WO2020000526A1 - Shp-1-knockout t-cell and construction method therefor - Google Patents

Shp-1-knockout t-cell and construction method therefor Download PDF

Info

Publication number
WO2020000526A1
WO2020000526A1 PCT/CN2018/095635 CN2018095635W WO2020000526A1 WO 2020000526 A1 WO2020000526 A1 WO 2020000526A1 CN 2018095635 W CN2018095635 W CN 2018095635W WO 2020000526 A1 WO2020000526 A1 WO 2020000526A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
car
shp
gene
cell
Prior art date
Application number
PCT/CN2018/095635
Other languages
French (fr)
Chinese (zh)
Inventor
胡边
张琳琳
Original Assignee
奥妙生物技术(广州)有限公司
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 奥妙生物技术(广州)有限公司 filed Critical 奥妙生物技术(广州)有限公司
Publication of WO2020000526A1 publication Critical patent/WO2020000526A1/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
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • 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
    • 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
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/10Vectors comprising a non-peptidic targeting moiety

Definitions

  • the invention relates to the technical fields of tumor immunology, molecular biology, cell biology and gene editing, and in particular relates to a method for preparing SHP-1 knockout T cells by using a CRISPR / Cas9 system.
  • Immune checkpoint blocking therapy has shown amazing results in the treatment of tumors. Clinical trials have found that immune checkpoint blocking therapy can reactivate the effector functions of depleted T cells and increase the anti-tumor activity of T cells. Immune checkpoint blocking therapy mainly uses antibodies to block the interaction between immune checkpoint molecules and their corresponding antibodies to improve the anti-tumor ability of T cells. The most common immune checkpoint molecules are PD-1 and CTLA-4. Immune checkpoint blocking therapy has achieved significant results in the treatment of a variety of tumors, including melanoma, non-small cell lung cancer, Hodgkin's lymphoma, head and neck cancer, ovarian cancer, kidney cancer, bladder cancer, and mismatch repair Defective tumors [1].
  • CTLA-4-targeting antibody ipilimumab has been approved for use in the treatment of patients with advanced melanoma, 20% of whom have received antibody therapy and have a survival period of more than three years.
  • PD-1 antibodies nivolumab and pembrolizumab and PD-L1 antibody atezolizumab have also been approved by the FDA for the treatment of advanced melanoma, non-small cell lung cancer and kidney cancer.
  • Some patients show partial or complete remission after receiving immune checkpoint blockade. However, not all patients respond to the treatment of CTLA-4 and PD-1 inhibitors.
  • T cells The presence of phosphatase in T cells has important regulatory effects on the function of T cells.
  • T cells recognize the antigen through the TCR-MHC complex, a large number of intracellular signaling molecules will be phosphorylated and activated by kinases such as Lck, ZAP70, and participate in signal transmission.
  • kinases such as Lck, ZAP70
  • a large number of inhibitory phosphatases in T cells will also be activated, causing dephosphorylation of signal molecules and inactivation, affecting the transmission of T cell activation signals, and ultimately weakening T cells. Killing function of cells [2].
  • PTP phosphatase is an intracellular protein, its function cannot be blocked by antibody blocking agents such as PD-1 and CTLA-4; small molecule inhibitors of phosphatase often lack the ability to inhibit enzymes or cells. Specificity, leading to great clinical side effects. These reasons also increase the difficulty for people to make choices and operations.
  • T cells are terminally differentiated primary cells, and it is difficult to perform genetic manipulations such as transfection or infection, coupled with their limited proliferation capacity, which limits the genetic manipulation of T cells in vitro, especially gene knockout.
  • gene editing technology has provided an opportunity for T cell immunotherapy.
  • Gene editing with CRISPR / Cas9 technology is simple, efficient, and specific, which makes gene editing of T cells in vitro gradually possible from possible.
  • the purpose of the present invention is to overcome the shortcomings of the prior art and provide an SHP-1 knockout T cell and a method for constructing the same.
  • Another object of the present invention is to provide a SHP-1 knockout CAR-T cell and a method for constructing the same.
  • the T cells of tumor cells are efficiently killed, and the SHP-1 gene of T cells is endogenously silenced.
  • the SHP-1 gene of T cells or its regulatory gene is at least partially knocked out.
  • the third exon of the SHP-1 gene of the T cells was knocked out.
  • the T cells are CAR-T cells.
