WO2021170089A1 - Engineering immune cells via simultaneous knock-in and gene disruption - Google Patents

Engineering immune cells via simultaneous knock-in and gene disruption Download PDF

Info

Publication number
WO2021170089A1
WO2021170089A1 PCT/CN2021/078164 CN2021078164W WO2021170089A1 WO 2021170089 A1 WO2021170089 A1 WO 2021170089A1 CN 2021078164 W CN2021078164 W CN 2021078164W WO 2021170089 A1 WO2021170089 A1 WO 2021170089A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
host cell
cells
cell
trac
Prior art date
Application number
PCT/CN2021/078164
Other languages
French (fr)
Inventor
Bo Feng
Chenzi ZHANG
Xiangjun HE
Jingyi Wang
Original Assignee
The Chinese University Of Hong Kong
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 The Chinese University Of Hong Kong filed Critical The Chinese University Of Hong Kong
Priority to CN202180031193.5A priority Critical patent/CN115552015A/en
Publication of WO2021170089A1 publication Critical patent/WO2021170089A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses

Definitions

  • Chimeric antigen receptor (CAR) -T cell therapy is a kind of treatment in which a donor’s T cells are genetically modified ex vivo to express a CAR specific to a tumor antigen, expanded in culture, and then infused back to patients to exert the programmed function.
  • CAR-T therapy represents an incredibly promising cellular immunotherapy approach for cancer treatment, and it has now been extensively applied not only in dealing with blood malignancy but also in treating some solid tumors [1, 2] .
  • the standard procedure of CAR-T treatment relies on autologous cell transfer, which is expensive, time-consuming and often difficult to generate CAR-T cells of sufficient quality and quantity for autologous transplantation from patients suffering from late-stage cancers.
  • the current CAR-T cells are generated through undefined integration of CAR cassettes in the genome of patients’ T cells using viral vectors (mainly the ⁇ -retrovirus and lentivirus) .
  • CAR-T cells produced via this strategy had been adopted in two FDA-approved autologous CAR-T therapies, Kymriah and Yescarta TM , and both showed very high response rate in treating patients with refractory/relapse ALL [2, 20] .
  • CAR-T therapies concerns about the unpredictable and varied CAR expression due to the random integrations at different genome regions still remains [1] .
  • GVHD graft-versus-host-disease
  • T cells derived from healthy donors T cells were genetically modified by integration of anti-CD19 CAR, and knockout of TRAC and CD52, to allow its application in non-HLA matched patients. Although the trials are still on going, the preliminary data showed mild and low incidence of GVHD and promising anti-tumor response [11] .
  • Described herein are methods and compositions for highly efficient production of modified host cells that can be used as universal CAR-T cells.
  • the host cells are engineered to simultaneously disrupt genetic loci that encode proteins that can trigger graft-versus-host-disease (GVHD) , and insert a polynucleotide encoding an antigen binding protein at the genetic locus, where the antigen binding protein specifically binds an antigen that is expressed by diseased cells, such as cancer or tumor cells.
  • GVHD graft-versus-host-disease
  • the host cells can be modified using genome editing methods, such as CRISPR technology.
  • the method is an vitro method of inserting a polynucleotide sequence at a pre-determined locus in a host cell genome, the method comprising:
  • a donor vector comprising: (1) a polynucleotide sequence encoding at least one polypeptide; (2) a polyA segment at the 3' end of the polynucleotide sequence; and (3) two homology fragments sharing identical sequences to the flanking regions of a target sequence in the host cell genome, one located at a 5'-end of the polynucleotide sequence and the other one located at a 3'-end of the polyA segment in the donor vector; and
  • a ribonucleoprotein complex comprising (1) a Cas nuclease, and (2) at least two small guide RNAs (sgRNA) that are complementary to at least two selected nucleic acid sequences within the pre-determined locus in the host cell genome.
  • RNP ribonucleoprotein complex
  • sgRNA small guide RNAs
  • the donor vector is selected from a plasmid, AAV viral particle, adenovirus particle, lentivirus particle, or a DNA-nanoparticle complex.
  • the donor vector further comprises at least one polynucleotide sequence encoding a CAR that has anti-tumor activity.
  • the donor vector further comprises at least one polynucleotide sequence encoding a CAR that specifically binds a tumor antigen, for example, an antigen expressed by a tumor cell.
  • the donor vector further comprises at least one polynucleotide sequence encoding an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or anti-BCMA CAR protein.
  • the donor vector further comprises at least one polynucleotide sequence encoding an immune checkpoint protein or an anti-immune checkpoint protein. In some embodiments, the donor vector further comprises at least one polynucleotide sequence encoding a reporter gene. In some embodiments, the donor vector further comprises a 2A self-cleaving sequence, an internal ribosome entry site (IRES) element, or a promoter at the 5' end of the at least one gene coding sequence. In some embodiments, the donor vector comprises additional regulatory or linker elements that control expression of a sequence encoding a CAR protein, for example, regulatory or linker elements located downstream or 3’ of the protein coding sequence.
  • the Cas nuclease is selected from a protein or an RNA molecule encoding the protein. In some embodiments, the Cas nuclease is a class 1 endonuclease or class 2 endonuclease. In some embodiments, the Cas nuclease is Cas9, Cpf1, or a Cas orthologue having genome editing function.
  • At least two small guide RNAs (sgRNA) targeting TRAC sequences listed in the Table 2 are used in the ribonucleoprotein complex.
  • the at least two small guide RNAs (sgRNA) in the ribonucleoprotein complex are selected from any sequences that are complementary to at least two selected nucleic acid sequences within a target locus in the host cell genome.
  • the method further comprises detecting an RNA transcribed from the polynucleotide sequence, or a protein encoded by the polynucleotide sequence, expressed by the host cell.
  • the method further comprises evaluating the functionality of the inserted polynucleotide via in vitro or in vivo assays.
  • the in vitro assay is a cytotoxicity assay.
  • the cytotoxicity assay comprises contacting a target cell expressing a target antigen with a modified host cell described herein, wherein the polypeptide specifically binds to the target antigen, and determining the number of cells lysed by the cell.
  • the in vivo assay comprises administering a modified host cell described herein to a living organism, wherein the living organism comprises cells expressing a target antigen that specifically binds the polypeptide, and detecting a decrease in the number of cells that express the target antigen.
  • the living organism is an animal, a mammal or a human.
  • the host cell is isolated from a human with cancer. In some embodiments, the host cell is isolated from a human carrying an inherited disease.
  • the pre-determined locus is TRAC, ACTB, or GAPDH.
  • the at least one guide RNA comprises a nucleic acid region of about 20 nucleotides that is complementary to the pre-determined polynucleotide sequence in the host cell genome.
  • the method further comprises administering the host cell to a subject to treat a disease in a subject.
  • the disease is a hematological cancer.
  • a modified host cell in another aspect, is provided.
  • the modified host cell is produced by the method described herein.
  • the modified host cell is an immune cell, such as a T cell.
  • a pharmaceutical composition comprsing a modified host cell described herein is provided.
  • a method for treating a disease in a subject comprising: administering a therapeutically effective amount of a one or more (or a plurality of) modified host cell (s) described herein to the subject.
  • the modified host cell is an autologous immune cell isolated from the subject.
  • the modified host cell is an allogeneic immune cell.
  • the method further comprises detecting expression of a RNA or protein encoded by the polynucleotide sequence in the donor vector. In some embodiments, the method further comprises confirming the expression of the RNA or protein encoded by the polynucleotide sequence is sufficient to treat the disease. In some embodiments, the the disease is a hematological cancer.
  • one or more (or a plurality of) modified host cell (s) described herein for use in treating a disease is provided.
  • the modified host cell is an autologous immune cell isolated from the subject.
  • the modified host cell is an allogeneic immune cell.
  • the disease is a hematological cancer.
  • modified host cell in another aspect, use of one or more (or a plurality of) modified host cell (s) described herein in the preparation of a pharmaceutical composition for the treatment of a disease is provided.
  • the modified host cell is an autologous immune cell isolated from the subject.
  • the modified host cell is an allogeneic immune cell.
  • the disease is a hematological cancer.
  • kits for treating a somatic tissue disease comprises:
  • a first container comprising a donor vector, wherein the donor vector comprises a polynucleotide encoding a CAR;
  • a second container comprising at least two sgRNA that is complementary to a selected nucleic acid sequence in the host cell genome
  • the kit further comprises a Cas or CfPl protein or an RNA encoding a Cas or Cfpl protein. In some embodiments, the kit further comprises an instruction manual.
  • Fig. 1 CRISPR/Cas9-mediated HDR targeting at TRAC locus by different sgRNAs.
  • Fig. 1A Schematics for Cas9-RNP based TRAC HDR editing using different sgRNAs, and the design of AAV donor TRAC-800bp for TRAC editing. Arrows indicate the primers for genome PCR amplification in T7E1 assay.
  • Fig. 1B T7E1 assay for different sgRNAs targeting at TRAC locus in Jurkat cells, and the control group was included without TRAC editing.
  • Fig. 1C Flow cytometry analysis for TRAC targeting using AAV6 TRAC-800bp donor and different sgRNAs in Jurkat cells.
  • Fig. 1D Flow cytometry analysis for TRAC targeting using AAV6 TRAC-800bp donor and different sgRNAs in Jurkat cells.
  • X axis indicates the GFP expression level, and the GFP percentages were shown in the flow plot.
  • Fig. 2 CRISPR/Cas9-mediated HDR targeting at TRAC locus by different AAV donors.
  • Fig. 2A Schematics for Cas9-RNP based TRAC targeting using different AAV HDR donors, including TRAC-800bp, TRAC-400bp and TRAC-2A-800bp donors.
  • Fig. 2B Flow cytometry analysis in Jurkat cells for TRAC targeting using different donors shown in A.
  • X axis indicates the GFP expression level, and the GFP percentage were shown in the flow plot.
  • Fig. 2C Schematics of Cas9 RNP based TRAC targeting using AAV donors of TRAC-800bp-LRA, TRAC-SFFV-GFP and TRAC-EF1 ⁇ -GFP donors.
  • Fig. 2D Schematics of Cas9 RNP based TRAC targeting using AAV donors of TRAC-800bp-LRA, TRAC-SFFV-GFP and TRAC-EF1 ⁇ -GFP donors.
  • Fig. 2E Flow cytometry for TRAC protein detection after TRAC targeting in Jurkat cells at day 15 post electroporation.
  • X axis indicates the GFP signal representing the TRAC donor Knock-in, while Y axis indicates the PE level suggesting the TRAC expression in Jurkat cells.
  • Fig. 2F Bar chart showing the TRAC positive percentages of Jurkat cells after Cas9 RNP targeting.
  • Fig. 3 CRISPR/Cas9-mediated HDR transgene integration at TRAC locus using AAV donor and paired sgRNAs for TRAC gene disruption in Jurkat cells and T cells.
  • Fig. 3A Schematics for Cas9-RNP based TRAC targeting using different paired sgRNAs combined with AAV donors.
  • Fig. 3B Flow cytometry analysis for TRAC targeting using either single or paired sgRNAs (sg19-19bp &24) and combined with TRAC-800bp HDR donor, in Jurkat cells.
  • Fig. 3D Flow cytometry analysis for TRAC staining targeted by paired sgRNAs of sg19-19bp &24 with TRAC-800bp-LRA donor, and paired sgRNAs of sg5 &24 with TRAC-2A-800bp donor, in Jurkat cells.
  • Fig. 3E Flow cytometry analysis for TRAC staining targeted by paired sgRNAs of sg19-19bp &24 with TRAC-800bp-LRA donor, and paired sgRNAs of sg5 &24 with TRAC-2A-800bp donor, in Jurkat cells.
  • Fig. 3F Flow cytometry analysis of TRAC targeting in T cells.
  • X axis indicates the GFP expression level, and the GFP percentages were shown in the flow plot.
  • Fig. 3G Flow cytometry analysis of the TRAC expression level using the electroporated cells in Fig. 3B.
  • Fig. 3H Flow cytometry analysis of TRAC targeting in T cells on day 4 or 4 weeks post electroporation when a higher MOI of AAV6 was applied for the transduction.
  • Fig. 4 CRISPR/Cas9-HDR-mediated CAR knockin at TRAC locus to generate universal CAR-T cells.
  • Fig. 4A (i) Schematics for Cas9-RNP mediated TRAC knock-in using AAV virus of TRAC-E2A-CAR and TRAC-SFFV-CAR. (ii) Schematics of Lenti-SFFV-CAR vector and Lenti-EF1 ⁇ -CAR vector for lentivirus transduction in human T cells.
  • Fig. 4B Flow cytometry analysis of TRAC targeting in human T cells using Cas9 RNP based targeting.
  • Fig. 4C Cytotoxicity analysis of different CAR-T cells targeting to CD19+ BV173-LUC (i) and CD19-OCI-AML3-LUC (ii) cells. T cells without genomic modification were included as control. “*” indicates that the p value is less than 0.05 when compare to control T cell group. The experiments were done in duplicates.
  • a “gene” or “reporter” refers to a polynucleotide sequence encoding a protein product that can generate, under appropriate conditions, a detectable signal that allows detection for indicating the presence and/or quantity of the reporter gene protein product.
  • cell refers to a microorganism and includes an individual cell or cell culture that can be or has been a recipient of any recombinant vector (s) or isolated polynucleotide (s) of the invention.
  • host cell refers to a cell that is capable of being modified by the methods described herein, and includes cells derived from animals and mammals, including cells derived from livestock, companion animals, rats, mice and humans. Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change.
  • a host cell includes cells into which a recombinant vector or a polynucleotide of the disclosure has been introduced, including by transformation, transfection, and the like.
  • Cas9 or (CRISPR associated protein 9) is an RNA-guided DNA endonuclease enzyme associated with the CRISPR (Clustered Regularly Interspersed Palindromic Repeats) adaptive immunity system in Streptococcus pyogenes, among other bacteria.
  • S. pyogenes utilizes Cas9 to memorize and later interrogate and cleave foreign DNA, such as the DNA of an invading bacteriophage.
  • Cas9 complexed with a small guide RNA (sgRNA) , performs this interrogation by unwinding foreign DNA and checking whether the DNA contains any sequence segment complementary to a 20 bp spacer region of the sgRNA. If the sgRNA finds sequence complementarity in the DNA, it is cleaved by Cas9.
  • sgRNA small guide RNA
  • sgRNA or “small guide RNA” refers to a short RNA molecule that is capable of forming a complex with Cas9 protein and contains a segment of about 20 nucleotides complementary to a target DNA sequence, such that the Cas9-sgRNA complex directs Cas9 cleavage of a target DNA sequence upon the sgRNA recognizing the complementary sequence in the target DNA sequence.
  • a sgRNA is approximately a 20-base sequence (ranging from about 10-50, 15-45, or 20-40, for example, 15, 20, 25, or 30 bases) specific to the target DNA 5’ of a non-variable scaffold sequence.
  • 3’-UTR is a term of the art understood by skilled persons and means the section of messenger RNA (mRNA) that immediately follows the translation termination codon.
  • mRNA messenger RNA
  • An mRNA molecule is transcribed from the DNA sequence and is later translated into protein.
  • IVS is a term of the art understood by skilled persons and means internal ribosome entry site segments which are known to attract eukaryotic ribosomal translation initiation complex and thus promote translation initiation independently of the presence of the commonly utilized 5'-terminal 7mG cap structure.
  • eGFP is a term of the art understood by skilled persons and means enhanced green fluorescent protein with F64L point mutation which folds the efficiency at 37 °C.
  • eGFP leads to the significant performance of GFPs in mammalian cells.
  • Luc is a term of the art understood by skilled persons and means firefly luciferase protein which is an enzyme catalyzing the oxidation of a luciferin and causing it to produce a visible glow.
  • GAPDH is a term of the art understood by skilled persons and means a housekeeping gene which produces Glyceraldehyde 3-phosphate dehydrogenase. GAPDH gene is often stably and constitutively expressed at high levels in most human tissues and cells. Thus, GAPDH is commonly used as control for western blot to check protein expression levels or for qPCR to check mRNA expression levels.
  • expression cassette or “construct” or “vector” or “donor plasmid” or “donor vector” are used interchangeably and refer to a nucleic acid construct that, when introduced into a host cell, results in transcription and/or translation of an RNA or polypeptide, respectively.
  • an expression cassette is a polynucleotide construct that comprises a polynucleotide sequence encoding a polypeptide of the invention protein operably linked to a promoter, e.g., its native promoter, where the expression cassette is introduced into a heterologous microorganism or host cell.
  • an expression cassette comprises a polynucleotide sequence encoding a polypeptide of the invention where the polynucleotide is targeted to a position in the genome of a microorganism or host cell such that expression of the polynucleotide sequence is driven by a promoter that is present in the microorganism or host cell.
  • target sequence or “target DNA sequence, ” when used to refer to a pre-determined segment of a genomic sequence or polynucleotide construct of this invention (e.g., a donor plasmid) , is similarly defined in regard to the percentage sequence identity between the target sequence and its corresponding sgRNA.
  • CRISPR system refers to a prokaryotic immune system that confers resistance to foreign genetic elements.
  • CRISPR is short for clustered regularly interspaced short palindromic repeats which are segments of prokaryotic DNA containing short, repetitive base sequences acquired from plasmid or phages. These segments could be transcribed into RNA and form as a scaffold to bind with CRISPR-associated protein (Cas protein, such as Cas9) . The combined complex would be directed to degrade the target sequence recognized by these transcribed segments to acquire immunity.
  • Cas protein CRISPR-associated protein
  • AAV adeno-associated virus
