[go: up one dir, main page]

WO2025140609A1 - Method for constructing cell which produces defucosylated protein and use of cell - Google Patents

Method for constructing cell which produces defucosylated protein and use of cell Download PDF

Info

Publication number
WO2025140609A1
WO2025140609A1 PCT/CN2024/143321 CN2024143321W WO2025140609A1 WO 2025140609 A1 WO2025140609 A1 WO 2025140609A1 CN 2024143321 W CN2024143321 W CN 2024143321W WO 2025140609 A1 WO2025140609 A1 WO 2025140609A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
zinc finger
cell
fut8
finger nuclease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2024/143321
Other languages
French (fr)
Chinese (zh)
Inventor
周杨钫
李颖
饶一帆
高荣
朱小彬
王利乐
廖善辉
王帅
章雨
张胜
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wuxi Biologics Shanghai Co Ltd
Wuxi Biologics Ireland Ltd
Original Assignee
Wuxi Biologics Shanghai Co Ltd
Wuxi Biologics Ireland 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 Wuxi Biologics Shanghai Co Ltd, Wuxi Biologics Ireland Ltd filed Critical Wuxi Biologics Shanghai Co Ltd
Publication of WO2025140609A1 publication Critical patent/WO2025140609A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor

Definitions

  • the present disclosure relates to the fields of molecular biology and biopharmaceuticals; in particular, to cells for producing defucosylated proteins constructed by gene editing using zinc finger protein nuclease technology and applications thereof.
  • ADCC antibody-dependent cellular cytotoxicity
  • NK natural killer cells
  • Fc constant region of antibodies primarily through interaction with NK cell Fc ⁇ RIII receptors.
  • NK cells then deposit perforins and granzymes on the surface of target cells, inducing cell lysis and apoptosis, respectively.
  • defucosylated protein production cells To meet the above needs, after long-term and in-depth exploration and research, the inventors have developed a new method for constructing defucosylated protein production cells. This method uses zinc finger nuclease to inactivate the FUT8 gene in cells, and the resulting cells produce defucosylated proteins (e.g., antibodies) with strong effector functions, which has broad application prospects in industry.
  • defucosylated proteins e.g., antibodies
  • the present invention provides a zinc finger protein capable of targeting and binding to the FUT8 gene, which comprises the zinc finger recognition region amino acid sequence of zinc finger F1 to F5 or to F6 shown in the same row in the following table in order from N to C terminus:
  • the present invention provides a zinc finger nuclease (ZFN) against the FUT8 gene, which is a fusion protein and comprises the ZFP described herein and at least one DNA cleavage domain or cleavage half-domain.
  • ZFN zinc finger nuclease
  • Yet another aspect herein provides a polynucleotide encoding a ZFP or ZFN as described herein.
  • Another aspect herein provides a vector comprising the polynucleotide as described above.
  • Another aspect herein provides a cell line in which the FUT8 gene is inactivated or partially inactivated, comprising a ZFP, ZFN, polynucleotide or vector described herein.
  • Another aspect herein provides a method of producing a FUT8-deficient cell, the method comprising:
  • Another aspect herein provides a method of generating a FUT8-deficient cell line, the method comprising:
  • Another aspect of the present invention provides a method for producing a target recombinant protein in a host cell, the method comprising:
  • the present invention provides a FUT8-deficient cell line, which comprises a nucleotide sequence as shown in one or more of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58 and SEQ ID NO: 59 at the FUT8 locus.
  • Reverse primer TGCCACTGCTTCTATATACTGATTC (SEQ ID NO: 47);
  • Reverse primer GGTCTGTTCCATCCCCAGAATG (SEQ ID NO: 49);
  • Reverse primer TGTTACTTAAGCCCCAGGC (SEQ ID NO: 51).
  • Another aspect of the present invention provides a method for cultivating a FUT8-deficient cell line, comprising:
  • FIG. 1 shows the binding site, cleavage site, and PCR primer sites of zinc finger nuclease targeting FUT8 exon 3 and exon 10 and their vicinity according to one of the exemplary embodiments herein.
  • FIG 2A shows a schematic diagram of the expression plasmid of the zinc finger protein nuclease according to one of the exemplary embodiments of the present invention.
  • the zinc finger protein and the restriction endonuclease FokI cleavage half domain are fused and cloned in the plasmid.
  • the fusion protein is under the control of the CMV promoter.
  • Figure 2B shows a schematic diagram of the specific structure of the zinc finger protein nuclease according to one of the exemplary embodiments of the present invention, in which each zinc finger binding domain is sequentially inserted between the framework sequence of the natural zinc finger protein and connected to the FokI cleavage half domain at the C-terminus.
  • Figure 3 shows the T7E1 digestion experiment to verify whether the ZFN plasmid can effectively remove the target gene sequence.
  • Lane 1 S2-L and S4-R transfection, the editing efficiency calculated according to the band brightness measured by ImageJ is 4.7%;
  • Lane 2 L3 and R3 transfection, the editing efficiency is 6.2%;
  • NC negative control group, that is, the cell genomic DNA without knockout of the FUT8 gene;
  • PC positive control group, that is, the cell genome containing 50% of the knockout FUT8 gene.
  • Figure 4 shows the results of flow cytometric analysis of cell pools stained with green fluorescent marker lentil agglutinin (LCA-FITC).
  • A Quantitative analysis of different sub-cell populations, where the circled cell population with lower green fluorescence is the FUT8 -/- cell population, which accounts for 27.44% of the cell pool.
  • B Peak shifts of different sub-cell populations.
  • Figure 5 shows the flow cytometry analysis results of clone 154-F8ZFN-09-08 and negative control after LCA-FITC staining. Solid line: 154-F8ZFN-09-08 cells; dotted line: negative control without knockout of FUT8 gene.
  • FIG6 shows the NGS validation results, demonstrating that a pair of alleles of clones 154-F8ZFN-09-08 and 154-F8ZFN-10-02 were knocked out.
  • Figure 7 shows the application of FUT8 -/- cell lines (i.e., 154-F8ZFN-09-08 and 154-F8ZFN-10-02) in the screening of monoclonal antibody expression.
  • NC Negative control in which the FUT8 gene was not knocked out.
  • A Changes in viability during FB culture after cell pool recovery
  • B Changes in viable cell density VCD during FB culture of cell pool
  • C Protein expression level titer on day 14 during fed-batch culture
  • D The harvested supernatant was purified by one-step ProA and then analyzed by size exclusion chromatography SEC-HPLC
  • E One-step purified samples were subjected to SDS Caliper NR and SDS Caliper R
  • F Glycoform detection results of one-step purified samples.
  • Figure 8 shows the performance evaluation results of the FUT8-/- cell line (154-F8ZFN-09-08) in antibody production.
  • A Cell viability of the 154-F8ZFN-09-08 group during FB culture;
  • B Changes in viable cell density (VCD);
  • C Protein expression titer;
  • D SEC-HPLC and SDS Caliper NR analysis results;
  • E Glycoform analysis results.
  • Figure 9 shows the results of gene editing efficiency detection.
  • Lane 1 31-L1/31-R1 knockout sample, editing efficiency is 9.4%
  • Lane 2 32-L1/32-R1 knockout sample, editing efficiency is 10.9%
  • NC wild-type CHO-K1.
  • Figure 10 shows the results of SCP monoclonal sorting.
  • the unstained sample is CHO K1;
  • Fut8 -/- clone C6 is the monoclonal clone selected after LCA-FITC staining after sorting monoclonal clones;
  • Fut8 wt is CHO K1 stained with LCA-FITC.
  • Figure 11 shows the results of SCP monoclonal sorting.
  • the unstained sample is CHO K1-N079;
  • Fut8 -/- clone F2 is a monoclonal selected after LCA-FITC staining after sorting;
  • Fut8 wt is CHO K1 stained with LCA-FITC.
  • FIG12 shows the NGS validation results, demonstrating that a pair of alleles in clones C6 and F2 were knocked out.
  • Fut8 attaches core fucose to oligosaccharides present on the Fc region of antibodies, which is generally considered to be important for the effector function of antibody-dependent cellular cytotoxicity.
  • Three-dimensional analysis of the structure of human Fut8 revealed that three ⁇ 2/ ⁇ 6 fucosyltransferase motifs form the catalytic core of the enzyme. See, Ihara et al. (2007) Glycobiology 17:455-66. In this region, many single residue point mutations to alanine result in complete inactivation of the enzyme. See, Ihara et al. (2007) Glycobiology 17:455-66; Takahashi et al. (2000) Glycobiology 10:503-10.
  • the present invention provides a method for constructing a defucosylated protein production cell, which inactivates the FUT8 gene in the cell by using a zinc finger nuclease targeting the FUT8 gene.
  • the zinc finger protein nuclease comprises a zinc finger protein (ZFP) and a nuclease cleavage domain or a cleavage partner domain.
  • ZFP zinc finger protein
  • zinc finger protein or "ZFP” refers to a protein that binds DNA in a sequence-specific manner via one or more zinc fingers (or zinc finger recognition regions).
  • the zinc fingers are amino acid sequence regions within the protein whose structure is stabilized by the coordination of zinc ions.
  • the zinc fingers in ZFP are named in the form of "F+sequential numbers from N to C".
  • the first zinc finger is named "F1”
  • the second zinc finger is named "F2”, and so on.
  • one or more zinc fingers of the ZFP have an engineered zinc finger recognition region.
  • the ZFP has the recognition region amino acid sequence of the zinc finger (F1 to F5 or to F6) shown in the same row in Table 1:
  • each row describes a single ZFP.
  • the name and DNA target sequence (in 5'-3' order) of each ZFP are shown in the first column, and the amino acid sequence of the recognition region of the zinc finger (F1 to F5 or to F6) in each ZFP is shown in columns 2 to 6 or 7.
  • the multiple zinc fingers of the ZFP can be connected by a suitable linker sequence.
  • the linker sequence can include a linker of 1, 2, 3, 4, 5 or more amino acids in length.
  • F2 TSGNLTE (SEQ ID NO: 31);
  • the left ZFP can have the following recognition region amino acid sequence (ZFP 31-L1):
  • F2 NQSNLLR (SEQ ID NO: 40);
  • F3 FHSNLLA (SEQ ID NO: 18);
  • F5 QSGNLSR (SEQ ID NO: 42).
  • the right ZFP can have the following recognition region amino acid sequence (ZFP 32-R1):
  • RKSHLTM SEQ ID NO: 19
  • F2 FHSGLLA (SEQ ID NO: 20);
  • F4 RKYVLLR (SEQ ID NO: 21);
  • RKDNLKN (SEQ ID NO: 33).
  • fusion protein refers to a protein molecule in which two or more subunit molecules (eg, a ZFP and a DNA cleavage domain or cleavage half-domain as described herein) are linked to each other (preferably covalently linked).
  • cleavage refers to the breakage of the covalent backbone of a DNA molecule.
  • Cutting can be initiated by various methods, including but not limited to enzymatic or chemical hydrolysis of phosphodiester bonds.
  • cutting can be single-stranded cutting or double-stranded cutting.
  • double-stranded cutting can be caused by two different single-stranded cutting events.
  • Cutting of DNA can result in the generation of flat ends or staggered ends.
  • the ZFN is used to target double-stranded DNA cutting.
  • cleavage half-domain refers to a polypeptide sequence that can be conjugated with another polypeptide (the same or different) to form a complex having cleavage activity (preferably double-chain cleavage activity).
  • cleavage activity preferably double-chain cleavage activity
  • two cleavage half-domains typically function as a dimer (in pairs) to exert cleavage activity.
  • the ZFP and at least one DNA cleavage domain or cleavage half-domain are operably linked.
  • operably linked may refer to a component being linked to another component in a functional manner.
  • a fusion protein in which a ZFP is fused to a cleavage domain if the ZFP in the fusion protein can bind to its target site and/or its binding site, and the cleavage domain can cleave DNA near the target site, then the ZFP and the cleavage domain are in an operably linked relationship.
  • the zinc finger nuclease described in this article can be used as a tool for targeted cutting and inactivation of a target gene (such as the FUT8 gene), which can achieve the purpose of targeted knockout of the target gene by introducing a double-stranded DNA break (DSB) at the target site of the target gene and utilizing non-homologous end joining (NHEJ)-mediated DSB repair.
  • a target gene such as the FUT8 gene
  • a cleavage domain or cleavage half-domain can be any portion of a protein that retains cleavage activity or retains the ability to multimerize (e.g., dimerize) to form a functional cleavage domain.
  • the type IIS restriction enzyme is FokI.
  • the enzyme is active as a dimer. Accordingly, in some embodiments, a portion of the FokI enzyme used in the fusion protein is considered to be a cleavage half-domain.
  • two fusion proteins each comprising a FokI cleavage half-domain, can be used to reconstitute a catalytically active cleavage domain.
  • a single polypeptide molecule containing a zinc finger protein and two FokI cleavage half-domains can also be used.
  • the cleavage half-domain is a FokI cleavage half-domain. In some embodiments, the cleavage half-domain is a wild-type FokI cleavage half-domain. In other embodiments, the cleavage half-domain is a modified FokI cleavage half-domain.
  • the cleavage half-domain is derived from Sharkey FokI nuclease obtained by amino acid mutation based on wild-type FokI, and its exemplary amino acid sequence is shown in SEQ ID NO: 3.
  • restriction enzymes comprising separable binding and cleavage domains are also contemplated herein. See, e.g., Roberts et al. (2003) Nucleic Acids Res. 31: 418-420.
  • the mutation at 490 replaces Glu (E) with Lys (K); the mutation at 538 replaces Iso (I) with Lys (K); the mutation at 486 replaces Gln (Q) with Glu (E); and the mutation at 499 replaces Iso (I) with Lys (K).
  • the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 52 and SEQ ID NO: 53 at the FUT8 locus. In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 54 and SEQ ID NO: 55 at the FUT8 locus. In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 56 and SEQ ID NO: 57 at the FUT8 locus. In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 58 and SEQ ID NO: 59 at the FUT8 locus.
  • FUT8-deficient cell line comprising an altered FUT8 locus, wherein the altered FUT8 locus can be detected by the following primer pair:
  • Reverse primer TGCCACTGCTTCTATATACTGATTC (SEQ ID NO: 47);
  • Reverse primer GGTCTGTTCCATCCCCAGAATG (SEQ ID NO: 49);
  • Reverse primer TGTTACTTAAGCCCCAGGC (SEQ ID NO: 51).
  • the detection uses an equivalent cell line with normal (non-defective) FUT8 as a control cell line.
  • the detection shows a difference in results between the FUT8-deficient cell line and the control cell line (e.g., a difference in molecular size and band brightness measured by electrophoresis, etc.).
  • inactivation comprises partial or complete inhibition of expression of the FUT8 gene in a cell.
  • inactivation of the FUT8 gene can be achieved, for example, by a single cleavage event, by cleavage followed by non-homologous end joining, by cleavage at two sites followed by joining to delete the sequence between the two cleavage sites, by targeted recombination of missense or nonsense codons to the coding region, by targeted recombination of an unrelated sequence (i.e., a "stuffer" sequence) to a gene or its regulatory region to disrupt a gene or regulatory region, or by targeted recombination of a splice acceptor sequence to an intron to cause mis-splicing of the transcript.
  • an unrelated sequence i.e., a "stuffer" sequence
  • the ZFN-mediated inactivation (knockout or inhibition) described herein has a variety of applications.
  • Another aspect herein provides a method of producing a FUT8-deficient cell, the method comprising:
  • a polynucleotide encoding one or more ZFNs into a cell, wherein the ZFN comprises: (i) a ZFP as described herein, which is capable of binding to a target site in a FUT8 gene in the cell; and (ii) a DNA cleavage domain or cleavage half-domain;
  • the method may further include introducing an additional nuclease having a target site in the FUT8 gene into the cell.
  • additional nuclease may include, for example, homing endonucleases and meganucleases.
  • Another aspect herein provides a method of generating a FUT8-deficient cell line, the method comprising:
  • Another aspect of the present invention provides a method for producing a target recombinant protein in a host cell, the method comprising:
  • the step of inactivating the endogenous FUT8 gene in the cell as described herein and the step of introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell may be performed in reverse order or simultaneously.
  • Suitable cells include, but are not limited to, eukaryotic and prokaryotic cells and/or cell lines.
  • Non-limiting examples of such cells or cell lines include: COS, CHO (e.g., CHO-S, CHO-K1, CHO-DG44, CHO-DUXB11, CHO-DUKX, CHOK1SV), VERO, MDCK, WI38, V79, B14AF28-G3, BHK, HaK, NS0, SP2/0-Ag14, HeLa, HEK293 (e.g., HEK293-F, HEK293-H, HEK293-T) and perC6 cells and insect cells such as fall armyworm (Sf) or fungal cells such as saccharomyces cerevisiae, pichia pastoris and fission yeast.
  • COS CHO
  • CHO-K1, CHO-DG44 CHO-DUXB11
  • CHO-DUKX CHOK1SV
  • VERO MDCK
  • the cell is a mammalian cell.
  • the cell is a Chinese hamster ovary cell (CHO cell). Progeny, variants and derivatives of these cell lines may also be used.
  • the cell is a mammalian cell.
  • the cell is a Chinese hamster ovary cell (CHO cell).
  • the target recombinant protein includes an Fc-containing recombinant protein. In some embodiments, the target recombinant protein is a defucosylated Fc-containing recombinant protein.
  • Another aspect herein provides the use of the ZFPs, ZFNs, polynucleotides, vectors described herein for generating FUT8-deficient cells.
  • Another aspect herein provides use of a ZFP, ZFN, polynucleotide, vector, cell or cell line described herein for producing a defucosylated Fc-containing recombinant protein.