  • CAR-T cells include CD133 CART, CD19 CART, CD20 CART, BMSA CART, MSLN CART, EGFRVIII CART, Her2 CART, GD2 CART, CEA CART.
  • the method for constructing the T cell includes introducing into the T cell a plasmid DNA expressing Cas9 and sgRNA, a Cas9 protein or a sgRNA-Cas9 protein complex, and knocking out at least a part of the sequence of SHP-1 or at least a part of its expression control sequence to make SHP-1 Gene endogenous silencing.
  • the sgRNA sequence targets the third exon of the SHP-1 gene.
  • CAR-T cells include CD19 CART, CD20 CART, BMSACART, MSLN CART, EGFRVIIICART, Her2CART, GD2CART, CEACART.
  • a method for treating a tumor includes the following steps:
  • the SHP-1 gene of T cells or its regulatory gene is at least partially knocked out.
  • the CAR modification is selected from the group consisting of CD133, CD19, CD20, BMSA, MSLN, EGFRVIII, Her2, GD2, and CEA modifications.
  • the invention breaks through the limitations of the prior art, creatively blocks SHP-1 in T cells endogenously, and effectively improves the killing effect of T cells on tumors.
  • CAR-T cells such as CD133 CAR-T, CD19 CAR T, CD20 CAR T, BMSA CAR T, MSLN CAR T, EGFRVIII CAR T, Her2 CAR T, GD2 CAR T, CEA SHP in CAR T cells -1 knockout can effectively improve the ability of CAR-T cells to kill tumor cells and provide a new target for tumor immunotherapy.
  • Cas9 and sgRNA-expressing plasmids were transferred to T cells to achieve efficient knockout of the SHP-1 gene. Compared with the previously reported use of Cas9 protein or Cas9 mRNA and electrotransduction of sgRNA transcribed in vitro , Saving time and cost of preparation.
  • FIG. 1 is a structure of a third-generation CAR used in the present invention, including a CSF2RA chimeric receptor signal peptide, an extracellular antigen-binding region (scFv), a c-Myc tag peptide, a CD8 hinge region, and an intracellular signaling region;
  • a CSF2RA chimeric receptor signal peptide an extracellular antigen-binding region (scFv), a c-Myc tag peptide, a CD8 hinge region, and an intracellular signaling region;
  • FIG. 2 shows the knockout of the SHP-1 gene in T cells after T cells were transfected by the plasmid expressing Cas9 and sgRNA in Example 1 of the present invention
  • 3 is the result of overexpression of CD133 CAR and knockout of the SHP-1 gene in T cells by plasmid electrotransformation in the present invention, and the expression of the CAR gene and the knockout of the SHP-1 gene in the T cells were detected, respectively;
  • Figure 4 is a comparison of the efficiency of different sgRNAs
  • FIG. 5 is a detection of the ability to kill tumor cells and detection of cytokine secretion by SHP-1 knockout CD133 CAR-T cells prepared in Example 3 of the present invention
  • Figure 6 shows the results of the safety experiment of SHP-1 gene knockout in vitro mediated by CRISPR / Cas9.
  • the T cells of tumor cells are efficiently killed, and the SHP-1 gene of T cells is endogenously silenced.
  • Gene silencing refers to a decrease in the expression level of a gene or a lack of expression. Endogenous silencing means that the expression of the gene itself is reduced or is not expressed.
  • the SHP-1 gene of T cells or its regulatory gene is at least partially knocked out. So that SHP-1 cannot be expressed normally, or active SHP-1 cannot be obtained after expression.
  • CAR-T cells have specific lethality to specific cells.
  • the T cells are CAR-T cells.
  • CAR-T cells include but are not limited to CD133 CAR T, CD19 CAR T, CD20 CAR T, BMSA CAR T, MSLN CAR T, EGFRVIII CAR T, Her2 CAR T, GD2 CAR T, CEA CAR T etc.
  • the method for constructing the T cell includes introducing into the T cell a plasmid DNA expressing Cas9 and sgRNA, a Cas9 protein or a sgRNA-Cas9 protein complex, and knocking out at least a part of the sequence of SHP-1 or at least a part of its expression control sequence to make SHP-1 Gene endogenous silencing.
  • the sgRNA sequence targets the third exon of the SHP-1 gene.
  • CAR-T cells include CD19 CART, CD20 CART, BMSACART, MSLN CART, EGFRVIIICART, Her2CART, GD2CART, CEACART.
  • Figure 1a shows the position of the sgRNA-targeted sequence on the SHP1 gene.