  • adeno-associated virus refers to a viral vector system developed from naturally prevalent nonpathogenic adeno-associated virus, a small virus which infects humans and some other mammalian species.
  • the AAV viral vector has a very low immunogenicity, can infect both dividing and quiescent cells and persist in an extrachromosomal state without integrating into the genome of the host cell.
  • AAV donor refers to a vector that is used as donor template and carries a variable set of elements required for subsequent genome editing and knock-in.
  • HDR or “homology directed recombination” or “homologous recombination” refers to a DNA repair mechanism. Facing DNA break, a cell could use the sister chromatin or any provided donors to repair it based on the intact allele or template through pairing homologous sequence around the break site.
  • NHEJ non-homologous end joining
  • Facing DNA break a cell could repair the break site through directly linking the DNA ends together, and during this process, small nucleotides may be inserted or deleted.
  • RNP is a term of the art understood by skilled persons and means a ribonucleoprotein complexes that comprising of Cas9 protein and guide RNA oligonucleotides, and is applied for genome editing in cells or in animal models.
  • CD19 is a term of the art understood by skilled persons and also known as CD19 molecule, and it encodes a transmembrane protein that is expressed in all B lineage cells, except for plasma cells and in follicular dendritic cells. It is a biomarker for B lymphocyte development, lymphoma diagnosis and can be utilized as a target for leukemia immunotherapies.
  • T cell is a term of the art understood by skilled persons and means constant region of T cell receptor alpha chain.
  • Alpha-beta T cell receptors are antigen specific receptors which are essential to the immune response and are present on the cell surface of T lymphocytes.
  • Jurkat cells As used herein the term “Jurkat cells” , “Jurkat cell” or “Jurkat” is a term of the art understood by skilled persons and means an immortalized cell line of human T lymphocytes, and it is widely used for studying acute T cell leukemia, T cell signaling and functions Luciferase.
  • activated T cell refers to a T cell that has been activated by CD3 and CD28 co-stimulation signals. Activated T cells typically divide rapidly in culture medium and secrete cytokines that regulate or assist the immune response.
  • BV173 is a term of the art understood by skilled persons, and the BV173 means a cell line derived from a patient with Philadelphia chromosome (Ph1) -positive acute leukemia.
  • BV173-Luc is a term of the art understood by skilled persons.
  • the BV173-Luc cells were derived from the BV173 cells by integrating a constant luciferase-GFP cassette into the cell genome through lentivirus transduction.
  • the BV173-luc cells have been confirmed with CD19 positive, and were used for the detecting the cytotoxicity of CAR-T cells.
  • OCI-AML3 is a term of the art understood by skilled persons.
  • Tthe OCI-AML3 is a cell line established from the peripheral blood of a 57-year-old man with acute myeloid leukemia at diagnosis in 1987, and the cells carry an NPM1 gene mutation (type A) and the DNMT3A R882C mutation.
  • OCI-AML3-Luc is a term of the art understood by skilled persons.
  • the OCI-AML3-Luc were derived from the OCI-AML3 cells by integrating a constant luciferase-GFP cassette into the cell genome through lentivirus transduction.
  • the BV173-luc cells have been confirmed with CD19 positive, and were used for the detecting the cytotoxicity of CAR-T cells.
  • antigen binding protein refers to a protein or polypeptide that binds with high affinity to a specific target antigen.
  • chimeric antigen receptor As used herein the term “chimeric antigen receptor” , “CAR” or “CARs” is a term of the art understood by skilled persons that refers to a type of antigen binding protein that comprises a fusion protein of a selected single-chain fragment variable domain from a specific monoclonal antibody and one or more T-cell receptor intracellular signaling domains.
  • CAR-T cell therapy is a term of the art understood by skilled persons and means a type of treatment in which involves genetic modification of patient's autologous T-cells to express a CAR specific for a tumor antigen, following by ex vivo cell expansion and re-infusion back to the patient.
  • This T-cell genetic modification can occur either via viral-based gene transfer methods or nonviral methods, such as DNA-based transposons, CRISPR/Cas9 technology or direct transfer of in vitro transcribed-mRNA by electroporation.
  • T cell is a term of the art understood by skilled persons and means a type of lymphocyte which develops in the thymus gland and plays a central role in the immune response. T cells can be distinguished from other lymphocytes by the presence of T-cell receptor on the cells surface.
  • GVHD is a term of the art understood by skilled persons and means a syndrome characterized by inflammation in different organs, with the specificity of epithelial cell apoptosis and crypt drop out.
  • GVDH is commonly associated with stem cell transplants such as those that occur with bone marrow transplants, and could also be induced by CAR therapies based on donor leukocyte infusion, virus-specific T cells, T-cell receptor–deficient T cells, lymphoid progenitor cells, and regulatory T cells.
  • HEK293T is a term of the art understood by skilled persons and means a variant of human embryonic kidney 293 cells (HEK293) that contains the SV40 large T-antigen.
  • the antigen allows episomal replication of transfected plasmids containing the SV40 origin of replication, which leads to the amplification of transfected plasmids and extended temporal expression of the desired gene products.
  • in vitro refers to methods that are performed outside of a living organism, typically using biological materials that are isolated from their normal biological host or usual biological context.
  • 2A self-cleaving sequence refers to a class of 18-22 amino acid peptides that were originally identified in viruses and can mediate ribosome-skipping events. Including one or more 2A self-cleaving sequences in a protein-coding sequence enables the generation of two or more separate peptide products from one mRNA.
  • orthologue or “orthologous sequences” refers to homologous nucleic acid or amino acid sequences that are descended from the same ancestral sequence separated by a speciation event.
  • homolog or “homologous sequences” refers to nucleic acid or amino acid sequences that descended from a common ancestral sequence.
  • percent identity refers to two or more nucleic acid or amino acid sequences or subsequences that have a specified percentage of nucleotides or amino acid residues that are the same, e.g., at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region, when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using a sequence comparison algorithm described herein or by manual alignment and visual inspection.
  • two or more nucleic acid or amino acid sequences or subsequences are considered “substantially identical” if they have at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region, or have 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region.
  • nucleic acid or amino acid sequences or subsequences can have greater than 90%identity but less than 100%identity, or any subrange thereof, for example 90%to 99%identity, or 95%to 99%indentity. These definitions also refer to the complement of a test sequence.
  • any sequence disclosed herein can include sequences that are substantially similar to the reference sequence, i.e., have at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region; or have 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region, unless otherwise clear from the context.
  • percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the sequence in the comparison window can comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • the sequence comparison algorithm calculates the percent sequence identities or similarities for the test sequences relative to the reference sequence, based on the program parameters. Default program parameters are commonly used, or alternative parameters can be designated.
  • Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith and Waterman (Adv. Appl. Math. 2: 482, 1970) , by the homology alignment algorithm of Needleman and Wunsch (J. Mol. Biol. 48: 443, 1970) , by the search for similarity method of Pearson and Lipman (Proc. Natl. Acad. Sci. USA 85: 2444, 1988) , by computerized implementations of these algorithms (e.g., GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis. ) , or by manual alignment and visual inspection (see, e.g., Ausubel et al., Current Protocols in Molecular Biology (1995 supplement) ) .
  • Algorithms suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (Nuc. Acids Res. 25: 3389-402, 1977) , and Altschul et al. (J. Mol. Biol. 215: 403-10, 1990) , respectively.
  • HSPs high scoring sequence pairs
  • Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0) .
  • M forward score for a pair of matching residues; always >0
  • N penalty score for mismatching residues; always ⁇ 0
  • W BLAST algorithm parameters
  • W wordlength
  • E expectation
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul, Proc. Natl. Acad. Sci. USA 90: 5873-87, 1993) .
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P (N) ) , which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P (N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, typically less than about 0.01, and more typically less than about 0.001.
  • Described herein is a gene editing method coupled with multiple newly designed AAV donors and paired sgRNAs for highly efficient generation of gene knockout universal CAR-T cells.
  • the method uses CRISPR/Cas9 mediated gene editing.
  • the method inserts a CAR into a genetic locus that encodes a protein that can trigger graft-versus-host-disease (GVHD) .
  • GVHD graft-versus-host-disease
  • the methods described herein produce highly efficient knock-in of transgenes into the TRAC locus and simultaneous disruption of TRAC expression in Jurkat cells.
  • the inventors also demonstrated excellent results using the methods to insert an anti-CD19 CAR at the TRAC locus in human T cells, which produced universal CAR-T cells with prominent anti-tumor activity.
  • the inventors unexpectedly showed that the CRISPR/Cas9 based HDR editing using optimal AAV donor designs combined with paired sgRNAs targeting the TRAC locus have greatly improved the editing efficiency, which provides the potential for the efficient generation of high-performance TRAC knockout universal CAR-T cells for further clinical use.
  • the current disclosure also provides a versatile system, which can be easily modified by changing homology arms and paired sgRNAs to a new target sequence to generate distinct CAR-T cells with a different gene disruption, or by changing to another CAR in the donor to target different tumor antigens.
  • the methods and compositions described herein provide the following advantages.
  • the instant methods provide high-efficiency generation of universal CAR-T cells that could overcome these limitations by enabling the generation of allogenic CAR-T cells using T-cells from healthy donors.
  • the instant methods can potentially provide universal “off-the-shelf” CAR-T products to benefit a broad range of patients in clinical treatments.
  • the instant methods result in targeted integration of CARs at specific genetic loci, which provides consistent expression of CARs from batch to batch, and could diminish the variance of CAR expression, provide reproducible cytotoxicity and therapeutic activities, and thus potentially produce standardized cells for off-shelf products.
  • the defined insertion of CARs can also reduce the clonal effect and risk for tumorigenesis, which is common for CAR-T cells generated via lentivirus transduction due to random integrations.
  • the CAR is inserted at the TRAC locus.
  • the instant methods also provide an optimal design of the donor vector and small guide RNAs (sgRNAs) that, compared to the current single sgRNA-based knock-in approach, could nearly double the donor integration.
  • the instant methods can significantly increase the yields and reduce the number of starting T cells for universal CAR-T cell generation.
  • the instant methods provide a versatile system, which can be easily modified by changing homology arms and paired sgRNAs to a new target sequence to generate distinct CAR-T cells with a different gene disruption, or by changing to another CAR in the donor vector to target different antigens.
  • this technology holds tremendous potential for generating universal CAR-T cells, or for enhancing CAR-T cell performance by disrupting specific genes while simultaneously inserting the CAR at high efficiency.
  • the instant methods provide great potential and value to improve CAR-T technology and broaden its potential to treat various diseases.
  • the genome editing system comprises the CRISPR/Cas9-mediated genome editing system.
  • the CRISPR/Cas9-mediated genome editing system was selected from multiple designs for its high-efficiency knock-in of CAR expression vectors into specific genes for producing universal CAR-T cells.
  • the methods insert a CAR at the TRAC locus, thereby generating CAR-T cells while simultaneously disrupting TRAC expression, which results in universal CAR-T cells that are disabled for recognizing the recipients’ target tissues as nonself and thereby evoke GVHD during transplantation.
  • the instant methods generate universal CAR-T cells by integrating a CAR in the TRAC locus and simultaneously disrupting TRAC expression using a one-step manipulation.
  • the instant disclosure provides an optimal design of the AAV donor and usage of sgRNAs after thorough examination of multiple sgRNAs targeting TRAC locus and several different AAV donors carrying distinct homology arms.
  • the AAV donor coupled with paired sgRNAs could nearly double the donor integration.
  • Using Cas9 RNP combined with HDR donors and paired sgRNAs targeting has successfully fulfilled the requirements of high efficiency in generating universal CAR-T cells, which significantly increased the yields and reduced the usage of starting T cells for universal CAR-T cell generation.
  • the methods described herein can include, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, and recombinant DNA techniques, which are within the skill of one of ordinary skill in the art.
  • General methods are described for example, in T.E. Creighton, Proteins: Structures and Molecular Properties (W.H. Freeman and Company, 1993) ; A.L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition) ; Green &Sambrook, et al., Molecular Cloning: A Laboratory Manual (4th Edition, 2012) ; Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.
  • the modified host cell is a modified immune cell, such as a modified T cell.
  • the modified host cell is an activated T cell.
  • the modified host cell is an autologous cell.
  • the modified host cell is an allogeneic cell.
  • the methods are in vitro methods.
  • the host cell is modified to insert an exogenous polynucleotide at a pre-determined target locus in the host cell genome.
  • the exogenous polynucleotide encodes a protein or polypeptide.
  • the polynucleotide encodes an antigen binding protein (ABP) or antigen-binding fragment thereof.
  • the antigen binding protein is a chimeric antigen receptor (CAR) .
  • the host cells can be modified using the methods described herein.
  • the method comprises inserting a polynucleotide encoding an ABP at a pre-determined locus (also referred to as a “selected locus” or “target locus” ) in the host cell genome.
  • the method comprises homology directed recombination or homologous recombination.
  • the method comprises CRISPR/Cas mediated gene editing.
  • the method comprises CRISPR/Cas mediated gene editing in combination with AAV donor vectors and paired sgRNAs.
  • the CRISPR-associated (Cas) nuclease is a class 1 endonuclease or class 2 endonuclease.
  • the Cas is Cas9.
  • the Cas is CPf1 (now known as Cas12a) .
  • the method comprises contacting a host cell with a donor vector comprising: (1) a polynucleotide sequence coding for at least one polypeptide; (2) a polyA segment at the 3' end of the polynucleotide; and (3) two homology fragments sharing identical sequences to the flanking regions of a target sequence in genome, one located at a 5'-end of the polynucleotide sequence and the other one located at a 3'-end of the polyA segment in the donor vector; followed by contacting the host cell with a ribonucleoprotein complex (RNP) comprising (1) a Cas nuclease, and (2) at least two small guide RNAs (sgRNA) that are complementary to at least two selected nucleic acid sequences within the pre-determined locus in the host cell genome.
  • RNP ribonucleoprotein complex
  • the donor vector comprises a promoter that regulates transcription of the polynucleotide sequence into mRNA in the host cell.
  • the mRNA is translated into a polypeptide in the host cell.
  • the polypeptide comprises an ABP, such as a CAR.
  • the CAR is an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or an anti-BCMA CAR.
  • the pre-determined locus where the exogenous polynucleotide is inserted is the constant region of a T cell receptor alpha chain (TRAC) .
  • the pre-determined locus is the beta ( ⁇ ) -actin (ACTB) locus, or the Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) locus.
  • the CRISPR-Cas gene-editing system is described, for example, in U.S. Patent No. 8,697,359 and U.S. Patent Publication 2014/0068797.
  • the CRISPR-Cas9 system originated from type II CRISPR-Cas systems, which provide bacteria with adaptive immunity to viruses and plasmids.
  • the CRISPR-associated protein Cas9 is an endonuclease that uses a guide sequence within an RNA duplex, tracrRNA: crRNA, to form base pairs with DNA target sequences, enabling Cas9 to introduce a site-specific double-strand break in the DNA.
  • crRNA was engineered as a single guide RNA (sgRNA) that retains two critical features: a sequence at the 5′ side that determines the DNA target site by Watson-Crick base-pairing and a duplex RNA structure at the 3′ side that binds to Cas9. This created a simple two-component system in which changes in the guide sequence of the sgRNA program Cas9 to target any DNA sequence of interest.
  • sgRNA single guide RNA
  • the CRISPR-associated protein (Cas) genes are grouped into two classes, Class 1 and Class 2, and further grouped into at least 35 families based on sequence similarity of the encoded proteins. Therefore, the instant methods can use any Cas protein having the required functional characteristics.
  • the Cas is Cas9, or a protein having at least 90% (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) amino acid sequence identity to Cas9 (SEQ ID NO: 2) .
  • the Cas9 is encoded by a nucleic acid having at least 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) sequence identity to SEQ ID NO: 1) .
