fusion protein that effectively binds to CD2 and prevents its interaction with CD58 expressed on APC, has been shown to be efficacious in the treatment of moderate-to-severe chronic plaque psoriasis in adult patients with psoriatic arthritis (PsA) .
  • PsA psoriatic arthritis
  • CD2-CD58 costimulatory blockade Organ transplantation and certain types of autoimmune disorders, conditions characterized by excessive activation of T cells and NK cells, constitute the natural target for CD2-CD58 costimulatory blockade (CoB) .
  • CD2-CD58 CoB could, e.g., be used in induction therapy and maintenance immunosuppression to prolong graft survival and minimize the risk for allograft rejection.
  • a substantial part of CD2-CD58 CoB clinical studies have been conducted using depleting anti-CD2 mAbs which provides two MoAs: depletion of CD2-expressing cells and CD2/CD58 costimulatory blockade.
  • CD2 immunobiology.
  • Immunosuppression by Co-stimulatory Molecules Inhibition of CD2-CD48/CD58 Interaction by Peptides from CD2 to Suppress Progression of Collagen-induced Arthritis in Mice.
  • Chemical Biology &Drug Design 82.1 (2013) : 106-118; and Liu, L. L., et al. Critical role of CD2 co-stimulation in adaptive natural killer cell responses revealed in NKG2C-deficient humans.
  • Cell Reports 15.5 (2016) : 1088-1099 each of which is incorporated by reference in its entirety.
  • CD2 gene locus has five exons, exon 1, exon 2, exon 3, exon 4, and exon 5 (FIG. 1) .
  • the CD2 protein also has, from N-terminus to C-terminus, a signal peptide, an extracellular region, a transmembrane region, and a cytoplasmic region.
  • the nucleotide sequence for human CD2 mRNA is NM_001767.5
  • the amino acid sequence for human CD2 is NP_001758.2 (SEQ ID NO: 2) .
  • the location for each exon and each region in human CD2 nucleotide sequence and amino acid sequence is listed below:
  • the human CD2 gene (Gene ID: 914) is located in Chromosome 1 of the human genome, which is located from 116754430 to 116769229 (GRCh38. p13 (GCF_000001405.39) ) .
  • the 5’-UTR is from 116754430 to 116754492
  • exon 1 is from 116754430 to 116754553
  • intron 1 is from 116754554 to 116754630
  • exon 2 is from 116754631 to 116754951
  • intron 2 is from 116754952 to 116760401
  • exon 3 is from 116760402 to 116760632
  • intron 3 is from 116760633 to 116764483
  • exon 4 is from 116764484 to 116764606, intron 4 is from 116764607 to 116768463
  • exon 5 is from 116768464 to 116769229
  • the 3’-UTR is from 116768784 to
  • CD2 gene locus has five exons, exon 1, exon 2, exon 3, exon 4, and exon 5 (FIG. 1) .
  • the mouse CD2 protein also has, from N-terminus to C-terminus, a signal peptide, an extracellular region, a transmembrane region, and a cytoplasmic region.
  • the nucleotide sequence for mouse CD2 mRNA is NM_013486.2
  • the amino acid sequence for mouse CD2 is NP_038514.1 (SEQ ID NO: 1) .
  • the location for each exon and each region in the mouse CD2 nucleotide sequence and amino acid sequence is listed below:
  • the mouse CD2 gene (Gene ID: 12481) is located in Chromosome 3 of the mouse genome, which is located from 101183224 to 101195255 (GRCm39 (GCF_000001635.27) ) .
  • the 5’-UTR is from 101195196 to 101195255
  • exon 1 is from 101195255 to 101195141
  • intron 1 is from 101195140 to 101195032
  • exon 2 is from 101195031 to 101194723
  • intron 2 is from 101194722 to 101190578
  • exon 3 is from 101190577 to 101190347
  • intron 3 is from 101190346 to 101187227
  • exon 4 is from 101187226 to 101187104
  • intron 4 is from 101187103 to 101183590
  • exon 5 is from 101183589 to 101183215
  • the 3’-UTR is from 101183215 to 101183272, based on transcript NM_013486.2. All relevant information for mouse CD2 locus can be
  • FIG. 10 shows the alignment between human CD2 amino acid sequence (NP_001758.2; SEQ ID NO: 2) and mouse CD2 amino acid sequence (NP_038514.1; SEQ ID NO: 1) .
  • NP_001758.2 SEQ ID NO: 2
  • mouse CD2 amino acid sequence NP_038514.1; SEQ ID NO: 1
  • CD2 genes, proteins, and locus of the other species are also known in the art.
  • the gene ID for CD2 in Rattus norvegicus (rat) is 497761
  • the gene ID for CD2 in Macaca mulatta (Rhesus monkey) is 677855
  • the gene ID for CD2 in Canis lupus familiaris (dog) is 607448
  • the gene ID for CD2 in Sus scrofa (pig) is 396662.
  • the relevant information for these genes e.g., intron sequences, exon sequences, amino acid residues of these proteins
  • NCBI database which is incorporated by reference herein in its entirety.
  • FIG. 11 shows the alignment between human CD2 amino acid sequence (NP_001758.2; SEQ ID NO: 2) and rat CD2 amino acid sequence (NP_036962.2; SEQ ID NO: 44) .
  • NP_001758.2 SEQ ID NO: 2
  • rat CD2 amino acid sequence NP_036962.2; SEQ ID NO: 44
  • the present disclosure provides human or chimeric (e.g., humanized) CD2 nucleotide sequence and/or amino acid sequences.
  • the entire sequence of mouse exon 1, exon 2, exon 3, exon 4, exon 5, signal peptide, extracellular region, transmembrane region, and/or cytoplasmic region are replaced by the corresponding human sequence.
  • a “region” or “portion” of mouse exon 1, exon 2, exon 3, exon 4, exon 5, signal peptide, extracellular region, transmembrane region, and/or cytoplasmic region are replaced by the corresponding human sequence.
  • region can refer to at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 550, 600, 606, 610, 620, or 624 nucleotides, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 201, 202, 203, 204, 205, 208, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 344, 350, or 351 amino acid residues.
  • the “region” or “portion” can be at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%identical to exon 1, exon 2, exon 3, exon 4, exon 5, signal peptide, extracellular region, transmembrane region, or cytoplasmic region.
  • a region, a portion, or the entire sequence of mouse exon 1, exon 2, exon 3, exon 4, and/or exon 5 are replaced by a region, a portion, or the entire sequence of the human exon 1, exon 2, exon 3, exon 4, and/or exon 5 (e.g., a portion of exon 1, exon 2, exon 3, and a portion of exon 4) .
  • a “region” or “portion” of the signal peptide, extracellular region, transmembrane region, cytoplasmic region, exon 1, exon 2, exon 3, and/or exon 4 is deleted.
  • the present disclosure is related to a genetically-modified, non-human animal whose genome comprises a chimeric (e.g., humanized ) CD2 nucleotide sequence.
  • the chimeric (e.g., humanized ) CD2 nucleotide sequence encodes a CD2 protein comprising a signal peptide, an extracellular region, a transmembrane region, and a cytoplasmic region.
  • the signal peptide comprises a sequence that is at least 80%, 85%, 90%, 95%, or 100%identical to amino acids 1-24 of SEQ ID NO: 2.
  • the signal peptide comprises all or part of human CD2 signal peptide.
  • the extracellular region comprises a sequence that is at least 80%, 85%, 90%, 95%, or 100%identical to amino acids 25-208 or 25-209 of SEQ ID NO: 2. In some embodiments, the extracellular region comprises all or part of human CD2 extracellular region. In some embodiments, the transmembrane region comprises a sequence that is at least 80%, 85%, 90%, 95%, or 100%identical to amino acids 204-229 of SEQ ID NO: 1. In some embodiments, the transmembrane region comprises all or part of endogenous CD2 transmembrane region.
  • the cytoplasmic region comprises a sequence that is at least 80%, 85%, 90%, 95%, or 100%identical to amino acids 230-344 of SEQ ID NO: 1. In some embodiments, the cytoplasmic region comprises all or part of endogenous CD2 cytoplasmic region. In some embodiments, the genome of the animal comprises a sequence that is at least 80%, 85%, 90%, 95%, or 100%identical to SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14.
  • the genetically-modified non-human animal described herein comprises a sequence encoding a human or humanized CD2 protein.
  • the CD2 protein comprises, from N-terminus to C-terminus, a signal peptide, an extracellular region, a transmembrane region, and a cytoplasmic region.
  • the humanized CD2 protein comprises a human or humanized signal peptide.
  • the humanized CD2 protein comprises an endogenous signal peptide.
  • the humanized CD2 protein comprises a human or humanized extracellular region.
  • the humanized CD2 protein comprises an endogenous extracellular region.
  • the humanized CD2 protein comprises a human or humanized transmembrane region. In some embodiments, the humanized CD2 protein comprises an endogenous transmembrane region. In some embodiments, the humanized CD2 protein comprises a human or humanized cytoplasmic region. In some embodiments, the humanized CD2 protein comprises an endogenous cytoplasmic region. In some embodiments, the humanized CD2 protein comprises a human or humanized signal peptide, a human or humanized extracellular region, an endogenous transmembrane region, and an endogenous cytoplasmic region. In some embodiments, the humanized CD2 protein comprises an endogenous sequence that corresponds to amino acids 203-344 of SEQ ID NO: 1.
  • the genetically-modified non-human animal described herein comprises a human or humanized CD2 gene.
  • the humanized CD2 gene comprises 5 exons.
  • the humanized CD2 gene comprises humanized exon 1, human exon 2, human exon 3, humanized exon 4, and/or endogenous exon 5.
  • the humanized CD2 gene comprises 4 introns.
  • the humanized CD2 gene comprises human intron 1, human intron 2, human intron 3, and/or endogenous intron 4.
  • the humanized CD2 gene comprises human or humanized 5’ UTR.
  • the humanized CD2 gene comprises human or humanized 3’ UTR.
  • the humanized CD2 gene comprises endogenous 5’ UTR.
  • the humanized CD2 gene comprises endogenous 3’ UTR.
  • the present disclosure also provides a chimeric (e.g., humanized) CD2 nucleotide sequence and/or amino acid sequences, wherein in some embodiments, at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%of the sequence are identical to or derived from mouse CD2 mRNA sequence (e.g., NM_013486.2) , mouse CD2 amino acid sequence (e.g., SEQ ID NO: 1) , or a portion thereof (e.g., 5’ UTR, a portion of exon 1, a portion of exon 4, exon 5, and 3’ UTR) ; and in some embodiments, at least 1%, 2%, 3%, 4%, 5%
  • sequence encoding amino acids 1-202 of mouse CD2 (SEQ ID NO:1) is replaced.
  • sequence is replaced by a sequence encoding a corresponding region of human CD2 (e.g., amino acids 1-208 of human CD2 (SEQ ID NO: 2) ) .
  • sequence encoding amino acids 1-203 of mouse CD2 (SEQ ID NO: 1) is replaced.
  • sequence is replaced by a sequence encoding a corresponding region of human CD2 (e.g., amino acids 1-209 of human CD2 (SEQ ID NO: 2) ) .
  • sequence encoding amino acids 23-202 of mouse CD2 (SEQ ID NO: 1) is replaced. In some embodiments, the sequence is replaced by a sequence encoding a corresponding region of human CD2 (e.g., amino acids 25-208 of human CD2 (SEQ ID NO: 2) ) .
  • sequence encoding amino acids 23-203 of mouse CD2 (SEQ ID NO: 1) is replaced.