  • T7EN1 digestion showed (Figure 1b) that cleavage of DNA occurred at the sgRNA targeting site.
  • Sanger sequencing results showed ( Figure 1c) that the mutation rate of SHP-1 gene was 87.5% (sevenths of eight).
  • the CAR-targeted antigen used in this example is CD133, and the structure of the CAR is shown in Figure 2. In this order, it includes the CSF2RA chimeric receptor signal peptide (SEQ ID NO: 1), the extracellular antigen-binding region (scFv, SEQ ID:
  • PBMCs were activated with human anti-CD3 / CD28 and Dynabeads, and cultured in AIM-V medium containing 10% FBS and 300U / ml IL-2. After 5 days of cell activation, Dynabeads were removed and cultured, and 0.5 ⁇ g / ml puromycin drug sieve to enrich CAR positive T cells;
  • the inventors also selected sgRNAs targeted at other sites for comparison ( Figure 4a and Figure 4b).
  • T7EN1 to detect and compare the cleavage effect of different sgRNAs
  • the sg1 used above has the best effect
  • the other sgRNA sg2- 4
  • the editing efficiency is lower than sg1 (editing efficiency is proportional to the brightness of the smaller bands generated, Figure 4b).
  • the experimental data show that the sequence selected and optimized in the present invention has better targeting and cutting efficiency, and has unexpected effects.
  • the method used for cell killing detection is the Luciferase-Luciferin chemiluminescence detection method. Luciferase is overexpressed in the target cells for killing. After T cells are killed, the number of target cells is reduced and the chemiluminescence detection value is reduced. By comparing the chemiluminescence detection values of target cells before and after killing, the killing ability of T cells is estimated.
  • the target cells used in the present invention are U251 cells (U251-CD133-luc) that overexpress CD133 antigen and Luciferase, and the non-target cells are U251 cells (U251-CD133) that overexpress luciferase.
  • the specific operations are as follows:
  • T cells un-transduced T cells
  • CD133 CAR CAR T cells
  • SHP-1 KO CD133 CAR gene-edited CAR T cells
  • the cultured cells were collected, washed once with PBS, and resuspended in 100 ⁇ l of PBS. The cells were transferred to an opaque 96-well plate, 100 ⁇ l of D-luciferin at a concentration of 150 ⁇ g / ml was added, and the luminal at 560 nm was detected by a microplate reader;
  • T cell electroporation uses plasmid electrotransformation, which may cause random insertion of the genome. Simultaneous insertion of Cas9 and sgRNA will cause multiple cuts of the genome and improve off-target effects. Because the Cas9 protein is fused with a GFP fluorescent protein, the integration of Cas9 can be roughly judged by detecting whether there is a GFP signal. The insertion of Cas9 was detected by flow cytometry one day and 24 days after electric rotation ( Figure 6a). The results showed that no Cas9 gene was integrated into the genome.
  • CAR T cells can be constructed in a similar way without creative effort.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Mycology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided are an SHP-1-knockout T-cell and a construction method therefor. The killing effect of the T-cell on tumors can be effectively improved by the endogenous blockade of SHP-1 in the T-cell, thereby providing a new target for tumor immunotherapy.

Description

SHP-1敲除的T细胞及其构建方法SHP-1 knockout T cells and construction method thereof 技术领域Technical field
本发明涉及肿瘤免疫学、分子生物学、细胞生物学和基因编辑技术领域,尤其涉及一种利用CRISPR/Cas9系统制备SHP-1敲除T细胞的方法。The invention relates to the technical fields of tumor immunology, molecular biology, cell biology and gene editing, and in particular relates to a method for preparing SHP-1 knockout T cells by using a CRISPR / Cas9 system.