  • the Cas is Cas12a (Cpf1) , or a protein having at least 90% (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) sequence identity to Cas12a (SEQ ID NO: 4) .
  • the CRISPR/Cas system is selected from Cas9 (Csn1) , Cas12a (Cpf1) , Cas13a (C2c2) and Cas13b (C2c6) systems or other CRISPR/Cas systems with similar activities.
  • Cas9 Casn1
  • Cas12a Cas12a
  • Cas13a Cas13a
  • Cas13b Cas13b
  • Chimeric antigen receptor (CAR) T-cell therapy is a type of immunotherapy called adoptive cell therapy.
  • CAR T therapy typically involves genetic modification of a patient's autologous T-cells to express a CAR specific for an antigen, followed by ex vivo cell expansion and re-infusion back into the patient.
  • Genetic modification of T-cells can occur using viral-based gene transfer methods or non-viral methods, such as DNA-based transposons, CRISPR/Cas9 technology or direct transfer of in vitro transcribed-mRNA by electroporation.
  • the methods described herein allow for the production of modified T cells that can be used as universal “off the shelf” CAR-T cells.
  • the methods allow for production of modified T cells that do not rely on autologous T-cells isolated from a particular subject or patient, and instead provide modified allogeneic host cells that can be administered to more than one subject or patient for treating a disease.
  • a CAR is a recombinant fusion protein comprising an antigen-specific extracellular domain coupled to an intracellular domain that provides an intracellular signal upon antigen binding to the extracellular domain.
  • CARs are different from other antigen binding agents because they can both bind MHC-independent antigen and transduce activation signals via the intracellular domain.
  • Specific binding of CARs to antigen is typically expressed as an affinity constant or affinity of interaction (KD) , where the K D is between about 0.1 pico Molar (pM) and about 10 micro Molar ( ⁇ M) , or about 0.1 pM to about 1 ⁇ M, or about 0.1 pM to about 100 nano Molar (nM) .
  • KD affinity constant or affinity of interaction
  • an antigen-specific extracellular domain suitable for use in a CAR of the present disclosure may be any antigen-binding polypeptide known in the art.
  • the antigen-binding domain comprises an antibody-based binding domain, for example a single chain Fv (scFv) , a single domain antibody variable domain (sdAb VH) , a camelid antibody variable domain (cAb VHH) or humanized versions thereof, or a shark antibody variable domain (IgNAR VH) or humanized versions thereof.
  • scFv single chain Fv
  • sdAb VH single domain antibody variable domain
  • cAb VHH camelid antibody variable domain
  • IgNAR VH shark antibody variable domain
  • the antigen-binding domain comprises a T-cell receptor (TCR) based binding domain, such as a single chain TCR (scTv) , or a single chain two-domain TCR containing the variable alpha and beta chains.
  • TCR T-cell receptor
  • the CAR recognizes or specifically binds to an antigen expressed on a tumor, cancer or malignant cell.
  • the tumor, cancer or malignant cell is from a tumor of the hematopoietic or lymphoid tissues.
  • the tumor or cancer is a leukemia, such as acute lymphoblastic leukemia (ALL) , acute myeloid leukemia (AML) , chronic lymphocytic leukemia (CLL) and chronic myeloid leukemia (CML) .
  • the tumor is a B cell tumor or T cell tumor.
  • B cell cancers and tumors include B-cell lymphomas, which may be either indolent (slow-growing) or aggressive (fast-growing) .
  • Most B-cell lymphomas are non-Hodgkin lymphomas, including Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) , diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma.
  • the CAR specifically binds to an antigen expressed by a B cell.
  • the CAR specifically binds to CD19, CD20, CD22, CD47, or B-cell maturation antigen (BCMA) .
  • the CAR is an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or anti-BCMA CAR.
  • the CAR comprises an amino acid sequence having at least 90%sequence identity (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%or 100%) to SEQ ID NO: 6 (anti-CD19 CAR) , SEQ ID NO: 8 (anti-CD20 CAR) , SEQ ID NO: 10 (anti-CD22 CAR) , or SEQ ID NO: 12 (anti-BCMA CAR) .
  • SEQ ID NO: 6 anti-CD19 CAR
  • SEQ ID NO: 8 anti-CD20 CAR
  • SEQ ID NO: 10 anti-CD22 CAR
  • SEQ ID NO: 12 anti-BCMA CAR
  • the CAR is encoded by a nucleic acid sequence having at least 90%sequence identity (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%or 100%) to SEQ ID NO: 5 (anti-CD19 CAR) , SEQ ID NO: 7 (anti-CD20 CAR) , SEQ ID NO: 9 (anti-CD22 CAR) , or SEQ ID NO: 11 (anti-BCMA CAR) . Sequences of some examples of various components of CARs of the instant invention, and nucleic acids that encode them are listed in Table 1, where (aa) stands for amino acids, and (na) stands for nucleic acids that encode the corresponding peptide. And in some embodiments, the CAR protein in this design can be changed to other CAR proteins with specific targeting.
  • T-cell cancers and tumors include T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) , T-cell large granular lymphocyte (LGL) leukemia, human T-cell leukemia virus type 1-positive (HTLV-1+) adult T-cell leukemia/lymphoma (ATL) , T-cell prolymphocytic leukemia (T-PLL) , and various peripheral T-cell lymphomas (PTCLs) , including angioimmunoblastic T-cell lymphoma (AITL) , ALK-positive anaplastic large cell lymophoma, and ALK-negative anaplastic large cell lymophoma.
  • the CAR specifically binds to an antigen expressed by a T cell.
  • the CAR specifically binds to CD9, CD7, CD5, CD2, CD30, or CD4.
  • the CAR comprises an intracellular domain that provides an intracellular signal to the T cell upon antigen binding to the antigen-specific extracellular domain.
  • the intracellular signaling domain activates an effector function of the modified T cell.
  • the effector function can include cytolytic activity or helper activity including the secretion of cytokines, or antigen-dependent proliferation.
  • the intracellular domain includes the zeta chain of the T-cell receptor or any of its homologs (e.g., beta, delta, gamma, or epsilon) , MB 1 chain, B29, Fc RIII, Fc RI, and combinations of signaling molecules, such as CD3.
  • zeta the intracellular domain that provides an intracellular signal to the T cell upon antigen binding to the antigen-specific extracellular domain.
  • the intracellular signaling domain activates an effector function of the modified T cell.
  • the effector function can include cytolytic activity or helper activity including the secretion of cytokines, or antigen-dependent proliferation.
  • Intracellular signaling portions of other members of the families of activating proteins may be used, such as Fc. gamma. RIII and Fc. epsilon. RI.
  • the intracellular signaling domain is a truncated portion of the signaling domain that retains functional signal transduction activity.
  • the antigen-specific extracellular domain is linked to the intracellular domain of the chimeric antigen receptor by a transmembrane domain.
  • the CAR further comprises one or more costimulatory domains, and/or one or more spacers.
  • a costimulatory domain can be derived from the intracellular signaling domains of costimulatory proteins that enhance cytokine production, proliferation, cytotoxicity, and/or persistence in vivo.
  • An amino acid spacer can be used to connect the different domains of the CAR together, for example, a spacer can connect the antigen-specific extracellular domain to the transmembrane domain, the transmembrane domain to a costimulatory domain, a costimulatory domain to the intracellular domain, or the transmembrane domain to the intracellular domain.
  • a spacer can connect the antigen-specific extracellular domain to the transmembrane domain, the transmembrane domain to a costimulatory domain, a costimulatory domain to the intracellular domain, or the transmembrane domain to the intracellular domain.
  • inclusion of a spacer domain between the antigen-specific extracellular domain and the transmembrane domain may affect flexibility of the antigen-binding domain and thereby CAR function.
  • the CAR comprises the CD8 hinge-transmembrane domain and/or the co-stimulatory signaling domain of 41bb fused to CD3 zeta.
  • Other co-stimulatory signaling domains e.g., 41bb, CD27, CD28 or a combination of two co-stimulatory domains can also be used to construct different CARs.
  • CAR-T cells described herein are deficient in endogenous T cell receptor (TCR) signaling.
  • TCR T cell receptor
  • decreasing or eliminating endogenous TCR signaling in CAR-T cells may prevent or reduce graft versus host disease (GvHD) when allogenic T cells are used to produce the CAR-T cells.
  • Methods for decreasing or eliminating endogenous TCR signaling include, but are not limited to, modifying a part of the TCR receptor (e.g., the TCR receptor alpha chain (TRAC) , etc. ) .
  • TRAC modification can be used to block TCR mediated signaling, which could allow allogeneic T cells to be modified to express various CARs and administered to patients without inducing life-threatening GvHD.
  • the modified host cells described herein can further comprise one or more suicide genes.
  • a suicide gene generally encodes an enzyme that selectively converts a nontoxic prodrug into highly toxic metabolites, specifically eliminating cells expressing the enzyme.
  • Suicide genes may allow effective tracking and elimination of the CAR-T cells in vivo if required.
  • One examples of suicide genes include herpes simplex virus thymidine kinase (HSV-tk) which phosphorylates the nontoxic antiviral prodrug ganciclovir (GCV) into GCV triphosphate. GCV triphosphate is incorporated into DNA in replicating cells, inhibiting DNA synthesis and resulting in cell death.
  • Other examples include inducible caspase 9 protein, or a CD34/thymidine kinase chimeric suicide gene.
  • the modified host cell is a modified immune cell, such as a modified T cell.
  • the modified host cell is an autologous cell.
  • the modified host cell is an allogeneic cell.
  • the modified host cell is included in a pharmaceutical compositon.
  • the subject is an animal, a mammal, a livestock animal such as a cow, horse, sheep, goat or pig, a companion animal such as a cat or dog, or a human.
  • the disease is a hematological cancer.
  • the disease is a cancer or tumor of the hematopoietic or lymphoid tissues, as described herein.
  • the disease is a leukemia, such as acute lymphoblastic leukemia (ALL) , acute myeloid leukemia (AML) , chronic lymphocytic leukemia (CLL) and chronic myeloid leukemia (CML) .
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myeloid leukemia
  • the disease is a B cell cancer or tumor.
  • B cell cancers and tumors include B-cell lymphomas, which may be either indolent (slow-growing) or aggressive (fast-growing) .
  • Most B-cell lymphomas are non-Hodgkin lymphomas, including .
  • Burkitt lymphoma chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) , diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma.
  • CLL/SLL chronic lymphocytic leukemia/small lymphocytic lymphoma
  • the disease is a tumor that expresses a B cell antigen or marker, such as CD19, CD20, CD22, CD47, or B-cell maturation antigen (BCMA) .
  • a B cell antigen or marker such as CD19, CD20, CD22, CD47, or B-cell maturation antigen (BCMA) .
  • the disease is a T-cell cancer or tumor, such as T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) , T-cell large granular lymphocyte (LGL) leukemia, human T-cell leukemia virus type 1-positive (HTLV-1+) adult T-cell leukemia/lymphoma (ATL) , T-cell prolymphocytic leukemia (T-PLL) , and various peripheral T-cell lymphomas (PTCLs) , including angioimmunoblastic T-cell lymphoma (AITL) , ALK-positive anaplastic large cell lymophoma, and ALK-negative anaplastic large cell lymophoma.
  • the disease is a tumor that expresses a T cell antigen or marker, such as CD7, CD5, CD2, CD30, and CD4.
  • the method is an in vitro method.
  • the in vitro method can be a cytotoxicity assay.
  • a modified host cell described herein, which expresses a polypeptide encoded by the exogenous polynucleotide is contacted with a target cell expressing an antigen that binds to the polypeptide, and the number of target cells that are lysed or killed by the modified host cell is determined.
  • the method is an in vivo method.
  • the method comprising administering, for example, by infusion, a modified host cell described herein to a living animal that expresses a target antigen that binds to the polypeptide encoded by exogenous polynucleotide.
  • the exogenous polynucleotide encodes a chimeric antigen receptor that specifically binds an antigen in the living organism.
  • the modified host cell expresses a CAR that specifically binds an antigen expressed in the living organism.
  • the modified host cell is a T cell.
  • the antigen is expressed by a tumor or cancer cell.
  • Example 1 Efficient CRISPR/Cas9-mediated transgene integration at TRAC locus using AAV donor and single sgRNA for TRAC gene disruption
  • Example 2 Efficient CRISPR/Cas9-mediated transgene integration at TRAC locus using AAV donor and paired sgRNAs for TRAC gene disruption
  • Flow cytometry analysis revealed that applying paired sgRNAs almost doubled editing efficiency compared to single-sgRNA editing, indicating that the paired sgRNAs targeting introduced more efficient donor integration at the TRAC locus (Fig. 3 B, C) . Consistently, the paired sgRNAs targeting also achieved the high efficient TRAC knockout in Jurkat cells (Fig. 3 D) . Furthermore, we tested this method in activated T cells, and obtained similar rates for donor integration and TRAC knockout in activated T cells (Fig. 3 E-G) , indicating the successful and high frequencies of simultaneous donor integration and TRAC disruption in T cells, which provide great value for using this new editing methods for potential clinical use.
  • Example 3 CRISPR/Cas9-mediated CAR knockin at TRAC locus to generate universal CAR-T cells using AAV donor and paired sgRNAs
  • AAV-6 system was used for virus packaging [18] .
  • Transfection was carried out in 293FT cells with 80%confluence, and 3 days after transfection, the virus was collected. Basically, the virus was extracted from the cytoplasm and nuclear, then gradient centrifugation was performed with ultra-speed. Finally, the viral particles were concentrated and further purified with ultra-centrifugal-100KD filter.
  • Transfection was carried out in 293FT cells with 80%confluence, and 3 days after transfection, the cell culture medium was collected, and the virus particles were concentrated and further purified with ultra-centrifugal-100KD filter.
  • Cas9 protein and sgRNA in vitro synthesis were purchased from Thermo Fisher (TrueCut TM Cas9 Protein v2, Thermo Fisher ) , and in vitro transcription (IVT) of sgRNAs used in electroporation were generated by GeneArt TM Precision gRNA Synthesis Kit (Thermo Fisher) according to manual instruction.
  • sgRNA target sequences were designed based on the output from website www. casblastr. org) . All the sgRNA targeting information is listed in Table 2.
  • anti-CD19 CAR-GFP cassette was constructed as described previously [19] . Briefly, the anti-CD19 CAR consisting a single chain variable fragment scFV specific for the human CD19 preceded by a CD8a leader peptide were amplified from plasmid (Addgene 113014) , and fused with 41bb hinge-transmembrane-intracellular regions and CD3 ⁇ intracellular domain amplified from T cell cDNA. The CAR cassette was then fused with a e2a-gfp cassette followed by a polyA element through overlapping PCR.
  • the entire CAR-GFP cassette was inserted into plasmid of CD19-FcGamma CAR (Addgene 113014) at Mlu1 and Not1 sites to generate Lenti-SFFV-CAR-GFP (termed Lenti-SFFV-CAR for short) plasmid.
  • AAV donors The backbone of AAV donors were purchased from addgene (addgene#87115) . Different homology arms of TRAC gene were amplified from human genome, and inserted into the backbone, and IRES-GFP-PA, 2A-GFP-PA, SFFV-GFP-PA, EF1 ⁇ -GFP-PA, 2A-anti-CD19 CAR-GFP-PA, SFFV-anti-CD19 CAR-GFP-PA, EF1 ⁇ -anti-CD19 CAR-GFP-PA cassettes were constructed separately, and inserted between different homology arms to generate AAV donors of TRAC-800bp (SEQ ID NO: 13) , TRAC-400bp (SEQ ID NO: 14) , TRAC-2A-800bp (SEQ ID NO: 15) , TRAC-800bp-LRA (SEQ ID NO: 16) , TRAC-SFFV-GFP (SEQ ID NO: 17) , TRAC-EF1 ⁇ -GFP (SEQ ID
  • Lentivirus vector the anti-CD19 CAR cassette was inserted into plasmid of CD19-FcGamma CAR (Addgene 113014) at Mlu1 and Not1 sites to generate Lenti-SFFV-CAR plasmid, and the EF1 ⁇ promoter was amplified from plasmid (Addgene 109049) , and inserted into backbone of Lenti-SFFV-CAR at MfeI and NcoI to replace the SFFV element, so as to generate Lenti-EF1 ⁇ -CAR vector.
  • EXAMPLE 7 Genomic DNA extraction and PCR detection of genomic integrations
  • Genome DNA from cultured cells was extracted using Genome DNA extraction Kit (Tiangen) following the manufacturer's instruction.
  • Genome DNA extraction Kit Tris buffer and proteinase K was used for overnight digestion at 37°C, then purified with 75%ethanol. 200 ng genomic DNA were generally used for PCR reaction using Phusion High-Fidelity DNA Polymerase (New England Biolabs) , following the manufacturer's instruction.
  • Fluorescence Activated Cell Sorting (FACS) analyzer (BD LSRFortessa Cell Analyzer) was configured with a single 488 nm argon ion laser (200 mW) .
  • the laser is used to induce light scattering by either the excitation of cellular fluorescent proteins (eGFP) or the granularity within the cell.
  • eGFP cellular fluorescent proteins
  • the recorded events within the gate on the FITC-A (GFP) log scale provided a good indication of the GFP expression level and the counts indicate the number of GFP-positive cells.
  • the ration of GFP-positive cells over the total counts in the gated area is defined as targeting efficiency.
  • T cells transduced with CAR can be detected by standard luciferase-assay described previously [8] .
  • ALL-cell lines that express CD19 and luciferase were used as target cells (T)
  • T cells with CAR integration were used as effector cells (E) .
  • 5x104 target cells were seeded in 96-well plates, and co-cultured with the effector cells at different ratios. The final volume in each well was adjusted to 200 ⁇ L.
  • Target cells cultured without effector cells were used to determine the maximal luciferase expression (relative light units, RLUmax) . After 18 hours’ co-culture, 100 ⁇ L luciferase substrate (Promega) was added to each well, and the luminescence was the detected by luciferase imaging system.
  • EXAMPLE 10 Cas9 ribonucleoprotein (RNP) assembly and electroporation
  • DNA fragment covering a targeting region in TRAC gene loci were amplified by genome PCR, and followed with DNA purification process using MEGAquick-spin Total Fragment DNA Purification Kit (iNtRON) according to manufacturer’s instructions.
  • the purified fragments were denatured and annealed and then were treated with T7E1 (T7 endonuclease 1, NEB) at 37 °C for 1 hour to completely cleave the unmatched nucleotide pairs. Subsequently, the editing efficiency was determined using Image J.