  • sequence is replaced by a sequence encoding a corresponding region of human CD2 (e.g., amino acids 25-209 of human CD2 (SEQ ID NO: 2) ) .
  • the nucleic acids as described herein are operably linked to a promotor or regulatory element, e.g., an endogenous mouse CD2 promotor, an inducible promoter, an enhancer, and/or mouse or human regulatory elements.
  • a promotor or regulatory element e.g., an endogenous mouse CD2 promotor, an inducible promoter, an enhancer, and/or mouse or human regulatory elements.
  • the nucleic acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides, e.g., contiguous or non-contiguous nucleotides) that are different from part of or the entire mouse CD2 nucleotide sequence (e.g., a portion of exon 1, exons 2-3, and a portion of exon 4 of NM_013486.2) .
  • the nucleic acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides, e.g., contiguous or non-contiguous nucleotides) that is the same as part of or the entire mouse CD2 nucleotide sequence (e.g., 5’ UTR, a portion of exon 1, a portion of exon 4, exon 5, and 3’ UTR of NM_013486.2) .
  • the nucleic acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides, e.g., contiguous or non-contiguous nucleotides) that is different from part of or the entire human CD2 nucleotide sequence (e.g., 5’ UTR, a portion of exon 1, a portion of exon 4, exon 5, and 3’ UTR of NM_001767.5) .
  • the nucleic acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides, e.g., contiguous or non-contiguous nucleotides) that is the same as part of or the entire human CD2 nucleotide sequence (e.g., a portion (at least 20 bp) of exon 1, exons 2-3, and a portion (at least 5 bp) of exon 4 of NM_001767.5) .
  • a portion e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 nucleotides, e.g., contiguous or non-contiguous nucleotides
  • the entire human CD2 nucleotide sequence e.g., a portion (at least 20 bp) of exon 1, ex
  • the amino acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid residues, e.g., contiguous or non-contiguous amino acid residues) that is different from part of or the entire mouse CD2 amino acid sequence (e.g., amino acids 1-202 of NP_038514.1 (SEQ ID NO: 1) ) .
  • the amino acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid residues, e.g., contiguous or non-contiguous amino acid residues) that is the same as part of or the entire mouse CD2 amino acid sequence (e.g., amino acids 203-344 of NP_038514.1 (SEQ ID NO: 1) ) .
  • the amino acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid residues, e.g., contiguous or non-contiguous amino acid residues) that is different from part of or the entire human CD2 amino acid sequence (e.g., amino acids 209-351 of NP_001758.2 (SEQ ID NO: 2) ) .
  • the amino acid sequence has at least a portion (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid residues, e.g., contiguous or non-contiguous amino acid residues) that is the same as part of or the entire human CD2 amino acid sequence (e.g., amino acids 1-208 of NP_001758.2 (SEQ ID NO: 2) ) .
  • the present disclosure also provides a humanized CD2 mouse amino acid sequence, wherein the amino acid sequence is selected from the group consisting of:
  • nucleic acid sequence an amino acid sequence encoded by a nucleic acid sequence, wherein the nucleic acid sequence is able to hybridize to a nucleotide sequence encoding the amino acid shown in SEQ ID NO: 1, 2, or 10 under a low stringency condition or a strict stringency condition;
  • amino acid sequence having a homology of at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, or at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to the amino acid sequence shown in SEQ ID NO: 1, 2, or 10;
  • amino acid sequence that is different from the amino acid sequence shown in SEQ ID NO: 1, 2, or 10 by no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or no more than 1 amino acid;
  • amino acid sequence that comprises a substitution, a deletion and /or insertion of one or more amino acids to the amino acid sequence shown in SEQ ID NO: 1, 2, or 10.
  • the present disclosure also provides a humanized CD2 amino acid sequence, wherein the amino acid sequence is selected from the group consisting of:
  • an amino acid sequence have a homology of at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%to amino acids 1-208 or 25-208 of SEQ ID NO: 2;
  • amino acid sequence that is different from amino acids 1-208 or 25-208 of SEQ ID NO: 2 by no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or no more than 1 amino acid;
  • amino acid sequence that comprises a substitution, a deletion and /or insertion of one or more amino acids to amino acids 1-208 or 25-208 of SEQ ID NO: 2.
  • the present disclosure also provides a humanized CD2 amino acid sequence, wherein the amino acid sequence is selected from the group consisting of:
  • an amino acid sequence have a homology of at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%to amino acids 203-344 of SEQ ID NO: 1;
  • amino acid sequence that is different from amino acids 203-344 of SEQ ID NO: 1 by no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or no more than 1 amino acid;
  • amino acid sequence that comprises a substitution, a deletion and /or insertion of one or more amino acids to amino acids 203-344 of SEQ ID NO: 1.
  • the present disclosure also relates to a CD2 nucleic acid (e.g., DNA or RNA) sequence, wherein the nucleic acid sequence can be selected from the group consisting of:
  • nucleic acid sequence as shown in SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14, or a nucleic acid sequence encoding a homologous CD2 amino acid sequence of a humanized mouse CD2;
  • nucleic acid sequence that is able to hybridize to the nucleotide sequence as shown in SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14 under a low stringency condition or a strict stringency condition;
  • nucleic acid sequence that has a homology of at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, or at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to the nucleotide sequence as shown in SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14;
  • nucleic acid sequence that encodes an amino acid sequence, wherein the amino acid sequence has a homology of at least 90%with or at least 90%identical to the amino acid sequence shown in SEQ ID NO: 1, 2, or 10;
  • nucleic acid sequence that encodes an amino acid sequence, wherein the amino acid sequence has a homology of at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%with, or at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%identical to the amino acid sequence shown in SEQ ID NO: 1, 2, or 10;
  • nucleic acid sequence that encodes an amino acid sequence, wherein the amino acid sequence is different from the amino acid sequence shown in SEQ ID NO: 1, 2, or 10 by no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or no more than 1 amino acid;
  • nucleic acid sequence that encodes an amino acid sequence, wherein the amino acid sequence comprises a substitution, a deletion and /or insertion of one or more amino acids to the amino acid sequence shown in SEQ ID NO: 1, 2, or 10.
  • the present disclosure further relates to a CD2 genomic DNA sequence of a humanized mouse.
  • the DNA sequence is obtained by reverse transcription of the mRNA obtained by transcription thereof is consistent with or complementary to the DNA sequence homologous to the sequence shown in SEQ ID NO: 7 or 9.
  • the disclosure also provides an amino acid sequence that has a homology of at least 90%with, or at least 90%identical to the sequence shown in SEQ ID NO: 1, 2, or 10, and has protein activity.
  • the homology with the sequence shown in SEQ ID NO: 1, 2, or 10 is at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99%.
  • the foregoing homology is at least about 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 80%, or 85%.
  • the percentage identity with the sequence shown in SEQ ID NO: 1, 2, or 10 is at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99%. In some embodiments, the foregoing percentage identity is at least about 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 80%, or 85%.
  • the disclosure also provides a nucleotide sequence that has a homology of at least 90%, or at least 90%identical to the sequence shown in SEQ ID NO: 7 or 9, and encodes a polypeptide that has protein activity.
  • the homology with the sequence shown in SEQ ID NO: 7 or 9 is at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99%.
  • the foregoing homology is at least about 50%, 55%, 60%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 80%, or 85%.
  • the percentage identity with the sequence shown in SEQ ID NO: 3, 4, 5, 6, 7, 9, 11, 12, 13, or 14 is at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99%. In some embodiments, the foregoing percentage identity is at least about 50%, 55%, 60%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 80%, or 85%.
  • the disclosure also provides a nucleic acid sequence that is at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%identical to any nucleotide sequence as described herein, and an amino acid sequence that is at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%identical to any amino acid sequence as described herein.
  • the disclosure relates to nucleotide sequences encoding any peptides that are described herein, or any amino acid sequences that are encoded by any nucleotide sequences as described herein.
  • the nucleic acid sequence is less than 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 150, 200, 250, 300, 350, 400, 500, or 600 nucleotides.
  • the amino acid sequence is less than 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 208 amino acid residues.
  • the amino acid sequence (i) comprises an amino acid sequence; or (ii) consists of an amino acid sequence, wherein the amino acid sequence is any one of the sequences as described herein.
  • the nucleic acid sequence (i) comprises a nucleic acid sequence; or (ii) consists of a nucleic acid sequence, wherein the nucleic acid sequence is any one of the sequences as described herein.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes) .
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percentage of residues conserved with similar physicochemical properties can also be used to measure sequence similarity. Families of amino acid residues having similar physicochemical properties have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • Cells, tissues, and animals are also provided that comprise the nucleotide sequences as described herein, as well as cells, tissues, and animals (e.g., mouse) that express human or chimeric (e.g., humanized) CD2 from an endogenous non-human CD2 locus.
  • the term “genetically-modified non-human animal” refers to a non-human animal having exogenous DNA in at least one chromosome of the animal’s genome.
  • at least one or more cells e.g., at least 1%, 2%, 3%, 4%, 5%, 10%, 20%, 30%, 40%, 50%of cells of the genetically-modified non-human animal have the exogenous DNA in its genome.
  • the cell having exogenous DNA can be various kinds of cells, e.g., an endogenous cell, a somatic cell, an immune cell, a T cell, a B cell, an antigen presenting cell, a macrophage, a dendritic cell, a germ cell, a blastocyst, or an endogenous tumor cell.
  • genetically-modified non-human animals are provided that comprise a modified endogenous CD2 locus that comprises an exogenous sequence (e.g., a human sequence) , e.g., a replacement of one or more non-human sequences with one or more human sequences.
  • the animals are generally able to pass the modification to progeny, i.e., through germline transmission.
  • chimeric gene or “chimeric nucleic acid” refers to a gene or a nucleic acid, wherein two or more portions of the gene or the nucleic acid are from different species, or at least one of the sequences of the gene or the nucleic acid does not correspond to the wild-type nucleic acid in the animal.
  • the chimeric gene or chimeric nucleic acid has at least one portion of the sequence that is derived from two or more different sources, e.g., sequences encoding different proteins or sequences encoding the same (or homologous) protein of two or more different species.
  • the chimeric gene or the chimeric nucleic acid is a humanized gene or humanized nucleic acid.
  • chimeric protein or “chimeric polypeptide” refers to a protein or a polypeptide, wherein two or more portions of the protein or the polypeptide are from different species, or at least one of the sequences of the protein or the polypeptide does not correspond to wild-type amino acid sequence in the animal.
  • the chimeric protein or the chimeric polypeptide has at least one portion of the sequence that is derived from two or more different sources, e.g., same (or homologous) proteins of different species.
  • the chimeric protein or the chimeric polypeptide is a humanized protein or a humanized polypeptide.
  • humanized protein or “humanized polypeptide” refers to a protein or a polypeptide, wherein at least a portion of the protein or the polypeptide is from the human protein or human polypeptide. In some embodiments, the humanized protein or polypeptide is a human protein or polypeptide.
  • humanized nucleic acid refers to a nucleic acid, wherein at least a portion of the nucleic acid is from the human. In some embodiments, the entire nucleic acid of the humanized nucleic acid is from human. In some embodiments, the humanized nucleic acid is a humanized exon. A humanized exon can be e.g., a human exon or a chimeric exon.