Landscapes

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

Abstract

Provided is a method for constructing a cell which produces defucosylated protein using a zinc finger nuclease which contains a zinc finger protein capable of binding to the FUT8 gene in a targeted manner and a DNA cleavage domain or cleavage half-domain. Also provided are a polynucleotide which encodes the zinc finger protein and the zinc finger nuclease, and a cell containing the polynucleotide and the zinc finger nuclease.

Description

一种构建生产去岩藻糖基化蛋白质的细胞的方法及其应用A method for constructing a cell for producing defucosylated protein and its application 技术领域Technical Field

本公开涉及分子生物学及生物制药领域;具体地,涉及通过利用锌指蛋白核酸酶技术进行基因编辑而构建的生产去岩藻糖基化蛋白质的细胞及其应用。The present disclosure relates to the fields of molecular biology and biopharmaceuticals; in particular, to cells for producing defucosylated proteins constructed by gene editing using zinc finger protein nuclease technology and applications thereof.

背景技术Background Art

近年来,随着基因工程抗体的成功制备和广泛应用,以及抗体生产水平的显著提高,抗体药物的研发进入了一个快速发展的新阶段。据文献报道,截止2019年12月,市场上已有79种治疗性抗体,主要用于肿瘤、自身免疫病、代谢及传染性疾病(Lu RM等(2020)J Biomed Sci.27(1):1-30)。2022年全球十大畅销抗肿瘤药物中包括四款抗体药,其总销售额达近424.92亿美元。In recent years, with the successful preparation and widespread application of genetically engineered antibodies and the significant improvement in antibody production levels, the research and development of antibody drugs has entered a new stage of rapid development. According to literature reports, as of December 2019, there were 79 therapeutic antibodies on the market, mainly used for tumors, autoimmune diseases, metabolic and infectious diseases (Lu RM et al. (2020) J Biomed Sci. 27(1): 1-30). The top ten best-selling anti-cancer drugs in the world in 2022 include four antibody drugs, with a total sales of nearly US$42.492 billion.

抗体通过两种机制激活免疫系统破坏攻击细胞:补体依赖性细胞毒性(CDC)和抗体依赖性细胞的细胞毒性(ADCC)。ADCC是主要由针对抗体包被的靶的天然杀伤(NK)细胞产生的免疫应答。在ADCC中,NK细胞主要经由与NK细胞FcγRIII受体的相互作用来识别抗体的恒定(Fc)区。然后,NK细胞沉积穿孔蛋白和粒酶于靶细胞表面,分别诱导细胞裂解和凋亡。Antibodies activate the immune system to destroy attacking cells through two mechanisms: complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). ADCC is an immune response produced primarily by natural killer (NK) cells against antibody-coated targets. In ADCC, NK cells recognize the constant (Fc) region of antibodies primarily through interaction with NK cell FcγRIII receptors. NK cells then deposit perforins and granzymes on the surface of target cells, inducing cell lysis and apoptosis, respectively.

大多数哺乳动物免疫球蛋白被岩藻糖基化,包括中国仓鼠卵巢细胞(CHO细胞)产生的免疫球蛋白。α-1,6岩藻糖转移酶(FUT8)催化岩藻糖残基由二磷酸鸟苷-岩藻糖(GDP-Fuc)转移至N-糖链核心最内侧乙酰氨基葡萄糖(GlcNAc)的末位以形成附着于抗体Fc核心区的岩藻糖。已有研究表明,当从IgG抗体Fc片段的CH2结构域Asn297位置处的糖链结构去除岩藻糖时,能够增强NK细胞FcγRIIIa与IgG抗体Fc片段之间的结合,从而增强抗体的ADCC效应(Shinkawa T等(2003)J Biol Chem.278(5):3466-73)。Most mammalian immunoglobulins are fucosylated, including those produced by Chinese hamster ovary cells (CHO cells). α-1,6-fucosyltransferase (FUT8) catalyzes the transfer of fucose residues from guanosine diphosphate-fucose (GDP-Fuc) to the end of the innermost acetylglucosamine (GlcNAc) of the N-glycan core to form fucose attached to the antibody Fc core region. Studies have shown that when fucose is removed from the sugar chain structure at the Asn297 position of the CH2 domain of the IgG antibody Fc fragment, it can enhance the binding between NK cell FcγRIIIa and the IgG antibody Fc fragment, thereby enhancing the ADCC effect of the antibody (Shinkawa T et al. (2003) J Biol Chem. 278 (5): 3466-73).

目前,工业生产去岩藻糖基化抗体的常见方法有三种:(1)在培养基中添加岩藻糖基化抑制剂,如2-氟岩藻糖(2FF)(Rillahan CD等(2012)Nat Chem Biol.8(7):661-8);(2)在工程细胞中稳定过表达细菌RMD蛋白(GDP-6-脱氧-D-来苏-4-己酮糖还原酶)(von Horsten HH等(2010)Glycobiology.20(12):1607-18),通过干扰底物GDP-岩藻糖的形成,阻断Fc区域的岩藻糖基化修饰;(3)干扰岩藻糖生成通路中的关键基因,如导入表达β(1,4)-N-乙酰氨基葡萄糖转移酶III(GnTIII)(Popp O等(2018)MAbs.10(2):290-303),敲除GDP-岩藻糖转运蛋白(GFT)(Omasa T等(2008)J Biosci Bioeng.106(2):168-73),或敲除α-1,6-岩藻糖基转移酶(FUT8)(Yamane-Ohnuki N等(2004)Biotechnol Bioeng.87(5):614-22)等。At present, there are three common methods for industrial production of defucosylated antibodies: (1) adding fucosylation inhibitors such as 2-fluorofucose (2FF) to the culture medium (Rillahan CD et al. (2012) Nat Chem Biol. 8(7): 661-8); (2) stably overexpressing bacterial RMD protein (GDP-6-deoxy-D-lyso-4-hexulose reductase) in engineered cells (von Horsten HH et al. (2010) Glycobiology. 20(12): 1607-18) to block the fucosylation modification of the Fc region by interfering with the formation of substrate GDP-fucose; 3) Interfering with key genes in the fucose production pathway, such as introducing expression of β(1,4)-N-acetylglucosamine transferase III (GnTIII) (Popp O et al. (2018) MAbs. 10(2): 290-303), knocking out GDP-fucose transporter (GFT) (Omasa T et al. (2008) J Biosci Bioeng. 106(2): 168-73), or knocking out α-1,6-fucosyltransferase (FUT8) (Yamane-Ohnuki N et al. (2004) Biotechnol Bioeng. 87(5): 614-22), etc.

本领域仍需构建去岩藻糖基化蛋白质生产细胞的新方法。There is still a need in the art for new methods to construct defucosylated protein-producing cells.

发明内容Summary of the invention

为满足上述需求,经过长期深入的探索研究,发明人开发出了用于构建去岩藻糖基化蛋白质生产细胞的新方法。该方法利用锌指核酸酶使细胞中的FUT8基因失活,所得细胞产生具有强效应功能的去岩藻糖基化蛋白质(例如,抗体),在工业上具有广泛的应用前景。To meet the above needs, after long-term and in-depth exploration and research, the inventors have developed a new method for constructing defucosylated protein production cells. This method uses zinc finger nuclease to inactivate the FUT8 gene in cells, and the resulting cells produce defucosylated proteins (e.g., antibodies) with strong effector functions, which has broad application prospects in industry.

一方面,本文提供一种能靶向结合FUT8基因的锌指蛋白,其以自N至C端顺序包含下表同行所示的锌指F1至F5或至F6的锌指识别区氨基酸序列:

In one aspect, the present invention provides a zinc finger protein capable of targeting and binding to the FUT8 gene, which comprises the zinc finger recognition region amino acid sequence of zinc finger F1 to F5 or to F6 shown in the same row in the following table in order from N to C terminus:

另一方面,本文提供一种针对FUT8基因的锌指核酸酶(ZFN),其为融合蛋白,且包含本文所述的ZFP和至少一个DNA切割域或切割半域。In another aspect, the present invention provides a zinc finger nuclease (ZFN) against the FUT8 gene, which is a fusion protein and comprises the ZFP described herein and at least one DNA cleavage domain or cleavage half-domain.

本文的又一方面提供一种多核苷酸,其编码本文所述的ZFP或ZFN。Yet another aspect herein provides a polynucleotide encoding a ZFP or ZFN as described herein.

本文的另一方面提供一种载体,其包含如上所述的多核苷酸。Another aspect herein provides a vector comprising the polynucleotide as described above.

本文的另一方面提供一种分离的细胞(例如,宿主细胞),其包含本文所述的ZFP、ZFN、多核苷酸或载体。Another aspect herein provides an isolated cell (eg, a host cell) comprising a ZFP, ZFN, polynucleotide or vector described herein.

本文的另一方面提供一种其中FUT8基因被失活或部分失活的细胞系,其包含本文所述的ZFP、ZFN、多核苷酸或载体。Another aspect herein provides a cell line in which the FUT8 gene is inactivated or partially inactivated, comprising a ZFP, ZFN, polynucleotide or vector described herein.

本文的另一方面提供一种产生FUT8缺陷型细胞的方法,所述方法包括:Another aspect herein provides a method of producing a FUT8-deficient cell, the method comprising:

(a)将编码一种或多种ZFN的多核苷酸引入细胞,其中所述ZFN包含:(i)本文所述的ZFP,所述ZFP能与细胞中FUT8基因中的靶位点结合;和(ii)DNA切割域或切割半域;(a) introducing a polynucleotide encoding one or more ZFNs into a cell, wherein the ZFN comprises: (i) a ZFP as described herein, which is capable of binding to a target site in a FUT8 gene in the cell; and (ii) a DNA cleavage domain or cleavage half-domain;

(b)使所述ZFN在所述细胞中表达,从而所述ZFN与所述靶位点结合,并切割所述FUT8基因。(b) expressing the ZFN in the cell, so that the ZFN binds to the target site and cleaves the FUT8 gene.

本文的另一方面提供一种产生FUT8缺陷型细胞系的方法,所述方法包括:Another aspect herein provides a method of generating a FUT8-deficient cell line, the method comprising:

(a)通过如本文所述的产生FUT8缺陷型细胞的方法使细胞中的内源FUT8基因失活;和(a) inactivating the endogenous FUT8 gene in the cell by the method for producing FUT8-deficient cells as described herein; and

(b)在适合产生FUT8缺陷型细胞系的条件下培养所述细胞。(b) culturing the cells under conditions suitable for generating a FUT8-deficient cell line.

本文的又一方面提供一种在宿主细胞中产生目标重组蛋白的方法,所述方法包括:Another aspect of the present invention provides a method for producing a target recombinant protein in a host cell, the method comprising:

(a)提供本文所述的FUT8缺陷型细胞,(a) providing a FUT8-deficient cell as described herein,

(b)将包含编码所述目标重组蛋白的核酸的表达载体引入所述细胞,由此产生所述目标重组蛋白。(b) introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell, thereby producing the target recombinant protein.

本文的另一方面提供一种在宿主细胞中产生目标重组蛋白的方法,所述方法包括:Another aspect of the present invention provides a method for producing a target recombinant protein in a host cell, the method comprising:

(a)提供包含内源FUT8基因的细胞;(a) providing a cell comprising an endogenous FUT8 gene;

(b)通过如本文所述的产生FUT8缺陷型细胞的方法使所述细胞中的内源FUT8基因失活;和(b) inactivating the endogenous FUT8 gene in the cell by the method for producing a FUT8-deficient cell as described herein; and

(c)将包含编码所述目标重组蛋白的核酸的表达载体引入所述细胞,由此产生所述目标重组蛋白。(c) introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell, thereby producing the target recombinant protein.

本文的另一方面提供本文所述的ZFP、ZFN、多核苷酸、载体用于产生FUT8缺陷型细胞的用途。Another aspect herein provides the use of the ZFPs, ZFNs, polynucleotides, vectors described herein for generating FUT8-deficient cells.

本文的另一方面提供本文所述的ZFP、ZFN、多核苷酸、载体、细胞或细胞系用于产生去岩藻糖基化的含Fc重组蛋白的用途。Another aspect herein provides use of a ZFP, ZFN, polynucleotide, vector, cell or cell line described herein for producing a defucosylated Fc-containing recombinant protein.

本文的另一方面提供本文所述的ZFP、ZFN、多核苷酸、载体、细胞或细胞系用于产生能引发增强ADCC效应的抗体(如单克隆抗体)的用途。Another aspect provided herein is the use of a ZFP, ZFN, polynucleotide, vector, cell or cell line described herein for producing an antibody (eg, a monoclonal antibody) capable of eliciting an enhanced ADCC effect.

本文的另一方面提供一种FUT8缺陷型细胞系,其根据本文所述的产生FUT8缺陷型细胞系的方法制备。Another aspect herein provides a FUT8-deficient cell line prepared according to the method for generating a FUT8-deficient cell line described herein.

本文的另一方面提供一种FUT8缺陷型细胞系,其在FUT8基因座处包含如SEQ ID NO:52、SEQ ID NO:53、SEQ ID NO:54、SEQ ID NO:55、SEQ ID NO:56、SEQ ID NO:57、SEQ ID NO:58和SEQ ID NO:59中一或多者所示的核苷酸序列。On the other hand, the present invention provides a FUT8-deficient cell line, which comprises a nucleotide sequence as shown in one or more of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58 and SEQ ID NO: 59 at the FUT8 locus.

本文的另一方面提供一种FUT8缺陷型细胞系,其包含经改造的FUT8基因座,所述经改造的FUT8基因座能通过如下引物被检测:Another aspect of the present invention provides a FUT8-deficient cell line comprising an altered FUT8 locus, wherein the altered FUT8 locus can be detected by the following primers:

引物对(i):Primer pair (i):

正向引物:AGCCTGAAGTACATAGCCGA(SEQ ID NO:46);和Forward primer: AGCCTGAAGTACATAGCCGA (SEQ ID NO: 46); and

反向引物:TGCCACTGCTTCTATATACTGATTC(SEQ ID NO:47);Reverse primer: TGCCACTGCTTCTATATACTGATTC (SEQ ID NO: 47);

和/或and/or

引物对(ii):Primer pair (ii):

正向引物:GACGCACTGACAAAGTGGGA(SEQ ID NO:48);和Forward primer: GACGCACTGACAAAGTGGGA (SEQ ID NO: 48); and

反向引物:GGTCTGTTCCATCCCCAGAATG(SEQ ID NO:49);Reverse primer: GGTCTGTTCCATCCCCAGAATG (SEQ ID NO: 49);

和/或and/or

引物对(iii):Primer pair (iii):

正向引物:CTGTTGATTCCAGGTTCCCA(SEQ ID NO:50);和Forward primer: CTGTTGATTCCAGGTTCCCA (SEQ ID NO: 50); and

反向引物:TGTTACTTAAGCCCCAGGC(SEQ ID NO:51)。Reverse primer: TGTTACTTAAGCCCCAGGC (SEQ ID NO: 51).

本文的另一方面提供一种培育FUT8缺陷型细胞系的方法,其包括:Another aspect of the present invention provides a method for cultivating a FUT8-deficient cell line, comprising:

(a)提供根据权利要求15-17中任一项所述的FUT8缺陷型细胞系;和(a) providing a FUT8-deficient cell line according to any one of claims 15 to 17; and

(b)培养或扩增所述细胞系的细胞。(b) culturing or expanding cells of said cell line.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

下面结合附图对本发明作进一步说明,其中这些显示仅为了图示说明本发明的实施方式,而不是为了局限本发明的范围。The present invention will be further described below in conjunction with the accompanying drawings, wherein these drawings are only for illustrating the embodiments of the present invention rather than for limiting the scope of the present invention.

图1显示根据本文的示例性实施方式之一的针对FUT8外显子3和外显子10及其附近区域的锌指蛋白核酸酶的结合位点、切割位点,及PCR引物位点。FIG. 1 shows the binding site, cleavage site, and PCR primer sites of zinc finger nuclease targeting FUT8 exon 3 and exon 10 and their vicinity according to one of the exemplary embodiments herein.

图2A显示根据本文的示例性实施方式之一的锌指蛋白核酸酶的表达质粒图谱示意图。其中,锌指蛋白和限制性内切酶FokI切割半域被融合并克隆在质粒中。该融合蛋白在CMV启动子控制下。图2B显示的是根据本文的示例性实施方式之一的锌指蛋白核酸酶的具体结构示意图,其中各个锌指结合域按顺序插入在天然锌指蛋白的框架(Framework)序列之间,并在C端和FokI切割半域连接。Figure 2A shows a schematic diagram of the expression plasmid of the zinc finger protein nuclease according to one of the exemplary embodiments of the present invention. In which the zinc finger protein and the restriction endonuclease FokI cleavage half domain are fused and cloned in the plasmid. The fusion protein is under the control of the CMV promoter. Figure 2B shows a schematic diagram of the specific structure of the zinc finger protein nuclease according to one of the exemplary embodiments of the present invention, in which each zinc finger binding domain is sequentially inserted between the framework sequence of the natural zinc finger protein and connected to the FokI cleavage half domain at the C-terminus.

图3显示T7E1酶切实验验证ZFN质粒是否有效切除目的基因序列。泳道1:S2-L和S4-R转染,根据ImageJ测算的条带亮度计算的编辑效率为4.7%;泳道2:L3和R3转染,编辑效率为6.2%;NC:阴性对照组,即未敲除FUT8基因的细胞基因组DNA;PC:阳性对照组,即含有50%比例的已敲除FUT8基因的细胞基因组。Figure 3 shows the T7E1 digestion experiment to verify whether the ZFN plasmid can effectively remove the target gene sequence. Lane 1: S2-L and S4-R transfection, the editing efficiency calculated according to the band brightness measured by ImageJ is 4.7%; Lane 2: L3 and R3 transfection, the editing efficiency is 6.2%; NC: negative control group, that is, the cell genomic DNA without knockout of the FUT8 gene; PC: positive control group, that is, the cell genome containing 50% of the knockout FUT8 gene.