Abstract

Methods for producing modified host cells that can be used as universal chimeric antigen receptor (CAR) T cells for treating diseases. The methods include inserting a polynucleotide that encodes a CAR at a specified locus, such as a TCR or HLA gene, in the host cell genome using gene editing technology, such as CRISPR/Cas9-based genome editing. The insertion can inactivate the TCR or HLA gene, thus reducing the expression of genes that can trigger graft-versus-host-disease (GVHD) in a patient.

Description

ENGINEERING IMMUNE CELLS VIA SIMULTANEOUS KNOCK-IN AND GENE DISRUPTION
SEQUENCE LISTING
The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on February 25, 2021, is named 080015-1233866_ (030010PC) _SL. txt and is 543, 002 bytes in size.
BACKGROUND OF THE INVENTION
Chimeric antigen receptor (CAR) -T cell therapy is a kind of treatment in which a donor’s T cells are genetically modified ex vivo to express a CAR specific to a tumor antigen, expanded in culture, and then infused back to patients to exert the programmed function. CAR-T therapy represents an incredibly promising cellular immunotherapy approach for cancer treatment, and it has now been extensively applied not only in dealing with blood malignancy but also in treating some solid tumors [1, 2] . To date, the standard procedure of CAR-T treatment relies on autologous cell transfer, which is expensive, time-consuming and often difficult to generate CAR-T cells of sufficient quality and quantity for autologous transplantation from patients suffering from late-stage cancers. These limitations have greatly hampered the applications of CAR-T therapy.
The main hurdle of using allogenic CAR-T cells is the high risk for graft-versus-host-disease (GVHD) associated with the infusion of immune-competent allogenic T-cells, which carry TCR or HLA to target the host cells. Evidence showed that this could be addressed by inhibiting the expression of TCR or HLA class I gene in CAR-T cells. The breakthrough of CRISPR technology enables targeted genome editing and holds great promise for the development of novel therapeutic strategies to treat human diseases. Recent studies have also attempted to employ CRISPR/Cas9-based genome editing to disrupt/knockout TCR or HLA class I genes to generate of universal CAR-T cell [2, 3] . Thereby the modified T cells without  TRAC/TRBC or HLA class I antigen were not able to identify allogeneic antigens, and avoided generating GVHD.
The current CAR-T cells are generated through undefined integration of CAR cassettes in the genome of patients’ T cells using viral vectors (mainly the γ-retrovirus and lentivirus) . CAR-T cells produced via this strategy had been adopted in two FDA-approved autologous CAR-T therapies, Kymriah and Yescarta TM, and both showed very high response rate in treating patients with refractory/relapse ALL [2, 20] . Despite the effectiveness of these CAR-T therapies, concerns about the unpredictable and varied CAR expression due to the random integrations at different genome regions still remains [1] .
Several studies have reported using ZFNs [4-6] , TALENs [7] and CRISPR/Cas9 [6, 8, 9] for generating universal CAR-T cells with defects in TCR or HLA class I genes. These studies showed the potential of using TRAC-deficient CAR-T cells for off-the-shelf therapy. Torikai et al. conducted a pioneer study to generate universal T cell by using ZFNs to specifically block the endogenous TCR expression [5] . Another study by Qasim et al. used TALEN-based editing for TRAC knockout in anti-CD19 CAR-T cell generation and evaluated the product in two patients [10] . Unfortunately, the therapeutic effects were not satisfactory in both studies. Moreover, one of the infant patient experienced the graft-versus-host-disease (GVHD) due to mismatch of the MHC derived from the unedited T cells with retained TRAC gene, suggesting that a more thorough editing is required for clinical therapy and also highlighting the significance of gene editing efficiency in clinical applications. Ren J et al. performed multiplex genome editing to disrupt TRAC and TRBC expression with high editing frequency, and reduced GVHD responses in animal models [9] . Another study by Eyquem et al. also employed CRISPR/Cas9 to target the TRAC locus, and coupled with CD19 CAR-containing adeno-associated virus (AAV) virus as DNA template for HDR repair. The integration of the anti-CD19 CAR was been driven under the endogenous TRAC promoter, allowing for expression more close to that under physiological regulation of the TRAC, which also led to superior anti-tumor performance [8] . Chen Sidi’s group used a similar AAV-CRISPR-Cpf1 system to integrate dual CARs (CD19 and CD22) into TRAC and PD1 locus to simultaneously disrupted the expression of the TRAC and PD1, and the dual-CAR knock-in system generated universal CAR-T cell with greater potency in cytokine production and cancer cell killing [6] . Two phase I clinical trials using universal anti-CD19  CAR-T cells in adult and pediatric patients have been initiated. Briefly, T cells derived from healthy donors T cells were genetically modified by integration of anti-CD19 CAR, and knockout of TRAC and CD52, to allow its application in non-HLA matched patients. Although the trials are still on going, the preliminary data showed mild and low incidence of GVHD and promising anti-tumor response [11] .
So far, generation of universal CAR-T cell by targeted disruption in TRAC locus has showed great potential in mediating cancer cell clearance, and at the same time, reducing the CAR-T cell immunogenicity. [6, 8] . Two approaches were mainly applied for generating universal TRAC knockout CAR-T cells. One approach combined the lentivirus or retrovirus delivery of the CAR with CRISPR/Cas9-mediated targeting for TRAC gene knockout [12-14] , and another method used CRISPR/Cas9 based TRAC targeting paired with CAR-containing AAV HDR template to knock-in a CAR into TRAC locus [6, 8] . Although both methods produced potent CAR-T cells, the former one required a two-step gene modification for CAR insertion and TRAC knockout, which pose additional difficulties in manufacturing and evaluation processes, and the randomly genome integration nature of the lentivirus may result in oncogenic transformation, diversified transgene expression and transcriptional silencing in CAR-T cells [15, 16] , Which limited its application in clinical trials. Whereas, the latter approach achieved CAR knock-in and TRAC knockout by one single gene targeting, and this greatly simplified the whole procedures. Moreover, the employment of AAV HDR template donor containing promoter-less CAR could result in a more safe and constant CAR expression under the control of endogenous TRAC promoter [17] . Therefore, directing a CAR to the TRAC locus through CRISPR/Cas9-mediated genome editing showed great prospect in generating universal CAR-T cells.
BRIEF SUMMARY OF THE INVENTION
Described herein are methods and compositions for highly efficient production of modified host cells that can be used as universal CAR-T cells. The host cells are engineered to simultaneously disrupt genetic loci that encode proteins that can trigger graft-versus-host-disease (GVHD) , and insert a polynucleotide encoding an antigen binding protein at the genetic locus, where the antigen binding protein specifically binds an antigen that is expressed by diseased cells,  such as cancer or tumor cells. The host cells can be modified using genome editing methods, such as CRISPR technology.
In one aspect, the method is an vitro method of inserting a polynucleotide sequence at a pre-determined locus in a host cell genome, the method comprising:
contacting a host cell sequentially with:
(i) a donor vector comprising: (1) a polynucleotide sequence encoding at least one polypeptide; (2) a polyA segment at the 3' end of the polynucleotide sequence; and (3) two homology fragments sharing identical sequences to the flanking regions of a target sequence in the host cell genome, one located at a 5'-end of the polynucleotide sequence and the other one located at a 3'-end of the polyA segment in the donor vector; and
(ii) a ribonucleoprotein complex (RNP) comprising (1) a Cas nuclease, and (2) at least two small guide RNAs (sgRNA) that are complementary to at least two selected nucleic acid sequences within the pre-determined locus in the host cell genome.
In some embodiments, the donor vector is selected from a plasmid, AAV viral particle, adenovirus particle, lentivirus particle, or a DNA-nanoparticle complex. In some embodiments, the donor vector further comprises at least one polynucleotide sequence encoding a CAR that has anti-tumor activity. In some embodiments, the donor vector further comprises at least one polynucleotide sequence encoding a CAR that specifically binds a tumor antigen, for example, an antigen expressed by a tumor cell. In some embodiments, the donor vector further comprises at least one polynucleotide sequence encoding an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or anti-BCMA CAR protein.
In some embodiments, the donor vector further comprises at least one polynucleotide sequence encoding an immune checkpoint protein or an anti-immune checkpoint protein. In some embodiments, the donor vector further comprises at least one polynucleotide sequence encoding a reporter gene. In some embodiments, the donor vector further comprises a 2A self-cleaving sequence, an internal ribosome entry site (IRES) element, or a promoter at the 5' end of the at least one gene coding sequence. In some embodiments, the donor vector comprises additional regulatory or linker elements that control expression of a sequence encoding a CAR protein, for example, regulatory or linker elements located downstream or 3’ of the protein coding sequence.
In some embodiments, the Cas nuclease is selected from a protein or an RNA molecule encoding the protein. In some embodiments, the Cas nuclease is a class 1 endonuclease or class 2 endonuclease. In some embodiments, the Cas nuclease is Cas9, Cpf1, or a Cas orthologue having genome editing function.
In some embodiments, at least two small guide RNAs (sgRNA) targeting TRAC sequences listed in the Table 2 are used in the ribonucleoprotein complex. In some embodiments, the at least two small guide RNAs (sgRNA) in the ribonucleoprotein complex are selected from any sequences that are complementary to at least two selected nucleic acid sequences within a target locus in the host cell genome.
In some embodiments, the method further comprises detecting an RNA transcribed from the polynucleotide sequence, or a protein encoded by the polynucleotide sequence, expressed by the host cell.
In some embodiments, the method further comprises evaluating the functionality of the inserted polynucleotide via in vitro or in vivo assays. In some embodiments, the in vitro assay is a cytotoxicity assay. In some embodiments, the cytotoxicity assay comprises contacting a target cell expressing a target antigen with a modified host cell described herein, wherein the polypeptide specifically binds to the target antigen, and determining the number of cells lysed by the cell.
In some embodiments, the in vivo assay comprises administering a modified host cell described herein to a living organism, wherein the living organism comprises cells expressing a target antigen that specifically binds the polypeptide, and detecting a decrease in the number of cells that express the target antigen. In some embodiments, the living organism is an animal, a mammal or a human.
In some embodiments, the host cell is isolated from a human with cancer. In some embodiments, the host cell is isolated from a human carrying an inherited disease.
In some embodiments, the pre-determined locus is TRAC, ACTB, or GAPDH.
In some embodiments, the at least one guide RNA comprises a nucleic acid region of about 20 nucleotides that is complementary to the pre-determined polynucleotide sequence in the host cell genome.
In some embodiments, the method further comprises administering the host cell to a subject to treat a disease in a subject. In some embodiments, the disease is a hematological cancer.
In another aspect, a modified host cell is provided. In some embodiments, the modified host cell is produced by the method described herein. In some embodiments, the modified host cell is an immune cell, such as a T cell. In some embodiments, a pharmaceutical composition comprsing a modified host cell described herein is provided.
In another aspect, a method for treating a disease in a subject is provided, the method comprising: administering a therapeutically effective amount of a one or more (or a plurality of) modified host cell (s) described herein to the subject. In some embodiments, the modified host cell is an autologous immune cell isolated from the subject. In some embodiments, the modified host cell is an allogeneic immune cell.
In some embodiments, the method further comprises detecting expression of a RNA or protein encoded by the polynucleotide sequence in the donor vector. In some embodiments, the method further comprises confirming the expression of the RNA or protein encoded by the polynucleotide sequence is sufficient to treat the disease. In some embodiments, the the disease is a hematological cancer.
In another aspect, one or more (or a plurality of) modified host cell (s) described herein for use in treating a disease is provided. In some embodiments, the modified host cell is an autologous immune cell isolated from the subject. In some embodiments, the modified host cell is an allogeneic immune cell. In some embodiments, the the disease is a hematological cancer.
In another aspect, use of one or more (or a plurality of) modified host cell (s) described herein in the preparation of a pharmaceutical composition for the treatment of a disease is provided. In some embodiments, the modified host cell is an autologous immune cell isolated from the subject. In some embodiments, the modified host cell is an allogeneic immune cell. In some embodiments, the the disease is a hematological cancer.
In another aspect, a kit for treating a somatic tissue disease is provided. In some embodiments, the kit comprises:
(i) a first container comprising a donor vector, wherein the donor vector comprises a polynucleotide encoding a CAR;
(ii) a second container comprising at least two sgRNA that is complementary to a selected nucleic acid sequence in the host cell genome; and
(iii) a third container comprising a Cas nuclease.
In some embodiments, the kit further comprises a Cas or CfPl protein or an RNA encoding a Cas or Cfpl protein. In some embodiments, the kit further comprises an instruction manual.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1. CRISPR/Cas9-mediated HDR targeting at TRAC locus by different sgRNAs.
Fig. 1A. Schematics for Cas9-RNP based TRAC HDR editing using different sgRNAs, and the design of AAV donor TRAC-800bp for TRAC editing. Arrows indicate the primers for genome PCR amplification in T7E1 assay. Fig. 1B. T7E1 assay for different sgRNAs targeting at TRAC locus in Jurkat cells, and the control group was included without TRAC editing. Fig. 1C. Flow cytometry analysis for TRAC targeting using AAV6 TRAC-800bp donor and different sgRNAs in Jurkat cells. Fig. 1D. Flow cytometry analysis for TRAC targeting using Cas9 assembled with different truncated or non-truncated sgRNAs, in combination with TRAC-800bp AAV6 donor, in Jurkat cells. X axis indicates the GFP expression level, and the GFP percentages were shown in the flow plot.
Fig. 2. CRISPR/Cas9-mediated HDR targeting at TRAC locus by different AAV donors.
Fig. 2A. Schematics for Cas9-RNP based TRAC targeting using different AAV HDR donors, including TRAC-800bp, TRAC-400bp and TRAC-2A-800bp donors. Fig. 2B. Flow cytometry analysis in Jurkat cells for TRAC targeting using different donors shown in A. X axis indicates the GFP expression level, and the GFP percentage were shown in the flow plot. Fig. 2C. Schematics of Cas9 RNP based TRAC targeting using AAV donors of TRAC-800bp-LRA,  TRAC-SFFV-GFP and TRAC-EF1α-GFP donors. Fig. 2D. Flow cytometry analysis in Jurkat cells for TRAC targeting using different AAV donors at day 3 (upper) and day 15 (lower) post electroporation. Fig. 2E. Flow cytometry for TRAC protein detection after TRAC targeting in Jurkat cells at day 15 post electroporation. X axis indicates the GFP signal representing the TRAC donor Knock-in, while Y axis indicates the PE level suggesting the TRAC expression in Jurkat cells. Fig. 2F. Bar chart showing the TRAC positive percentages of Jurkat cells after Cas9 RNP targeting.
Fig. 3. CRISPR/Cas9-mediated HDR transgene integration at TRAC locus using AAV donor and paired sgRNAs for TRAC gene disruption in Jurkat cells and T cells.
Fig. 3A. Schematics for Cas9-RNP based TRAC targeting using different paired sgRNAs combined with AAV donors. (i) TRAC targeting using paired sgRNAs (sg19-19bp &sg24) combined with TRAC-800bp-LRA or TRAC-800bp HDR AAV donor. (ii) TRAC targeting using paired sgRNAs (sg19-19bp &sg24) combined with TRAC-2A-800bp donor. Fig. 3B. Flow cytometry analysis for TRAC targeting using either single or paired sgRNAs (sg19-19bp &24) and combined with TRAC-800bp HDR donor, in Jurkat cells. Fig. 3C. Flow cytometry analysis for TRAC targeting using paired sgRNAs of (sg19-19bp &24) with TRAC-800bp donor or TRAC-800bp-LRA AAV donor, and paired sgRNAs of sg5 &24 with TRAC-2A-800bp AAV donor, in Jurkat cells. Fig. 3D. Flow cytometry analysis for TRAC staining targeted by paired sgRNAs of sg19-19bp &24 with TRAC-800bp-LRA donor, and paired sgRNAs of sg5 &24 with TRAC-2A-800bp donor, in Jurkat cells. Fig. 3E. Schematics of Cas9 RNP based TRAC targeting using paired sg19-19bp &sg24 and combined with TRAC-800bp-LRA AAV donor in activated human T cells. Fig. 3F. Flow cytometry analysis of TRAC targeting in T cells. X axis indicates the GFP expression level, and the GFP percentages were shown in the flow plot. Fig. 3G. Flow cytometry analysis of the TRAC expression level using the electroporated cells in Fig. 3B. Fig. 3H. Flow cytometry analysis of TRAC targeting in T cells on  day  4 or 4 weeks post electroporation when a higher MOI of AAV6 was applied for the transduction.
Fig. 4. CRISPR/Cas9-HDR-mediated CAR knockin at TRAC locus to generate universal CAR-T cells.
Fig. 4A. (i) Schematics for Cas9-RNP mediated TRAC knock-in using AAV virus of TRAC-E2A-CAR and TRAC-SFFV-CAR. (ii) Schematics of Lenti-SFFV-CAR vector and Lenti-EF1α-CAR vector for lentivirus transduction in human T cells. Fig. 4B. Flow cytometry analysis of TRAC targeting in human T cells using Cas9 RNP based targeting. Fig. 4C. Cytotoxicity analysis of different CAR-T cells targeting to CD19+ BV173-LUC (i) and CD19-OCI-AML3-LUC (ii) cells. T cells without genomic modification were included as control. “*” indicates that the p value is less than 0.05 when compare to control T cell group. The experiments were done in duplicates.
DEFINITIONS
As used herein, the term “comprises” is a term of art that is open ended, and does not exclude additional elements or features.
As used herein, a “gene” or “reporter” refers to a polynucleotide sequence encoding a protein product that can generate, under appropriate conditions, a detectable signal that allows detection for indicating the presence and/or quantity of the reporter gene protein product.
The term “cell” as used herein refers to a microorganism and includes an individual cell or cell culture that can be or has been a recipient of any recombinant vector (s) or isolated polynucleotide (s) of the invention.