  • the chimeric gene or the chimeric nucleic acid is a humanized CD2 gene or a humanized CD2 nucleic acid. In some embodiments, at least one or more portions of the gene or the nucleic acid is from the human CD2 gene, at least one or more portions of the gene or the nucleic acid is from a non-human CD2 gene. In some embodiments, the gene or the nucleic acid comprises a sequence that encodes an CD2 protein. The encoded CD2 protein is functional or has at least one activity of the human CD2 protein or the non-human CD2 protein, e.g., binding to its ligand CD58 on APC in humans and on CD48 in rodents.
  • the chimeric protein or the chimeric polypeptide is a humanized CD2 protein or a humanized CD2 polypeptide. In some embodiments, at least one or more portions of the amino acid sequence of the protein or the polypeptide is from a human CD2 protein, and at least one or more portions of the amino acid sequence of the protein or the polypeptide is from a non-human CD2 protein.
  • the humanized CD2 protein or the humanized CD2 polypeptide is functional or has at least one activity of the human CD2 protein or the non-human CD2 protein.
  • the extracellular region is human or humanized.
  • the signal peptide is human or humanized.
  • the cytoplasmic region is human or humanized.
  • the transmembrane region is human or humanized.
  • both the extracellular region and signal peptide are human or humanized.
  • both the transmembrane and cytoplasmic regions are endogenous.
  • the genetically modified non-human animal can be various animals, e.g., a mouse, rat, rabbit, pig, bovine (e.g., cow, bull, buffalo) , deer, sheep, goat, chicken, cat, dog, ferret, primate (e.g., marmoset, rhesus monkey) .
  • ES embryonic stem
  • Such methods include, e.g., modifying a non-ES cell genome (e.g., a fibroblast or an induced pluripotent cell) and employing nuclear transfer to transfer the modified genome to a suitable cell, e.g., an oocyte, and gestating the modified cell (e.g., the modified oocyte) in a non-human animal under suitable conditions to form an embryo.
  • a suitable cell e.g., an oocyte
  • gestating the modified cell e.g., the modified oocyte
  • the animal is a mammal, e.g., of the superfamily Dipodoidea or Muroidea.
  • the genetically modified animal is a rodent.
  • the rodent can be selected from a mouse, a rat, and a hamster.
  • the genetically modified animal is from a family selected from Calomyscidae (e.g., mouse-like hamsters) , Cricetidae (e.g., hamster, New World rats and mice, voles) , Muridae (true mice and rats, gerbils, spiny mice, crested rats) , Nesomyidae (climbing mice, rock mice, with-tailed rats, Malagasy rats and mice) , Platacanthomyidae (e.g., spiny dormice) , and Spalacidae (e.g., mole rates, bamboo rats, and zokors) .
  • Calomyscidae e.g., mouse-like hamsters
  • Cricetidae e.g., hamster, New World rats and mice, voles
  • Muridae true mice and rats, gerbils, spiny mice, crested rats
  • the genetically modified rodent is selected from a true mouse or rat (family Muridae) , a gerbil, a spiny mouse, and a crested rat.
  • the non-human animal is a mouse.
  • the animal is a mouse of a C57BL strain selected from C57BL/A, C57BL/An, C57BL/GrFa, C57BL/KaLwN, C57BL/6, C57BL/6J, C57BL/6ByJ, C57BL/6NJ, C57BL/10, C57BL/10ScSn, C57BL/10Cr, and C57BL/Ola.
  • a C57BL strain selected from C57BL/A, C57BL/An, C57BL/GrFa, C57BL/KaLwN, C57BL/6, C57BL/6J, C57BL/6ByJ, C57BL/6NJ, C57BL/10, C57BL/10ScSn, C57BL/10Cr, and C57BL/Ola.
  • the mouse is a 129 strain selected from the group consisting of a strain that is 129P1, 129P2, 129P3, 129X1, 129S1 (e.g., 129S1/SV, 129S1/SvIm) , 129S2, 129S4, 129S5, 129S9/SvEvH, 129S6 (129/SvEvTac) , 129S7, 129S8, 129T1, 129T2.
  • a strain that is 129P1, 129P2, 129P3, 129X1, 129S1 (e.g., 129S1/SV, 129S1/SvIm) , 129S2, 129S4, 129S5, 129S9/SvEvH, 129S6 (129/SvEvTac) , 129S7, 129S8, 129T1, 129T2.
  • the genetically modified mouse is a mix of the 129 strain and the C57BL/6 strain. In some embodiments, the mouse is a mix of the 129 strains, or a mix of the BL/6 strains.
  • the mouse is a BALB strain, e.g., BALB/c strain. In some embodiments, the mouse is a mix of a BALB strain and another strain. In some embodiments, the mouse is from a hybrid line (e.g., 50%BALB/c-50%12954/Sv; or 50%C57BL/6-50%129) . In some embodiments, the non-human animal is a rodent.
  • the non-human animal is a mouse having a BALB/c, A, A/He, A/J, A/WySN, AKR, AKR/A, AKR/J, AKR/N, TA1, TA2, RF, SWR, C3H, C57BR, SJL, C57L, DBA/2, KM, NIH, ICR, CFW, FACA, C57BL/A, C57BL/An, C57BL/GrFa, C57BL/KaLwN, C57BL/6, C57BL/6J, C57BL/6ByJ, C57BL/6NJ, C57BL/10, C57BL/10ScSn, C57BL (C57BL/10Cr and C57BL/Ola) , C58, CBA/Br, CBA/Ca, CBA/J, CBA/st, or CBA/H background.
  • the animal is a rat.
  • the rat can be selected from a Wistar rat, an LEA strain, a Sprague Dawley strain, a Fischer strain, F344, F6, and Dark Agouti.
  • the rat strain is a mix of two or more strains selected from the group consisting of Wistar, LEA, Sprague Dawley, Fischer, F344, F6, and Dark Agouti.
  • the animal can have one or more other genetic modifications, and/or other modifications, that are suitable for the particular purpose for which the humanized CD2 animal is made.
  • suitable mice for maintaining a xenograft e.g., a human cancer or tumor
  • mice for maintaining a xenograft can have one or more modifications that compromise, inactivate, or destroy the immune system of the non-human animal in whole or in part.
  • Compromise, inactivation, or destruction of the immune system of the non-human animal can include, for example, destruction of hematopoietic cells and/or immune cells by chemical means (e.g., administering a toxin) , physical means (e.g., irradiating the animal) , and/or genetic modification (e.g., knocking out one or more genes) .
  • Non-limiting examples of such mice include, e.g., NOD mice, SCID mice, NOD/SCID mice, IL2R ⁇ knockout mice, NOD/SCID/ ⁇ c null mice (Ito, M.
  • a genetically modified mouse can include a humanization of at least a portion of an endogenous non-human CD2 locus, and further comprises a modification that compromises, inactivates, or destroys the immune system (or one or more cell types of the immune system) of the non-human animal in whole or in part.
  • modification is, e.g., selected from the group consisting of a modification that results in NOD mice, SCID mice, NOD/SCID mice, IL-2R ⁇ knockout mice, NOD/SCID/ ⁇ c null mice, nude mice, Rag1 and/or Rag2 knockout mice, NOD-Prkdc scid IL-2r ⁇ null mice, NOD-Rag 1 -/- -IL2rg -/- (NRG) mice, Rag 2 -/- -IL2rg -/- (RG) mice, and a combination thereof.
  • NSG NSG
  • RG Rag 2 -/- -IL2rg -/-
  • the mouse can include a replacement of all or part of mature CD2 coding sequence with human mature CD2 coding sequence.
  • Genetically modified non-human animals can comprise a modification at an endogenous non-human CD2 locus.
  • the modification can comprise a human nucleic acid sequence encoding at least a portion of a mature CD2 protein (e.g., at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, or 99%identical to the mature CD2 protein sequence) .
  • genetically modified cells are also provided that can comprise the modifications described herein (e.g., ES cells, somatic cells)
  • the genetically modified non-human animals comprise the modification of the endogenous CD2 locus in the germline of the animal.
  • Genetically modified animals can express a human CD2 and/or a chimeric (e.g., humanized) CD2 from endogenous mouse loci, wherein the endogenous mouse CD2 gene has been replaced with a human CD2 gene and/or a nucleotide sequence that encodes a region of human CD2 sequence or an amino acid sequence that is at least 10%, 20%, 30%, 40%, 50%, 60%, 70&, 80%, 90%, 95%, 96%, 97%, 98%, or 99%identical to the human CD2 sequence.
  • an endogenous non-human CD2 locus is modified in whole or in part to comprise human nucleic acid sequence encoding at least one protein-coding sequence of a mature CD2 protein.
  • the genetically modified mice can express the human CD2 and/or chimeric CD2 (e.g., humanized CD2) from endogenous loci that are under control of mouse promoters and/or mouse regulatory elements.
  • the replacement (s) at the endogenous mouse loci provide non-human animals that express human CD2 or chimeric CD2 (e.g., humanized CD2) in appropriate cell types and in a manner that does not result in the potential pathologies observed in some other transgenic mice known in the art.
  • the human CD2 or the chimeric CD2 (e.g., humanized CD2) expressed in animal can maintain one or more functions of the wild-type mouse or human CD2 in the animal.
  • the expressed CD2 can bind to human CD58 or non-human (e.g., mouse) CD48.
  • the animal does not express endogenous CD2.
  • the animal expresses a decreased level of endogenous CD2 as compared to CD2 expression level in a wild-type animal.
  • endogenous CD2 refers to CD2 protein that is expressed from an endogenous CD2 nucleotide sequence of the non-human animal (e.g., mouse) before any genetic modification.
  • the genome of the animal can comprise a sequence encoding an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%identical to human CD2 (NP_001758.2) (SEQ ID NO: 2) .
  • the genome comprises a sequence encoding an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%identical to SEQ ID NO: 7 or 9.
  • the genome of the genetically modified animal can comprise a replacement at an endogenous CD2 gene locus of a sequence encoding a region of endogenous CD2 with a sequence encoding a corresponding region of human CD2.
  • the sequence that is replaced is any sequence within the endogenous CD2 gene locus, e.g., exon 1, exon 2, exon 3, exon 4, exon 5, 5’-UTR, 3’-UTR, intron 1, intron 2, intron 3, intron 4, or any combination thereof.
  • the sequence that is replaced is within the regulatory region of the endogenous CD2 gene.
  • the sequence that is replaced is a portion of exon 1, exon 2, exon 3, and a portion of exon 4, of an endogenous mouse CD2 gene locus.
  • the genetically modified animal can have one or more cells expressing a human or chimeric CD2 (e.g., humanized CD2) having, from N-terminus to C-terminus, a signal peptide, an extracellular region, a transmembrane region, and a cytoplasmic region.
  • a human or chimeric CD2 e.g., humanized CD2
  • the signal peptide comprises a sequence that is at least 50%, 60%, 70%, 80%, 90%, 95%, 99%identical to the signal peptide of human CD2.
  • the signal peptide of the humanized CD2 has a sequence that has at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 amino acids (e.g., contiguously or non-contiguously) that are identical to the signal peptide of human CD2.
  • the extracellular region comprises a sequence that is at least 50%, 60%, 70%, 80%, 90%, 95%, 99%identical to the extracellular region of human CD2.
  • the extracellular region of the humanized CD2 has a sequence that has at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 181, 182, 183, 184, or 185 amino acids (e.g., contiguously or non-contiguously) that are identical to the extracellular region of human CD2.
  • the extracellular region of the humanized CD2 has a sequence that has at least 1, 2, 3, 4, or 5 amino acids (contiguously or non-contiguously) that are identical to the C-terminal 1-5 amino acids in the extracellular region of endogenous CD2 (e.g., mouse CD2) .