图4显示采用流式细胞仪分析经绿色荧光标记小扁豆凝集素(LCA-FITC)染色后的细胞池的结果。(A)不同亚细胞群的定量分析,其中,被圈出的绿色荧光更低的细胞群即为FUT8-/-细胞群,在该细胞池中所占比例为27.44%。(B)不同亚细胞群的峰位移。Figure 4 shows the results of flow cytometric analysis of cell pools stained with green fluorescent marker lentil agglutinin (LCA-FITC). (A) Quantitative analysis of different sub-cell populations, where the circled cell population with lower green fluorescence is the FUT8 -/- cell population, which accounts for 27.44% of the cell pool. (B) Peak shifts of different sub-cell populations.

图5显示克隆154-F8ZFN-09-08和阴性对照经过LCA-FITC染色后的流式分析结果。实线:154-F8ZFN-09-08细胞;虚线:未敲除FUT8基因的阴性对照。Figure 5 shows the flow cytometry analysis results of clone 154-F8ZFN-09-08 and negative control after LCA-FITC staining. Solid line: 154-F8ZFN-09-08 cells; dotted line: negative control without knockout of FUT8 gene.

图6显示NGS验证结果,证明克隆154-F8ZFN-09-08和154-F8ZFN-10-02的一对等位基因被敲除。FIG6 shows the NGS validation results, demonstrating that a pair of alleles of clones 154-F8ZFN-09-08 and 154-F8ZFN-10-02 were knocked out.

图7显示FUT8-/-细胞株(即154-F8ZFN-09-08和154-F8ZFN-10-02)在单抗表达筛选中的应用。NC:FUT8基因未经敲除的阴性对照。(A)细胞池恢复后进行FB培养期间活率变化;(B)细胞池FB培养期间活细胞密度VCD变化;(C)流加式培养过程中第14天蛋白表达水平效价;(D)收获的上清一步ProA纯化后进行分子排阻层析分析SEC-HPLC;(E)一步纯化样品进行SDS Caliper NR及SDS Caliper R;(F)一步纯化样品糖型检测结果。Figure 7 shows the application of FUT8 -/- cell lines (i.e., 154-F8ZFN-09-08 and 154-F8ZFN-10-02) in the screening of monoclonal antibody expression. NC: Negative control in which the FUT8 gene was not knocked out. (A) Changes in viability during FB culture after cell pool recovery; (B) Changes in viable cell density VCD during FB culture of cell pool; (C) Protein expression level titer on day 14 during fed-batch culture; (D) The harvested supernatant was purified by one-step ProA and then analyzed by size exclusion chromatography SEC-HPLC; (E) One-step purified samples were subjected to SDS Caliper NR and SDS Caliper R; (F) Glycoform detection results of one-step purified samples.

图8显示FUT8-/-细胞株(154-F8ZFN-09-08)在抗体生产中的性能评估结果。(A)在FB培养过程中,154-F8ZFN-09-08组的细胞活力;(B)活细胞密度(VCD)变化;(C)蛋白质表达效价;(D)SEC-HPLC与SDS Caliper NR分析结果;(E)糖型分析结果。Figure 8 shows the performance evaluation results of the FUT8-/- cell line (154-F8ZFN-09-08) in antibody production. (A) Cell viability of the 154-F8ZFN-09-08 group during FB culture; (B) Changes in viable cell density (VCD); (C) Protein expression titer; (D) SEC-HPLC and SDS Caliper NR analysis results; (E) Glycoform analysis results.

图9显示基因编辑效率检测结果。泳道1:31-L1/31-R1敲除样品,编辑效率为9.4%;泳道2:32-L1/32-R1敲除样品,编辑效率为10.9%;NC:野生型CHO-K1。Figure 9 shows the results of gene editing efficiency detection. Lane 1: 31-L1/31-R1 knockout sample, editing efficiency is 9.4%; Lane 2: 32-L1/32-R1 knockout sample, editing efficiency is 10.9%; NC: wild-type CHO-K1.

图10显示SCP单克隆分选结果。未染色样品为CHO K1;Fut8-/-克隆C6为分选单克隆后进行LCA-FITC染色后挑选的单克隆;Fut8 wt为CHO K1进行LCA-FITC染色。Figure 10 shows the results of SCP monoclonal sorting. The unstained sample is CHO K1; Fut8 -/- clone C6 is the monoclonal clone selected after LCA-FITC staining after sorting monoclonal clones; Fut8 wt is CHO K1 stained with LCA-FITC.

图11显示SCP单克隆分选结果。未染色样品为CHO K1-N079;Fut8-/-克隆F2为分选后进行LCA-FITC染色后挑选的单克隆;Fut8 wt为CHO K1进行LCA-FITC染色。Figure 11 shows the results of SCP monoclonal sorting. The unstained sample is CHO K1-N079; Fut8 -/- clone F2 is a monoclonal selected after LCA-FITC staining after sorting; Fut8 wt is CHO K1 stained with LCA-FITC.

图12显示NGS验证结果,证明克隆C6和F2的一对等位基因被敲除。FIG12 shows the NGS validation results, demonstrating that a pair of alleles in clones C6 and F2 were knocked out.

图13显示敲除Fut8基因后的宿主细胞生产的阿达木单抗的ADCC效应比野生型的对照显著增强。FIG. 13 shows that the ADCC effect of adalimumab produced by host cells after knocking out the Fut8 gene is significantly enhanced compared with the wild-type control.

具体实施方式DETAILED DESCRIPTION

本申请中科技术语的含意与本领域技术人员的普遍理解一致,除非另做说明。本申请中,“一”或其与各种量词的组合既包括单数含意也包括复数含意,除非特别说明。本申请中,对于同一参数或变量,当给出多个数值、数值范围、或其组合予以说明时,相当于具体揭示了这些数值、范围端值以及由它们任意组合而成的数值范围。本申请中,任一数值不论是否带有“约”之类的修饰词,一律涵盖本领域技术人员能够理解的约略范围,例如正负10%、5%等。本文中,每一“实施方式”均同等地指代且涵盖了本申请各项方法和各项系统的实施方式。本申请中,任意实施方式中一项或多项技术特征可以与任何一个或多个其它实施方式中的一项或多项技术特征自由组合,由此得到的实施方式同样属于本申请公开的内容。The meaning of scientific and technological terms in this application is consistent with the general understanding of those skilled in the art, unless otherwise specified. In this application, "one" or its combination with various quantifiers includes both singular and plural meanings, unless otherwise specified. In this application, for the same parameter or variable, when multiple numerical values, numerical ranges, or combinations thereof are given for description, it is equivalent to specifically revealing these numerical values, range ends, and numerical ranges formed by any combination thereof. In this application, any numerical value, whether or not it carries a modifier such as "about", covers an approximate range that can be understood by those skilled in the art, such as plus or minus 10%, 5%, etc. In this article, each "implementation method" refers to and covers the implementation methods of various methods and systems of this application equally. In this application, one or more technical features in any implementation method can be freely combined with one or more technical features in any one or more other implementation methods, and the implementation methods obtained thereby also belong to the content disclosed in this application.

在哺乳动物细胞中,Fut8将核心岩藻糖附着于存在于抗体Fc区上的寡糖上,其一般被认为对抗体依赖性细胞的细胞毒性的效应功能非常重要。人类Fut8结构的三维分析显示3个α2/α6岩藻糖基转移酶基序形成酶的催化核心。参见,Ihara等(2007)Glycobiology 17:455-66。该区域中,许多单个残基点突变为丙氨酸导致酶的完全失活。参见,Ihara等(2007)Glycobiology 17:455-66;Takahashi等(2000)Glycobiology 10:503-10。已有研究表明,其中FUT8的表达降低或消除的细胞(例如敲除细胞系或使用siRNA)能产生具有更强效应功能的非岩藻糖基化的抗体。参见,例如,Kanada等(2007)Biotechnol.130(3):300-310;Kanada等(2007)Glycobiology 18:104-118;Mori等(2004)Biotechnol.Bioeng.88:901-908。In mammalian cells, Fut8 attaches core fucose to oligosaccharides present on the Fc region of antibodies, which is generally considered to be important for the effector function of antibody-dependent cellular cytotoxicity. Three-dimensional analysis of the structure of human Fut8 revealed that three α2/α6 fucosyltransferase motifs form the catalytic core of the enzyme. See, Ihara et al. (2007) Glycobiology 17:455-66. In this region, many single residue point mutations to alanine result in complete inactivation of the enzyme. See, Ihara et al. (2007) Glycobiology 17:455-66; Takahashi et al. (2000) Glycobiology 10:503-10. It has been shown that cells in which the expression of FUT8 is reduced or eliminated (e.g., knockout cell lines or using siRNA) can produce non-fucosylated antibodies with enhanced effector function. See, for example, Kanada et al. (2007) Biotechnol. 130(3): 300-310; Kanada et al. (2007) Glycobiology 18: 104-118; Mori et al. (2004) Biotechnol. Bioeng. 88: 901-908.

本文提供了一种构建去岩藻糖基化蛋白质生产细胞的方法,该方法利用靶向FUT8基因的锌指核酸酶使细胞中的FUT8基因失活。所述锌指蛋白核酸酶(ZFN)包含锌指蛋白(ZFP)和核酸酶切割域或切割伴域。发明人惊奇地发现,利用根据本文所述特别设计的锌指核酸酶,能够高度有效地敲除细胞中的FUT8基因,所得细胞产生具有更强效应功能的去岩藻糖基化蛋白质(例如,抗体)。The present invention provides a method for constructing a defucosylated protein production cell, which inactivates the FUT8 gene in the cell by using a zinc finger nuclease targeting the FUT8 gene. The zinc finger protein nuclease (ZFN) comprises a zinc finger protein (ZFP) and a nuclease cleavage domain or a cleavage partner domain. The inventors surprisingly found that the FUT8 gene in the cell can be highly effectively knocked out using the zinc finger nuclease specially designed as described herein, and the resulting cell produces a defucosylated protein (e.g., antibody) with stronger effector function.

A.锌指蛋白A. Zinc finger proteins

一方面,本文提供一种能够靶向结合FUT8基因的锌指蛋白(ZFP)。In one aspect, the present invention provides a zinc finger protein (ZFP) capable of targeting and binding to the FUT8 gene.

本文中,术语“锌指蛋白”或“ZFP”指这样的蛋白质,其通过一个或多个锌指(或锌指识别区)以序列特异性的方式结合DNA。所述锌指是所述蛋白质内的结构由锌离子的配位而稳定的氨基酸序列区。As used herein, the term "zinc finger protein" or "ZFP" refers to a protein that binds DNA in a sequence-specific manner via one or more zinc fingers (or zinc finger recognition regions). The zinc fingers are amino acid sequence regions within the protein whose structure is stabilized by the coordination of zinc ions.

本文所述的ZFP是非天然存在的ZFP。所述ZFP可具有1、2、3、4、5、6个或更多个锌指,其各自具有能够结合FUT8基因中靶位点的锌指识别区。在一些实施方式中,所述ZFP包含五或六个锌指。The ZFP described herein is a non-naturally occurring ZFP. The ZFP may have 1, 2, 3, 4, 5, 6 or more zinc fingers, each of which has a zinc finger recognition region capable of binding to a target site in the FUT8 gene. In some embodiments, the ZFP comprises five or six zinc fingers.

本文中,术语“靶位点”指的是本文所述的ZFP或ZFN将要结合的靶基因(例如FUT8基因)的DNA的部分的核苷酸序列。Herein, the term "target site" refers to a nucleotide sequence of a portion of the DNA of a target gene (eg, FUT8 gene) to which the ZFP or ZFN described herein is to bind.

在一些实施方式中,靶位点可包括FUT8基因的外显子3(相关序列示于SEQ ID NO:1)。在一些实施方式中,靶位点可包括FUT8基因的外显子10及其附近区域(相关序列示于SEQ ID NO:2)。In some embodiments, the target site may include exon 3 of the FUT8 gene (the relevant sequence is shown in SEQ ID NO: 1). In some embodiments, the target site may include exon 10 of the FUT8 gene and its vicinity (the relevant sequence is shown in SEQ ID NO: 2).

本文中,ZFP中的锌指以“F+自N至C端顺序数字编号”方式命名。例如,就某ZFP而言,以白N至C端顺序,第一锌指命名为“F1”、第二锌指命名为“F2”,以此类推。Herein, the zinc fingers in ZFP are named in the form of "F+sequential numbers from N to C". For example, for a certain ZFP, in the order from N to C, the first zinc finger is named "F1", the second zinc finger is named "F2", and so on.

在一些实施方式中,所述ZFP的一个或多个锌指具有工程化锌指识别区。在一些实施方式中,所述ZFP具有表1中同行所示锌指(F1至F5或至F6)的识别区氨基酸序列:In some embodiments, one or more zinc fingers of the ZFP have an engineered zinc finger recognition region. In some embodiments, the ZFP has the recognition region amino acid sequence of the zinc finger (F1 to F5 or to F6) shown in the same row in Table 1:

表1.靶向FUT8基因的锌指蛋白

Table 1. Zinc finger proteins targeting FUT8 gene

表1中,各行分别描述了单一ZFP。各ZFP的名称和DNA靶序列(以5’-3’顺序)显示于第1列,第2至6或7列显示了各ZFP中锌指(F1至F5或至F6)的识别区氨基酸序列。In Table 1, each row describes a single ZFP. The name and DNA target sequence (in 5'-3' order) of each ZFP are shown in the first column, and the amino acid sequence of the recognition region of the zinc finger (F1 to F5 or to F6) in each ZFP is shown in columns 2 to 6 or 7.

在一些实施方式中,所述ZFP的多个锌指可由适合的连接体序列连接。在一些实施方式中,所述连接体序列可包括长度为1、2、3、4、5个或更多个氨基酸的连接体。In some embodiments, the multiple zinc fingers of the ZFP can be connected by a suitable linker sequence. In some embodiments, the linker sequence can include a linker of 1, 2, 3, 4, 5 or more amino acids in length.

进一步地,本文还提供ZFP对,其包括左ZFP和右ZFP。所述ZFP对中的左ZFP的靶位点与右ZFP的靶位点位于靶基因DNA序列上相近位置处(例如,相隔5至10个核苷酸碱基对)的相对链上。例如,所述ZFP对中的左ZFP能够结合至靶基因DNA序列的一条链(例如,模板链或编码链)上的靶位点,而右ZFP能够结合至该靶基因DNA序列的相反链(例如,编码链或模板链)上位于左ZFP靶位点附近(例如,相隔5至20个核苷酸碱基对)处的靶位点。在一些实施方式中,左ZFP的靶位点和右ZFP的靶位点之间相隔5至20个核苷酸碱基对,例如,相隔5、6、7、8、9、10、11、12、13、14、15、16、17、18、19、20个核苷酸碱基对。Further, ZFP pairs are also provided herein, which include left ZFP and right ZFP. The target site of the left ZFP in the ZFP pair is located on the relative strand of the target gene DNA sequence at a close position (e.g., 5 to 10 nucleotide base pairs apart). For example, the left ZFP in the ZFP pair can bind to a target site on a strand (e.g., template strand or coding strand) of the target gene DNA sequence, and the right ZFP can bind to a target site located near the left ZFP target site (e.g., 5 to 20 nucleotide base pairs apart) on the opposite strand (e.g., coding strand or template strand) of the target gene DNA sequence. In some embodiments, the target site of the left ZFP and the target site of the right ZFP are separated by 5 to 20 nucleotide base pairs, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 nucleotide base pairs.

在一些实施方式中,左ZFP可具有以下识别区氨基酸序列(ZFP 32-L1):In some embodiments, the left ZFP can have the following recognition region amino acid sequence (ZFP 32-L1):

F1:QLATLNR(SEQ ID NO:13);F1: QLATLNR (SEQ ID NO: 13);

F2:TRWNLRA(SEQ ID NO:14);F2: TRWNLRA (SEQ ID NO: 14);

F3:SRRDLRR(SEQ ID NO:15);F3: SRRDLRR (SEQ ID NO: 15);

F4:WRRRLLS(SEQ ID NO:16);F4: WRRRLLS(SEQ ID NO: 16);

F5:RSDYLTN(SEQ ID NO:17),和F5:RSDYLTN(SEQ ID NO:17), and

F6:FHSNLLA(SEQ ID NO:18)。F6: FHSNLLA (SEQ ID NO: 18).

在一些实施方式中,左ZFP可具有以下识别区氨基酸序列(ZFP L3):In some embodiments, the left ZFP can have the following recognition region amino acid sequence (ZFP L3):

F1:SKWNLRS(SEQ ID NO:24);F1:SKWNLRS(SEQ ID NO:24);

F2:AQSNLLS(SEQ ID NO:25);F2: AQSNLLS(SEQ ID NO: 25);

F3:LRHQLRR(SEQ ID NO:26);F3: LRHQLRR (SEQ ID NO: 26);

F4:RSDYLTN(SEQ ID NO:17);F4: RSDYLTN (SEQ ID NO: 17);

F5:RSDYLTN(SEQ ID NO:17)。F5: RSDYLTN (SEQ ID NO: 17).