The term “host cell” refers to a cell that is capable of being modified by the methods described herein, and includes cells derived from animals and mammals, including cells derived from livestock, companion animals, rats, mice and humans. Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change. A host cell includes cells into which a recombinant vector or a polynucleotide of the disclosure has been introduced, including by transformation, transfection, and the like.
“Cas9” or (CRISPR associated protein 9) is an RNA-guided DNA endonuclease enzyme associated with the CRISPR (Clustered Regularly Interspersed Palindromic Repeats) adaptive immunity system in Streptococcus pyogenes, among other bacteria. S. pyogenes utilizes Cas9 to memorize and later interrogate and cleave foreign DNA, such as the DNA of an  invading bacteriophage. Cas9, complexed with a small guide RNA (sgRNA) , performs this interrogation by unwinding foreign DNA and checking whether the DNA contains any sequence segment complementary to a 20 bp spacer region of the sgRNA. If the sgRNA finds sequence complementarity in the DNA, it is cleaved by Cas9.
As used herein, “sgRNA” or “small guide RNA” refers to a short RNA molecule that is capable of forming a complex with Cas9 protein and contains a segment of about 20 nucleotides complementary to a target DNA sequence, such that the Cas9-sgRNA complex directs Cas9 cleavage of a target DNA sequence upon the sgRNA recognizing the complementary sequence in the target DNA sequence. Accordingly, a sgRNA is approximately a 20-base sequence (ranging from about 10-50, 15-45, or 20-40, for example, 15, 20, 25, or 30 bases) specific to the target DNA 5’ of a non-variable scaffold sequence.
As used herein the term “3’-UTR” is a term of the art understood by skilled persons and means the section of messenger RNA (mRNA) that immediately follows the translation termination codon. An mRNA molecule is transcribed from the DNA sequence and is later translated into protein.
As used herein the term “IRES” is a term of the art understood by skilled persons and means internal ribosome entry site segments which are known to attract eukaryotic ribosomal translation initiation complex and thus promote translation initiation independently of the presence of the commonly utilized 5'-terminal 7mG cap structure.
As used herein the term “eGFP” is a term of the art understood by skilled persons and means enhanced green fluorescent protein with F64L point mutation which folds the efficiency at 37 ℃. Thus, eGFP leads to the significant performance of GFPs in mammalian cells.
As used herein the term “Luc” is a term of the art understood by skilled persons and means firefly luciferase protein which is an enzyme catalyzing the oxidation of a luciferin and causing it to produce a visible glow.
As used herein the term “GAPDH” is a term of the art understood by skilled persons and means a housekeeping gene which produces Glyceraldehyde 3-phosphate dehydrogenase. GAPDH gene is often stably and constitutively expressed at high levels in most human tissues  and cells. Thus, GAPDH is commonly used as control for western blot to check protein expression levels or for qPCR to check mRNA expression levels.
The terms “expression cassette” or “construct” or “vector” or “donor plasmid” or “donor vector” are used interchangeably and refer to a nucleic acid construct that, when introduced into a host cell, results in transcription and/or translation of an RNA or polypeptide, respectively. One example of an expression cassette is a polynucleotide construct that comprises a polynucleotide sequence encoding a polypeptide of the invention protein operably linked to a promoter, e.g., its native promoter, where the expression cassette is introduced into a heterologous microorganism or host cell. In some embodiments, an expression cassette comprises a polynucleotide sequence encoding a polypeptide of the invention where the polynucleotide is targeted to a position in the genome of a microorganism or host cell such that expression of the polynucleotide sequence is driven by a promoter that is present in the microorganism or host cell.
The term “target sequence” or “target DNA sequence, ” when used to refer to a pre-determined segment of a genomic sequence or polynucleotide construct of this invention (e.g., a donor plasmid) , is similarly defined in regard to the percentage sequence identity between the target sequence and its corresponding sgRNA.
As used herein the term “CRISPR system” refers to a prokaryotic immune system that confers resistance to foreign genetic elements. CRISPR is short for clustered regularly interspaced short palindromic repeats which are segments of prokaryotic DNA containing short, repetitive base sequences acquired from plasmid or phages. These segments could be transcribed into RNA and form as a scaffold to bind with CRISPR-associated protein (Cas protein, such as Cas9) . The combined complex would be directed to degrade the target sequence recognized by these transcribed segments to acquire immunity.
As used herein the term “AAV” or “adeno-associated virus” refers to a viral vector system developed from naturally prevalent nonpathogenic adeno-associated virus, a small virus which infects humans and some other mammalian species. The AAV viral vector has a very low immunogenicity, can infect both dividing and quiescent cells and persist in an extrachromosomal state without integrating into the genome of the host cell. These features make the AAV system a  useful tool for in vivo gene delivery and gene therapy. Recent human clinical trials with AAV for gene therapy have proven this approach.
The term “AAV donor” as used herein refers to a vector that is used as donor template and carries a variable set of elements required for subsequent genome editing and knock-in.
As used herein the term “HDR” or “homology directed recombination” or “homologous recombination” refers to a DNA repair mechanism. Facing DNA break, a cell could use the sister chromatin or any provided donors to repair it based on the intact allele or template through pairing homologous sequence around the break site.
As used herein the term “NHEJ” or “non-homologous end joining” refers to a DNA repair mechanism. Facing DNA break, a cell could repair the break site through directly linking the DNA ends together, and during this process, small nucleotides may be inserted or deleted.
As used herein the term “RNP” is a term of the art understood by skilled persons and means a ribonucleoprotein complexes that comprising of Cas9 protein and guide RNA oligonucleotides, and is applied for genome editing in cells or in animal models.
As used herein the term “CD19” is a term of the art understood by skilled persons and also known as CD19 molecule, and it encodes a transmembrane protein that is expressed in all B lineage cells, except for plasma cells and in follicular dendritic cells. It is a biomarker for B lymphocyte development, lymphoma diagnosis and can be utilized as a target for leukemia immunotherapies.
As used herein the term “TRAC” is a term of the art understood by skilled persons and means constant region of T cell receptor alpha chain. Alpha-beta T cell receptors are antigen specific receptors which are essential to the immune response and are present on the cell surface of T lymphocytes.
As used herein the term “Jurkat cells” , “Jurkat cell” or “Jurkat” is a term of the art understood by skilled persons and means an immortalized cell line of human T lymphocytes, and it is widely used for studying acute T cell leukemia, T cell signaling and functions Luciferase.
As used herein, the term “activated T cell” refers to a T cell that has been activated by CD3 and CD28 co-stimulation signals. Activated T cells typically divide rapidly in culture medium and secrete cytokines that regulate or assist the immune response.
As used herein the term “BV173” is a term of the art understood by skilled persons, and the BV173 means a cell line derived from a patient with Philadelphia chromosome (Ph1) -positive acute leukemia.
As used herein the term “BV173-Luc” is a term of the art understood by skilled persons. The BV173-Luc cells were derived from the BV173 cells by integrating a constant luciferase-GFP cassette into the cell genome through lentivirus transduction. The BV173-luc cells have been confirmed with CD19 positive, and were used for the detecting the cytotoxicity of CAR-T cells.
As used herein the term “OCI-AML3” is a term of the art understood by skilled persons. Tthe OCI-AML3 is a cell line established from the peripheral blood of a 57-year-old man with acute myeloid leukemia at diagnosis in 1987, and the cells carry an NPM1 gene mutation (type A) and the DNMT3A R882C mutation.
As used herein the term “OCI-AML3-Luc” is a term of the art understood by skilled persons. The OCI-AML3-Luc were derived from the OCI-AML3 cells by integrating a constant luciferase-GFP cassette into the cell genome through lentivirus transduction. The BV173-luc cells have been confirmed with CD19 positive, and were used for the detecting the cytotoxicity of CAR-T cells.
As used herein, the term “antigen binding protein” refers to a protein or polypeptide that binds with high affinity to a specific target antigen.
As used herein the term “chimeric antigen receptor” , “CAR” or “CARs” is a term of the art understood by skilled persons that refers to a type of antigen binding protein that comprises a fusion protein of a selected single-chain fragment variable domain from a specific monoclonal antibody and one or more T-cell receptor intracellular signaling domains.
As used herein the term “CAR-T cell therapy” is a term of the art understood by skilled persons and means a type of treatment in which involves genetic modification of patient's  autologous T-cells to express a CAR specific for a tumor antigen, following by ex vivo cell expansion and re-infusion back to the patient. This T-cell genetic modification can occur either via viral-based gene transfer methods or nonviral methods, such as DNA-based transposons, CRISPR/Cas9 technology or direct transfer of in vitro transcribed-mRNA by electroporation.
As used herein the term “T cell” is a term of the art understood by skilled persons and means a type of lymphocyte which develops in the thymus gland and plays a central role in the immune response. T cells can be distinguished from other lymphocytes by the presence of T-cell receptor on the cells surface.
As used herein the term “GVHD” is a term of the art understood by skilled persons and means a syndrome characterized by inflammation in different organs, with the specificity of epithelial cell apoptosis and crypt drop out. GVDH is commonly associated with stem cell transplants such as those that occur with bone marrow transplants, and could also be induced by CAR therapies based on donor leukocyte infusion, virus-specific T cells, T-cell receptor–deficient T cells, lymphoid progenitor cells, and regulatory T cells.
As used herein the term “HEK293T” is a term of the art understood by skilled persons and means a variant of human embryonic kidney 293 cells (HEK293) that contains the SV40 large T-antigen. The antigen allows episomal replication of transfected plasmids containing the SV40 origin of replication, which leads to the amplification of transfected plasmids and extended temporal expression of the desired gene products.
As used herein the term “in vitro” refers to methods that are performed outside of a living organism, typically using biological materials that are isolated from their normal biological host or usual biological context.
As used herein the term “2A self-cleaving sequence” refers to a class of 18-22 amino acid peptides that were originally identified in viruses and can mediate ribosome-skipping events. Including one or more 2A self-cleaving sequences in a protein-coding sequence enables the generation of two or more separate peptide products from one mRNA.
As used herein the term “orthologue” or “orthologous sequences” refers to homologous nucleic acid or amino acid sequences that are descended from the same ancestral sequence separated by a speciation event.
As used herein the term “homolog” or “homologous sequences” refers to nucleic acid or amino acid sequences that descended from a common ancestral sequence.
As used herein the term “percent identity” refers to two or more nucleic acid or amino acid sequences or subsequences that have a specified percentage of nucleotides or amino acid residues that are the same, e.g., at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region, when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using a sequence comparison algorithm described herein or by manual alignment and visual inspection. Thus, two or more nucleic acid or amino acid sequences or subsequences are considered “substantially identical” if they have at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region, or have 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region. In addition, two or more nucleic acid or amino acid sequences or subsequences can have greater than 90%identity but less than 100%identity, or any subrange thereof, for example 90%to 99%identity, or 95%to 99%indentity. These definitions also refer to the complement of a test sequence. It will be understood that any sequence disclosed herein can include sequences that are substantially similar to the reference sequence, i.e., have at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region; or have 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%or 99%sequence identity over a specified region, unless otherwise clear from the context.
As used herein the term "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the sequence in the comparison window can comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. The sequence comparison algorithm calculates the percent sequence identities or similarities for the test sequences relative to the reference sequence, based on the program parameters. Default program parameters are commonly used, or alternative parameters can be designated.
Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith and Waterman (Adv. Appl. Math. 2: 482, 1970) , by the homology alignment algorithm of Needleman and Wunsch (J. Mol. Biol. 48: 443, 1970) , by the search for similarity method of Pearson and Lipman (Proc. Natl. Acad. Sci. USA 85: 2444, 1988) , by computerized implementations of these algorithms (e.g., GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis. ) , or by manual alignment and visual inspection (see, e.g., Ausubel et al., Current Protocols in Molecular Biology (1995 supplement) ) .
Algorithms suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (Nuc. Acids Res. 25: 3389-402, 1977) , and Altschul et al. (J. Mol. Biol. 215: 403-10, 1990) , respectively.
Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http: //www. ncbi. nlm. nih. gov/) . This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra) . These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0) . For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the  cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) or 10, M=5, N=-4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA 89: 10915, 1989) alignments (B) of 50, expectation (E) of 10, M=5, N=-4.
The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul, Proc. Natl. Acad. Sci. USA 90: 5873-87, 1993) . One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P (N) ) , which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, typically less than about 0.01, and more typically less than about 0.001.
DETAILED DESCRIPTION OF THE INVENTION
Described herein is a gene editing method coupled with multiple newly designed AAV donors and paired sgRNAs for highly efficient generation of gene knockout universal CAR-T cells. In some embodiments, the method uses CRISPR/Cas9 mediated gene editing. In some embodiments, the method inserts a CAR into a genetic locus that encodes a protein that can trigger graft-versus-host-disease (GVHD) . In some embodiments, the methods described herein produce highly efficient knock-in of transgenes into the TRAC locus and simultaneous disruption of TRAC expression in Jurkat cells. The inventors also demonstrated excellent results using the methods to insert an anti-CD19 CAR at the TRAC locus in human T cells, which produced universal CAR-T cells with prominent anti-tumor activity. The inventors unexpectedly showed that the CRISPR/Cas9 based HDR editing using optimal AAV donor designs combined with paired sgRNAs targeting the TRAC locus have greatly improved the editing efficiency, which provides the potential for the efficient generation of high-performance TRAC knockout universal CAR-T cells for further clinical use. The current disclosure also provides a versatile  system, which can be easily modified by changing homology arms and paired sgRNAs to a new target sequence to generate distinct CAR-T cells with a different gene disruption, or by changing to another CAR in the donor to target different tumor antigens.
The methods and compositions described herein provide the following advantages. First, the methods simultaneously disrupt the expression of genes that express proteins that can trigger GVHD, such as proteins expressed by immune-competent allogenic T cells. Unlike the conventional autologous CAR-T therapy, which is expensive, time-consuming and often difficult due to the insufficient numbers of T cells in patients suffering from late stage cancers, the instant methods provide high-efficiency generation of universal CAR-T cells that could overcome these limitations by enabling the generation of allogenic CAR-T cells using T-cells from healthy donors. The instant methods can potentially provide universal “off-the-shelf” CAR-T products to benefit a broad range of patients in clinical treatments.
Second, the instant methods result in targeted integration of CARs at specific genetic loci, which provides consistent expression of CARs from batch to batch, and could diminish the variance of CAR expression, provide reproducible cytotoxicity and therapeutic activities, and thus potentially produce standardized cells for off-shelf products. Moreover, the defined insertion of CARs can also reduce the clonal effect and risk for tumorigenesis, which is common for CAR-T cells generated via lentivirus transduction due to random integrations. In some embodiments, the CAR is inserted at the TRAC locus.
Third, current approaches produce universal CAR-T cells through two steps, where the first is random CAR integration in the genome by lentivirus transduction, and the second step is a separate gene disruption or knockout step using a CRISPR-based method. The current methods do not avoid the undefined nature of CAR-T integration via lentivirus and introduce additional manipulation and delay due to the separate CRISPR-based knockout of specific genes that may contribute to GVHD. Compared to current approaches, the methods described herein greatly simplify the manipulation and shorten the process for generating universal CAR-T cells. The instant methods generate universal CAR-T cells by integrating a CAR at a specific genetic locus and simultaneously disrupting expression of the gene at the locus through a one-step manipulation. Moreover, the targeted integration of CARs in a pre-selected gene locus using the  instant methods also significantly reduces the risk for potential T cell tumorigenesis due to random and uncontrollable CAR integration using current lentivirus transduction methods.
Fourth, the instant methods also provide an optimal design of the donor vector and small guide RNAs (sgRNAs) that, compared to the current single sgRNA-based knock-in approach, could nearly double the donor integration. The instant methods can significantly increase the yields and reduce the number of starting T cells for universal CAR-T cell generation.