  • the extracellular region described herein includes the signal peptide. In some embodiments, the extracellular region described herein does not include the signal peptide. Because human CD2 and non-human CD2 (e.g., mouse CD2) sequences, in many cases, are different, antibodies that bind to human CD2 will not necessarily have the same binding affinity with non-human CD2 or have the same effects to non-human CD2. Therefore, the genetically modified animal having a human or a humanized extracellular region can be used to better evaluate the effects of anti-human CD2 antibodies in an animal model.
  • human CD2 and non-human CD2 e.g., mouse CD2
  • the transmembrane comprises a sequence that is at least 50%, 60%, 70%, 80%, 90%, 95%, 99%identical to the transmembrane region of endogenous CD2 (e.g., amino acids 204-229 of SEQ ID NO: 1) .
  • the transmembrane region of the humanized CD2 has a sequence that has at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or 26 amino acids (contiguously or non-contiguously) that are identical to the transmembrane region of endogenous CD2 (e.g., mouse CD2) .
  • the cytoplasmic comprises a sequence that is at least 50%, 60%, 70%, 80%, 90%, 95%, 99%identical to the cytoplasmic of endogenous CD2 (e.g., amino acids 230-344 of SEQ ID NO: 1) .
  • the cytoplasmic region of the humanized CD2 has a sequence that has at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 111, 112, 113, 114, or 115 amino acids (contiguously or non-contiguously) that are identical to the cytoplasmic region of endogenous CD2 (e.g., mouse CD2) .
  • the entire transmembrane region and the entire cytoplasmic region of the humanized CD2 described herein are derived from endogenous sequence.
  • the genome of the genetically modified animal comprises a sequence encoding an amino acid sequence that corresponds to a portion or the entire sequence of exon 1, exon 2, exon 3, exon 4, and/or exon 5 of human CD2; a portion or the entire sequence of the extracellular region, and/or a portion or the entire sequence of the signal peptide of human CD2; or a portion or the entire sequence of amino acids 1-208 or 1-209 of SEQ ID NO: 2.
  • the genome of the genetically modified animal comprises a portion of exon 1, exons 2-3, and a portion of exon 4 of human CD2 gene.
  • the portion of exon 1 includes at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 46, 47, 48, 49, 50, 55, 60, 61, 62, 63, 64, 65, 70, 80, 90, 100, 110, 120, 121, 122, 123, or 124 nucleotides.
  • the portion of exon 1 includes 61 nucleotides.
  • the portion of exon 1 includes a nucleotide of at least 20 bp.
  • the portion of exon 4 includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 , 13, 14, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 121, 122, or 123 nucleotides. In some embodiments, the portion of exon 4 includes 11 nucleotides. In some embodiments, the portion of exon 4 includes a nucleotide of at least 5 bp.
  • the non-human animal can have, at an endogenous CD2 gene locus, a nucleotide sequence encoding a chimeric human/non-human CD2 polypeptide, wherein a human portion of the chimeric human/non-human CD2 polypeptide comprises the entire human CD2 signal peptide and all or a portion of the human CD2 extracellular region, and wherein the animal expresses a functional CD2 on a surface of a cell of the animal.
  • the human portion of the chimeric human/non-human CD2 polypeptide can comprise an amino acid sequence encoded by a portion of exon 1, exons 2-3, and/or a portion of exon 4 of human CD2.
  • the human portion of the chimeric human/non-human CD2 polypeptide can comprise a sequence that is at least 80%, 85%, 90%, 95%, or 99%identical to amino acids 1-208 of SEQ ID NO: 2.
  • the transmembrane region includes a sequence corresponding to the entire or part of amino acids 204-229 of SEQ ID NO: 1.
  • the cytoplasmic region includes a sequence corresponding to the entire or part of amino acids 230-344 of SEQ ID NO: 1.
  • the chimeric human/non-human CD2 polypeptide comprises a signal peptide, which includes a sequence corresponding to the entire or part of amino acids 1-24 of SEQ ID NO: 2.
  • the non-human portion of the chimeric human/non-human CD2 polypeptide comprises the entire transmembrane region and/or the entire cytoplasmic region of an endogenous non-human CD2 polypeptide.
  • the non-human animal described herein can generate a soluble CD2.
  • the soluble CD2 is cleaved from any of the CD2 protein (e.g., humanized CD2) described herein.
  • the genetically modified animal can be heterozygous with respect to the replacement at the endogenous CD2 locus, or homozygous with respect to the replacement at the endogenous CD2 locus.
  • the humanized CD2 locus lacks a human CD2 5’-UTR.
  • the humanized CD2 locus comprises an endogenous (e.g., mouse) 5’-UTR.
  • the humanization comprises an endogenous (e.g., mouse) 3’-UTR.
  • mouse and human CD2 genes appear to be similarly regulated based on the similarity of their 5’-flanking sequence.
  • humanized CD2 mice that comprise a replacement at an endogenous mouse CD2 locus which retain mouse regulatory elements but comprise a humanization of CD2 encoding sequence, do not exhibit pathologies. Both genetically modified mice that are heterozygous or homozygous for humanized CD2 are grossly normal.
  • the present disclosure further relates to a non-human mammal generated through the method mentioned above.
  • the genome thereof contains human gene (s) .
  • the non-human mammal is a rodent, and preferably, the non-human mammal is a mouse.
  • the non-human mammal expresses a protein encoded by a humanized CD2 gene.
  • the present disclosure also relates to a tumor bearing non-human mammal model, characterized in that the non-human mammal model is obtained through the methods as described herein.
  • the non-human mammal is a rodent (e.g., a mouse) .
  • the present disclosure further relates to a cell or cell line, or a primary cell culture thereof derived from the non-human mammal or an offspring thereof, or the tumor bearing non-human mammal; the tissue, organ or a culture thereof derived from the non-human mammal or an offspring thereof, or the tumor bearing non-human mammal; and the tumor tissue derived from the non-human mammal or an offspring thereof when it bears a tumor, or the tumor bearing non-human mammal.
  • the present disclosure also provides non-human mammals produced by any of the methods described herein.
  • a non-human mammal is provided; and the genetically modified animal contains the DNA encoding human or humanized CD2 in the genome of the animal.
  • the non-human mammal comprises the genetic construct as described herein (e.g., gene construct as shown in FIGS. 2, 3, 4, and 5) .
  • a non-human mammal expressing human or humanized CD2 is provided.
  • the tissue-specific expression of human or humanized CD2 protein is provided.
  • the expression of human or humanized CD2 in a genetically modified animal is controllable, as by the addition of a specific inducer or repressor substance.
  • the specific inducer is selected from Tet-Off System/Tet-On System, or Tamoxifen System.
  • Non-human mammals can be any non-human animal known in the art and which can be used in the methods as described herein.
  • Preferred non-human mammals are mammals, (e.g., rodents) .
  • the non-human mammal is a mouse.
  • the present disclosure also relates to the progeny produced by the non-human mammal provided by the present disclosure mated with the same or other genotypes.
  • the present disclosure also provides a cell line or primary cell culture derived from the non-human mammal or a progeny thereof.
  • a model based on cell culture can be prepared, for example, by the following methods.
  • Cell cultures can be obtained by way of isolation from a non-human mammal, alternatively cells can be obtained from the cell culture established using the same constructs and the standard cell transfection techniques.
  • the integration of genetic constructs containing DNA sequences encoding human CD2 protein can be detected by a variety of methods.
  • RNA quantification approaches using reverse transcriptase polymerase chain reaction (RT-PCR) or Southern blotting, and in situ hybridization
  • protein level including histochemistry, immunoblot analysis and in vitro binding studies
  • RT-PCR reverse transcriptase polymerase chain reaction
  • protein level including histochemistry, immunoblot analysis and in vitro binding studies
  • the expression level of the gene of interest can be quantified by ELISA techniques well known to those skilled in the art.
  • Many standard analysis methods can be used to complete quantitative measurements. For example, transcription levels can be measured using RT-PCR and hybridization methods including RNase protection, Southern blot analysis, RNA dot analysis (RNAdot) analysis. Immunohistochemical staining, flow cytometry, Western blot analysis can also be used to assess the presence of human or humanized CD2 protein.
  • the disclosure also provides a genetically-modified, non-human animal whose genome comprise a disruption in the animal’s endogenous CD2 gene, wherein the disruption of the endogenous CD2 gene comprises deletion of exon 1, exon 2, exon 3, exon 4, and/or exon 5, or part thereof of the endogenous CD2 gene.
  • the disruption of the endogenous CD2 gene comprises deletion of one or more exons or part of exons selected from the group consisting of exon 1, exon 2, exon 3, exon 4, and exon 5 of the endogenous CD2 gene.
  • the disruption of the endogenous CD2 gene further comprises deletion of one or more introns or part of introns selected from the group consisting of intron 1, intron 2, intron 3, and intron 4 of the endogenous CD2 gene.
  • deletion can comprise deleting at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2500, 3000, 5000, 10000, 11000, 12000, 13000, 14000, 15000, or more nucleotides.
  • the disruption of the endogenous CD2 gene comprises the deletion of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 10, 220, 230, 240, 250, 260, 270, 280, 290, or 300 nucleotides of exon 1, exon 2, exon 3, exon 4, and/or exon 5 (e.g., deletion of at least 20 nucleotides from exon 1, exons 2-3, and at least 5 nucleotides from exon 4) .
  • the present disclosure relates to a targeting vector, comprising: a) a DNA fragment homologous to the 5’ end of a region to be altered (5’ arm) , which is selected from the CD2 gene genomic DNAs in the length of 100 to 10,000 nucleotides; b) a desired/donor DNA sequence encoding a donor region; and c) a second DNA fragment homologous to the 3’ end of the region to be altered (3’ arm) , which is selected from the CD2 gene genomic DNAs in the length of 100 to 10,000 nucleotides.
  • a) the DNA fragment homologous to the 5’ end of a conversion region to be altered (5’ arm) is selected from the nucleotide sequences that have at least 90%homology to the NCBI accession number NC_000069.7; c) the DNA fragment homologous to the 3’ end of the region to be altered (3’ arm) is selected from the nucleotide sequences that have at least 90%homology to the NCBI accession number NC_000069.7.
  • a) the DNA fragment homologous to the 5’ end of a region to be altered (5’ arm) is selected from the nucleotides from the position 101195196 to the position 101198704 of the NCBI accession number NC_000069.7; c) the DNA fragment homologous to the 3’ end of the region to be altered (3’ arm) is selected from the nucleotides from the position 101183839 to the position 101187215 of the NCBI accession number NC_000069.7.
  • a) the DNA fragment homologous to the 5’ end of a region to be altered (5’ arm) is selected from the nucleotides from the position 101195196 to the position 101196555 of the NCBI accession number NC_000069.7; c) the DNA fragment homologous to the 3’ end of the region to be altered (3’ arm) is selected from the nucleotides from the position 101185740 to the position 101187215 of the NCBI accession number NC_000069.7.
  • the length of the selected genomic nucleotide sequence in the targeting vector can be more than about 8 kb, about 8.5 kb, about 9 kb, about 9.5 kb, about 10 kb, about 10.5 kb, about 11 kb, about 12 kb, or about 13 kb.
  • the region to be altered is exon 1, exon 2, exon 3, and/or exon 4 of CD2 gene (e.g., a portion of exon 1, exons 2-3, and a portion of exon 4 of mouse CD2 gene) .
  • the targeting vector can further include one or more selectable markers, e.g., positive or negative selectable markers.