在一些实施方式中,左ZFP可具有以下识别区氨基酸序列(ZFP S2-L):In some embodiments, the left ZFP can have the following recognition region amino acid sequence (ZFP S2-L):

F1:RADNLTE(SEQ ID NO:30);F1: RADNLTE(SEQ ID NO: 30);

F2:TSGNLTE(SEQ ID NO:31);F2: TSGNLTE (SEQ ID NO: 31);

F3:TSGHLVR(SEQ ID NO:32);F3: TSGHLVR (SEQ ID NO: 32);

F4:RKDNLKN(SEQ ID NO:33);和F4: RKDNLKN (SEQ ID NO: 33); and

F5:RKDNLKN(SEQ ID NO:33)。F5: RKDNLKN (SEQ ID NO: 33).

在一些实施方式中,左ZFP可具有以下识别区氨基酸序列(ZFP 31-L1):In some embodiments, the left ZFP can have the following recognition region amino acid sequence (ZFP 31-L1):

F1:DRSNLLS(SEQ ID NO:39);F1: DRSNLLS(SEQ ID NO: 39);

F2:NQSNLLR(SEQ ID NO:40);F2: NQSNLLR (SEQ ID NO: 40);

F3:FHSNLLA(SEQ ID NO:18);F3: FHSNLLA (SEQ ID NO: 18);

F4:QLSTLNY(SEQ ID NO:41);和F4: QLSTLNY(SEQ ID NO: 41); and

F5:QSGNLSR(SEQ ID NO:42)。F5: QSGNLSR (SEQ ID NO: 42).

在一些实施方式中,右ZFP可具有以下识别区氨基酸序列(ZFP 32-R1):In some embodiments, the right ZFP can have the following recognition region amino acid sequence (ZFP 32-R1):

F1:RKSHLTM(SEQ ID NO:19);F1: RKSHLTM (SEQ ID NO: 19);

F2:FHSGLLA(SEQ ID NO:20);F2: FHSGLLA (SEQ ID NO: 20);

F3:WRRRLLS(SEQ ID NO:16);F3: WRRRLLS(SEQ ID NO: 16);

F4:RKYVLLR(SEQ ID NO:21);F4: RKYVLLR (SEQ ID NO: 21);

F5:RKDYLVL(SEQ ID NO:22);和F5: RKDYLVL (SEQ ID NO: 22); and

F6:QQAGLIN(SEQ ID NO:23)。F6: QQAGLIN (SEQ ID NO: 23).

在一些实施方式中,右ZFP可具有以下识别区氨基酸序列(ZFP R3):In some embodiments, the right ZFP can have the following recognition region amino acid sequence (ZFP R3):

RSDYLTN(SEQ ID NO:17);RSDYLTN (SEQ ID NO: 17);

QKITLVR(SEQ ID NO:27);QKITLVR (SEQ ID NO: 27);

RSDYLTN(SEQ ID NO:17);RSDYLTN (SEQ ID NO: 17);

QLATLNR(SEQ ID NO:13);QLATLNR (SEQ ID NO: 13);

SRFNLTR(SEQ ID NO:28);和SRFNLTR (SEQ ID NO: 28); and

TKYILTN(SEQ ID NO:29)。TKYILTN (SEQ ID NO: 29).

在一些实施方式中,右ZFP可具有以下识别区氨基酸序列(ZFP S4-R):In some embodiments, the right ZFP can have the following recognition region amino acid sequence (ZFP S4-R):

RSDNLSV(SEQ ID NO:34);RSDNLSV (SEQ ID NO: 34);

SPADLTR(SEQ ID NO:35);SPADLTR (SEQ ID NO: 35);

RSDHLSQ(SEQ ID NO:36);RSDHLSQ (SEQ ID NO: 36);

QSGDLRR(SEQ ID NO:37);QSGDLRR (SEQ ID NO: 37);

RSDNLVR(SEQ ID NO:38);和RSDNLVR (SEQ ID NO: 38); and

RKDNLKN(SEQ ID NO:33)。RKDNLKN (SEQ ID NO: 33).

在一些实施方式中,右ZFP可具有以下识别区氨基酸序列(ZFP 31-R1):In some embodiments, the right ZFP can have the following recognition region amino acid sequence (ZFP 31-R1):

FHSGLLA(SEQ ID NO:20);FHSGLLA (SEQ ID NO: 20);

HPSTLSK(SEQ ID NO:43);HPSTLSK (SEQ ID NO: 43);

ARWTLDC(SEQ ID NO:44);ARWTLDC (SEQ ID NO: 44);

HPSTLSK(SEQ ID NO:43);HPSTLSK (SEQ ID NO: 43);

HPSTLSK(SEQ ID NO:43);和HPSTLSK (SEQ ID NO: 43); and

RKFTLTN(SEQ ID NO:45)。RKFTLTN (SEQ ID NO: 45).

在一些实施方式中,ZFP对可由ZFP 32-L1与ZFP 32-R1组成。在一些实施方式中,ZFP对可由ZFP L3与ZFP R3组成。在一些实施方式中,ZFP对可由ZFP S2-L与S4-R组成。在一些实施方式中,ZFP对可由ZFP 31-L1与ZFP 31-R1组成。In some embodiments, the ZFP pair may consist of ZFP 32-L1 and ZFP 32-R1. In some embodiments, the ZFP pair may consist of ZFP L3 and ZFP R3. In some embodiments, the ZFP pair may consist of ZFP S2-L and S4-R. In some embodiments, the ZFP pair may consist of ZFP 31-L1 and ZFP 31-R1.

B.锌指核酸酶B. Zinc Finger Nuclease

另一方面,本文提供一种针对FUT8基因的锌指核酸酶(ZFN),其为融合蛋白,且包含本文所述的ZFP和至少一个DNA切割域或切割半域。In another aspect, the present invention provides a zinc finger nuclease (ZFN) against the FUT8 gene, which is a fusion protein and comprises the ZFP described herein and at least one DNA cleavage domain or cleavage half-domain.

本文中,术语“融合蛋白”指其中两个或更多个亚基分子(例如本文所述的ZFP和DNA切割域或切割半域)彼此连接(优选共价连接)而形成的蛋白质分子。As used herein, the term "fusion protein" refers to a protein molecule in which two or more subunit molecules (eg, a ZFP and a DNA cleavage domain or cleavage half-domain as described herein) are linked to each other (preferably covalently linked).

本文中,术语“切割”指DNA分子共价骨架的断裂。切割可由各种方法来起始,这些方法包括但不限于磷酸二酯键的酶促或化学水解。在一些实施方式中,切割可以是单链切割或双链切割。在一些实施方式中,双链切割可由两个不同的单链切割事件所致。DNA的切割可导致平末端或交错末端的产生。在某些实施方式中,所述ZFN用于靶向双链DNA切割。As used herein, the term "cleavage" refers to the breakage of the covalent backbone of a DNA molecule. Cutting can be initiated by various methods, including but not limited to enzymatic or chemical hydrolysis of phosphodiester bonds. In some embodiments, cutting can be single-stranded cutting or double-stranded cutting. In some embodiments, double-stranded cutting can be caused by two different single-stranded cutting events. Cutting of DNA can result in the generation of flat ends or staggered ends. In certain embodiments, the ZFN is used to target double-stranded DNA cutting.

本文中,术语“切割半结构域”是能与(相同或不同的)另一多肽轭合形成具有切割活性(优选双链切割活性)的复合体的多肽序列。在一些实施方式中,两个切割半结构域通常以二聚化形式(成对)发挥切割作用。As used herein, the term "cleavage half-domain" refers to a polypeptide sequence that can be conjugated with another polypeptide (the same or different) to form a complex having cleavage activity (preferably double-chain cleavage activity). In some embodiments, two cleavage half-domains typically function as a dimer (in pairs) to exert cleavage activity.

在一些实施方式中,ZFP和至少一个DNA切割域或切割半域操作性地连接。In some embodiments, the ZFP and at least one DNA cleavage domain or cleavage half-domain are operably linked.

本文中,术语“操作性地连接”可指某组分与其它组分以有功能的方式彼此连接。例如,就其中ZFP与切割域融合的融合蛋白而言,如果融合蛋白中ZFP能结合其靶位点和/或其结合位点,而切割域能在靶位点附近切割DNA,那么该ZFP与切割域为操作性连接关系。As used herein, the term "operably linked" may refer to a component being linked to another component in a functional manner. For example, in the case of a fusion protein in which a ZFP is fused to a cleavage domain, if the ZFP in the fusion protein can bind to its target site and/or its binding site, and the cleavage domain can cleave DNA near the target site, then the ZFP and the cleavage domain are in an operably linked relationship.

本文所述的锌指核酸酶可用作靶向切割并使靶基因(如FUT8基因)失活的工具,其通过在靶基因的靶位点处引入双链DNA断裂(DSB),并利用非同源末端连接(NHEJ)介导的DSB修复可达到靶基因定向敲除的目的。The zinc finger nuclease described in this article can be used as a tool for targeted cutting and inactivation of a target gene (such as the FUT8 gene), which can achieve the purpose of targeted knockout of the target gene by introducing a double-stranded DNA break (DSB) at the target site of the target gene and utilizing non-homologous end joining (NHEJ)-mediated DSB repair.

进一步地,本文还提供ZFN对,其包括左ZFN和右ZFN。所述左ZFN和右ZFN各自由本文所述的ZFP与至少一个DNA切割域或切割半域融合而成。例如,ZFN 32-L1为包含ZFP 32-L1与至少一个DNA切割域或切割半域的融合蛋白。Further, the present invention also provides a ZFN pair, which includes a left ZFN and a right ZFN. The left ZFN and the right ZFN are each fused with the ZFP described herein and at least one DNA cleavage domain or cleavage half-domain. For example, ZFN 32-L1 is a fusion protein comprising ZFP 32-L1 and at least one DNA cleavage domain or cleavage half-domain.

所述ZFN对中的左ZFN的靶位点与右ZFN的靶位点位于靶基因DNA序列上相近位置处(例如,相隔5至10个核苷酸碱基对)的相对链上。例如,所述ZFN对中的左ZFN能够结合至靶基因DNA序列的一条链(例如,模板链或编码链)上的靶位点,而右ZFN能够结合至该靶基因DNA序列的相反链(例如,编码链或模板链)上位于左ZFN靶位点附近(例如,相隔5至20个核苷酸碱基对)处的靶位点。在一些实施方式中,左ZFN的靶位点和右ZFN的靶位点之间相隔5至20个核苷酸碱基对,例如,相隔5、6、7、8、9、10、11、12、13、14、15、16、17、18、19、20个核苷酸碱基对。The target site of the left ZFN in the ZFN pair and the target site of the right ZFN are located on opposite strands at a close position (e.g., 5 to 10 nucleotide base pairs apart) on the target gene DNA sequence. For example, the left ZFN in the ZFN pair can bind to a target site on one strand (e.g., template strand or coding strand) of the target gene DNA sequence, while the right ZFN can bind to a target site on the opposite strand (e.g., coding strand or template strand) of the target gene DNA sequence that is located near the left ZFN target site (e.g., 5 to 20 nucleotide base pairs apart). In some embodiments, the target site of the left ZFN and the target site of the right ZFN are separated by 5 to 20 nucleotide base pairs, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 nucleotide base pairs.

在一些实施方式中,ZFN对可由ZFN 32-L1与ZFN 32-R1组成。在一些实施方式中,ZFN对可由ZFN L3与ZFN R3组成。在一些实施方式中,ZFN对可由ZFN S2-L与S4-R组成。在一些实施方式中,ZFN对可由ZFN 31-L1与ZFN 31-R1组成。In some embodiments, the ZFN pair may consist of ZFN 32-L1 and ZFN 32-R1. In some embodiments, the ZFN pair may consist of ZFN L3 and ZFN R3. In some embodiments, the ZFN pair may consist of ZFN S2-L and S4-R. In some embodiments, the ZFN pair may consist of ZFN 31-L1 and ZFN 31-R1.

本文公开的融合蛋白的切割域部分可获自任何内切核酸酶或外切核酸酶。可衍生切割域的示例性内切核酸酶包括但不限于限制性内切核酸酶和归巢内切核酸酶。参见,例如,新英格兰生物实验室公司(New England Biolabs)2002-2003目录,马萨诸塞州贝弗利;和Belfort等(1997)Nucleic Acids Res.25:3379-3388。能够切割DNA的其它酶是已知的(例如,S1核酸酶;绿豆核酸酶;胰腺DNA酶I;微球菌核酸酶;酵母HO内切核酸酶;还参见Linn等(编)《核酸酶》(Nucleases),冷全港出版社,1993)。这些酶(或其功能性片段)的一种或多种可用作切割域和切割半域的来源。The cleavage domain portion of the fusion protein disclosed herein can be obtained from any endonuclease or exonuclease. Exemplary endonucleases from which the cleavage domain can be derived include, but are not limited to, restriction endonucleases and homing endonucleases. See, e.g., New England Biolabs 2002-2003 Catalog, Beverly, Massachusetts; and Belfort et al. (1997) Nucleic Acids Res. 25: 3379-3388. Other enzymes capable of cleaving DNA are known (e.g., S1 nuclease; mung bean nuclease; pancreatic DNase I; micrococcal nuclease; yeast HO endonuclease; see also Linn et al. (eds.) Nucleases (Nucleases), Cold Spring Harbor Press, 1993). One or more of these enzymes (or functional fragments thereof) can be used as a source of cleavage domains and cleavage half-domains.

相似地,切割半域可衍生自如上文提到其切割活性需要二聚化的任何核酸酶或其片段。一般而言,如果融合蛋白包含切割半域,则需要两个融合蛋白来执行切割。任选地,可使用包含两个切割半域的单个蛋白质。两个切割半域可衍生自相同的内切核酸酶(或其功能性片段),或者各切割半域可衍生自不同的内切核酸酶(或其功能性片段)。另外,两个融合蛋白的靶位点设置为使得这两个融合蛋白与其各自靶位点的结合将切割半域置于允许切割半域(例如通过二聚化)形成功能性切割域的彼此的空间定向。因此,在某些实施方式中,靶位点可相隔5-20个或更多个核苷酸碱基对。一般而言,切割位点位于靶位点之间。Similarly, the cleavage half-domains may be derived from any nuclease or fragment thereof whose cleavage activity requires dimerization as mentioned above. In general, if a fusion protein comprises a cleavage half-domain, two fusion proteins are required to perform cleavage. Optionally, a single protein comprising two cleavage half-domains may be used. The two cleavage half-domains may be derived from the same endonuclease (or a functional fragment thereof), or each cleavage half-domain may be derived from a different endonuclease (or a functional fragment thereof). In addition, the target sites of the two fusion proteins are arranged so that the binding of the two fusion proteins to their respective target sites places the cleavage half-domains in a spatial orientation relative to each other that allows the cleavage half-domains to form a functional cleavage domain (e.g., by dimerization). Thus, in certain embodiments, the target sites may be separated by 5-20 or more nucleotide base pairs. In general, the cleavage site is located between the target sites.

限制性内切核酸酶(限制酶)存在于许多物种,并且能序列特异地结合DNA(在识别位点),并在结合位点或接近结合位点处切割DNA。某些限制酶(例如,IIS型)在远离识别位点的位点处切割DNA,并具有可分的结合结构域和切割域。示例性的IIS型限制性内切酶描述于国际公布WO 07/014275,其全文通过引用方式纳入本文。因此,在一个实施方式中,所述融合蛋白包含所述锌指蛋白和来自至少一个IIS型限制酶的切割域(或切割半域)。Restriction endonucleases (restriction enzymes) are present in many species and can sequence-specifically bind to DNA (at a recognition site) and cleave the DNA at or near the binding site. Certain restriction enzymes (e.g., Type IIS) cleave DNA at a site distal to the recognition site and have separable binding domains and cleavage domains. Exemplary Type IIS restriction endonucleases are described in International Publication WO 07/014275, the entirety of which is incorporated herein by reference. Thus, in one embodiment, the fusion protein comprises the zinc finger protein and a cleavage domain (or cleavage half-domain) from at least one Type IIS restriction enzyme.

切割域或切割半域可以是保留切割活性或保留多聚化(例如二聚化)以形成功能性切割域的能力的蛋白质的任何部分。A cleavage domain or cleavage half-domain can be any portion of a protein that retains cleavage activity or retains the ability to multimerize (e.g., dimerize) to form a functional cleavage domain.

在一些实施方式中,所述IIS型限制酶是FokI。该酶以二聚体形式发挥活性。相应地,在一些实施方式中,融合蛋白中所用的FokI酶的一部分被认为是切割半域。因此,对使用锌指蛋白-FokI融合体的细胞序列的靶向双链切割和/或靶向替代而言,各包含FokI切割半域的两个融合蛋白可用来重构催化上有活性的切割域。任选地,也可使用含有锌指蛋白和两个FokI切割半域的单个多肽分子。在一些实施方式中,切割半域是FokI切割半域。在一些实施方式中,切割半域是野生型FokI切割半结构域。在另一些实施方式中,切割半域是改造的FokI切割半结构域。In some embodiments, the type IIS restriction enzyme is FokI. The enzyme is active as a dimer. Accordingly, in some embodiments, a portion of the FokI enzyme used in the fusion protein is considered to be a cleavage half-domain. Thus, for targeted double-stranded cleavage and/or targeted replacement of cellular sequences using zinc finger protein-FokI fusions, two fusion proteins, each comprising a FokI cleavage half-domain, can be used to reconstitute a catalytically active cleavage domain. Optionally, a single polypeptide molecule containing a zinc finger protein and two FokI cleavage half-domains can also be used. In some embodiments, the cleavage half-domain is a FokI cleavage half-domain. In some embodiments, the cleavage half-domain is a wild-type FokI cleavage half-domain. In other embodiments, the cleavage half-domain is a modified FokI cleavage half-domain.