Fifth, the instant methods provide a versatile system, which can be easily modified by changing homology arms and paired sgRNAs to a new target sequence to generate distinct CAR-T cells with a different gene disruption, or by changing to another CAR in the donor vector to target different antigens. Thus, this technology holds tremendous potential for generating universal CAR-T cells, or for enhancing CAR-T cell performance by disrupting specific genes while simultaneously inserting the CAR at high efficiency. The instant methods provide great potential and value to improve CAR-T technology and broaden its potential to treat various diseases.
In some embodiments, the genome editing system comprises the CRISPR/Cas9-mediated genome editing system. The CRISPR/Cas9-mediated genome editing system was selected from multiple designs for its high-efficiency knock-in of CAR expression vectors into specific genes for producing universal CAR-T cells. In some embodiments, the methods insert a CAR at the TRAC locus, thereby generating CAR-T cells while simultaneously disrupting TRAC expression, which results in universal CAR-T cells that are disabled for recognizing the recipients’ target tissues as nonself and thereby evoke GVHD during transplantation. In some embodiments, the instant methods generate universal CAR-T cells by integrating a CAR in the TRAC locus and simultaneously disrupting TRAC expression using a one-step manipulation.
Previous studies have attempted one-step generation of universal CAR-T using single sgRNA, but the efficiency is relative low.
In contrast, the instant disclosure provides an optimal design of the AAV donor and usage of sgRNAs after thorough examination of multiple sgRNAs targeting TRAC locus and several different AAV donors carrying distinct homology arms. Compared to the current single sgRNA-based knock-in approach, the AAV donor coupled with paired sgRNAs could nearly  double the donor integration. Using Cas9 RNP combined with HDR donors and paired sgRNAs targeting has successfully fulfilled the requirements of high efficiency in generating universal CAR-T cells, which significantly increased the yields and reduced the usage of starting T cells for universal CAR-T cell generation.
General Methods
The methods described herein can include, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, and recombinant DNA techniques, which are within the skill of one of ordinary skill in the art. General methods are described for example, in T.E. Creighton, Proteins: Structures and Molecular Properties (W.H. Freeman and Company, 1993) ; A.L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition) ; Green &Sambrook, et al., Molecular Cloning: A Laboratory Manual (4th Edition, 2012) ; Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc. ) ; Remington's Pharmaceutical Sciences, 18th Edition (Easton, Pennsylvania: Mack Publishing Company, 1990) ; Carey and Sundberg Advanced Organic Chemistry 3rd Ed. (Plenum Press) Vols A and B (1992) ; Current Protocols in Molecular Biology (2002- ; Wiley; Online ISBN: 9780471142720; DOI: 10. 1002/04711142727) ; and Current Protocols in Immunology (2001-; Wiley; Online ISBN: 9780471142737; DOI: 10. 1002/0471142735) .
Methods for Modifying a Host Cell
In one aspect, described herein are methods for modifying a host cell. In some embodiments, the modified host cell is a modified immune cell, such as a modified T cell. In some embodiments, the modified host cell is an activated T cell. In some embodiments, the modified host cell is an autologous cell. In some embodiments, the modified host cell is an allogeneic cell. In some embodiments, the methods are in vitro methods.
In some embodiments, the host cell is modified to insert an exogenous polynucleotide at a pre-determined target locus in the host cell genome. In some embodiments, the exogenous polynucleotide encodes a protein or polypeptide. In some embodiments, the polynucleotide encodes an antigen binding protein (ABP) or antigen-binding fragment thereof. In some embodiments, the antigen binding protein is a chimeric antigen receptor (CAR) .
The host cells can be modified using the methods described herein. In some embodiments, the method comprises inserting a polynucleotide encoding an ABP at a pre-determined locus (also referred to as a “selected locus” or “target locus” ) in the host cell genome. In some embodiments, the method comprises homology directed recombination or homologous recombination. In some embodiments, the method comprises CRISPR/Cas mediated gene editing. In some embodiments, the method comprises CRISPR/Cas mediated gene editing in combination with AAV donor vectors and paired sgRNAs. In some embodiments, the CRISPR-associated (Cas) nuclease is a class 1 endonuclease or class 2 endonuclease. In some embodiments, the Cas is Cas9. In some embodiments, the Cas is CPf1 (now known as Cas12a) .
In some embodiments, the method comprises contacting a host cell with a donor vector comprising: (1) a polynucleotide sequence coding for at least one polypeptide; (2) a polyA segment at the 3' end of the polynucleotide; and (3) two homology fragments sharing identical sequences to the flanking regions of a target sequence in genome, one located at a 5'-end of the polynucleotide sequence and the other one located at a 3'-end of the polyA segment in the donor vector; followed by contacting the host cell with a ribonucleoprotein complex (RNP) comprising (1) a Cas nuclease, and (2) at least two small guide RNAs (sgRNA) that are complementary to at least two selected nucleic acid sequences within the pre-determined locus in the host cell genome.
In some embodiments, the donor vector comprises a promoter that regulates transcription of the polynucleotide sequence into mRNA in the host cell. In some embodiments, the mRNA is translated into a polypeptide in the host cell. In some embodiments, the polypeptide comprises an ABP, such as a CAR. In some embodiments, the CAR is an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or an anti-BCMA CAR.
In some embodiments, the pre-determined locus where the exogenous polynucleotide is inserted is the constant region of a T cell receptor alpha chain (TRAC) . In some embodiments, the pre-determined locus is the beta (β) -actin (ACTB) locus, or the Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) locus.
CRISPR/Cas Gene-editing System
The CRISPR-Cas gene-editing system is described, for example, in U.S. Patent No. 8,697,359 and U.S. Patent Publication 2014/0068797. The CRISPR-Cas9 system originated  from type II CRISPR-Cas systems, which provide bacteria with adaptive immunity to viruses and plasmids. The CRISPR-associated protein Cas9 is an endonuclease that uses a guide sequence within an RNA duplex, tracrRNA: crRNA, to form base pairs with DNA target sequences, enabling Cas9 to introduce a site-specific double-strand break in the DNA. The dual tracrRNA: crRNA was engineered as a single guide RNA (sgRNA) that retains two critical features: a sequence at the 5′ side that determines the DNA target site by Watson-Crick base-pairing and a duplex RNA structure at the 3′ side that binds to Cas9. This created a simple two-component system in which changes in the guide sequence of the sgRNA program Cas9 to target any DNA sequence of interest. The simplicity of CRISPR-Cas9 programming, together with a unique DNA cleaving mechanism, the capacity for multiplexed target recognition, and the existence of many natural type II CRISPR-Cas system variants, has allowed scientists to precisely and efficiently target, edit, modify, regulate, and mark genomic loci of a wide array of cells and organisms. (see Jennifer A. Doudna and Emmanuelle Charpentier, Science, 28 Nov 2014; Vol. 346, Issue 6213, 1258096; DOI: 10.1126/science. 1258096) .
It will be understood by one of ordinary skill in the art that the CRISPR-associated protein (Cas) genes are grouped into two classes, Class 1 and Class 2, and further grouped into at least 35 families based on sequence similarity of the encoded proteins. Therefore, the instant methods can use any Cas protein having the required functional characteristics. In some embodiments, the Cas is Cas9, or a protein having at least 90% (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) amino acid sequence identity to Cas9 (SEQ ID NO: 2) . In some embodiments, the Cas9 is encoded by a nucleic acid having at least 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) sequence identity to SEQ ID NO: 1) . In some embodiments, the Cas is Cas12a (Cpf1) , or a protein having at least 90% (e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) sequence identity to Cas12a (SEQ ID NO: 4) . In some embodiments, the CRISPR/Cas system is selected from Cas9 (Csn1) , Cas12a (Cpf1) , Cas13a (C2c2) and Cas13b (C2c6) systems or other CRISPR/Cas systems with similar activities. (see Tang, Y. et al. “Class 2 CRISPR/Cas: an expanding biotechnology toolbox for and beyond genome editing. ” Cell Biosci 8, 59 (2018) ) .
CAR T-cell Therapy
Chimeric antigen receptor (CAR) T-cell therapy (CAR T Therapy) is a type of immunotherapy called adoptive cell therapy. CAR T therapy typically involves genetic modification of a patient's autologous T-cells to express a CAR specific for an antigen, followed by ex vivo cell expansion and re-infusion back into the patient. Genetic modification of T-cells can occur using viral-based gene transfer methods or non-viral methods, such as DNA-based transposons, CRISPR/Cas9 technology or direct transfer of in vitro transcribed-mRNA by electroporation. However, the methods described herein allow for the production of modified T cells that can be used as universal “off the shelf” CAR-T cells. In other words, the methods allow for production of modified T cells that do not rely on autologous T-cells isolated from a particular subject or patient, and instead provide modified allogeneic host cells that can be administered to more than one subject or patient for treating a disease.
A CAR is a recombinant fusion protein comprising an antigen-specific extracellular domain coupled to an intracellular domain that provides an intracellular signal upon antigen binding to the extracellular domain. Thus, CARs are different from other antigen binding agents because they can both bind MHC-independent antigen and transduce activation signals via the intracellular domain. Specific binding of CARs to antigen is typically expressed as an affinity constant or affinity of interaction (KD) , where the K D is between about 0.1 pico Molar (pM) and about 10 micro Molar (μM) , or about 0.1 pM to about 1 μM, or about 0.1 pM to about 100 nano Molar (nM) .
An antigen-specific extracellular domain suitable for use in a CAR of the present disclosure may be any antigen-binding polypeptide known in the art. For example, in some embodiments, the antigen-binding domain comprises an antibody-based binding domain, for example a single chain Fv (scFv) , a single domain antibody variable domain (sdAb VH) , a camelid antibody variable domain (cAb VHH) or humanized versions thereof, or a shark antibody variable domain (IgNAR VH) or humanized versions thereof. In some embodiments, the antigen-binding domain comprises a T-cell receptor (TCR) based binding domain, such as a single chain TCR (scTv) , or a single chain two-domain TCR containing the variable alpha and beta chains.
In some embodiments, the CAR recognizes or specifically binds to an antigen expressed on a tumor, cancer or malignant cell. In some embodiments, the tumor, cancer or malignant cell is from a tumor of the hematopoietic or lymphoid tissues. In some embodiments, the tumor or cancer is a leukemia, such as acute lymphoblastic leukemia (ALL) , acute myeloid leukemia (AML) , chronic lymphocytic leukemia (CLL) and chronic myeloid leukemia (CML) . In some embodiments, the tumor is a B cell tumor or T cell tumor. Non-limiting examples of B cell cancers and tumors include B-cell lymphomas, which may be either indolent (slow-growing) or aggressive (fast-growing) . Most B-cell lymphomas are non-Hodgkin lymphomas, including Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) , diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma. In some embodiments, the CAR specifically binds to an antigen expressed by a B cell.
In some embodiments, the CAR specifically binds to CD19, CD20, CD22, CD47, or B-cell maturation antigen (BCMA) . In some embodiments, the CAR is an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or anti-BCMA CAR. In some embodiments, the CAR comprises an amino acid sequence having at least 90%sequence identity (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%or 100%) to SEQ ID NO: 6 (anti-CD19 CAR) , SEQ ID NO: 8 (anti-CD20 CAR) , SEQ ID NO: 10 (anti-CD22 CAR) , or SEQ ID NO: 12 (anti-BCMA CAR) . In some embodiments, the CAR is encoded by a nucleic acid sequence having at least 90%sequence identity (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%or 100%) to SEQ ID NO: 5 (anti-CD19 CAR) , SEQ ID NO: 7 (anti-CD20 CAR) , SEQ ID NO: 9 (anti-CD22 CAR) , or SEQ ID NO: 11 (anti-BCMA CAR) . Sequences of some examples of various components of CARs of the instant invention, and nucleic acids that encode them are listed in Table 1, where (aa) stands for amino acids, and (na) stands for nucleic acids that encode the corresponding peptide. And in some embodiments, the CAR protein in this design can be changed to other CAR proteins with specific targeting.
Non-limiting examples of T-cell cancers and tumors include T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) , T-cell large granular lymphocyte (LGL) leukemia, human T-cell leukemia virus type 1-positive (HTLV-1+) adult T-cell leukemia/lymphoma (ATL) , T-cell prolymphocytic leukemia (T-PLL) , and various peripheral T-cell lymphomas (PTCLs) , including angioimmunoblastic T-cell lymphoma (AITL) , ALK-positive anaplastic large cell lymophoma,  and ALK-negative anaplastic large cell lymophoma. In some embodiments, the CAR specifically binds to an antigen expressed by a T cell. In some embodiments, the CAR specifically binds to CD9, CD7, CD5, CD2, CD30, or CD4.
In some embodiments, the CAR comprises an intracellular domain that provides an intracellular signal to the T cell upon antigen binding to the antigen-specific extracellular domain. Upon antigen binding, the intracellular signaling domain activates an effector function of the modified T cell. The effector function can include cytolytic activity or helper activity including the secretion of cytokines, or antigen-dependent proliferation. In some embodiments, the intracellular domain includes the zeta chain of the T-cell receptor or any of its homologs (e.g., beta, delta, gamma, or epsilon) , MB 1 chain, B29, Fc RIII, Fc RI, and combinations of signaling molecules, such as CD3. zeta. and CD28, CD27, 4-1 BB, DAP-10, OX40, and combinations thereof, as well as other similar molecules and fragments. Intracellular signaling portions of other members of the families of activating proteins may be used, such as Fc. gamma. RIII and Fc. epsilon. RI. In some embodiments, the intracellular signaling domain is a truncated portion of the signaling domain that retains functional signal transduction activity.
In some embodiments, the antigen-specific extracellular domain is linked to the intracellular domain of the chimeric antigen receptor by a transmembrane domain. In some embodiments, the CAR further comprises one or more costimulatory domains, and/or one or more spacers. A costimulatory domain can be derived from the intracellular signaling domains of costimulatory proteins that enhance cytokine production, proliferation, cytotoxicity, and/or persistence in vivo. An amino acid spacer can be used to connect the different domains of the CAR together, for example, a spacer can connect the antigen-specific extracellular domain to the transmembrane domain, the transmembrane domain to a costimulatory domain, a costimulatory domain to the intracellular domain, or the transmembrane domain to the intracellular domain. For example, inclusion of a spacer domain between the antigen-specific extracellular domain and the transmembrane domain may affect flexibility of the antigen-binding domain and thereby CAR function.
In some embodiments, the CAR comprises the CD8 hinge-transmembrane domain and/or the co-stimulatory signaling domain of 41bb fused to CD3 zeta. Other co-stimulatory  signaling domains (e.g., 41bb, CD27, CD28 or a combination of two co-stimulatory domains) can also be used to construct different CARs.
In some embodiments, CAR-T cells described herein are deficient in endogenous T cell receptor (TCR) signaling. In some embodiments, it may be desirable to decrease or eliminate endogenous TCR signaling in CAR-T cells disclosed herein. For example, decreasing or eliminating endogenous TCR signaling in CAR-T cells may prevent or reduce graft versus host disease (GvHD) when allogenic T cells are used to produce the CAR-T cells. Methods for decreasing or eliminating endogenous TCR signaling include, but are not limited to, modifying a part of the TCR receptor (e.g., the TCR receptor alpha chain (TRAC) , etc. ) . TRAC modification can be used to block TCR mediated signaling, which could allow allogeneic T cells to be modified to express various CARs and administered to patients without inducing life-threatening GvHD.
In some embodiments, the modified host cells described herein can further comprise one or more suicide genes. A suicide gene generally encodes an enzyme that selectively converts a nontoxic prodrug into highly toxic metabolites, specifically eliminating cells expressing the enzyme. Suicide genes may allow effective tracking and elimination of the CAR-T cells in vivo if required. One examples of suicide genes include herpes simplex virus thymidine kinase (HSV-tk) which phosphorylates the nontoxic antiviral prodrug ganciclovir (GCV) into GCV triphosphate. GCV triphosphate is incorporated into DNA in replicating cells, inhibiting DNA synthesis and resulting in cell death. Other examples include inducible caspase 9 protein, or a CD34/thymidine kinase chimeric suicide gene.
Methods of Treatment
Described herein are methods of treating a disease in a subject in need thereof by administering a modified host cell described herein to the subject. In some embodiments, the modified host cell is a modified immune cell, such as a modified T cell. In some embodiments, the modified host cell is an autologous cell. In some embodiments, the modified host cell is an allogeneic cell. In some embodiments, the modified host cell is included in a pharmaceutical compositon. In some embodiments, the subject is an animal, a mammal, a livestock animal such as a cow, horse, sheep, goat or pig, a companion animal such as a cat or dog, or a human.
In some embodiments, the disease is a hematological cancer.
In some embodiments, the disease is a cancer or tumor of the hematopoietic or lymphoid tissues, as described herein. For example, in some embodiments, the disease is a leukemia, such as acute lymphoblastic leukemia (ALL) , acute myeloid leukemia (AML) , chronic lymphocytic leukemia (CLL) and chronic myeloid leukemia (CML) . In some embodiments, the disease is a B cell cancer or tumor. Non-limiting examples of B cell cancers and tumors include B-cell lymphomas, which may be either indolent (slow-growing) or aggressive (fast-growing) . Most B-cell lymphomas are non-Hodgkin lymphomas, including . Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) , diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma.