  • the positive selectable marker is a Neo gene or Neo cassette.
  • the negative selectable marker is a DTA gene.
  • sequence of the 5’ arm is shown in SEQ ID NO: 3; and the sequence of the 3’ arm is shown in SEQ ID NO: 4. In some embodiments, the sequence of the 5’ arm is shown in SEQ ID NO: 5; and the sequence of the 3’ arm is shown in SEQ ID NO: 6.
  • the sequence is derived from human (e.g., 116754493-116764494 of NC_000001.11; or 61-666 of NM_013486.2) .
  • the target region in the targeting vector is a part or entirety of the nucleotide sequence of a human CD2, preferably exon 1, exon 2, exon 3, exon 4, and/or exon 5 of the human CD2.
  • the nucleotide sequence of the humanized CD2 encodes the entire or the part of human CD2 protein with the NCBI accession number NP_001758.2 (SEQ ID NO: 2) .
  • the disclosure also provides vectors for constructing a humanized animal model or a knock-out model.
  • the vectors comprise a sgRNA sequence, wherein the sgRNA sequence targets CD2 gene, and the sgRNA is unique on the target sequence of the gene to be altered, and meets the sequence arrangement rule of 5’-NNN (20) -NGG3’ or 5’-CCN-N (20) -3’; and in some embodiments, the targeting site of the sgRNA in the mouse CD2 gene is located on the exon 1, exon 2, exon 3, exon 4, exon 5, intron 1, intron 2, intron 3, intron 4, upstream of exon 1, or downstream of exon 5 of the mouse CD2 gene.
  • the targeting sequences are shown as SEQ ID NOs: 17, 18, 19, 20, 21, 22, 23, and 24.
  • the disclosure provides sgRNA sequences for constructing a genetic modified animal model.
  • the oligonucleotide sgRNA sequences are set forth in SEQ ID NOs: 17 and 18.
  • the disclosure relates to a plasmid construct (e.g., pT7-sgRNA) including the sgRNA sequence, and/or a cell including the construct.
  • a plasmid construct e.g., pT7-sgRNA
  • the disclosure also relates to a cell comprising the targeting vectors as described above.
  • the present disclosure further relates to a non-human mammalian cell, having any one of the foregoing targeting vectors, and one or more in vitro transcripts of the construct as described herein.
  • the cell includes Cas9 mRNA or an in vitro transcript thereof.
  • the genes in the cell are heterozygous. In some embodiments, the genes in the cell are homozygous.
  • the non-human mammalian cell is a mouse cell. In some embodiments, the cell is a fertilized egg cell. In some embodiments, the cell is an embryonic stem cell.
  • Genetically modified animals can be made by several techniques that are known in the art, including, e.g., nonhomologous end-joining (NHEJ) , homologous recombination (HR) , zinc finger nucleases (ZFNs) , transcription activator-like effector-based nucleases (TALEN) , and the clustered regularly interspaced short palindromic repeats (CRISPR) -Cas system.
  • NHEJ nonhomologous end-joining
  • HR homologous recombination
  • ZFNs zinc finger nucleases
  • TALEN transcription activator-like effector-based nucleases
  • CRISPR clustered regularly interspaced short palindromic repeats
  • homologous recombination is used.
  • CRISPR-Cas9 genome editing is used to generate genetically modified animals.
  • genome editing techniques are known in the art, and is described, e.g., in Yin et al., "Delivery technologies for genome editing, " Nature Reviews Drug Discovery 16.6 (2017) : 387-399, which is incorporated by reference in its entirety.
  • Many other methods are also provided and can be used in genome editing, e.g., micro-injecting a genetically modified nucleus into an enucleated oocyte, and fusing an enucleated oocyte with another genetically modified cell.
  • the disclosure provides replacing in at least one cell of the animal, at an endogenous CD2 gene locus, a sequence encoding a region of an endogenous CD2 with a sequence encoding a corresponding region of human or chimeric CD2.
  • the replacement occurs in a germ cell, a somatic cell, a blastocyst, or a fibroblast, etc.
  • the nucleus of a somatic cell or the fibroblast can be inserted into an enucleated oocyte.
  • FIG. 3 and FIG. 5 show humanization strategies for a mouse CD2 locus.
  • the targeting strategies involve a vector comprising a 5’ homologous arm, a human CD2 gene fragment, and a 3’ homologous arm.
  • the process can involve replacing endogenous CD2 sequence with human sequence by homologous recombination.
  • the cleavage at the upstream and the downstream of the target site e.g., by zinc finger nucleases, TALEN or CRISPR
  • the homologous recombination is used to replace endogenous CD2 sequence with human CD2 sequence.
  • the methods for making a genetically modified, humanized animal can include the step of replacing at an endogenous CD2 locus (or site) , a nucleic acid sequence encoding a region of endogenous CD2 with a sequence encoding a corresponding region of human CD2.
  • the sequence can include a region (e.g., a part or the entire region) of exon 1, exon 2, exon 3, exon 4, and/or exon 5 of a human CD2 gene.
  • the sequence includes a portion of exon 1, exons 2-3, and a portion of exon 4 of a human CD2 gene (e.g., nucleic acids 64-687 of NM_001767.5) .
  • the region includes the signal peptide of human CD2 (e.g., amino acids 1-24 of SEQ ID NO: 2) , and/or the extracellular region of human CD2 (e.g., amino acids 25-208 or 25-209 of SEQ ID NO: 2) .
  • the endogenous CD2 locus is exon 1, exon 2, exon 3, exon 4, and/or exon 5 of mouse CD2.
  • the sequence includes a portion of exon 1, a portion of exon 4, and exon 5 of mouse CD2 gene (e.g., nucleic acids 1-60 and 667-1144 of NM_013486.2) .
  • the methods of modifying a CD2 locus of a mouse to express a chimeric human/mouse CD2 peptide can include the steps of replacing at the endogenous mouse CD2 locus a nucleotide sequence encoding a mouse CD2 with a nucleotide sequence encoding a human CD2, thereby generating a sequence encoding a chimeric human/mouse CD2.
  • the nucleotide sequence encoding the chimeric human/mouse CD2 can include a first nucleotide sequence encoding the signal peptide and all or a portion of the extracellular region of human CD2; and a second nucleotide sequence encoding the transmembrane region and the cytoplasmic region of mouse CD2, optionally the C-terminal 1, 2, 3, 4, or 5 amino acids in the extracellular region of mouse CD2.
  • the nucleotide sequences as described herein do not overlap with each other (e.g., the first nucleotide sequence, the second nucleotide sequence, and/or the third nucleotide sequence do not overlap) .
  • the amino acid sequences as described herein do not overlap with each other.
  • the present disclosure further provides a method for establishing a CD2 gene humanized animal model, involving the following steps:
  • step (d) identifying the germline transmission in the offspring genetically modified humanized non-human mammal of the pregnant female in step (c) .
  • the non-human mammal in the foregoing method is a mouse (e.g., a C57BL/6 mouse) .
  • the non-human mammal in step (c) is a female with pseudopregnancy (or false pregnancy) .
  • the fertilized eggs for the methods described above are C57BL/6 fertilized eggs.
  • Other fertilized eggs that can also be used in the methods as described herein include, but are not limited to, FVB/N fertilized eggs, BALB/c fertilized eggs, DBA/1 fertilized eggs and DBA/2 fertilized eggs.
  • Fertilized eggs can come from any non-human animal, e.g., any non-human animal as described herein.
  • the fertilized egg cells are derived from rodents.
  • the genetic construct can be introduced into a fertilized egg by microinjection of DNA. For example, by way of culturing a fertilized egg after microinjection, a cultured fertilized egg can be transferred to a false pregnant non-human animal, which then gives birth of a non-human mammal, so as to generate the non-human mammal mentioned in the methods described above.
  • methods of making the genetically modified animal comprises modifying the coding frame of the non-human animal’s CD2 gene, e.g., by inserting a nucleotide sequence (e.g., DNA or cDNA sequence) encoding human or humanized CD2 protein, e.g., immediately after the endogenous regulatory element of the non-human animal’s CD2 gene.
  • a nucleotide sequence e.g., DNA or cDNA sequence
  • one or more functional region sequences of the non-human animal’s CD2 gene can be knocked out, or inserted with a sequence, such that the non-human animal cannot express or expresses a decreased level of endogenous CD2 protein.
  • the coding frame of the modified non-human animal’s CD2 gene can be all or part of the nucleotide sequence from exon 1 to exon 5 of the non-human animal’s CD2 gene.
  • methods of making the genetically modified animal comprises inserting a nucleotide sequence encoding human or humanized CD2 protein and/or an auxiliary sequence after the endogenous regulatory element of the non-human animal’s CD2 gene.
  • the auxiliary sequence can be a stop codon, such that the CD2 gene humanized animal model can express human or humanized CD2 protein in vivo, but does not express non-human animal’s CD2 protein.
  • the auxiliary sequence includes WPRE (WHP Posttranscriptional Response Element) , loxP, and/or polyA.
  • the method for making the genetically modified animal comprises:
  • plasmid comprising a human CD2 gene fragment, flanked by a 5’ homologous arm and a 3’ homologous arm, wherein the 5’ and 3’ homologous arms target an endogenous CD2 gene;
  • sgRNAs small guide RNAs
  • step (3) modifying genome of a fertilized egg or an embryonic stem cell by using the plasmid of step (1) , the sgRNAs of step (2) , and Cas9;
  • step (2) mating the child mouse obtained in step (2) to obtain a homozygote mouse
  • the fertilized egg is modified by CRISPR with sgRNAs that target a 5’-terminal targeting site and a 3’-terminal targeting site.
  • sequence encoding the humanized CD2 protein is operably linked to an endogenous regulatory element at the endogenous CD2 gene locus.
  • the genetically-modified animal does not express an endogenous CD2 protein.
  • the method for making the genetically modified animal comprises:
  • plasmid comprising a human or chimeric CD2 gene fragment, flanked by a 5’ homologous arm and a 3’ homologous arm, wherein the 5’ and 3’ homologous arms target an endogenous CD2 gene;
  • sgRNAs small guide RNAs
  • the transgene with human regulatory elements expresses in a manner that is unphysiological or otherwise unsatisfactory, and can be actually detrimental to the animal.
  • the disclosure demonstrates that a replacement with human sequence at an endogenous locus under control of endogenous regulatory elements provides a physiologically appropriate expression pattern and level that results in a useful humanized animal whose physiology with respect to the replaced gene are meaningful and appropriate in the context of the humanized animal's physiology.
  • Genetically modified animals that express human or humanized CD2 protein provide a variety of uses that include, but are not limited to, developing therapeutics for human diseases and disorders, and assessing the toxicity and/or the efficacy of these human therapeutics in the animal models.
  • genetically modified animals are provided that express human or humanized CD2, which are useful for testing therapeutic agents that can decrease or block the interaction between the interaction between CD2 and anti-human CD2 antibodies, testing whether an therapeutic agent can increase or decrease the immune response, and/or determining whether an agent is an CD2 agonist or antagonist.
  • the genetically modified animals can be, e.g., an animal model of a human disease, e.g., the disease is induced genetically (a knock-in or knockout) .
  • the genetically modified non-human animals further comprise an impaired immune system, e.g., a non-human animal genetically modified to sustain or maintain a human xenograft, e.g., a human solid tumor (e.g., breast cancer) or a blood cell tumor (e.g., a lymphocyte tumor, a B or T cell tumor) .