背景技术Background technique
免疫检查点阻断治疗在肿瘤的治疗中显示出惊人的疗效。临床试验发现,免疫检查点阻断治疗可以重新激活耗竭T细胞的效应功能,提高T细胞的抗肿瘤活性。免疫检查点阻断治疗主要利用抗体来阻断免疫检查点分子与其相应抗体之间的相互作用来提高T细胞的抗肿瘤能力。最常见的免疫检查点分子是PD-1和CTLA-4。免疫检查点阻断治疗已经在多种肿瘤的治疗中取得显著成效,其中包括黑色素瘤、非小细胞性肺癌、霍奇金淋巴瘤、头颈癌、卵巢癌、肾癌、膀胱癌以及错配修复缺陷的肿瘤[1]。靶向CTLA-4的抗体ipilimumab已经获批用于晚期黑色素瘤病人的治疗中,其中20%接受过抗体治疗的病人,生存期在三年以上。近日,PD-1的抗体nivolumab和pembrolizumab以及PD-L1的抗体atezolizumab也获得FDA批准,用于晚期黑色素瘤、非小细胞性肺癌以及肾癌的治疗中。部分患者在接受免疫检查点阻断治疗后,表现出部分缓解或者是完全缓解,然而,并不是所有的病人对CTLA-4和PD-1抑制剂的治疗都有反应。Immune checkpoint blocking therapy has shown amazing results in the treatment of tumors. Clinical trials have found that immune checkpoint blocking therapy can reactivate the effector functions of depleted T cells and increase the anti-tumor activity of T cells. Immune checkpoint blocking therapy mainly uses antibodies to block the interaction between immune checkpoint molecules and their corresponding antibodies to improve the anti-tumor ability of T cells. The most common immune checkpoint molecules are PD-1 and CTLA-4. Immune checkpoint blocking therapy has achieved significant results in the treatment of a variety of tumors, including melanoma, non-small cell lung cancer, Hodgkin's lymphoma, head and neck cancer, ovarian cancer, kidney cancer, bladder cancer, and mismatch repair Defective tumors [1]. CTLA-4-targeting antibody ipilimumab has been approved for use in the treatment of patients with advanced melanoma, 20% of whom have received antibody therapy and have a survival period of more than three years. Recently, PD-1 antibodies nivolumab and pembrolizumab and PD-L1 antibody atezolizumab have also been approved by the FDA for the treatment of advanced melanoma, non-small cell lung cancer and kidney cancer. Some patients show partial or complete remission after receiving immune checkpoint blockade. However, not all patients respond to the treatment of CTLA-4 and PD-1 inhibitors.
T细胞中磷酸酶的存在对T细胞的功能发挥具有重要的调节作用。T细胞通过TCR-MHC复合物识别抗原之后,胞内大量信号分子会被Lck、ZAP70等激酶磷酸化而激活,参与信号的传递。然而为了维持稳态、防止T细胞的过度激活,T细胞中大量抑制性磷酸酶也会被激活,造成信号分子的去磷酸化而失活,影响了T细胞激活信号的传递,最终削弱了T细胞的杀伤功能[2]。细胞中有一百多种PTP磷酸酶,即使人类T细胞也含有至少45种,并且越来越多的实验证据表明这些磷酸酶往往具有独立而无可替代的作用。因此,虽然理论上适当抑制T细胞中磷酸酶的活性有可能会提高T细胞清除肿瘤细胞的能力,但是同样的,不当抑制T细胞中磷酸酶的活性,特别是关键磷酸酶的活性,可能会导致难以预见的不利后果,对免疫系统的影响未知,甚至引发自身免疫性疾病的发生。选择其中的哪一种作为我们的靶点也是一个共认的难题[2]。另外,由于PTP磷酸酶是胞内蛋白,无法通过如PD-1、CTLA-4那样的抗体阻断剂实现对其功能的阻断;磷酸酶的小分子抑制剂也往往缺乏对酶或者细胞的特异性,导致临床上毒副作用很大。这些原因也都增加了人们进行选择和操作的难度。The presence of phosphatase in T cells has important regulatory effects on the function of T cells. After T cells recognize the antigen through the TCR-MHC complex, a large number of intracellular signaling molecules will be phosphorylated and activated by kinases such as Lck, ZAP70, and participate in signal transmission. However, in order to maintain homeostasis and prevent excessive activation of T cells, a large number of inhibitory phosphatases in T cells will also be activated, causing dephosphorylation of signal molecules and inactivation, affecting the transmission of T cell activation signals, and ultimately weakening T cells. Killing function of cells [2]. There are more than a hundred types of PTP phosphatases in cells, even human T cells contain at least 45 species, and there is increasing experimental evidence that these phosphatases often have independent and irreplaceable effects. Therefore, although theoretically inhibiting phosphatase activity in T cells may increase the ability of T cells to clear tumor cells, the same, improper inhibition of phosphatase activity in T cells, especially key phosphatase activity, may Leading to unpredictable adverse consequences, the impact on the immune system is unknown, and even trigger the occurrence of autoimmune diseases. Choosing which one to use as our target is also a common problem [2]. In addition, because PTP phosphatase is an intracellular protein, its function cannot be blocked by antibody blocking agents such as PD-1 and CTLA-4; small molecule inhibitors of phosphatase often lack the ability to inhibit enzymes or cells. Specificity, leading to great clinical side effects. These reasons also increase the difficulty for people to make choices and operations.