In some embodiments, the disease is a tumor that expresses a B cell antigen or marker, such as CD19, CD20, CD22, CD47, or B-cell maturation antigen (BCMA) .
In some embodiments, the disease is a T-cell cancer or tumor, such as T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) , T-cell large granular lymphocyte (LGL) leukemia, human T-cell leukemia virus type 1-positive (HTLV-1+) adult T-cell leukemia/lymphoma (ATL) , T-cell prolymphocytic leukemia (T-PLL) , and various peripheral T-cell lymphomas (PTCLs) , including angioimmunoblastic T-cell lymphoma (AITL) , ALK-positive anaplastic large cell lymophoma, and ALK-negative anaplastic large cell lymophoma. In some embodiments, the disease is a tumor that expresses a T cell antigen or marker, such as CD7, CD5, CD2, CD30, and CD4.
In some aspects, the method is an in vitro method. For example, the in vitro method can be a cytotoxicity assay. In some embodiments, a modified host cell described herein, which expresses a polypeptide encoded by the exogenous polynucleotide, is contacted with a target cell expressing an antigen that binds to the polypeptide, and the number of target cells that are lysed or killed by the modified host cell is determined.
In some aspects, the method is an in vivo method. In some embodiments, the method comprising administering, for example, by infusion, a modified host cell described herein to a living animal that expresses a target antigen that binds to the polypeptide encoded by exogenous polynucleotide. In some embodiments, the exogenous polynucleotide encodes a chimeric  antigen receptor that specifically binds an antigen in the living organism. Thus, in some embodiments, the modified host cell expresses a CAR that specifically binds an antigen expressed in the living organism. In some embodiments, the modified host cell is a T cell. In some embodiments, the antigen is expressed by a tumor or cancer cell.
Table 1. Sequences of various components of Cas9, CAR proteins and AAV donors. (aa -amino acids, na -nucleic acids that encodes the corresponding protein)
Figure PCTCN2021078164-appb-000001
Figure PCTCN2021078164-appb-000002
Figure PCTCN2021078164-appb-000003
Figure PCTCN2021078164-appb-000004
Figure PCTCN2021078164-appb-000005
Figure PCTCN2021078164-appb-000006
Figure PCTCN2021078164-appb-000007
Figure PCTCN2021078164-appb-000008
Figure PCTCN2021078164-appb-000009
Figure PCTCN2021078164-appb-000010
Figure PCTCN2021078164-appb-000011
Figure PCTCN2021078164-appb-000012
Figure PCTCN2021078164-appb-000013
Figure PCTCN2021078164-appb-000014
Figure PCTCN2021078164-appb-000015
Figure PCTCN2021078164-appb-000016
Figure PCTCN2021078164-appb-000017
Figure PCTCN2021078164-appb-000018
Figure PCTCN2021078164-appb-000019
EXAMPLES
The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results.
Example 1: Efficient CRISPR/Cas9-mediated transgene integration at TRAC locus using AAV donor and single sgRNA for TRAC gene disruption
We first achieved efficient CRISPR/Cas9-HDR-mediated GFP transgene integration at TRAC locus using AAV donor and single sgRNA, which disrupts the TRAC gene expression at the same time. We synthesized five different in vitro transcribed sgRNAs (sg19, sg5, sg15, sg11, sg12) targeting the first coding exon of TRAC gene (Fig. 1 A) , and confirmed the high editing efficiencies by T7E1 assay in Jurkat cells (Fig. 1 B) . Then, we constructed AAV-TRAC-HDR-ires-GFP-800bp donor carrying 800 bp homologous arms, each at one side of a promoter-less ires-GFP cassette (Fig. 1 A) . We performed the TRAC HDR targeting using different sgRNAs along with TRAC-800bp donor, and observed efficient GFP expression (Fig. 1 C) , indicating successful transgene integration in the TRAC locus. We also confirmed efficient editing rates when using truncated version of selected sgRNAs (Fig. 1 D) .
To determine the optimal length of homology arms, we constructed different AAV donors with various homologous arm lengths, different co-expression cassette signal, with or without promoters (Fig. 2 A-D) . All the donors showed efficient editing after performing the TRAC targeting, among which, the TRAC-800bp-LRA showed highest editing rates compared to other promoter-less donors (Fig. 2 A-D) . Moreover, the SFFV and EF1α promoter produced higher GFP intensity compared to the endogenous TRAC promoter (Fig. 2 C-E) . These data indicate that Cas9-RNP based AAV donor and single sgRNA based knock-in could successfully disrupt TRAC expression in human Jurkat cells.
Example 2: Efficient CRISPR/Cas9-mediated transgene integration at TRAC locus using AAV donor and paired sgRNAs for TRAC gene disruption
We then confirmed efficient CRISPR/Cas9-HDR-mediated GFP transgene integration at TRAC locus using AAV donor and paired sgRNAs. We designed another sgRNA (sg24) targeting the 3’ region of TRAC exon-1, and we performed TRAC editing by applying Cas9  RNP assembled with paired sgRNAs (sg19-19bp &sg24 or sg5 &sg24) , along with different TRAC-800bp-LRA, TRAC-800bp and TRAC-2A-800bp AAV donor (Fig. 3A) . Flow cytometry analysis revealed that applying paired sgRNAs almost doubled editing efficiency compared to single-sgRNA editing, indicating that the paired sgRNAs targeting introduced more efficient donor integration at the TRAC locus (Fig. 3 B, C) . Consistently, the paired sgRNAs targeting also achieved the high efficient TRAC knockout in Jurkat cells (Fig. 3 D) . Furthermore, we tested this method in activated T cells, and obtained similar rates for donor integration and TRAC knockout in activated T cells (Fig. 3 E-G) , indicating the successful and high frequencies of simultaneous donor integration and TRAC disruption in T cells, which provide great value for using this new editing methods for potential clinical use.
Example 3: CRISPR/Cas9-mediated CAR knockin at TRAC locus to generate universal CAR-T cells using AAV donor and paired sgRNAs
We then validated the successful and efficient generation of universal CAR-T cells through CRISPR/Cas9-HDR-mediated anti-CD19 CAR integration at TRAC locus using AAV donor and paired sgRNAs. We constructed TRAC-2A-CAR and TRAC-SFFV-CAR based on the backbone of the TRAC-800bp-LRA donor, and performed the TRAC targeting using paired sgRNAs (sg19-19bp &sg24) and different AAV donors (Fig 4. A) . We confirmed the efficient editing in the T cells throughCas9-RNP based editing approach using TRAC-2A-CAR AAV donors and paired sgRNAs (Fig 4. B) . We also constructed new lentivirus vectors of Lenti-SFFV-CAR and Lenti-EF1α-CAR carry the CAR sequence under control by an active promoter We then produced the universal CAR-T cells by Cas9 RNP targeting and conventional CAR-T cells by lentivirus transduction (Fig. 4 A) . The CAR-T cells produced by TRAC targeting using AAV donors of TRAC-E2A-CAR and TRAC-SFFV-CAR showed comparable or even higher lysis percentages than the conventional CAR-T cells produced from lentivirus transduction (Fig 4. C) . Collectively, we have successfully employed Cas9 RNP combined with AAV donor and paired sgRNAs method to produce universal CAR-T cells by blocking the TRAC gene expression while simultaneously inserting an anti-CD19 CAR fragment, and have verified the potential anti-cancer effects of the TRAC negative CAR-T cells by in vitro cytotoxicity assays.
EXAMPLE 4: AAV package
AAV-6 system was used for virus packaging [18] . Transfection was carried out in 293FT cells with 80%confluence, and 3 days after transfection, the virus was collected. Basically, the virus was extracted from the cytoplasm and nuclear, then gradient centrifugation was performed with ultra-speed. Finally, the viral particles were concentrated and further purified with ultra-centrifugal-100KD filter.
EXAMPLE 5: Lentivirus package
Transfection was carried out in 293FT cells with 80%confluence, and 3 days after transfection, the cell culture medium was collected, and the virus particles were concentrated and further purified with ultra-centrifugal-100KD filter.
EXAMPLE 6: Plasmid construction
a) Cas9 protein and sgRNA in vitro synthesis: Cas9 proteins were purchased from Thermo Fisher (TrueCut TM Cas9 Protein v2, Thermo Fisher ) , and in vitro transcription (IVT) of sgRNAs used in electroporation were generated by GeneArt TM Precision gRNA Synthesis Kit (Thermo Fisher) according to manual instruction. sgRNA target sequences were designed based on the output from website www. casblastr. org) . All the sgRNA targeting information is listed in Table 2.
Table 2. sgRNA information of CRISPR/Cas9-HDR-mediated CAR knockin at TRAC locus
Figure PCTCN2021078164-appb-000020
Figure PCTCN2021078164-appb-000021
Figure PCTCN2021078164-appb-000022
Figure PCTCN2021078164-appb-000023
Figure PCTCN2021078164-appb-000024
Figure PCTCN2021078164-appb-000025
Figure PCTCN2021078164-appb-000026
Figure PCTCN2021078164-appb-000027
Figure PCTCN2021078164-appb-000028
Figure PCTCN2021078164-appb-000029
Figure PCTCN2021078164-appb-000030
Figure PCTCN2021078164-appb-000031
Figure PCTCN2021078164-appb-000032
Figure PCTCN2021078164-appb-000033
Figure PCTCN2021078164-appb-000034
Figure PCTCN2021078164-appb-000035
Figure PCTCN2021078164-appb-000036
Figure PCTCN2021078164-appb-000037
Figure PCTCN2021078164-appb-000038
Figure PCTCN2021078164-appb-000039
Figure PCTCN2021078164-appb-000040
Figure PCTCN2021078164-appb-000041
Figure PCTCN2021078164-appb-000042
Figure PCTCN2021078164-appb-000043
Figure PCTCN2021078164-appb-000044
Figure PCTCN2021078164-appb-000045
Figure PCTCN2021078164-appb-000046
Figure PCTCN2021078164-appb-000047
Figure PCTCN2021078164-appb-000048
Figure PCTCN2021078164-appb-000049
Figure PCTCN2021078164-appb-000050
Figure PCTCN2021078164-appb-000051
Figure PCTCN2021078164-appb-000052
Figure PCTCN2021078164-appb-000053
Figure PCTCN2021078164-appb-000054
Figure PCTCN2021078164-appb-000055
Figure PCTCN2021078164-appb-000056
Figure PCTCN2021078164-appb-000057
Figure PCTCN2021078164-appb-000058
Figure PCTCN2021078164-appb-000059
Figure PCTCN2021078164-appb-000060
Figure PCTCN2021078164-appb-000061
Figure PCTCN2021078164-appb-000062
b) anti-CD19 CAR: The anti-CD19-CAR-GFP cassette was constructed as described previously [19] . Briefly, the anti-CD19 CAR consisting a single chain variable fragment scFV specific for the human CD19 preceded by a CD8a leader peptide were amplified from plasmid (Addgene 113014) , and fused with 41bb hinge-transmembrane-intracellular regions and CD3ζ intracellular domain amplified from T cell cDNA. The CAR cassette was then fused with a e2a-gfp cassette followed by a polyA element through overlapping PCR. Subsequently, the entire CAR-GFP cassette was inserted into plasmid of CD19-FcGamma CAR (Addgene 113014) at Mlu1 and Not1 sites to generate Lenti-SFFV-CAR-GFP (termed Lenti-SFFV-CAR for short) plasmid.
c) AAV donors: The backbone of AAV donors were purchased from addgene (addgene#87115) . Different homology arms of TRAC gene were amplified from human genome, and inserted into the backbone, and IRES-GFP-PA, 2A-GFP-PA, SFFV-GFP-PA, EF1α-GFP-PA, 2A-anti-CD19 CAR-GFP-PA, SFFV-anti-CD19 CAR-GFP-PA, EF1α-anti-CD19 CAR-GFP-PA cassettes were constructed separately, and inserted between different homology arms to generate AAV donors of TRAC-800bp (SEQ ID NO: 13) , TRAC-400bp (SEQ ID NO: 14) , TRAC-2A-800bp (SEQ ID NO: 15) , TRAC-800bp-LRA (SEQ ID NO: 16) , TRAC-SFFV-GFP (SEQ ID NO: 17) , TRAC-EF1α-GFP (SEQ ID NO: 19) , and TRAC-SFFV-CAR (SEQ ID NO: 23) , respectively. All the donor information is listed in Table 1 and Figs. 2A, 2C and 4A.
d) Lentivirus vector: the anti-CD19 CAR cassette was inserted into plasmid of CD19-FcGamma CAR (Addgene 113014) at Mlu1 and Not1 sites to generate Lenti-SFFV-CAR plasmid, and the EF1α promoter was amplified from plasmid (Addgene 109049) , and inserted into backbone of Lenti-SFFV-CAR at MfeI and NcoI to replace the SFFV element, so as to generate Lenti-EF1α-CAR vector.
EXAMPLE 7: Genomic DNA extraction and PCR detection of genomic integrations
Genome DNA from cultured cells was extracted using Genome DNA extraction Kit (Tiangen) following the manufacturer's instruction. To extract genome DNA from liver tissues, the Tris buffer and proteinase K was used for overnight digestion at 37℃, then purified with 75%ethanol. 200 ng genomic DNA were generally used for PCR reaction using Phusion High-Fidelity DNA Polymerase (New England Biolabs) , following the manufacturer's instruction.
EXAMPLE 8: Fluorescence-activated cell sorting analysis
Fluorescence Activated Cell Sorting (FACS) analyzer (BD LSRFortessa Cell Analyzer) was configured with a single 488 nm argon ion laser (200 mW) . The laser is used to induce light scattering by either the excitation of cellular fluorescent proteins (eGFP) or the granularity within the cell. The recorded events within the gate on the FITC-A (GFP) log scale provided a good indication of the GFP expression level and the counts indicate the number of GFP-positive cells. The ration of GFP-positive cells over the total counts in the gated area is defined as targeting efficiency.
EXAMPLE 9: Cytotoxicity assays
The cytotoxicity of T cells transduced with CAR can be detected by standard luciferase-assay described previously [8] . ALL-cell lines that express CD19 and luciferase were used as target cells (T) , and T cells with CAR integration were used as effector cells (E) . 5x104 target cells were seeded in 96-well plates, and co-cultured with the effector cells at different ratios. The final volume in each well was adjusted to 200 μL. Target cells cultured without effector cells were used to determine the maximal luciferase expression (relative light units, RLUmax) . After 18 hours’ co-culture, 100 μL luciferase substrate (Promega) was added to each well, and the luminescence was the detected by luciferase imaging system.
EXAMPLE 10: Cas9 ribonucleoprotein (RNP) assembly and electroporation
Purified Cas9 (5 μg) and in vitro transcribed sgRNA were mixed at ratios of 1: 2, and incubated at room temperature for around 5-10 mins for Cas9 RNP assembly. Human Jurkat cells or activated human T cells were collected and electroporated with Cas9 RNP complex. After electroporation, the cells were added different types of donors were then directly accordingly. All the AAV donors were added at different MOI (104-106 vg/cell) .
EXAMPLE 11: T7 endonuclease 1 (T7E1) assay
DNA fragment covering a targeting region in TRAC gene loci were amplified by genome PCR, and followed with DNA purification process using MEGAquick-spin Total Fragment DNA Purification Kit (iNtRON) according to manufacturer’s instructions. First, the purified fragments were denatured and annealed and then were treated with T7E1 (T7  endonuclease 1, NEB) at 37 ℃ for 1 hour to completely cleave the unmatched nucleotide pairs. Subsequently, the editing efficiency was determined using Image J.
References
1. Perales MA, K.P., Kean LS, Sadelain M., Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR.. Biol Blood Marrow Transplant., 2018. 24 (1) : p. 27-31.
2. Wang Z, W.Z., Liu Y, Han W., New development in CAR-T cell therapy. J Hematol Oncol., 2017. 10: p. 53.
3. Mollanoori H, S.H., Rahmati Y, Teimourian S., CRISPR/Cas9 and CAR-T cell, collaboration of two revolutionary technologies in cancer immunotherapy, an instruction for successful cancer treatment.. Hum Immunol., 2018. 79 (12) : p. 876-882.
4. Torikai H, R.A., Liu PQ, Zhou Y, Zhang L, Maiti S, Huls H, Miller JC, Kebriaei P, Rabinovich B, Lee DA, Champlin RE, Bonini C, Naldini L, Rebar EJ, Gregory PD, Holmes MC, Cooper LJ., A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood., 2012. 119 (24) : p. 5697-705.
5. Torikai H, R.A., Soldner F, Warren EH, Yuen C, Zhou Y, et al. 2013; 122 (8) : 1341–9., Toward eliminating HLA class I expression to generate universal cells from allogeneic donors. Blood, 2013. 122 (8) : p. 1341-9.
6. Dai X, P.J., Du Y, Kim HR, Wang G, Errami Y, Chen S., One-step generation of modular CAR-T cells with AAV-Cpf1. Nat Methods., 2019. 16 (3) : p. 247-254.
7. Poirot L, P. B., Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, Potrel P, Bas C, Lemaire L, Galetto R, Lebuhotel C, Eyquem J, Cheung GW, Duclert A, Gouble A, Arnould S, Peggs K, Pule M, Scharenberg AM, Smith J., Multiplex Genome-Edited T-cell Manufacturing Platform for "Off-the-Shelf" Adoptive T-cell Immunotherapies. Cancer Res., 2015. 75 (18) : p. 3853-64.
8. Eyquem J, M. -S.J., Giavridis T, van der Stegen SJ, Hamieh M, Cunanan KM, Odak A, 
Figure PCTCN2021078164-appb-000063
M, Sadelain M., Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature, 2017. 543: p. 113-117.
9. Ren J, L.X., Fang C, Jiang S, June CH, Zhao Y, Multiplex Genome Editing toGenerate Universal CAR T Cells Resistant to PD1 Inhibition.. Clin Cancer Res., 2017. 23: p. 2255-2266.
10. Qasim W, Z.H., Samarasinghe S, Adams S, Amrolia P, Stafford S, Butler K, Rivat C, Wright G, Somana K, Ghorashian S, Pinner D, Ahsan G, Gilmour K, Lucchini G, Inglott S, Mifsud W, Chiesa R, Peggs KS, Chan L, Farzeneh F, Thrasher AJ, Vora A, Pule M, Veys P., Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells.. Sci Transl Med., 2017. 25: p. 374.
11. Benjamin R, G.C., Yallop D, Jozwik A, Ciocarlie O, Jain N, Jabbour EJ, Maus MV, Frigault M, Boissel N, Larghero J, Baruchel A, Mohty M, De Moerloose B, Bloor A, Frey NV, 
Figure PCTCN2021078164-appb-000064
A, Balandraud S, Philippe A, Fouliard S, Gauthier L, Pauly J, Konto C, Bermingham C, Veys P, Qasim W., Preliminary data on safety, cellular kinetics and anti-leukemic activity of UCART19, an allogeneic anti-CD19 CAR T-cell product, in a pool of adult and pediatric patients with high-risk CD19+ relapsed/refractory B-cell acute lymphoblastic leukemia.. Blood, 2018. 132 ( (Suppl 1) ) : p. 896.
12. Ren J, L.X., Fang C, Jiang S, June CH, Zhao Y., Multiplex Genome Editing to Generate Universal CAR T Cells Resistant to PD1 Inhibition.. Clin Cancer Res., 2017. 23 (9) : p. 2255-2266.
13. Cooper ML, C.J., Staser K, Ritchey JK, Devenport JM, Eckardt K, Rettig MP, Wang B, Eissenberg LG, Ghobadi A, Gehrs LN, Prior JL, Achilefu S, Miller CA, Fronick CC, O'Neal J, Gao F, Weinstock DM, Gutierrez A, Fulton RS, DiPersio JF., An "off-the-shelf" fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies.. Leukemia., 2018. 32 (9) : p. 1970-1983.
14. Ren J, Z.X., Liu X, Fang C, Jiang S, June CH, et al., A versatile system for rapid multiplex genome-edited CAR T cell generation.. Oncotarget., 2017. 8 (10) : p. 17002–11.
15. J., E., Silencing and variegation of gammaretrovirus and lentivirus vectors.. Hum Gene Ther., 2005. 16 (11) : p. 1241-6.
16. von Kalle C, D.A., Schmidt M. 2014 Jun; 25 (6) : 475-81., Vector integration and tumorigenesis.. Hum Gene Ther., 2014. 25 (6) : p. 475-81.
17. MacLeod DT, A.J., Martin AJ, Moser RJ, Hekele A, Wetzel KJ, Brown AE, Triggiano MA, Hux JA, Pham CD, Bartsevich VV, Turner CA, Lape J, Kirkland S, Beard CW, Smith J, Hirsch ML, Nicholson MG, Jantz D, McCreedy B., Integration of a CD19 CAR into the TCR Alpha Chain Locus Streamlines Production of Allogeneic Gene-Edited CAR T Cells. Mol Ther., 2017. 25 (4) : p. 949-961.
18. Grieger, J.C., V.W. Choi, and R.J. Samulski, Production and characterization of adeno-associated viral vectors. Nat Protoc, 2006. 1 (3) : p. 1412-28.
19. Kochenderfer JN, F.S., Zhao Y, Xu H, Black MA, Morgan RA, Wilson WH, Rosenberg SA., Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother, 2009. 32 (7) : p. 689-702.
20. June CH, S.M., Chimeric antigen receptor therapy.. N Engl J Med., 2018. 379: p. 64–73.
21. Yin H, X.W., Anderson DG., CRISPR-Cas: a tool for cancer research and therapeutics. Nat Rev Clin Oncol., 2019. 16 (5) : p. 281-295.
All patents, patent applications, and other publications, including GenBank Accession Numbers, cited in this application are incorporated by reference in the entirety for all purposes.