  • an impaired immune system e.g., a non-human animal genetically modified to sustain or maintain a human xenograft, e.g., a human solid tumor (e.g., breast cancer) or a blood cell tumor (e.g., a lymphocyte tumor, a B or T cell tumor) .
  • the therapeutic agent e.g., an anti-CD2 antibody or a fusion protein containing a CD2 ligand (e.g., CD58 or LFA-3) ) described herein can bind to CD2 expressed on the surface of immune cells (e.g., effector T cells) and induce apoptosis of the immune cells, thereby treating autoimmune diseases (e.g., psoriasis) .
  • the therapeutic agent e.g., an anti-CD2 antibody
  • the genetically modified animals can be used for determining effectiveness of a therapeutic agent (e.g., an anti-CD2 antibody or a CD2-targeting drug) for the treatment of cancer.
  • the methods involve administering the therapeutic agent (e.g., an anti-human CD2 antibody or a CD2-targeting drug) to the animal as described herein, wherein the animal has a cancer or tumor; and determining inhibitory effects of the therapeutic agent to the cancer or tumor.
  • the inhibitory effects that can be determined include, e.g., a decrease of tumor size or tumor volume, a decrease of tumor growth, a reduction of the increase rate of tumor volume in a subject (e.g., as compared to the rate of increase in tumor volume in the same subject prior to treatment or in another subject without such treatment) , a decrease in the risk of developing a metastasis or the risk of developing one or more additional metastasis, an increase of survival rate, and an increase of life expectancy, etc.
  • the tumor volume in a subject can be determined by various methods, e.g., as determined by direct measurement, MRI or CT. In addition, a delicate balance is required for these antibodies, as CD2 is also expressed on many other cells.
  • the humanized CD2 functions in a largely similar way as compared to the endogenous CD2, so that the results in the humanized animals can be used to predict the efficacy or toxicity of these therapeutic agents in the human.
  • the anti-CD2 antibody can directly target cancer cells or tumor-associated cells expressing CD2, e.g., by inducing complement mediated cytotoxicity (CMC) or antibody dependent cellular cytotoxicity (ADCC) to kill the cancer cells.
  • CMC complement mediated cytotoxicity
  • ADCC antibody dependent cellular cytotoxicity
  • the tumor comprises one or more cancer cells (e.g., human or mouse cancer cells) that are injected into the animal.
  • the anti-CD2 antibody inhibits CD2 signaling pathways. In some embodiments, the anti-CD2 antibody does not inhibit CD2 signaling pathways. In some embodiments, the anti-CD2 antibody activates CD2 signaling pathways.
  • the genetically modified animals can be used for determining whether an anti-CD2 antibody is a CD2 agonist or antagonist.
  • the methods as described herein are also designed to determine the effects of the therapeutic agent (e.g., anti-CD2 antibodies) on CD2, e.g., whether the agent can block the interaction of CD2 and CD58, whether the agent can deplete CD2-expressing cells (e.g., T cells) , whether the agent can induce apoptosis of CD2-expressing immune cells, whether the agent can upregulate the immune response or downregulate immune response, and/or whether the agent can induce complement mediated cytotoxicity (CMC) or antibody dependent cellular cytotoxicity (ADCC) .
  • the genetically modified animals can be used for determining the effective dosage of a therapeutic agent for treating a disease in the subject, e.g., an autoimmune disease or cancer.
  • the inhibitory effects on tumors can also be determined by methods known in the art, e.g., measuring the tumor volume in the animal, and/or determining tumor (volume) inhibition rate (TGI TV ) .
  • the therapeutic agent e.g., an anti-CD2 antibody or a CD2-targeting drug
  • cancer refers to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth. The term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness.
  • tumor refers to cancerous cells, e.g., a mass of cancerous cells.
  • Cancers that can be treated or diagnosed using the methods described herein include malignancies of the various organ systems, such as affecting lung, breast, thyroid, lymphoid, gastrointestinal, and genito-urinary tract, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • the agents described herein are designed for treating or diagnosing a carcinoma in a subject.
  • carcinoma is art recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas.
  • the cancer is renal carcinoma or melanoma.
  • Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary.
  • carcinosarcomas e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues.
  • an “adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
  • the term “sarcoma” is art recognized and refers to malignant tumors of mesenchymal derivation.
  • the cancer described herein is lymphoma, non-small cell lung cancer, cervical cancer, leukemia, ovarian cancer, nasopharyngeal cancer, breast cancer, endometrial cancer, colon cancer, rectal cancer, gastric cancer, bladder cancer, glioma, lung cancer, bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, liver and bile duct cancer, esophageal cancer, kidney cancer, thyroid cancer, head and neck cancer, testicular cancer, glioblastoma, astrocytoma, melanoma, myeloproliferation abnormal syndromes, and sarcomas.
  • the leukemia is selected from acute lymphocytic (lymphoblastic) leukemia, acute myeloid leukemia, myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, plasma cell leukemia, and chronic myelogenous leukemia.
  • the lymphoma is selected from Hodgkin's lymphoma and non-Hodgkin's lymphoma, including B-cell lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B-cell lymphoma, T-cell lymphoma, and Waldenstrom macroglobulinemia.
  • the sarcoma is selected from the group consisting of osteosarcoma, Ewing sarcoma, leiomyosarcoma, synovial sarcoma, soft tissue sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma , and chondrosarcoma.
  • the tumor is breast cancer, ovarian cancer, endometrial cancer, melanoma, kidney cancer, lung cancer, or liver cancer.
  • the cancer described herein is glioma, thyroid cancer, lung cancer, colorectal cancer, head and neck cancer, stomach cancer, liver cancer, pancreatic cancer, renal cancer, urothelial cancer, prostate cancer, testis cancer, breast cancer, cervical cancer, endometrial cancer, ovarian cancer, or melanoma. In some embodiments, the cancer described herein is carcinoid or lymphoma.
  • the therapeutic agent e.g., an anti-CD2 antibody or a CD2-targeting drug
  • the therapeutic agent is designed for treating various autoimmune diseases, including rheumatoid arthritis, Crohn’s disease, systemic lupus erythematosus, ankylosing spondylitis, inflammatory bowel diseases (IBD) , ulcerative colitis, or scleroderma.
  • the anti-CD2 antibody is designed for treating various immune disorders, including allergy, asthma, and/or atopic dermatitis.
  • the methods as described herein can be used to determine the effectiveness of an therapeutic agent (e.g., an anti-CD2 antibody or a CD2-targeting drug) in inhibiting immune response.
  • the immune disorders described herein is graft versus host disease (GVHD) , psoriasis, allergy, asthma, myocarditis, nephritis, hepatitis, systemic lupus erythematosus, rheumatoid arthritis, scleroderma, hyperthyroidism, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, ulcerative colitis, autoimmune liver disease, diabetes, pain or neurological disorders, etc.
  • the methods as described herein can be used to determine the effectiveness of an therapeutic agent (e.g., an anti-CD2 antibody or a CD2-targeting drug) in preventing and/or ameliorating immune recognition and attack to transplanted organs.
  • an therapeutic agent e.g., an anti-CD2 antibody or a CD2-targeting drug
  • the present disclosure also provides methods of determining toxicity of an antibody (e.g., anti-CD2 antibody) .
  • the methods involve administering the antibody to the animal as described herein.
  • the animal is then evaluated for its weight change, red blood cell count, hematocrit, and/or hemoglobin.
  • the antibody can decrease the red blood cells (RBC) , hematocrit, or hemoglobin by more than 20%, 30%, 40%, or 50%.
  • the animals can have a weight that is at least 5%, 10%, 20%, 30%, or 40%smaller than the weight of the control group (e.g., average weight of the animals that are not treated with the antibody) .
  • the present disclosure also relates to the use of the animal model generated through the methods as described herein in the development of a product related to an immunization processes of human cells, the manufacturing of a human antibody, or the model system for a research in pharmacology, immunology, microbiology and medicine.
  • the disclosure provides the use of the animal model generated through the methods as described herein in the production and utilization of an animal experimental disease model of an immunization processes involving human cells, the study on a pathogen, or the development of a new diagnostic strategy and/or a therapeutic strategy.
  • the disclosure also relates to the use of the animal model generated through the methods as described herein in the screening, verifying, evaluating or studying the CD2 gene function, human CD2 antibodies, drugs for human CD2 targeting sites, the drugs or efficacies for human CD2 targeting sites, the drugs for immune-related diseases and antitumor drugs.
  • the disclosure provides a method to verify in vivo efficacy of TCR-T, CAR-T, and/or other immunotherapies (e.g., T-cell adoptive transfer therapies) .
  • the methods include transplanting human tumor cells into the animal described herein, and applying human CAR-T to the animal with human tumor cells. Effectiveness of the CAR-T therapy can be determined and evaluated.
  • the animal is selected from the CD2 gene humanized non-human animal prepared by the methods described herein, the CD2 gene humanized non-human animal described herein, the double-or multi-humanized non-human animal generated by the methods described herein (or progeny thereof) , a non-human animal expressing the human or humanized CD2 protein, or the tumor-bearing or inflammatory animal models described herein.
  • the TCR-T, CAR-T, and/or other immunotherapies can treat the CD2-associated diseases described herein.
  • the TCA-T, CAR-T, and/or other immunotherapies provides an evaluation method for treating the CD2-associated diseases described herein.
  • the disclosure provides methods of determining effectiveness of a therapeutic agent for preventing and/or ameliorating immune recognition and attack to transplanted organs.
  • the methods involve administering the therapeutic agent (e.g., an anti-human CD2 antibody or a CD2-targeting drug) to the animal as described herein.
  • the animal is used to establish a xenograft model.
  • the present disclosure further relates to methods for generating genetically modified animal model with two or more human or chimeric genes.
  • the animal can comprise a human or chimeric CD2 gene and a sequence encoding an additional human or chimeric protein.
  • the additional human or chimeric protein can be CD3e molecule, epsilon (CD3E) , CD28, CD226, IL2, IL4, IL6, programmed cell death protein 1 (PD-1) , tumor necrosis factor receptor superfamily member 9 (4-1BB) , CD40, programmed cell death ligand 1 (PD-L1) , lymphocyte-activation gene 3 (LAG3) , tumor necrosis factor alpha (TNF- ⁇ ) , T cell immunoreceptor with Ig and ITIM domains (TIGIT) , cytotoxic T-lymphocyte-associated protein 4 (CTLA4) , tumor necrosis factor receptor superfamily, member 4 (OX40) , T-cell immunoglobulin and mucin-domain containing-3 (TIM3) , CD73, B And T Lymphocyte Associated (BTLA) , CD27, CD47, CD154, Glucocorticoid-Induced TNFR-Related Protein (GITR) , and/
  • the methods of generating genetically modified animal model with two or more human or chimeric genes can include the following steps:
  • the genetically modified animal in step (b) of the method, can be mated with a genetically modified non-human animal with human or chimeric CD3E, CD28, CD226, IL2, IL4, IL6, PD-1, 4-1BB, CD40, PD-L1, LAG3, TNF- ⁇ , TIGIT, CTLA4, OX40, TIM3, CD73, BTLA, CD27, CD47, CD154, GITR, and/or SIRP ⁇ .
  • a genetically modified non-human animal with human or chimeric CD3E, CD28, CD226, IL2, IL4, IL6, PD-1, 4-1BB, CD40, PD-L1, LAG3, TNF- ⁇ , TIGIT, CTLA4, OX40, TIM3, CD73, BTLA, CD27, CD47, CD154, GITR, and/or SIRP ⁇ .