在一些示例性的实施方式中,所述切割半域来自在野生型FokI基础上通过氨基酸突变而得的Sharkey FokI核酸酶,其示例性氨基酸序列示于SEQ ID NO:3。In some exemplary embodiments, the cleavage half-domain is derived from Sharkey FokI nuclease obtained by amino acid mutation based on wild-type FokI, and its exemplary amino acid sequence is shown in SEQ ID NO: 3.

本文描述的经改造切割半域可使用任何适合的方法来制备,例如,通过位点定向诱变。The engineered cleavage half-domains described herein can be prepared using any suitable method, for example, by site-directed mutagenesis.

本文也考虑包含可分的结合结构域和切割域的其它限制酶。参见例如,Roberts等(2003)Nucleic Acids Res.31:418-420。Other restriction enzymes comprising separable binding and cleavage domains are also contemplated herein. See, e.g., Roberts et al. (2003) Nucleic Acids Res. 31: 418-420.

在一些实施方式中,切割域可包含例如在美国专利公布第20050064474;20060188987和20080131962号(其所有公开内容通过引用整体并入本文)中所述最小化或阻止同源二聚化的一个或多个改造的切割半域(也称为二聚化结构域突变体)。位于FokI的446、447、479、483、484、486、487、490、491、496、498、499、500、531、534、537和538位置的氨基酸残基都是影响FokI切割半域二聚化的靶。In some embodiments, the cleavage domain may comprise one or more engineered cleavage half-domains (also referred to as dimerization domain mutants) that minimize or prevent homodimerization, such as described in U.S. Patent Publication Nos. 20050064474; 20060188987 and 20080131962 (the disclosures of all of which are incorporated herein by reference in their entireties). Amino acid residues located at positions 446, 447, 479, 483, 484, 486, 487, 490, 491, 496, 498, 499, 500, 531, 534, 537, and 538 of FokI are all targets for influencing dimerization of the FokI cleavage half-domain.

在一些实施方式中,形成专性异源二聚体的FokI的示例性经改造切割半域包括其中第一切割半域包括在FokI的490和538位置的氨基酸残基的突变,而第二切割半域包括在氨基酸残基486和499处的突变的对。In some embodiments, exemplary engineered cleavage half-domains of FokI that form obligate heterodimers include pairs wherein a first cleavage half-domain includes mutations in amino acid residues at positions 490 and 538 of FokI and a second cleavage half-domain includes mutations in amino acid residues 486 and 499.

在一个实施方式中,在490处的突变用Lys(K)替代Glu(E);在538处的突变用Lys(K)替代Iso(I);在486处的突变用Glu(E)替代Gln(Q);而在499处的突变用Lys(K)替代Iso(I)。具体地,通过突变一个切割半域中的位置490(E→K)和538(I→K)来产生命名为“E490K:I538K”的经改造切割半域和通过突变另一个切割半域中的位置486(Q→E)和499(I→L)来产生命名为“Q486E:I499L”的经改造切割半域制备本文所述的经改造切割半域。本文所述的经改造切割半域是其中当使用含有这些切割半域的一对或多对核酸酶用于切割时异常切割被最小化或消除的专性异源二聚体突变体。参见,例如,美国专利公布第20080131962号,为所有目的其公开内容通过引用整体并入。In one embodiment, the mutation at 490 replaces Glu (E) with Lys (K); the mutation at 538 replaces Iso (I) with Lys (K); the mutation at 486 replaces Gln (Q) with Glu (E); and the mutation at 499 replaces Iso (I) with Lys (K). Specifically, the engineered cleavage half-domains described herein are prepared by mutating positions 490 (E→K) and 538 (I→K) in one cleavage half-domain to generate an engineered cleavage half-domain designated "E490K:I538K" and by mutating positions 486 (Q→E) and 499 (I→L) in the other cleavage half-domain to generate an engineered cleavage half-domain designated "Q486E:I499L". The engineered cleavage half-domains described herein are obligate heterodimer mutants in which aberrant cleavage is minimized or eliminated when one or more pairs of nucleases containing these cleavage half-domains are used for cleavage. See, for example, US Patent Publication No. 20080131962, the disclosure of which is incorporated by reference in its entirety for all purposes.

C.递送C. Delivery

可将本文所述的ZFN递送至细胞以使细胞中FUT8基因的失活。可通过将编码所述ZFN的多核苷酸引入细胞来实现所述ZFN向细胞的递送,其中该多核苷酸在细胞中被转录,转录物被翻译产生所述ZFN融合蛋白。细胞中蛋白质的表达还可涉及反式剪接、多肽切割和多肽连接。The ZFNs described herein can be delivered to cells to inactivate the FUT8 gene in the cells. The delivery of the ZFNs to cells can be achieved by introducing a polynucleotide encoding the ZFN into the cells, wherein the polynucleotide is transcribed in the cells and the transcript is translated to produce the ZFN fusion protein. The expression of proteins in cells can also involve trans-splicing, polypeptide cleavage, and polypeptide ligation.

本文的又一方面提供一种多核苷酸,其编码本文所述的ZFP或ZFN。在一些实施方式中,所述多核苷酸可以是mRNA。Another aspect of the present invention provides a polynucleotide encoding a ZFP or ZFN as described herein. In some embodiments, the polynucleotide may be mRNA.

本文所述的ZFN可通过多种适合的方法被递送至靶细胞。The ZFNs described herein can be delivered to target cells by a variety of suitable methods.

在一些实施方式中,可采用载体(或载体系统)将所述ZFN递送至靶细胞。In some embodiments, a vector (or vector system) can be used to deliver the ZFN to a target cell.

本文的另一方面提供一种载体,其包含如上所述的多核苷酸。可用的载体包括但不限于以下一种或多种:质粒载体、反转录病毒载体、慢病毒载体、腺病毒载体、腺相关病毒载体、痘病毒载体、疱疹病毒载体等。Another aspect of the present invention provides a vector comprising a polynucleotide as described above. Available vectors include, but are not limited to, one or more of the following: plasmid vectors, retroviral vectors, lentiviral vectors, adenoviral vectors, adeno-associated viral vectors, poxvirus vectors, herpesvirus vectors, and the like.

在一些实施方式中,所述载体可包含编码一种或多种ZFN的多核苷酸。在一些实施方式中,编码一种或多种ZFN的多核苷酸可位于一种(相同)或多种(不同)载体。在采用多种载体的实施方式中,各载体可包含编码一种或多种ZFN的多核苷酸。在采用多种载体的实施方式中,各载体可同时或先后被递送至靶细胞。In some embodiments, the vector may contain a polynucleotide encoding one or more ZFNs. In some embodiments, the polynucleotide encoding one or more ZFNs may be located on one (same) or multiple (different) vectors. In embodiments using multiple vectors, each vector may contain a polynucleotide encoding one or more ZFNs. In embodiments using multiple vectors, each vector may be delivered to the target cell simultaneously or sequentially.

在一些实施方式中,可采用非载体递送方法将所述ZFN递送至靶细胞。可用的非载体递送方法包括但不限于,电穿孔、脂质转染、显微注射、基因枪、脂质体、免疫脂质体、聚阳离子或脂类:核酸轭合物、裸DNA和增强DNA摄取的试剂等。还可使用采用例如Sonitron 2000系统(Rich-Mar)的声孔效应来递送编码所述ZFN的多核苷酸。In some embodiments, the ZFN can be delivered to the target cell using a non-vector delivery method. Available non-vector delivery methods include, but are not limited to, electroporation, lipofection, microinjection, gene guns, liposomes, immunoliposomes, polycations or lipids: nucleic acid conjugates, naked DNA, and agents that enhance DNA uptake. The polynucleotide encoding the ZFN can also be delivered using, for example, the Sonitron 2000 system (Rich-Mar).

本文的另一方面提供一种分离的细胞(例如,宿主细胞),其包含本文所述的ZFP、ZFN、多核苷酸或载体。Another aspect herein provides an isolated cell (eg, a host cell) comprising a ZFP, ZFN, polynucleotide or vector described herein.

适合的细胞包括但不限于真核和原核细胞和/或细胞系。此类细胞或细胞系的非限制性实例包括:COS、CHO(例如,CHO-S、CHO-K1、CHO-DG44、CHO-DUXB11、CHO-DUKX、CHOK1SV)、VERO、MDCK、WI38、V79、B14AF28-G3、BHK、HaK、NS0、SP2/0-Ag14、HeLa、HEK293(例如,HEK293-F、HEK293-H、HEK293-T)和perC6细胞以及诸如草地贪夜蛾(Sf)的昆虫细胞或诸如酿酒酵母、毕赤酵母和裂殖酵母的真菌细胞。在一些实施方式中,所述细胞为哺乳动物细胞。在一个具体实施方式中,所述细胞为中国仓鼠卵巢细胞(CHO细胞)。也可使用这些细胞系的子代、变体和衍生物。Suitable cells include, but are not limited to, eukaryotic and prokaryotic cells and/or cell lines. Non-limiting examples of such cells or cell lines include: COS, CHO (e.g., CHO-S, CHO-K1, CHO-DG44, CHO-DUXB11, CHO-DUKX, CHOK1SV), VERO, MDCK, WI38, V79, B14AF28-G3, BHK, HaK, NS0, SP2/0-Ag14, HeLa, HEK293 (e.g., HEK293-F, HEK293-H, HEK293-T) and perC6 cells and insect cells such as fall armyworm (Sf) or fungal cells such as saccharomyces cerevisiae, pichia pastoris and fission yeast. In some embodiments, the cell is a mammalian cell. In a specific embodiment, the cell is a Chinese hamster ovary cell (CHO cell). Progeny, variants and derivatives of these cell lines can also be used.

本文的另一方面提供一种其中FUT8基因被失活或部分失活的细胞系,其包含本文所述的ZFP、ZFN、多核苷酸或载体。Another aspect herein provides a cell line in which the FUT8 gene is inactivated or partially inactivated, comprising a ZFP, ZFN, polynucleotide or vector described herein.

本文还提供一种其中FUT8基因被失活或部分失活的细胞系,其通过使用本文所述的ZFP、ZFN、多核苷酸和/或载体生成。Also provided herein is a cell line in which the FUT8 gene is inactivated or partially inactivated, which is generated by using the ZFP, ZFN, polynucleotide and/or vector described herein.

本文还提供一种FUT8缺陷型细胞系,其在FUT8基因座处包含如SEQ ID NO:52、SEQ ID NO:53、SEQ ID NO:54、SEQ ID NO:55、SEQ ID NO:56、SEQ ID NO:57、SEQ ID NO:58和SEQ ID NO:59中一或多者所示的核苷酸序列。The present invention also provides a FUT8-deficient cell line, which comprises a nucleotide sequence as shown in one or more of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58 and SEQ ID NO: 59 at the FUT8 locus.

在一些实施方式中,所述FUT8缺陷型细胞系在FUT8基因座处包含SEQ ID NO:52和SEQ ID NO:53所示的核苷酸序列。在一些实施方式中,所述FUT8缺陷型细胞系在FUT8基因座处包含SEQ ID NO:54和SEQ ID NO:55所示的核苷酸序列。在一些实施方式中,所述FUT8缺陷型细胞系在FUT8基因座处包含SEQ ID NO:56和SEQ ID NO:57所示的核苷酸序列。在一些实施方式中,所述FUT8缺陷型细胞系在FUT8基因座处包含SEQ ID NO:58和SEQ ID NO:59所示的核苷酸序列。In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 52 and SEQ ID NO: 53 at the FUT8 locus. In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 54 and SEQ ID NO: 55 at the FUT8 locus. In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 56 and SEQ ID NO: 57 at the FUT8 locus. In some embodiments, the FUT8-deficient cell line comprises the nucleotide sequences set forth in SEQ ID NO: 58 and SEQ ID NO: 59 at the FUT8 locus.

本文还提供一种FUT8缺陷型细胞系,其包含经改造的FUT8基因座,所述经改造的FUT8基因座能通过如下引物对被检测:Also provided herein is a FUT8-deficient cell line comprising an altered FUT8 locus, wherein the altered FUT8 locus can be detected by the following primer pair:

引物对(i):Primer pair (i):

正向引物:AGCCTGAAGTACATAGCCGA(SEQ ID NO:46);和Forward primer: AGCCTGAAGTACATAGCCGA (SEQ ID NO: 46); and

反向引物:TGCCACTGCTTCTATATACTGATTC(SEQ ID NO:47);Reverse primer: TGCCACTGCTTCTATATACTGATTC (SEQ ID NO: 47);

和/或and/or

引物对(ii):Primer pair (ii):

正向引物:GACGCACTGACAAAGTGGGA(SEQ ID NO:48);和Forward primer: GACGCACTGACAAAGTGGGA (SEQ ID NO: 48); and

反向引物:GGTCTGTTCCATCCCCAGAATG(SEQ ID NO:49);Reverse primer: GGTCTGTTCCATCCCCAGAATG (SEQ ID NO: 49);

和/或and/or

引物对(iii):Primer pair (iii):

正向引物:CTGTTGATTCCAGGTTCCCA(SEQ ID NO:50);和Forward primer: CTGTTGATTCCAGGTTCCCA (SEQ ID NO: 50); and

反向引物:TGTTACTTAAGCCCCAGGC(SEQ ID NO:51)。Reverse primer: TGTTACTTAAGCCCCAGGC (SEQ ID NO: 51).

在一些实施方式中,所述检测采用FUT8正常(非缺陷)的等同细胞系作为对照细胞系。在一些实施方式中,所述检测显示FUT8缺陷型细胞系与对照细胞系之间的结果差异(例如,电泳所测分子大小及条带亮度差异等)。In some embodiments, the detection uses an equivalent cell line with normal (non-defective) FUT8 as a control cell line. In some embodiments, the detection shows a difference in results between the FUT8-deficient cell line and the control cell line (e.g., a difference in molecular size and band brightness measured by electrophoresis, etc.).

D.应用D. Application

本文所述的ZFN可用于使细胞中的FUT8基因失活。在一些实施方式中,失活包括部分或完全抑制细胞中FUT8基因的表达。可例如通过单一切割事件、通过切割然后非同源末端连接、通过在两个位点切割然后连接以便删除两个切割位点之间的序列、通过错义或无义密码子靶向重组到编码区、通过不相关序列(即“填充”序列)靶向重组到基因或其调控区以便破坏基因或调控区,或通过剪接受体序列靶向重组到内含子而引起转录物的错误剪接来实现FUT8基因的失活。The ZFNs described herein can be used to inactivate the FUT8 gene in a cell. In some embodiments, inactivation comprises partial or complete inhibition of expression of the FUT8 gene in a cell. Inactivation of the FUT8 gene can be achieved, for example, by a single cleavage event, by cleavage followed by non-homologous end joining, by cleavage at two sites followed by joining to delete the sequence between the two cleavage sites, by targeted recombination of missense or nonsense codons to the coding region, by targeted recombination of an unrelated sequence (i.e., a "stuffer" sequence) to a gene or its regulatory region to disrupt a gene or regulatory region, or by targeted recombination of a splice acceptor sequence to an intron to cause mis-splicing of the transcript.

本文所述的ZFN介导的失活(敲除或抑制)有多种应用。The ZFN-mediated inactivation (knockout or inhibition) described herein has a variety of applications.

本文的另一方面提供一种产生FUT8缺陷型细胞的方法,所述方法包括:Another aspect herein provides a method of producing a FUT8-deficient cell, the method comprising:

(a)将编码一种或多种ZFN的多核苷酸引入细胞,其中所述ZFN包含:(i)本文所述的ZFP,所述ZFP能与细胞中FUT8基因中的靶位点结合;和(ii)DNA切割域或切割半域;(a) introducing a polynucleotide encoding one or more ZFNs into a cell, wherein the ZFN comprises: (i) a ZFP as described herein, which is capable of binding to a target site in a FUT8 gene in the cell; and (ii) a DNA cleavage domain or cleavage half-domain;

(b)使所述ZFN在所述细胞中表达,从而所述ZFN与所述靶位点结合,并切割所述FUT8基因。(b) expressing the ZFN in the cell, so that the ZFN binds to the target site and cleaves the FUT8 gene.

在一些实施方式中,所述一种或多种ZFN包括本文所述的ZFN对。In some embodiments, the one or more ZFNs include a ZFN pair described herein.

在一些实施方式中,所述方法还可包括将在FUT8基因中具有靶位点的附加核酸酶引入所述细胞中。所述附加核酸酶的示例可包括,例如,归巢内切核酸酶和大范围核酸酶。In some embodiments, the method may further include introducing an additional nuclease having a target site in the FUT8 gene into the cell. Examples of the additional nuclease may include, for example, homing endonucleases and meganucleases.

本文的另一方面提供一种产生FUT8缺陷型细胞系的方法,所述方法包括:Another aspect herein provides a method of generating a FUT8-deficient cell line, the method comprising:

(a)通过如本文所述的产生FUT8缺陷型细胞的方法使细胞中的内源FUT8基因失活;和(a) inactivating the endogenous FUT8 gene in a cell by the method for producing a FUT8-deficient cell as described herein; and

(b)在适合产生FUT8缺陷型细胞系的条件下培养所述细胞。(b) culturing the cells under conditions suitable for generating a FUT8-deficient cell line.