Claims (32)

  1. An in vitro method of inserting a polynucleotide sequence at a pre-determined locus in a host cell genome, the method comprising:
    contacting a host cell sequentially with:
    (i) a donor vector comprising: (1) a polynucleotide sequence encoding at least one polypeptide; (2) a polyA segment at the 3' end of the polynucleotide sequence; and (3) two homology fragments sharing identical sequences to the flanking regions of a target sequence in the host cell genome, one located at a 5'-end of the polynucleotide sequence and the other one located at a 3'-end of the polyA segment in the donor vector; and
    (ii) a ribonucleoprotein complex (RNP) comprising (1) a Cas nuclease, and (2) at least two small guide RNAs (sgRNA) that are complementary to at least two selected nucleic acid sequences within the pre-determined locus in the host cell genome.
  2. The method of claim 1, wherein the donor vector is selected from a plasmid, AAV viral particle, adenovirus particle, lentivirus particle, or a DNA-nanoparticle complex.
  3. The method of claim 1, wherein the donor vector further comprises at least one polynucleotide sequence encoding a CAR that specifically binds a tumor antigen.
  4. The method of claim 1, wherein the donor vector further comprises at least one polynucleotide sequence encoding an anti-CD19 CAR, anti-CD20 CAR, anti-CD22 CAR, anti-CD47 CAR, or anti-BCMA CAR protein.
  5. The method of claim 1, wherein the donor vector further comprises at least one polynucleotide sequence encoding an immune checkpoint protein or an anti-immune checkpoint protein.
  6. The method of claim 1, wherein the donor vector further comprises at least one polynucleotide sequence encoding a reporter gene.
  7. The method of claim 1, wherein the donor vector further comprises a 2A self-cleaving sequence, an internal ribosome entry site (IRES) element, a promoter at the 5' end of the at least one gene coding sequence, or a 3’ regulatory sequence.
  8. The method of claim 1, wherein the Cas nuclease is selected from a protein or an RNA molecule encoding the protein.
  9. The method of claim 1, wherein the Cas nuclease is a class 1 endonuclease or class 2 endonuclease.
  10. The method of claim 1, wherein the Cas nuclease is Cas9, Cpf1, or another Cas orthologue having genome editing activity.
  11. The method of claim 1, wherein at least two small guide RNAs (sgRNA) targeting TRAC sequences listed in Table 2 are used in the ribonucleoprotein complex.
  12. The method of claim 1, wherein at least two small guide RNAs (sgRNA) in the ribonucleoprotein complex are selected from any sequences that are complementary to at least two selected nucleic acid sequences within a target locus in the host cell genome.
  13. The method of claim 1, further comprising detecting an RNA transcribed from the polynucleotide sequence, or a protein encoded by the polynucleotide sequence, expressed by the host cell..
  14. The method of claim 1, further comprising evaluating the functionality of the inserted polynucleotide via in vitro or in vivo assays.
  15. The method of claim 14, wherein the in vitro assay is a cytotoxicity assay comprising contacting a target cell expressing a target antigen with the host cell of claim 1, wherein the polypeptide specifically binds to the target antigen, and determining the number of cells lysed by the cell.
  16. The method of claim 14, wherein the in vivo assay comprises administering the host cell of claim 1 to a living organism comprising cells expressing a target  antigen that specifically binds the polypeptide, and detecting a decrease in the number of cells that express the target antigen.
  17. The method of claim 1, wherein the host cell is isolated from a human with cancer.
  18. The method of claim 1, wherein the host cell is isolated from a human carrying an inherited disease.
  19. The method of claim 1, wherein the pre-determined locus is TRAC, ACTB, or GAPDH.
  20. The method of claim 1, wherein the at least one guide RNA comprises a nucleic acid region of about 20 nucleotides that is complementary to the pre-determined polynucleotide sequence in the host cell genome.
  21. The method of claim 1, further comprising administering the host cell to a subject to treat a disease in a subject.
  22. The method of claim 21, wherein the disease is a hematological cancer.
  23. A modified host cell produced by the method of claim 1.
  24. A method for treating a disease in a subject, the method comprising:
    administering the host cell of claim 1 or the modified host cell of claim 23 to the subject.
  25. The method of claim 24, wherein the host cell is an autologous immune cell isolated from the subject.
  26. The method of claim 24, wherein the host cell is an allogeneic immune cell.
  27. The method of claim 24, wherein the method further comprises detecting expression of a RNA or protein encoded by the polynucleotide sequence in the donor vector.
  28. The method of claim 24, wherein the method further comprises confirming the expression of the RNA or protein encoded by the desired gene is sufficient to treat the disease.
  29. The method of claim 24, wherein the disease is a hematological cancer.
  30. A kit for treating a somatic tissue disease comprising:
    (i) a first container comprising a donor vector, wherein the donor vector comprises a polynucleotide encoding a CAR;
    (ii) a second container comprising at least two sgRNA that is complementary to a selected nucleic acid sequence in the host cell genome; and
    (iii) a third container comprising a Cas nuclease.
  31. The kit of claim 30, further comprising a Cas or CfPl protein or a RNA encoding a Cas or Cfpl protein.
  32. The kit of claim 30, further comprising an instruction manual.
PCT/CN2021/078164 2020-02-28 2021-02-26 Engineering immune cells via simultaneous knock-in and gene disruption WO2021170089A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180031193.5A CN115552015A (en) 2020-02-28 2021-02-26 Engineering immune cells via simultaneous knock-in and gene disruption

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202062983024P 2020-02-28 2020-02-28
US62/983,024 2020-02-28

Publications (1)

Publication Number Publication Date
WO2021170089A1 true WO2021170089A1 (en) 2021-09-02

Family

ID=77490728

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/078164 WO2021170089A1 (en) 2020-02-28 2021-02-26 Engineering immune cells via simultaneous knock-in and gene disruption

Country Status (2)

Country Link
CN (1) CN115552015A (en)
WO (1) WO2021170089A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116732099B (en) * 2023-08-07 2023-11-24 北赛泓升(北京)生物科技有限公司 Stem cell multiple CRISPR/Cas genome editing method

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016049024A2 (en) * 2014-09-24 2016-03-31 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for modeling competition of multiple cancer mutations in vivo
CN107523588A (en) * 2017-08-18 2017-12-29 深圳大学 A kind of tetracycline slow virus inducible expression carrier and its method for building up and application
US10150985B2 (en) * 2014-02-13 2018-12-11 Takara Bio Usa, Inc. Methods of depleting a target molecule from an initial collection of nucleic acids, and compositions and kits for practicing the same
CN109112159A (en) * 2018-09-06 2019-01-01 西北农林科技大学 Based on the Cas9 site-directed integration FABP4 gene mediated and MSTN point mutation targeting vector and recombinant cell
CN109312316A (en) * 2016-02-15 2019-02-05 本森希尔生物系统股份有限公司 The composition and method of modifier group
WO2019113499A1 (en) * 2017-12-07 2019-06-13 The Broad Institute, Inc. High-throughput methods for identifying gene interactions and networks
WO2019134561A1 (en) * 2018-01-05 2019-07-11 The Chinese University Of Hong Kong High efficiency in vivo knock-in using crispr
CN110484549A (en) * 2018-04-20 2019-11-22 北京大学 Genome targeting modification method

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10150985B2 (en) * 2014-02-13 2018-12-11 Takara Bio Usa, Inc. Methods of depleting a target molecule from an initial collection of nucleic acids, and compositions and kits for practicing the same
WO2016049024A2 (en) * 2014-09-24 2016-03-31 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for modeling competition of multiple cancer mutations in vivo
CN109312316A (en) * 2016-02-15 2019-02-05 本森希尔生物系统股份有限公司 The composition and method of modifier group
CN107523588A (en) * 2017-08-18 2017-12-29 深圳大学 A kind of tetracycline slow virus inducible expression carrier and its method for building up and application
WO2019113499A1 (en) * 2017-12-07 2019-06-13 The Broad Institute, Inc. High-throughput methods for identifying gene interactions and networks
WO2019134561A1 (en) * 2018-01-05 2019-07-11 The Chinese University Of Hong Kong High efficiency in vivo knock-in using crispr
CN110484549A (en) * 2018-04-20 2019-11-22 北京大学 Genome targeting modification method
CN109112159A (en) * 2018-09-06 2019-01-01 西北农林科技大学 Based on the Cas9 site-directed integration FABP4 gene mediated and MSTN point mutation targeting vector and recombinant cell

Also Published As

Publication number Publication date
CN115552015A (en) 2022-12-30

Similar Documents

Publication Publication Date Title
JP7236398B2 (en) Donor repair template multiplex genome editing
US11421228B2 (en) Gene-regulating compositions and methods for improved immunotherapy
KR20200133218A (en) Gene-modulating compositions and methods for improved immunotherapy
TW202003845A (en) Modified immune cells having enhanced function and methods for screening for same
AU2023202582A1 (en) Targeted replacement of endogenous T cell receptors
JP2023179469A (en) Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
AU2016379393A1 (en) Engineered meganucleases with recognition sequences found in the human beta-2 microglobulin gene
EP3519562A2 (en) Hla class i-deficient nk-92 cells with decreased immunogenicity
KR20200133219A (en) Gene-modulating compositions and methods for improved immunotherapy
WO2019005957A1 (en) Genetically-modified t cells comprising a modified intron in the t cell receptor alpha gene
KR20200130826A (en) Gene-modulating compositions and methods for improved immunotherapy
CN111263808B (en) gRNA of target HPK1 and method for editing HPK1 gene
KR102338993B1 (en) artificially engineered immune cells
JP2022519595A (en) Combination of gene targets for improved immunotherapy
JP2023182711A (en) Methods, compositions, and components for crispr-cas9 editing of cblb in t cells for immunotherapy
Huang et al. CRISPR/Cas systems to overcome challenges in developing the next generation of T cells for cancer therapy
EP2670848B1 (en) Affinity maturated t cell receptors and use thereof
WO2021170089A1 (en) Engineering immune cells via simultaneous knock-in and gene disruption
US20220242929A1 (en) Immune cells expressing modified cell receptors and methods of making
US20230340139A1 (en) Immune cells having co-expressed shrnas and logic gate systems
WO2021198163A1 (en) Cd3-fusion protein and uses thereof
WO2022152266A1 (en) Composition for gene editing, and use thereof
Palma et al. HLA-G gene editing: a novel therapeutic alternative in cancer immunotherapy
WO2022266538A2 (en) Compositions and methods for targeting, editing or modifying human genes

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: 21760361

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21760361

Country of ref document: EP

Kind code of ref document: A1