  • the CD2 humanization is directly performed on a genetically modified animal having a human or chimeric CD3E, CD28, CD226, IL2, IL4, IL6, PD-1, 4-1BB, CD40, PD-L1, LAG3, TNF- ⁇ , TIGIT, CTLA4, OX40, TIM3, CD73, BTLA, CD27, CD47, CD154, GITR, and/or SIRP ⁇ gene.
  • a combination therapy that targets two or more of these proteins thereof may be a more effective treatment.
  • many related clinical trials are in progress and have shown a good effect.
  • the genetically modified animal model with two or more human or humanized genes can be used for determining effectiveness of a combination therapy that targets two or more of these proteins, e.g., an anti-CD2 antibody and an additional therapeutic agent for the treatment of cancer.
  • the methods include administering the anti-CD2 antibody and the additional therapeutic agent to the animal, wherein the animal has a tumor; and determining the inhibitory effects of the combined treatment to the tumor.
  • the additional therapeutic agent is an antibody that specifically binds to CD3E, CD28, CD226, IL2, IL4, IL6, PD-1, 4-1BB, CD40, PD-L1, LAG3, TNF- ⁇ , TIGIT, CTLA4, OX40, TIM3, CD73, BTLA, CD27, CD47, CD154, GITR, and/or SIRP ⁇ .
  • the additional therapeutic agent is an anti-CTLA4 antibody (e.g., ipilimumab) , an anti-PD-1 antibody (e.g., nivolumab) , or an anti-PD-L1 antibody.
  • the animal further comprises a sequence encoding a human or humanized PD-1, a sequence encoding a human or humanized PD-L1, or a sequence encoding a human or humanized CTLA-4.
  • the additional therapeutic agent is an anti-PD-1 antibody (e.g., nivolumab, pembrolizumab) , an anti-PD-L1 antibody, or an anti-CTLA-4 antibody.
  • the tumor comprises one or more tumor cells that express CD80, CD86, PD-L1, and/or PD-L2.
  • the combination treatment is designed for treating various cancers as described herein, e.g., a solid tumor, gynecologic cancer, breast cancer, colorectal cancer, gastric adenocarcinoma, lung adenocarcinoma, pancreatic cancer, or head and neck cancer.
  • a solid tumor e.g., a solid tumor, gynecologic cancer, breast cancer, colorectal cancer, gastric adenocarcinoma, lung adenocarcinoma, pancreatic cancer, or head and neck cancer.
  • the methods described herein can be used to evaluate the combination treatment with some other methods.
  • the methods of treating a cancer that can be used alone or in combination with methods described herein, include, e.g., treating the subject with chemotherapy, e.g., campothecin, doxorubicin, cisplatin, carboplatin, procarbazine, mechlorethamine, cyclophosphamide, adriamycin, ifosfamide, melphalan, chlorambucil, bisulfan, nitrosurea, dactinomycin, daunorubicin, bleomycin, plicomycin, mitomycin, etoposide, verampil, podophyllotoxin, tamoxifen, taxol, transplatinum, 5-flurouracil, vincristin, vinblastin, and/or methotrexate.
  • the methods can include performing surgery on the subject to remove at least a portion of the subject to remove at least
  • ScaI and AseI restriction enzymes were purchased from NEB (Catalog numbers: R3122S and R0526S, respectively) .
  • C57BL/6 mice and Flp transgenic mice were purchased from the China Food and Drugs Research Institute National Rodent Experimental Animal Center.
  • PE anti-mouse CD2 Antibody was purchased from BioLegend (Catalog number: 100107) .
  • APC anti-human CD2 Antibody was purchased from BioLegend (Catalog number: 300213) .
  • PerCP/Cyanine5.5 anti-mouse TCR ⁇ chain Antibody was purchased from BioLegend (Catalog number: 109228) .
  • PE/Cy TM 7 Mouse anti-mouse NK1.1 was purchased from BD Pharmingen (Catalog number: 552878) .
  • Purified anti-mouse CD16/32 Antibody was purchased from BioLegend (Catalog number: 101302) .
  • EXAMPLE 1 Mice with humanized CD2 gene
  • a non-human animal e.g., a mouse
  • a non-human animal was modified to include a nucleotide sequence encoding human CD2 protein
  • the obtained genetically-modified non-human animal can express a human or humanized CD2 protein in vivo.
  • the mouse CD2 gene (NCBI Gene ID: 12481, Primary source: MGI: 88320, UniProt ID: P08920) is located at 101183224 to 101195255 of chromosome 3 (NC_000069.7)
  • the human CD2 gene NCBI Gene ID: 914, Primary source: HGNC: 1639, UniProt ID: P06729) is located at 116754430 to 116769229 of chromosome 1 (NC_000001.11) .
  • the mouse CD2 transcript is NM_013486.2, and the corresponding protein sequence NP_038514.1 is set forth in SEQ ID NO: 1.
  • the human CD2 transcript is NM_001767.5, and the corresponding protein sequence NP_001758.2 is set forth in SEQ ID NO: 2.
  • Mouse and human CD2 gene loci are shown in FIG. 1.
  • nucleotide sequences encoding human CD2 protein can be introduced into the mouse endogenous CD2 locus, so that the mouse expresses human or humanized CD2 protein.
  • a sequence (about 8.0 kb) starting from within exon 1 and ending within exon 4 of mouse CD2 gene was replaced with a corresponding sequence (about 10.0 kb) starting from within exon 1 and ending within exon 4 of human CD2 gene, to obtain a humanized CD2 gene locus as shown in FIG. 2, thereby humanizing mouse CD2 gene.
  • the targeting vector contains homologous arm sequences upstream and downstream of the mouse CD2 gene, and an “A Fragment” containing DNA sequences of human CD2 gene.
  • sequence of the upstream homologous arm (5’ homologous arm, SEQ ID NO: 3) is identical to nucleotide sequence of 101195196-101198704 of NCBI accession number NC_000069.7
  • sequence of the downstream homologous arm (3’ homologous arm, SEQ ID NO: 4) is identical to nucleotide sequence of 101183839-101187215 of NCBI accession number NC_000069.7.
  • the genomic DNA sequence from human CD2 gene (SEQ ID NO: 7) is identical to nucleotide sequence of 116754493-116764494 of NCBI accession number NC_000001.11.
  • the connection between the 5’end of the A fragment and the mouse sequence was designed as: wherein the “G” in sequence is the last nucleotide of the mouse sequence, the first “A” in sequence “ ATGA ” is the first nucleotide of the human sequence.
  • the connection between the 3’ end of the A fragment and the mouse sequence was designed as: wherein the “G” in sequence is the last nucleotide of the human sequence, and the first “T” in sequence “ TCCT ” is the first nucleotide of the mouse sequence.
  • the targeting vector also includes an antibiotic resistance gene for positive clone screening (neomycin phosphotransferase gene, or Neo) , and two Frt recombination sites flanking the antibiotic resistance gene, that formed a Neo cassette (within the A Fragment) .
  • Neo cassette within the A Fragment
  • the connection between the 5’ end of the Neo cassette and the human sequence was designed as: wherein the “C” in sequence is the last nucleotide of the human sequence, and the first “G” in sequence “ GTCG ” is the first nucleotide of the Neo cassette.
  • the connection between the 3’ end of the Neo cassette and the human sequence was designed as: wherein the last “C” in sequence is the last nucleotide of the Neo cassette, and the first “C” in sequence “ CCCA ” is the first nucleotide of the human sequence.
  • a coding gene with a negative selectable marker (a gene encoding diphtheria toxin A subunit (DTA) ) was also constructed downstream of the 3' homologous arm of the targeting vector.
  • DTA diphtheria toxin A subunit
  • the targeting vector was constructed, e.g., by restriction enzyme digestion and ligation.
  • the constructed targeting vector sequences were preliminarily confirmed by restriction enzyme digestion, and then verified by sequencing.
  • Embryonic stem cells of C57BL/6 mice were transfected with the correct targeting vector by electroporation.
  • the positive selectable marker genes were used to screen the cells, and the integration of exogenous genes was confirmed by PCR and Southern Blot.
  • the clones identified as positive by PCR (primers shown in the table below) were then verified by Southern Blot.
  • the clones that were positive in Southern Blot detection were further sequenced. Clones verified to have no random insertion were subjected for subsequent experiments.
  • the positive clones that had been screened were introduced into isolated blastocysts (white mice) , and the resulted chimeric blastocysts were transferred to a culture medium for short-term culture and then transplanted to the fallopian tubes of the recipient mother (white mice) to produce the F0 chimeric mice (black and white) .
  • the F2 generation homozygous mice were obtained by backcrossing the F0 generation chimeric mice with wild-type mice to obtain the F1 generation mice, and then breeding the F1 generation heterozygous mice with each other.
  • the positive mice were also bred with the Flp transgenic mice to remove the positive selectable marker genes (schematic diagram shown in FIG. 4) , and then the humanized homozygous mice with a humanized CD2 gene were obtained by breeding the heterozygous mice with each other.
  • the CRISPR/Cas system can also be used for gene editing, and the targeting strategy shown in FIG. 5 was designed.
  • the targeting vector contains homologous arm sequences upstream and downstream of the mouse CD2 gene, and a human CD2 gene fragment. Specifically, sequence of the upstream homologous arm (5’ homologous arm, SEQ ID NO: 5) is identical to nucleotide sequence of 101195196-101196555 of NCBI accession number NC_000069.7, and sequence of the downstream homologous arm (3’ homologous arm, SEQ ID NO: 6) is identical to nucleotide sequence of 101185740-101187215 of NCBI accession number NC_000069.7.
  • the human CD2 gene fragment (SEQ ID NO: 7) is identical to identical to nucleotide sequence of 116754493-116764494 of NCBI accession number NC_000001.11. The connection between the 3’ end of the human CD2 gene fragment and the mouse sequence was designed as SEQ ID NO: 12.
  • the targeting vector was constructed, e.g., by restriction enzyme digestion, ligation, or direct synthesis.
  • the constructed targeting vector sequences were preliminarily confirmed by restriction enzyme digestion, and then verified by sequencing. Targeting vectors with verified sequences were used for subsequent experiments.
  • the targeting site sequences of the sgRNAs on the CD2 gene locus are as follows:
  • sgRNA1 targeting site (SEQ ID NO: 17) : 5’-CTGAGGTCTCGGGACTACGGAGG-3’;
  • sgRNA2 targeting site (SEQ ID NO: 18) : 5’-GCAGATTTAAGTAGCACTCCAGG-3’.
  • UCA kit was used to detect the activity of the sgRNAs. After confirming that the sgRNAs can induce efficient Cas9 cleavage, restriction enzyme cleavage sites were added to its 5' end and a complementary strand to obtain a forward oligonucleotide and a reverse oligonucleotide, as shown in the table below. After annealing, the products were ligated to the pT7-sgRNA plasmid (the plasmid was first linearized with BbsI) , to obtain expression vector pT7-CD2-1 and pT7-CD2-2.
  • the pT7-sgRNA vector was synthesized, which included a DNA fragment containing the T7 promoter and sgRNA scaffold (SEQ ID NO: 25) , and was ligated to the backbone vector (Takara, Catalog number: 3299) after restriction enzyme digestion (EcoRI and BamHI) . The resulting plasmid was confirmed by sequencing.
  • the pre-mixed Cas9 mRNA, the targeting vector, and in vitro transcription products of the pT7-CD2-1 and pT7-CD2-2 plasmids were injected into the cytoplasm or nucleus of fertilized eggs of C57BL/6 mice with a microinjection instrument.