示例性归巢内切核酸酶包括I-SceI、I-CeuI、PI-PspI、PI-Sce、I-SceIV、I-CsmI、I-PanI、I-SceII、I-PpoI、I-SceIII、I-CreI、I-TevI、I-TevII和I-TevIII。其识别序列已知。还参见美国专利第5,420,032号;美国专利第6,833,252号;Belfort等(1997)Nucleic Acids Res.25:3379-3388;Dujon等(1989)Gene 82:115-118;Perler等(1994)Nucleic Acids Res.22,1125-1127;Jasin(1996)Trends Genet.12:224-228;Gimble等(1996)J Mol.Biol.263:163-180;Argast等(1998)J. Mol.Biol.280:345-353和New England Biolabs目录。Exemplary homing endonucleases include I-Scel, I-Ceul, PI-PspI, PI-Sce, I-SceIV, I-Csml, I-Panl, I-Scel, I-Ppol, I-Scelll, I-Crel, I-Tevl, I-Tevll, and I-TevIII. Their recognition sequences are known. See also U.S. Pat. No. 5,420,032; U.S. Pat. No. 6,833,252; Belfort et al. (1997) Nucleic Acids Res. 25:3379-3388; Dujon et al. (1989) Gene 82:115-118; Perler et al. (1994) Nucleic Acids Res. 22, 1125-1127; Jasin (1996) Trends Genet. 12:224-228; Gimble et al. (1996) J Mol. Biol. 263:163-180; Argast et al. (1998) J. Mol. Biol. 280:345-353 and the New England Biolabs catalog.

尽管大多数归巢内切核酸酶的切割特异性关于其识别位点不是绝对的,这些位点具有足够的长度使得能通过在含有单拷贝其识别位点的细胞中表达归巢内切核酸酶来获得每个哺乳动物大小的基因组的单个切割事件。另据报道,归巢内切核酸酶和大范围核酸酶的特异性可被改造以便结合非天然的靶位点。参见,例如,Chevalier等(2002)Molec.Cell 10:895-905;Epinat等(2003)Nucleic Acids Res.31:2952-2962;Ashworth等(2006)Nature 441:656-659;Paques等(2007)Current Gene Therapy 7:49-66。Although the cleavage specificity of most homing endonucleases is not absolute with respect to their recognition sites, these sites are of sufficient length to enable a single cleavage event per mammalian-sized genome to be obtained by expressing the homing endonuclease in cells containing a single copy of its recognition site. It has also been reported that the specificity of homing endonucleases and meganucleases can be engineered to bind to non-natural target sites. See, e.g., Chevalier et al. (2002) Molec. Cell 10:895-905; Epinat et al. (2003) Nucleic Acids Res. 31:2952-2962; Ashworth et al. (2006) Nature 441:656-659; Paques et al. (2007) Current Gene Therapy 7:49-66.

本文还提供通过本文所述的产生FUT8缺陷型细胞系的方法制备的FUT8缺陷型细胞系。Also provided herein are FUT8-deficient cell lines produced by the methods of generating FUT8-deficient cell lines described herein.

本文的另一方面提供一种培育FUT8缺陷型细胞系的方法,其包括:Another aspect of the present invention provides a method for cultivating a FUT8-deficient cell line, comprising:

(a)提供根据本文所述获得的FUT8缺陷型细胞系;和(a) providing a FUT8-deficient cell line obtained as described herein; and

(b)培养或扩增所述细胞系的细胞。(b) culturing or expanding cells of said cell line.

本文的又一方面提供一种在宿主细胞中产生目标重组蛋白的方法,所述方法包括:Another aspect of the present invention provides a method for producing a target recombinant protein in a host cell, the method comprising:

(a)提供本文所述的FUT8缺陷型细胞,(a) providing a FUT8-deficient cell as described herein,

(b)将包含编码所述目标重组蛋白的核酸的表达载体引入所述细胞,由此产生所述目标重组蛋白。(b) introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell, thereby producing the target recombinant protein.

本文的另一方面提供一种在宿主细胞中产生目标重组蛋白的方法,所述方法包括:Another aspect of the present invention provides a method for producing a target recombinant protein in a host cell, the method comprising:

(a)提供包含内源FUT8基因的细胞;(a) providing a cell comprising an endogenous FUT8 gene;

(b)通过如本文所述的产生FUT8缺陷型细胞的方法使所述细胞中的内源FUT8基因失活;和(b) inactivating the endogenous FUT8 gene in the cell by the method for producing a FUT8-deficient cell as described herein; and

(c)将包含编码所述目标重组蛋白的核酸的表达载体引入所述细胞,由此产生所述目标重组蛋白。(c) introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell, thereby producing the target recombinant protein.

在一些实施方式中,如本文所述使细胞中的内源FUT8基因失活的步骤与将包含编码所述目标重组蛋白的核酸的表达载体引入细胞的步骤可彼此调换次序或同时进行。In some embodiments, the step of inactivating the endogenous FUT8 gene in the cell as described herein and the step of introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell may be performed in reverse order or simultaneously.

适合的细胞包括但不限于真核和原核细胞和/或细胞系。此类细胞或细胞系的非限制性实例包括:COS、CHO(例如,CHO-S、CHO-K1、CHO-DG44、CHO-DUXB11、CHO-DUKX、CHOK1SV)、VERO、MDCK、WI38、V79、B14AF28-G3、BHK、HaK、NS0、SP2/0-Ag14、HeLa、HEK293(例如,HEK293-F、HEK293-H、HEK293-T)和perC6细胞以及诸如草地贪夜蛾(Sf)的昆虫细胞或诸如酿酒酵母、毕赤酵母和裂殖酵母的真菌细胞。在一些实施方式中,所述细胞为哺乳动物细胞。在一个具体实施方式中,所述细胞为中国仓鼠卵巢细胞(CHO细胞)。也可使用这些细胞系的子代、变体和衍生物。在一些实施方式中,所述细胞为哺乳动物细胞。在一个具体实施方式中,所述细胞为中国仓鼠卵巢细胞(CHO细胞)。Suitable cells include, but are not limited to, eukaryotic and prokaryotic cells and/or cell lines. Non-limiting examples of such cells or cell lines include: COS, CHO (e.g., CHO-S, CHO-K1, CHO-DG44, CHO-DUXB11, CHO-DUKX, CHOK1SV), VERO, MDCK, WI38, V79, B14AF28-G3, BHK, HaK, NS0, SP2/0-Ag14, HeLa, HEK293 (e.g., HEK293-F, HEK293-H, HEK293-T) and perC6 cells and insect cells such as fall armyworm (Sf) or fungal cells such as saccharomyces cerevisiae, pichia pastoris and fission yeast. In some embodiments, the cell is a mammalian cell. In a specific embodiment, the cell is a Chinese hamster ovary cell (CHO cell). Progeny, variants and derivatives of these cell lines may also be used. In some embodiments, the cell is a mammalian cell. In a specific embodiment, the cell is a Chinese hamster ovary cell (CHO cell).

在一些实施方式中,所述目标重组蛋白包括含Fc重组蛋白。在一些实施方式中,所述目标重组蛋白为去岩藻糖基化的含Fc重组蛋白。In some embodiments, the target recombinant protein includes an Fc-containing recombinant protein. In some embodiments, the target recombinant protein is a defucosylated Fc-containing recombinant protein.

本文的另一方面提供本文所述的ZFP、ZFN、多核苷酸、载体用于产生FUT8缺陷型细胞的用途。Another aspect herein provides the use of the ZFPs, ZFNs, polynucleotides, vectors described herein for generating FUT8-deficient cells.

本文的另一方面提供本文所述的ZFP、ZFN、多核苷酸、载体、细胞或细胞系用于产生去岩藻糖基化的含Fc重组蛋白的用途。Another aspect herein provides use of a ZFP, ZFN, polynucleotide, vector, cell or cell line described herein for producing a defucosylated Fc-containing recombinant protein.

本文的另一方面提供本文所述的ZFP、ZFN、多核苷酸、载体、细胞或细胞系用于产生能引发增强的ADCC效应的抗体(如单克隆抗体)的用途。Another aspect provided herein is the use of a ZFP, ZFN, polynucleotide, vector, cell or cell line described herein for producing an antibody (eg, a monoclonal antibody) capable of eliciting an enhanced ADCC effect.

在一些实施方式中,含Fc重组蛋白可以是抗体(例如单克隆抗体、多克隆抗体)或Fc融合蛋白。经本文所述方法提供的去岩藻糖基化的含Fc重组蛋白表现出更强的效应功能,特别是在ADCC的诱导中。In some embodiments, the Fc-containing recombinant protein can be an antibody (eg, monoclonal antibody, polyclonal antibody) or an Fc fusion protein. The defucosylated Fc-containing recombinant protein provided by the method described herein exhibits stronger effector function, particularly in the induction of ADCC.

实施例Example

下面将结合具体的实施例对本发明的技术方案进行更为详细地阐述。以下实施例仅为示例,对本发明技术方案不构成任何限定或限制。以下实施例中具体的材料、步骤、条件、数值或数值范围等技术参数仅是举例,并非穷举亦非限定。The technical scheme of the present invention will be described in more detail below in conjunction with specific embodiments. The following embodiments are only examples and do not constitute any limitation or restriction on the technical scheme of the present invention. The specific materials, steps, conditions, values or numerical ranges and other technical parameters in the following embodiments are only examples and are not exhaustive or limiting.

除实施例中使用的具体方法、设备、材料外,根据本技术领域的技术人员对现有技术的掌握及本发明的记载,还可以使用与本发明实施例中所述的方法、设备、材料相似或等同的现有技术的任何方法、设备和材料来实现本发明。In addition to the specific methods, equipment, and materials used in the embodiments, based on the understanding of the prior art by those skilled in the art and the description of the present invention, any methods, equipment, and materials of the prior art that are similar or equivalent to the methods, equipment, and materials described in the embodiments of the present invention may also be used to implement the present invention.

实施例中所用的设备和试剂分别列于表2和表3。The equipment and reagents used in the examples are listed in Table 2 and Table 3, respectively.

表2:实验设备列表

Table 2: Experimental equipment list

表3:实验试剂列表

Table 3: List of experimental reagents

实施例1 FUT8-/-细胞株在开发稳定表达阿达木单抗单抗分子的应用Example 1 Application of FUT8 -/- cell line in developing stable expression of adalimumab monoclonal antibody molecules

1表达载体构建1 Expression vector construction

1.1如图2A中示例性地显示,将ZFP蛋白L3/R3、S2-L/S4-R各为一组,分别构建在表达载体上,由CMV作为启动子启动转录,ZFP蛋白作为DNA结合域和下游的FokI切割半域连接在一起,组成一个完整蛋白质,通过同一个ORF进行表达。该表达载体在转染进CHO细胞后不通过抗性筛选,故质粒构建中未插入真核细胞筛选标志物。图2B显示了示例性序列结构,其中5-6个锌指识别区按顺序插入天然锌指蛋白框架序列中(如ZIF268或SP1C),并在C端连接至FokI切割半域。本实施例中所用S2-L-FokI及S4-R-FokI全长序列如SEQ ID NO:60及SEQ ID NO:61示例性地显示。所用其它ZFP蛋白均以类似的方式将对应的锌指识别区构建至相同的框架序列中并与FokI连接。1.1 As shown in Figure 2A, ZFP proteins L3/R3 and S2-L/S4-R are each grouped and constructed on expression vectors, and transcription is initiated by CMV as a promoter. ZFP protein is connected to the downstream FokI cleavage half domain as a DNA binding domain to form a complete protein, which is expressed through the same ORF. The expression vector does not pass the resistance screening after transfection into CHO cells, so no eukaryotic cell screening marker is inserted in the plasmid construction. Figure 2B shows an exemplary sequence structure, in which 5-6 zinc finger recognition regions are sequentially inserted into the natural zinc finger protein framework sequence (such as ZIF268 or SP1C) and connected to the FokI cleavage half domain at the C-terminus. The full-length sequences of S2-L-FokI and S4-R-FokI used in this example are shown as SEQ ID NO: 60 and SEQ ID NO: 61. The other ZFP proteins used are constructed in a similar manner to construct the corresponding zinc finger recognition regions into the same framework sequence and connected to FokI.

1.2将阿达木单抗(Adalimumab)的HC序列(SEQ ID NO:4)构建至表达载体pWX039-HC-Z-105上,和博来霉素(Zeocin)抗性基因通过EMCV IRES连接。阿达木单抗单抗LC序列(SEQ ID NO 5)构建至表达载体pWX040-LC-B-105上,置于杀稻瘟菌素(Blasticidin)抗性基因上游,LC和抗性基因元件属于两个独立的阅读框,杀稻瘟菌素抗性基因由SV40启动子驱动表达。1.2 The HC sequence of Adalimumab (SEQ ID NO: 4) was constructed into the expression vector pWX039-HC-Z-105 and connected to the Zeocin resistance gene through EMCV IRES. The LC sequence of Adalimumab (SEQ ID NO: 5) was constructed into the expression vector pWX040-LC-B-105 and placed upstream of the Blasticidin resistance gene. The LC and resistance gene elements belong to two independent reading frames. The Blasticidin resistance gene is driven by the SV40 promoter.

2细胞转染和细胞池筛选2. Cell transfection and cell pool screening

2.1取1E7个CHO-K1宿主细胞悬液,取30μg质粒到250μL DPBS溶液中混合均匀,将质粒溶液和细胞悬液充分混匀后加到电转杯中。将电转杯置于Biorad电转仪卡槽中按照宿主细胞相应的程序进行电转。转染后将细胞悬液从电转杯吸出转移到7.5mL提前预热好的无压培养基的T25细胞培养瓶,置于36.5℃,5%CO2,85%湿度的二氧化碳培养箱中静置培养。2.1 Take 1E7 CHO-K1 host cell suspension, take 30μg plasmid and mix it evenly in 250μL DPBS solution, mix the plasmid solution and cell suspension thoroughly and add them to the electroporation cup. Place the electroporation cup in the card slot of the Biorad electroporator and perform electroporation according to the corresponding program of the host cell. After transfection, aspirate the cell suspension from the electroporation cup and transfer it to a T25 cell culture bottle with 7.5mL preheated pressure-free culture medium, and place it in a carbon dioxide incubator at 36.5℃, 5% CO2 , and 85% humidity for static culture.

2.2转染48小时后,将T25培养瓶转移至36.5℃条件的二氧化碳培养箱中,再经过24小时的静置培养后,对细胞池取样计数取1E6的细胞量进行基因组DNA的抽提,并用微量紫外分光光度计定量。选择100ng~200ng基因组DNA用高保真DNA聚合酶选择合适的引物(引物序列为:引物2F:CTGTTGATTCCAGGTTCCCA(SEQ ID NO 50);引物R:TGTTACTTAAGCCCCAGGC(SEQ ID NO 51))进行PCR扩增目的基因序列,然后将体系重新变性退火,退火完成后体系中加入1μL T7E1酶(10U/μL),37℃孵育15分钟。酶切后进行2%琼脂糖凝胶电泳检测酶切条带和编辑效率。2.2 After 48 hours of transfection, the T25 culture flask was transferred to a carbon dioxide incubator at 36.5℃. After 24 hours of static culture, the cell pool was sampled and counted to extract 1E6 cells for genomic DNA extraction and quantified using a micro-UV spectrophotometer. 100ng~200ng genomic DNA was selected and high-fidelity DNA polymerase was used to select appropriate primers (primer sequences: primer 2F: CTGTTGATTCCAGGTTCCCA (SEQ ID NO 50); primer R: TGTTACTTAAGCCCCAGGC (SEQ ID NO 51)) to PCR amplify the target gene sequence, and then the system was re-denatured and annealed. After annealing, 1μL T7E1 enzyme (10U/μL) was added to the system and incubated at 37℃ for 15 minutes. After enzyme digestion, 2% agarose gel electrophoresis was performed to detect the enzyme digestion bands and editing efficiency.

3克隆筛选3. Clone Screening

将活率恢复到90%以上的细胞池通过有限稀释法进行单克隆分选,将细胞静置96孔板培养约2周后,将克隆转移到24孔板中,培养约2-3天后扩增至摇管中培养,同时通过NGS方法检验细胞株中FUT8基因的序列被敲除。在摇管中进行每2-4天的传代,培养条件为36.5℃、6%CO2、85%湿度,转速为225rpm。The cell pool with a viability of more than 90% was sorted by limiting dilution method for monoclonal selection. After the cells were cultured in 96-well plates for about 2 weeks, the clones were transferred to 24-well plates and cultured for about 2-3 days before being expanded to shake tubes for culture. At the same time, the sequence of FUT8 gene in the cell line was checked by NGS method. The cells were subcultured every 2-4 days in shake tubes under the culture conditions of 36.5°C, 6% CO 2 , 85% humidity, and 225 rpm.

4目的细胞株在表达单抗分子方面的应用4. Application of target cell lines in expressing monoclonal antibody molecules

4.1待筛选到的目的细胞株(本实施例选择单克隆154-F8ZFN-09-08及单克隆154-F8ZFN-10-02)生长恢复后,分别将目的细胞株和未被敲除FUT8基因的细胞株(阴性对照组)作为宿主细胞,取1E7数量的细胞,和阿达木单抗单抗HC和LC质粒20ug。将细胞和质粒溶液混合均匀后转移到电转杯中,使用Biorad电转仪按相应细胞类型选择合适电转参数进行电转。电转之后将细胞全部转移到装有预热好培养基的摇管中,置于摇床中培养,培养条件为36.5℃、6%CO2、85%湿度,转速为225rpm。4.1 After the growth of the target cell strain to be screened (monoclonal 154-F8ZFN-09-08 and monoclonal 154-F8ZFN-10-02 were selected in this example), the target cell strain and the cell strain without knockout of FUT8 gene (negative control group) were used as host cells, and 1E7 cells and 20ug of adalimumab monoclonal antibody HC and LC plasmids were taken. The cells and plasmid solution were mixed evenly and transferred to an electroporation cup, and the Biorad electroporator was used to select appropriate electroporation parameters according to the corresponding cell type for electroporation. After electroporation, all cells were transferred to a shaking tube filled with preheated culture medium and placed in a shaking incubator for culture. The culture conditions were 36.5°C, 6% CO 2 , 85% humidity, and a rotation speed of 225rpm.

4.2在转染24小时后,加入筛选压力培养基后继续培养,每隔2-4天进行传代。4.2 24 hours after transfection, add screening stress medium and continue culturing, subculturing every 2-4 days.

4.3连续传代3-4周后,待细胞池活率恢复至95%以上,以4E5/mL的细胞密度接种到生产培养基中进行流加式(fed-batch)培养,在培养第3、5、7、9和11天取样检测活细胞密度及活率,并进行补料补糖。培养14天后离心收获上清,利用ProA-HPLC方法检测上清中单抗的效价水平。此外,将收获的上清样品进进行一步proA纯化后进行SEC-HPLC高聚含量检测,SDS Caliper和N-聚糖-LC糖型分析。在本实施例中,阴性对照组为未被敲除FUT8基因的宿主细胞进行单抗载体的转染。4.3 After 3-4 weeks of continuous subculturing, when the viability of the cell pool recovers to more than 95%, it is inoculated into the production medium at a cell density of 4E5/mL for fed-batch culture. Samples are taken on the 3rd, 5th, 7th, 9th and 11th days of culture to detect the density and viability of live cells, and feed and sugar are supplemented. After 14 days of culture, the supernatant is harvested by centrifugation, and the titer level of the monoclonal antibody in the supernatant is detected by the ProA-HPLC method. In addition, the harvested supernatant samples are further purified by proA and then subjected to SEC-HPLC high polymer content detection, SDS Caliper and N-glycan-LC glycoform analysis. In this embodiment, the negative control group is a host cell in which the FUT8 gene has not been knocked out and the monoclonal antibody vector is transfected.

5实验结果5 Experimental results

酶切实验结果成功验证了ZFN质粒能够有效切除目的基因序列(图3),细胞池使用流式分析(图4),可发现一定比例的FUT8-/-细胞群。通过克隆筛选流程,克隆154-F8ZFN-09-08和154-F8ZFN-10-02被成功验证一对等位基因被敲除(图5、图6)。从图7A和7B可看出,154-F8ZFN-09-08和154-F8ZFN-10-02在FB培养过程中的活细胞密度及峰值VCD与对照组相当;图7C表明154-F8ZFN-09-08组单抗表达效价与对照组无明显差异。并且SEC-HPLC和Caliper R/NR结果表明,154-F8ZFN-09-08及154-F8ZFN-10-02组细胞池一步纯化样品高聚水平和对照组无明显差异(图7D、7E)。最后,糖型结果(图7F)可看出,154-F8ZFN-09-08和154-F8ZFN-10-02组的岩藻糖水平要远远低于未被敲除FUT8基因的阴性对照组,降到了接近零的水平。The results of the enzyme digestion experiment successfully verified that the ZFN plasmid can effectively remove the target gene sequence (Figure 3). The cell pool was analyzed by flow cytometry (Figure 4), and a certain proportion of FUT8 -/- cell populations were found. Through the clone screening process, clones 154-F8ZFN-09-08 and 154-F8ZFN-10-02 were successfully verified to have a pair of alleles knocked out (Figures 5 and 6). As can be seen from Figures 7A and 7B, the live cell density and peak VCD of 154-F8ZFN-09-08 and 154-F8ZFN-10-02 during FB culture were comparable to those of the control group; Figure 7C showed that there was no significant difference in the monoclonal antibody expression titer of the 154-F8ZFN-09-08 group and the control group. And the results of SEC-HPLC and Caliper R/NR showed that there was no significant difference in the high polymer level of the one-step purified samples of the cell pool of the 154-F8ZFN-09-08 and 154-F8ZFN-10-02 groups and the control group (Figure 7D, 7E). Finally, the glycoform results (Figure 7F) showed that the fucose level of the 154-F8ZFN-09-08 and 154-F8ZFN-10-02 groups was much lower than that of the negative control group in which the FUT8 gene was not knocked out, and dropped to a level close to zero.

实施例2 FUT8-/-细胞株在开发稳定表达融合蛋白分子Dulaglutide的应用Example 2 Application of FUT8 -/- cell line in developing stable expression of fusion protein molecule Dulaglutide

1表达载体构建1 Expression vector construction

Dulaglutide为一种胰高血糖素样肽1(GLP-1)Fc融合蛋白。Dulaglutide is a glucagon-like peptide 1 (GLP-1) Fc fusion protein.

将Dulaglutide的序列(SEQ ID NO 6)构建至表达载体pWX039-Pr-Z-126A3和pWX040-Pr-B-126A3上。在pWX039-Pr-Z-126A3载体上,Dulaglutide的序列通过EMCV IRES和Zeocin抗性基因连接。在pWX040-Pr-B-126A3上,Dulaglutide的序列置于杀稻瘟菌素抗性基因上游,目的基因和抗性基因元件属于两个独立的阅读框,杀稻瘟菌素抗性基因由SV40启动子驱动表达。The sequence of dulaglutide (SEQ ID NO 6) was constructed into the expression vectors pWX039-Pr-Z-126A3 and pWX040-Pr-B-126A3. In the pWX039-Pr-Z-126A3 vector, the sequence of dulaglutide was connected by EMCV IRES and Zeocin resistance gene. In pWX040-Pr-B-126A3, the sequence of dulaglutide was placed upstream of the blasticidin resistance gene, and the target gene and resistance gene elements belonged to two independent reading frames. The expression of blasticidin resistance gene was driven by SV40 promoter.

2细胞转染及流加式培养2 Cell transfection and fed-batch culture

2.1将实施例1中筛选到的154-F8ZFN-09-08作为宿主细胞,取1E7数量的细胞和20μg表达载体混合均匀后转移到电转杯中,使用Biorad电转仪按相应细胞类型选择合适电转参数进行电转。电转之后将细胞全部转移到装有预热好培养基的摇管中,置于摇床中培养,培养条件为36.5℃、6%CO2、85%湿度,转速为225rpm。2.1 The 154-F8ZFN-09-08 selected in Example 1 was used as a host cell, 1E7 cells and 20 μg of the expression vector were mixed evenly and then transferred to an electroporation cup, and electroporated using a Biorad electroporator with appropriate electroporation parameters selected according to the corresponding cell type. After electroporation, all cells were transferred to a shaking tube filled with preheated culture medium and cultured in a shaking incubator at 36.5°C, 6% CO 2 , 85% humidity, and a rotation speed of 225 rpm.

2.2在转染24小时后,加入等体积筛选压力培养基后继续培养,每隔2-4天进行传代。2.2 24 hours after transfection, add an equal volume of screening stress medium and continue culturing, subculturing every 2-4 days.

2.3连续传代3-4周后,待细胞池活率恢复至95%以上,以4E5/mL的细胞密度接种到生产培养基中进行流加式培养,在培养第3、5、7、9和11天取样检测活细胞密度及活率,并进行补料补糖。培养14天后离心收获上清,利用ProA-HPLC方法检测上清中单抗的效价水平。此外,将收获的上清样品进进行一步proA纯化后进行SEC HPLC高聚含量检测,SDS CaliperNR纯度检测和N-聚糖糖型分析。在本实施例中,阳性对照组为被敲除FUT8基因的宿主细胞进行融合蛋白载体的转染。2.3 After 3-4 weeks of continuous subculturing, when the viability of the cell pool recovers to more than 95%, it is inoculated into the production culture medium at a cell density of 4E5/mL for fed-batch culture. Samples are taken on the 3rd, 5th, 7th, 9th and 11th days of culture to detect the density and viability of live cells, and feed and sugar are supplemented. After 14 days of culture, the supernatant is harvested by centrifugation, and the titer level of the monoclonal antibody in the supernatant is detected by the ProA-HPLC method. In addition, the harvested supernatant samples are further purified by proA and then subjected to SEC HPLC high polymer content detection, SDS CaliperNR purity detection and N-glycan glycotype analysis. In this embodiment, the positive control group is a host cell in which the FUT8 gene is knocked out and transfected with a fusion protein vector.

3实验结果3 Experimental results

图8A显示,154-F8ZFN-09-08组在FB培养过程中的活细胞密度与活率数据在可接受范围之内;图8C、D和E表明154-F8ZFN-09-08组蛋白质表达效价、SEC-HPLC主峰,及SDS CaliperNR主峰均与对照组差异不大。最后,糖型结果(图8E)显示,154-F8ZFN-09-08组的岩藻糖水平要远远低于未被敲除FUT8基因的阴性对照组,降到了接近零的水平。Figure 8A shows that the live cell density and viability data of the 154-F8ZFN-09-08 group during FB culture are within the acceptable range; Figures 8C, D, and E show that the protein expression titer, SEC-HPLC main peak, and SDS CaliperNR main peak of the 154-F8ZFN-09-08 group are not much different from those of the control group. Finally, the glycoform results (Figure 8E) show that the fucose level of the 154-F8ZFN-09-08 group is much lower than that of the negative control group in which the FUT8 gene has not been knocked out, and has dropped to a level close to zero.

实施例3在外显子3处进行ZFN编辑Example 3 ZFN editing at exon 3

1.表达载体构建1. Expression vector construction

1.1如图2A中示例性地显示,与实施例1中所述类似地,将ZFP蛋白31-L1/31-R1、32-L1/32-R1各为一组,分别构建在表达载体上。1.1 As shown in FIG. 2A , similarly to Example 1, the ZFP proteins 31-L1/31-R1 and 32-L1/32-R1 were each grouped and constructed on expression vectors.

2.细胞转染和细胞群(库)筛选2. Cell transfection and cell population (library) screening

2.2将1×107个CHO-K1细胞与32-L1/32-R1和31-L1/31-R1质粒混合均匀,使用程序WE在Bio-rad电转仪中转染。转染后将细胞在5mL CD CHO培养基中恢复,于30℃静置培养箱中培养。2.2 1×10 7 CHO-K1 cells were mixed evenly with 32-L1/32-R1 and 31-L1/31-R1 plasmids and transfected in a Bio-rad electroporator using program WE. After transfection, the cells were recovered in 5 mL CD CHO medium and cultured in a static incubator at 30°C.

2.3转染后第2天收集细胞,制备基因组DNA,使用引物Fut8-EX3-F1(SEQ ID NO:46)和Fut8-EX3-R1(SEQ ID NO:47)扩增FUT8基因的靶向区域;最后使用T7E1试剂盒检测基因编辑效率。如图9泳道2所示,根据ImageJ对条带亮度的计算,32-L1/32-R1修饰FUT8外显子3区域,导致了10.9%的DNA突变;如泳道1所示,31-L1/31-R1修饰FUT8外显子3区域,导致了9.4%的DNA突变。2.3 On the second day after transfection, cells were collected, genomic DNA was prepared, and primers Fut8-EX3-F1 (SEQ ID NO: 46) and Fut8-EX3-R1 (SEQ ID NO: 47) were used to amplify the targeted region of the FUT8 gene; finally, the gene editing efficiency was detected using the T7E1 kit. As shown in lane 2 of Figure 9, according to the calculation of the band brightness by ImageJ, 32-L1/32-R1 modified the FUT8 exon 3 region, resulting in 10.9% DNA mutation; as shown in lane 1, 31-L1/31-R1 modified the FUT8 exon 3 region, resulting in 9.4% DNA mutation.

3单克隆筛选3 Monoclonal screening

转染后第34天,进行SCP单克隆分选。对所有生长起来的单克隆,进行LCA-FITC染色,最终获得2个FUT8-/-表型的单克隆C6及F2(图10、11)。通过序列验证,这两个克隆的测序结果如图12所示,两个等位基因均被敲除。On the 34th day after transfection, SCP monoclonal sorting was performed. All grown monoclones were stained with LCA-FITC, and finally two FUT8 -/- phenotype monoclones C6 and F2 were obtained (Figures 10 and 11). Through sequence verification, the sequencing results of these two clones are shown in Figure 12, and both alleles were knocked out.

实施例4通过ZFN敲除的FUT8-/-克隆表达的抗体ADCC活性显著提高Example 4 The ADCC activity of the antibody expressed by the FUT8 −/− clone knocked out by ZFN was significantly improved

1.蛋白质表达1. Protein Expression

以根据本文所述方法获得的FUT8-/-双敲除的克隆(154-F8ZFN-09-08)作为宿主细胞,以没有进行突变的野生型CHO作为对照,按照如前所述的方法表达阿达木单抗(Adalimumab)并纯化。The FUT8 -/- double knockout clone (154-F8ZFN-09-08) obtained according to the method described herein was used as a host cell, and wild-type CHO cells without mutation were used as a control. Adalimumab was expressed and purified according to the method described above.

2.靶细胞标记2. Target Cell Labeling

表达TNFα的HT1080细胞首先用DELFIA BATDA(Cat#C136-100,Perkin Elmer,中国)在37℃、5%CO2湿润培养箱中在37℃下标记30~40分钟。孵育后,将HT1080细胞离心并用测定培养基洗涤,然后重悬于测定培养基(RPMI 1640+10%HI-FBS)中。HT1080 cells expressing TNFα were first labeled with DELFIA BATDA (Cat# C136-100, Perkin Elmer, China) at 37°C in a humidified incubator with 5% CO2 for 30-40 minutes. After incubation, HT1080 cells were centrifuged and washed with assay medium and then resuspended in assay medium (RPMI 1640 + 10% HI-FBS).

3.靶细胞和效应细胞共孵育3. Co-incubation of target cells and effector cells

外周血单核细胞(PBMC)重悬于测定培养基中。将1×106个PBMC与1×104个表达TNFα的HT1080细胞充分混合,加入0至50ng/mL抗TNFα治疗性抗体样品,96孔板中以200μL/孔在37℃、5%CO2培养箱中共培养1~2小时。Peripheral blood mononuclear cells (PBMC) were resuspended in assay medium. 1×10 6 PBMCs were thoroughly mixed with 1×10 4 HT1080 cells expressing TNFα, and 0 to 50 ng/mL anti-TNFα therapeutic antibody samples were added. The samples were co-cultured in a 96-well plate at 200 μL/well in a 37°C, 5% CO 2 incubator for 1 to 2 hours.

4.上清液反应4. Supernatant reaction

96孔板以500×g离心3~5分钟后,从每个孔中取出20μL上清液,并转移到96孔白色微孔板(3912,Corning,美国)中的相应孔中。随后,向各孔中加入200μL DELFIA铕溶液(Cat#C135-100,Perkin Elmer,中国),并将板孵育20~30分钟。After centrifugation of the 96-well plate at 500 × g for 3–5 min, 20 μL of supernatant was removed from each well and transferred to the corresponding well in a 96-well white microplate (3912, Corning, USA). Subsequently, 200 μL of DELFIA Europium solution (Cat# C135-100, Perkin Elmer, China) was added to each well, and the plate was incubated for 20–30 min.

5.数据处理5. Data processing

在酶标仪M5e(Molecular Devices,美国加利福尼亚州)上采集数据。将相对荧光单位(RFU)与抗体浓度作图,以生成4参数逻辑斯蒂(logistic)响应曲线。当满足所有系统和样品适用性时,通过EC50比率计算样品的相对效力。如图13所示,FUT8-/-双敲除的宿主生产的阿达木单抗的ADCC活性显著高于对照组野生型CHO宿主生产的相同蛋白质。



Data was collected on a microplate reader M5e (Molecular Devices, California, USA). Relative fluorescence units (RFU) were plotted against antibody concentration to generate a 4-parameter logistic response curve. When all system and sample suitability were met, the relative potency of the sample was calculated by the EC50 ratio. As shown in Figure 13, the ADCC activity of adalimumab produced by the FUT8 -/- double knockout host was significantly higher than that of the same protein produced by the wild-type CHO host in the control group.