  • the embryo microinjection was carried out according to the method described, e.g., in A. Nagy, et al., “Manipulating the Mouse Embryo: A Laboratory Manual (Third Edition) , ” Cold Spring Harbor Laboratory Press, 2006.
  • the injected fertilized eggs were then transferred to a culture medium to culture for a short time and then was transplanted into the oviduct of the recipient mouse to produce the genetically modified mice (F0 generation) .
  • the mouse population was further expanded by cross-breeding and self-breeding to establish stable homozygous mouse lines.
  • the genotype of the somatic cells of the F0 generation mice can be identified by PCR analysis. The PCR primers are shown in the table below.
  • mice identified as positive by PCR were then subjected to Southern Blot detection. Those mice identified as positive by Southern Blot were further sequenced to confirm there was no random insertion.
  • CD2 gene humanized mice Many methods can be used to verify the success of generating CD2 gene humanized mice. For example, the genotype of the somatic cells of the F1 generation mice can be identified by PCR analysis. The PCR primers are shown in the table below.
  • FIGS. 6A-6B The identification results of some F1 generation mice are shown in FIGS. 6A-6B. The results indicate that mice numbered F1-01, F1-02, F1-03, F1-04, F1-05, and F1-06 were positive heterozygous mice.
  • the F1 generation mice identified as positive by PCR were further verified by Southern Blot to confirm whether there was random insertion. Specifically, genomic DNA from the mouse tail was extracted, which was digested with ScaI or AseI restriction enzyme. The digested genomic DNA was then transferred to a membrane and hybridized with respective probes. The restriction enzymes, probes, and the size of target fragment sizes are shown in the table below.
  • the Southern Blot detection results are shown in FIG. 7.
  • the results showed that mice numbered F1-03, F1-04, and F1-06 were verified as positive heterozygous mice.
  • the results indicate that the CD2 gene humanized mice constructed using the methods described herein can be stably passaged without random insertions. Finally, the F1 generation heterozygous mice were bred with each other to obtain the F2 generation homozygous mice.
  • humanized CD2 mRNA in positive mice can be confirmed, e.g., by RT-PCR. Specifically, one 6-week-old male C57BL/6 wild-type mouse and one 6-week-old male CD2 gene humanized homozygous mouse generated using the methods described herein were selected, and spleen tissues were collected after euthanasia by cervical dislocation. Cellular RNA was extracted according to the instructions of the TRIzol TM kit, reverse transcribed into cDNA, and then detected by RT-PCR. As shown in FIGS.
  • PCR-F1 5’-CCTGAAGATAAAGAAGCCGAT-3’ (SEQ ID NO: 33) ,
  • PCR-R1 5’-TCTTAGACTCCTTGCTGACCG -3’ (SEQ ID NO: 34) ;
  • PCR-F2 5’-AATCCAGTGTCGAGCCTGTC-3’ (SEQ ID NO: 35) ,
  • PCR-R2 5’-TAATTAGGGGGCGGCAGTGA -3’ (SEQ ID NO: 36) ;
  • GAPDH-F 5’-TCACCATCTTCCAGGAGCGAGA-3’ (SEQ ID NO: 37) ,
  • GAPDH-R 5’-GAAGGCCATGCCAGTGAGCTT-3’ (SEQ ID NO: 38) .
  • human or humanized CD2 protein in positive mice can also be confirmed, e.g., by flow cytometry. Specifically, one 6-week-old male C57BL/6 wild-type mouse and one 6-week-old male CD2 gene humanized homozygous mouse were selected, and spleen tissues were collected after euthanasia by cervical dislocation.
  • mice numbered F-01, F-02, F-03, F-04, F-05, F-06, and F-07 were identified as positive heterozygous mice, indicating that CD2 gene knockout mice were successfully obtained.
  • the CD2 gene humanized mice prepared in Example 1 can be used to construct a variety of human disease models, including models of tumors, autoimmune diseases (e.g., psoriasis and atopic dermatitis) and inflammatory diseases (e.g., enteritis) , to test the in vivo efficacy of anti-human CD2 antibodies.
  • the CD2 gene humanized homozygous mice can be subcutaneously inoculated with tumor cells (e.g., mouse colon cancer cells MC38) . When the tumor volume reaches about 100 mm 3 , the mice can be placed into a control group and one or more treatment groups according to the tumor volume.
  • the treatment group mice can be administered with randomly selected drugs targeting human CD2 (e.g., an anti-human CD2 antibody) , and the control group mice can be injected with an equal volume of saline.
  • the tumor volume and body weight of the mice can be measured regularly, and the in vivo safety and efficacy of the drugs can be effectively assessed by comparing the changes in the body weight of the mice and the tumor size.
  • CD2 gene humanized mice can also be used to prepare various disease models such as autoimmune diseases and inflammatory diseases. By regularly measuring disease-related indicators, the safety and in vivo efficacy of antibody drugs in CD2 gene humanized mice can be effectively evaluated.
  • the CD2 gene humanized mice generated using the methods described herein can also be used to generate double-or multi-gene humanized mouse models.
  • the embryonic stem (ES) cells for blastocyst microinjection can be selected from mice comprising other genetic modifications such as modified (e.g., human or humanized) CD3E, CD28, CD226, IL2, IL4, IL6, PD-1, 41BB, CD40, PD-L1, LAG3, TNF- ⁇ and/or TIGIT genes.
  • embryonic stem cells from humanized CD2 mice described herein can be isolated, and gene recombination targeting technology can be used to obtain double-gene or multi-gene-modified mouse models of CD2 and other gene modifications.
  • the homozygous or heterozygous CD2 gene humanized mice obtained by the methods described herein with other genetically modified homozygous or heterozygous mice, and the offspring can be screened.
  • Mendel it is possible to generate double-gene or multi-gene modified heterozygous mice comprising modified (e.g., human or humanized) CD2 gene and other genetic modifications.
  • the heterozygous mice can be bred with each other to obtain homozygous double-gene or multi-gene modified mice.
  • These double-gene or multi-gene modified mice can be used for in vivo validation of gene regulators targeting human CD2 and other genes.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Environmental Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne des animaux non humains génétiquement modifiés qui expriment un CD2 humain ou chimérique (par exemple, humanisé), ainsi que des procédés d'utilisation de ceux-ci.
PCT/CN2022/131092 2021-11-10 2022-11-10 Animal non humain génétiquement modifié comportant un cd2 humain ou chimérique WO2023083250A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111328705.2 2021-11-10
CN202111328705 2021-11-10

Publications (1)

Publication Number Publication Date
WO2023083250A1 true WO2023083250A1 (fr) 2023-05-19

Family

ID=86335109

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/131092 WO2023083250A1 (fr) 2021-11-10 2022-11-10 Animal non humain génétiquement modifié comportant un cd2 humain ou chimérique

Country Status (1)

Country Link
WO (1) WO2023083250A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107529739A (zh) * 2015-04-30 2018-01-02 株式会社特殊免疫研究所 表达人特定分子和人Fcγ受体家族的转基因非人动物
US20190373869A1 (en) * 2017-12-12 2019-12-12 Beijing Biocytogen Co., Ltd. GENETICALLY MODIFIED NON-HUMAN ANIMAL WITH HUMAN OR CHIMERIC CD3e

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107529739A (zh) * 2015-04-30 2018-01-02 株式会社特殊免疫研究所 表达人特定分子和人Fcγ受体家族的转基因非人动物
US20190373869A1 (en) * 2017-12-12 2019-12-12 Beijing Biocytogen Co., Ltd. GENETICALLY MODIFIED NON-HUMAN ANIMAL WITH HUMAN OR CHIMERIC CD3e

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DATABASE Nucleotide NCBI; 16 May 2021 (2021-05-16), ANONYMOUS : "Homo sapiens chromosome 1, GRCh38.p13 Primary Assembly ", XP093065682, Database accession no. NC_000001.11 *
DATABASE Nucleotide NCBI; 22 September 2020 (2020-09-22), ANONYMOUS : "Mus musculus strain C57BL/6J chromosome 3, GRCm39 ", XP093065674, Database accession no. NC_000069.7 *
DATABASE Nucleotide NCBI; 26 October 2021 (2021-10-26), ANONYMOUS : "Mus musculus CD2 antigen (Cd2), mRNA ", XP093065670, Database accession no. NM_013486.2 *
DATABASE Nucleotide NCBI; 8 November 2021 (2021-11-08), ANONYMOUS : "Homo sapiens CD2 molecule (CD2), transcript variant 2, mRNA", XP093065672, Database accession no. NM_001767.5 *
LANG G., WOTTON D., OWEN M. J., SEWELL W. A., BROWN M. H., MASON D. Y., CRUMPTON M. J., KIOUSSIS D.: "The structure of the human CD2 gene and its expression in transgenic mice.", THE EMBO JOURNAL / EUROPEAN MOLECULAR BIOLOGY ORGANIZATION, IRL PRESS, OXFORD, vol. 7, no. 6, 1 June 1988 (1988-06-01), Oxford , pages 1675 - 1682, XP093065667, ISSN: 0261-4189, DOI: 10.1002/j.1460-2075.1988.tb02995.x *

Similar Documents

Publication Publication Date Title
US11723348B2 (en) Genetically modified mice expressing humanized CD47
US11279948B2 (en) Genetically modified non-human animal with human or chimeric OX40
US10973212B2 (en) Genetically modified non-human animal with human or chimeric SIRPa
US20220071185A1 (en) Genetically modified non-human animal with human or chimeric cd137
US10945418B2 (en) Genetically modified non-human animal with human or chimeric PD-L1
US11505806B2 (en) Genetically modified non-human animal with human or chimeric OX40
US11071290B2 (en) Genetically modified non-human animal with human or chimeric CTLA-4
WO2018068756A1 (fr) Animal non humain génétiquement modifié à btla humaine ou chimérique
US20190281798A1 (en) Genetically Modified Non-Human Animal With Human Or Chimeric PD-L1
US11497198B2 (en) Genetically modified mice expressing humanized CD40
WO2018086583A1 (fr) Animal non humain génétiquement modifié, doté d'un gène lag-3 humain ou chimérique
US11350614B2 (en) Genetically modified non-human animal with human or chimeric CD28
US11464876B2 (en) Genetically modified mouse comprising a chimeric TIGIT
US11272695B2 (en) Genetically modified non-human animal with human or chimeric PD-1
US10945419B2 (en) Genetically modified non-human animal with human or chimeric GITR
US20190343095A1 (en) Genetically modified non-human animal with human or chimeric cd137
WO2018233607A1 (fr) Animal non humain génétiquement modifié avec cd40 humain ou chimérique
US10925264B2 (en) Genetically modified non-human animal with human or chimeric LAG-3
WO2023284850A1 (fr) Animal non humain génétiquement modifié comportant un tfr1 humain ou chimérique
WO2018233606A1 (fr) Animal non humain génétiquement modifié avec gitr humain ou chimérique
US11564381B2 (en) Genetically modified non-human animal with human or chimeric LAG3
WO2023083250A1 (fr) Animal non humain génétiquement modifié comportant un cd2 humain ou chimérique
WO2018233608A1 (fr) Animal non humain génétiquement modifié à cd28 humaine ou chimérique
WO2023109956A1 (fr) Animal non humain génétiquement modifié comportant un cd98hc humain ou chimérique
WO2023072081A1 (fr) Animal non humain génétiquement modifié comportant un tgfbr2 humain ou chimérique

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: 22892044

Country of ref document: EP

Kind code of ref document: A1