Claims (18)

一种能靶向结合FUT8基因的锌指蛋白,其以自N至C端顺序包含下表同行所示的锌指F1至F5或至F6的锌指识别区氨基酸序列:
A zinc finger protein capable of targeting and binding to the FUT8 gene, comprising the amino acid sequences of the zinc finger recognition regions of zinc fingers F1 to F5 or to F6 shown in the same row in the following table in order from N to C terminus:
一种锌指核酸酶,其包含根据权利要求1所述的锌指蛋白和至少一个DNA切割域或切割半域。A zinc finger nuclease comprising the zinc finger protein according to claim 1 and at least one DNA cleavage domain or cleavage half-domain. 一种多核苷酸,其编码根据权利要求1所述的锌指蛋白。A polynucleotide encoding the zinc finger protein according to claim 1. 一种多核苷酸,其编码根据权利要求2所述的锌指核酸酶。A polynucleotide encoding the zinc finger nuclease according to claim 2. 一种载体,其包含根据权利要求3或4所述的多核苷酸。A vector comprising the polynucleotide according to claim 3 or 4. 一种分离的细胞,其包含根据权利要求1所述的锌指蛋白、根据权利要求2所述的锌指核酸酶、根据权利要求3或4所述的多核苷酸,或根据权利要求5所述的载体。An isolated cell comprising the zinc finger protein according to claim 1, the zinc finger nuclease according to claim 2, the polynucleotide according to claim 3 or 4, or the vector according to claim 5. 一种产生FUT8缺陷型细胞的方法,所述方法包括:A method for generating a FUT8-deficient cell, the method comprising: (a)将编码一种或多种根据权利要求2所述的锌指核酸酶的多核苷酸引入细胞;(a) introducing a polynucleotide encoding one or more zinc finger nucleases according to claim 2 into a cell; (b)使所述锌指核酸酶在所述细胞中表达,从而所述锌指核酸酶与所述靶位点结合并切割FUT8基因。(b) expressing the zinc finger nuclease in the cell, so that the zinc finger nuclease binds to the target site and cleaves the FUT8 gene. 根据权利要求7所述的方法,其中,在步骤(a)中,将包括左锌指核酸酶和右锌指核酸酶的锌指核酸酶对引入所述细胞,其中,所述锌指核酸酶对包括:The method according to claim 7, wherein, in step (a), a zinc finger nuclease pair comprising a left zinc finger nuclease and a right zinc finger nuclease is introduced into the cell, wherein the zinc finger nuclease pair comprises: 锌指核酸酶对(a):Zinc finger nuclease pair (a): 具有以下锌指识别区氨基酸序列的左锌指核酸酶:Left zinc finger nuclease having the following zinc finger recognition region amino acid sequence: F1:QLATLNR(SEQ ID NO:13);F1: QLATLNR (SEQ ID NO: 13); F2:TRWNLRA(SEQ ID NO:14);F2: TRWNLRA (SEQ ID NO: 14); F3:SRRDLRR(SEQ ID NO:15);F3: SRRDLRR (SEQ ID NO: 15); F4:WRRRLLS(SEQ ID NO:16);F4: WRRRLLS(SEQ ID NO: 16); F5:RSDYLTN(SEQ ID NO:17);和F5:RSDYLTN (SEQ ID NO:17); and F6:FHSNLLA(SEQ ID NO:18);F6: FHSNLLA (SEQ ID NO: 18); and 具有以下锌指识别区氨基酸序列的右锌指核酸酶:A right zinc finger nuclease having the following zinc finger recognition region amino acid sequence: F1:RKSHLTM(SEQ ID NO:19);F1: RKSHLTM (SEQ ID NO: 19); F2:FHSGLLA(SEQ ID NO:20);F2: FHSGLLA (SEQ ID NO: 20); F3:WRRRLLS(SEQ ID NO:16);F3: WRRRLLS(SEQ ID NO: 16); F4:RKYVLLR(SEQ ID NO:21);F4: RKYVLLR (SEQ ID NO: 21); F5:RKDYLVL(SEQ ID NO:22);和F5: RKDYLVL (SEQ ID NO: 22); and F6:QQAGLIN(SEQ ID NO:23);F6: QQAGLIN(SEQ ID NO: 23); 和/或and/or 锌指核酸酶对(b):Zinc finger nuclease pair (b): 具有以下锌指识别区氨基酸序列的左锌指核酸酶:Left zinc finger nuclease having the following zinc finger recognition region amino acid sequence: F1:SKWNLRS(SEQ ID NO:24);F1:SKWNLRS(SEQ ID NO:24); F2:AQSNLLS(SEQ ID NO:25);F2: AQSNLLS(SEQ ID NO: 25); F3:LRHQLRR(SEQ ID NO:26);F3: LRHQLRR (SEQ ID NO: 26); F4:RSDYLTN(SEQ ID NO:17);和F4:RSDYLTN (SEQ ID NO:17); and F5:RSDYLTN(SEQ ID NO:17);F5: RSDYLTN (SEQ ID NO: 17); and 具有以下锌指识别区氨基酸序列的右锌指核酸酶:A right zinc finger nuclease having the following zinc finger recognition region amino acid sequence: RSDYLTN(SEQ ID NO:17);RSDYLTN (SEQ ID NO: 17); QKITLVR(SEQ ID NO:27);QKITLVR (SEQ ID NO: 27); RSDYLTN(SEQ ID NO:17);RSDYLTN (SEQ ID NO: 17); QLATLNR(SEQ ID NO:13);QLATLNR (SEQ ID NO: 13); SRFNLTR(SEQ ID NO:28);和SRFNLTR (SEQ ID NO: 28); and TKYILTN(SEQ ID NO:29);TKYILTN (SEQ ID NO: 29); 和/或and/or 锌指核酸酶对(c):Zinc finger nuclease pair (c): 具有以下锌指识别区氨基酸序列的左锌指核酸酶:Left zinc finger nuclease having the following zinc finger recognition region amino acid sequence: F1:RADNLTE(SEQ ID NO:30);F1: RADNLTE(SEQ ID NO: 30); F2:TSGNLTE(SEQ ID NO:31);F2: TSGNLTE (SEQ ID NO: 31); F3:TSGHLVR(SEQ ID NO:32);F3: TSGHLVR (SEQ ID NO: 32); F4:RKDNLKN(SEQ ID NO:33);和F4: RKDNLKN (SEQ ID NO: 33); and F5:RKDNLKN(SEQ ID NO:33);F5: RKDNLKN (SEQ ID NO: 33); and 具有以下锌指识别区氨基酸序列的右锌指核酸酶:A right zinc finger nuclease having the following zinc finger recognition region amino acid sequence: RSDNLSV(SEQ ID NO:34);RSDNLSV (SEQ ID NO: 34); SPADLTR(SEQ ID NO:35);SPADLTR (SEQ ID NO: 35); RSDHLSQ(SEQ ID NO:36);RSDHLSQ (SEQ ID NO: 36); QSGDLRR(SEQ ID NO:37);QSGDLRR (SEQ ID NO: 37); RSDNLVR(SEQ ID NO:38);和RSDNLVR (SEQ ID NO: 38); and RKDNLKN(SEQ ID NO:33);RKDNLKN (SEQ ID NO: 33); 和/或and/or 锌指核酸酶对(d):Zinc finger nuclease pair (d): 具有以下锌指识别区氨基酸序列的左锌指核酸酶:Left zinc finger nuclease having the following zinc finger recognition region amino acid sequence: F1:DRSNLLS(SEQ ID NO:39);F1: DRSNLLS(SEQ ID NO: 39); F2:NQSNLLR(SEQ ID NO:40);F2: NQSNLLR (SEQ ID NO: 40); F3:FHSNLLA(SEQ ID NO:18);F3: FHSNLLA (SEQ ID NO: 18); F4:QLSTLNY(SEQ ID NO:41);和F4: QLSTLNY(SEQ ID NO: 41); and F5:QSGNLSR(SEQ ID NO:42);F5: QSGNLSR (SEQ ID NO: 42); or 具有以下锌指识别区氨基酸序列的右锌指核酸酶:A right zinc finger nuclease having the following zinc finger recognition region amino acid sequence: FHSGLLA(SEQ ID NO:20);FHSGLLA (SEQ ID NO: 20); HPSTLSK(SEQ ID NO:43);HPSTLSK (SEQ ID NO: 43); ARWTLDC(SEQ ID NO:44);ARWTLDC (SEQ ID NO: 44); HPSTLSK(SEQ ID NO:43);HPSTLSK (SEQ ID NO: 43); HPSTLSK(SEQ ID NO:43);和HPSTLSK (SEQ ID NO: 43); and RKFTLTN(SEQ ID NO:45)。RKFTLTN (SEQ ID NO: 45). 一种产生FUT8缺陷型细胞系的方法,所述方法包括:A method for generating a FUT8-deficient cell line, the method comprising: (a)通过根据权利要求7所述的方法使细胞中的FUT8基因失活;和(a) inactivating the FUT8 gene in a cell by the method according to claim 7; and (b)在适合产生FUT8缺陷型细胞系的条件下培养所述细胞;(b) culturing the cells under conditions suitable for generating a FUT8-deficient cell line; 具体地,所述细胞为哺乳动物细胞;更具体地,所述细胞为CHO细胞。Specifically, the cell is a mammalian cell; more specifically, the cell is a CHO cell. 一种在宿主细胞中产生目标重组蛋白的方法,所述方法包括:A method for producing a target recombinant protein in a host cell, the method comprising: (a)提供根据权利要求6所述的细胞,所述细胞为FUT8缺陷型细胞,(a) providing the cell according to claim 6, wherein the cell is a FUT8-deficient cell, (b)将包含编码所述目标重组蛋白的核酸的表达载体引入所述细胞,由此产生所述目标重组蛋白;(b) introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell, thereby producing the target recombinant protein; 具体地,所述细胞为哺乳动物细胞;更具体地,所述细胞为CHO细胞。Specifically, the cell is a mammalian cell; more specifically, the cell is a CHO cell. 一种在宿主细胞中产生目标重组蛋白的方法,所述方法包括:A method for producing a target recombinant protein in a host cell, the method comprising: (a)提供包含内源FUT8基因的细胞;(a) providing a cell comprising an endogenous FUT8 gene; (b)通过根据权利要求7所述的方法使所述细胞中的内源FUT8基因失活;和(b) inactivating the endogenous FUT8 gene in the cell by the method according to claim 7; and (c)将包含编码所述目标重组蛋白的核酸的表达载体引入所述细胞,由此产生所述目标重组蛋白;(c) introducing an expression vector comprising a nucleic acid encoding the target recombinant protein into the cell, thereby producing the target recombinant protein; 更具体地,步骤(b)在步骤(c)之前、同时或之后进行。More specifically, step (b) is performed before, simultaneously with or after step (c). 根据权利要求1所述的锌指蛋白、根据权利要求2所述的锌指核酸酶、根据权利要求3或4所述的多核苷酸或根据权利要求5所述的载体用于产生FUT8缺陷型细胞的用途。Use of the zinc finger protein according to claim 1, the zinc finger nuclease according to claim 2, the polynucleotide according to claim 3 or 4, or the vector according to claim 5 for producing FUT8-deficient cells. 根据权利要求1所述的锌指蛋白、根据权利要求2所述的锌指核酸酶、根据权利要求3或4所述的多核苷酸、根据权利要求5所述的载体或根据权利要求6所述的细胞用于产生去岩藻糖基化的含Fc重组蛋白的用途。Use of the zinc finger protein according to claim 1, the zinc finger nuclease according to claim 2, the polynucleotide according to claim 3 or 4, the vector according to claim 5 or the cell according to claim 6 for producing a defucosylated Fc-containing recombinant protein. 根据权利要求1所述的锌指蛋白、根据权利要求2所述的锌指核酸酶、根据权利要求3或4所述的多核苷酸、根据权利要求5所述的载体或根据权利要求6所述的细胞用于产生能引发增强ADCC效应的抗体(如单克隆抗体)的用途。Use of the zinc finger protein according to claim 1, the zinc finger nuclease according to claim 2, the polynucleotide according to claim 3 or 4, the vector according to claim 5 or the cell according to claim 6 for producing an antibody (such as a monoclonal antibody) that can induce an enhanced ADCC effect. 一种FUT8缺陷型细胞系,其根据权利要求9所述的方法制备。A FUT8-deficient cell line prepared according to the method of claim 9. 一种FUT8缺陷型细胞系,其在FUT8基因座处包含如SEQ ID NO:52、SEQ ID NO:53、SEQ ID NO:54、SEQ ID NO:55、SEQ ID NO:56、SEQ ID NO:57、SEQ ID NO:58和SEQ ID NO:59中一或多者所示的核苷酸序列。A FUT8-deficient cell line, which comprises a nucleotide sequence as shown in one or more of SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58 and SEQ ID NO: 59 at the FUT8 locus. 一种FUT8缺陷型细胞系,其包含经改造的FUT8基因座,所述经改造的FUT8基因座能通过以下引物对被检测:A FUT8-deficient cell line comprising an altered FUT8 locus, wherein the altered FUT8 locus can be detected by the following primer pair: 引物对(i):Primer pair (i): 正向引物:AGCCTGAAGTACATAGCCGA(SEQ ID NO:46);和Forward primer: AGCCTGAAGTACATAGCCGA (SEQ ID NO: 46); and 反向引物:TGCCACTGCTTCTATATACTGATTC(SEQ ID NO:47);Reverse primer: TGCCACTGCTTCTATATACTGATTC (SEQ ID NO: 47); 和/或and/or 引物对(ii):Primer pair (ii): 正向引物:GACGCACTGACAAAGTGGGA(SEQ ID NO:48);和Forward primer: GACGCACTGACAAAGTGGGA (SEQ ID NO: 48); and 反向引物:GGTCTGTTCCATCCCCAGAATG(SEQ ID NO:49);Reverse primer: GGTCTGTTCCATCCCCAGAATG (SEQ ID NO: 49); 和/或and/or 引物对(iii):Primer pair (iii): 正向引物:CTGTTGATTCCAGGTTCCCA(SEQ ID NO:50);和Forward primer: CTGTTGATTCCAGGTTCCCA (SEQ ID NO: 50); and 反向引物:TGTTACTTAAGCCCCAGGC(SEQ ID NO:51)。Reverse primer: TGTTACTTAAGCCCCAGGC (SEQ ID NO: 51). 一种培育FUT8缺陷型细胞系的方法,其包括:A method for cultivating a FUT8-deficient cell line, comprising: (a)提供根据权利要求15-17中任一项所述的FUT8缺陷型细胞系;和(a) providing a FUT8-deficient cell line according to any one of claims 15 to 17; and (b)培养或扩增所述细胞系的细胞。(b) culturing or expanding cells of said cell line.
PCT/CN2024/143321 2023-12-28 2024-12-27 Method for constructing cell which produces defucosylated protein and use of cell Pending WO2025140609A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2023142729 2023-12-28
CNPCT/CN2023/142729 2023-12-28

Publications (1)

Publication Number Publication Date
WO2025140609A1 true WO2025140609A1 (en) 2025-07-03

Family

ID=96216878

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2024/143321 Pending WO2025140609A1 (en) 2023-12-28 2024-12-27 Method for constructing cell which produces defucosylated protein and use of cell

Country Status (2)

Country Link
TW (1) TW202525834A (en)
WO (1) WO2025140609A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6453242B1 (en) * 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US20070042378A1 (en) * 2003-12-23 2007-02-22 Kim Jin-Soo Regulation of prokaryotic gene expression with zinc finger proteins
CN101883850A (en) * 2007-07-12 2010-11-10 桑格摩生物科学股份有限公司 Methods and compositions for inactivating alpha-1,6 fucosyltransferase (FUT8) gene expression
CN102264915A (en) * 2008-10-29 2011-11-30 桑格摩生物科学股份有限公司 Methods and compositions for inactivating glutamine synthetase gene expression
US20120058102A1 (en) * 2010-03-29 2012-03-08 The Trustees Of The University Of Pennsylvania Pharmacologically induced transgene ablation system
CN102858985A (en) * 2009-07-24 2013-01-02 西格马-奥尔德里奇有限责任公司 Method for genome editing
US20140249044A1 (en) * 2013-03-01 2014-09-04 Technophage, Investigaçâo E Desenvolvimento Em Biotecnologia, SA Cell-Based Methods for Coupling Protein Interactions and Binding Molecule Selection and Diversification

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6453242B1 (en) * 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US20070042378A1 (en) * 2003-12-23 2007-02-22 Kim Jin-Soo Regulation of prokaryotic gene expression with zinc finger proteins
CN101883850A (en) * 2007-07-12 2010-11-10 桑格摩生物科学股份有限公司 Methods and compositions for inactivating alpha-1,6 fucosyltransferase (FUT8) gene expression
CN102264915A (en) * 2008-10-29 2011-11-30 桑格摩生物科学股份有限公司 Methods and compositions for inactivating glutamine synthetase gene expression
CN102858985A (en) * 2009-07-24 2013-01-02 西格马-奥尔德里奇有限责任公司 Method for genome editing
US20120058102A1 (en) * 2010-03-29 2012-03-08 The Trustees Of The University Of Pennsylvania Pharmacologically induced transgene ablation system
US20140249044A1 (en) * 2013-03-01 2014-09-04 Technophage, Investigaçâo E Desenvolvimento Em Biotecnologia, SA Cell-Based Methods for Coupling Protein Interactions and Binding Molecule Selection and Diversification

Also Published As

Publication number Publication date
TW202525834A (en) 2025-07-01

Similar Documents

Publication Publication Date Title
US11932883B2 (en) CRISPR-associated (Cas) protein
CN118726313A (en) CAS9 mutant gene of Streptococcus pyogenes and polypeptide encoded thereby
KR20240036729A (en) Class ii, type v crispr systems
KR20210152597A (en) Chimeric genome engineering molecules and methods
JP2012514595A (en) Novel zinc finger nuclease and its use
CN112301024A (en) Improving the specificity of RNA-guided genome editing using RNA-guided FokI nuclease (RFN)
CN112654702A (en) Compositions and methods for improved nucleases
JP7561232B2 (en) Efficient selection of recombinant proteins
JP5662363B2 (en) Method for clarifying protein fusion factor (TFP) for secretion of difficult-to-express protein, method for producing protein fusion factor (TFP) library, and method for recombinant production of difficult-to-express protein
CN116179513B (en) A Cpf1 protein and its application in gene editing
EP4530351A2 (en) Compositions and methods for improved gene editing
JP2023062130A (en) Cell selection methods based on CRISPR/Cas-controlled incorporation of detectable tags into target proteins
KR20200066340A (en) Method of making protein
CN111954713A (en) Organisms and methods for producing sugar molecules with low sulfation
WO2025140609A1 (en) Method for constructing cell which produces defucosylated protein and use of cell
EP4271805A1 (en) Novel nucleic acid-guided nucleases
US20230242922A1 (en) Gene editing tools
CN109963946B (en) Complexes and methods for inactivation of the FUT8 gene
WO2024200857A1 (en) Cho cells with optimized host cell protein profile
WO2025132815A1 (en) Novel cas nucleases and polynucleotides encoding the same
CN120731221A (en) CHO cells with optimized ECM profile
HK40055450A (en) Efficient selectivity of recombinant proteins
Yu et al. Ustilago maydis

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: 24911525

Country of ref document: EP

Kind code of ref document: A1