T细胞是终末分化的原代细胞,很难进行转染或感染等基因操作,再加上其增殖能力有 限,这就限制了在体外对T细胞进行基因操作,尤其是基因的敲除。近几年,基因编辑技术的革命性发展为T细胞免疫治疗提供了契机。利用CRISPR/Cas9技术进行基因编辑操作简单、效率高、特异性强,这使得在体外对T细胞进行基因编辑逐渐由可能变为现实。T cells are terminally differentiated primary cells, and it is difficult to perform genetic manipulations such as transfection or infection, coupled with their limited proliferation capacity, which limits the genetic manipulation of T cells in vitro, especially gene knockout. In recent years, the revolutionary development of gene editing technology has provided an opportunity for T cell immunotherapy. Gene editing with CRISPR / Cas9 technology is simple, efficient, and specific, which makes gene editing of T cells in vitro gradually possible from possible.
参考文献:references:
1.Topalian SL,Drake CG,Pardoll DM.(2015)Immune checkpoint blockade:a common denominator approach to cancer therapy.Cancer Cell.27(4):450-4611.Topalian SL, Drake CG, Pardoll DM. (2015) Immune checkpoint blockade: a common denominator approach therapy. Cancer Cell. 27 (4): 450-461
2.Mustelin T,Vang T,Bottini N.(2005)Protein tyrosine phosphatases and the immune response.Nat Rev Immunol.5(1):43-57.。2. Mustelin T, Vang T, Bottini N. (2005) Protein tyrosine phosphatises and the immune response. Nat Rev Immunol. 5 (1): 43-57.
发明内容Summary of the invention
本发明的目的在于克服现有技术的不足,提供一种SHP-1敲除的T细胞及其构建方法。The purpose of the present invention is to overcome the shortcomings of the prior art and provide an SHP-1 knockout T cell and a method for constructing the same.
本发明的另一个目的在于提供一种SHP-1敲除的CAR-T细胞及其构建方法。Another object of the present invention is to provide a SHP-1 knockout CAR-T cell and a method for constructing the same.
本发明所采取的技术方案是:The technical solutions adopted by the present invention are:
高效杀伤肿瘤细胞的T细胞,T细胞的SHP-1基因被内源性沉默。The T cells of tumor cells are efficiently killed, and the SHP-1 gene of T cells is endogenously silenced.
作为上述T细胞的进一步改进,T细胞的SHP-1基因或其调控基因被至少部分敲除。As a further improvement of the above T cells, the SHP-1 gene of T cells or its regulatory gene is at least partially knocked out.
作为上述T细胞的进一步改进,T细胞的SHP-1基因的第3个外显子被敲除。As a further improvement of the aforementioned T cells, the third exon of the SHP-1 gene of the T cells was knocked out.
作为上述T细胞的进一步改进,T细胞为CAR-T细胞。As a further improvement of the above T cells, the T cells are CAR-T cells.
作为上述T细胞的进一步改进,CAR-T细胞包括CD133 CAR T,CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T。As a further improvement of the above T cells, CAR-T cells include CD133 CART, CD19 CART, CD20 CART, BMSA CART, MSLN CART, EGFRVIII CART, Her2 CART, GD2 CART, CEA CART.
上述T细胞的构建方法,包括向T细胞导入表达Cas9和sgRNA的质粒DNA、Cas9蛋白或sgRNA-Cas9蛋白复合体,敲除SHP-1的至少部分序列或其至少部分表达调控序列使SHP-1基因内源性沉默。The method for constructing the T cell includes introducing into the T cell a plasmid DNA expressing Cas9 and sgRNA, a Cas9 protein or a sgRNA-Cas9 protein complex, and knocking out at least a part of the sequence of SHP-1 or at least a part of its expression control sequence to make SHP-1 Gene endogenous silencing.
作为上述T细胞的构建方法的进一步改进,sgRNA序列靶向SHP-1基因的第3个外显子。As a further improvement of the above T-cell construction method, the sgRNA sequence targets the third exon of the SHP-1 gene.
作为上述T细胞的构建方法的进一步改进,CAR-T细胞包括CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T。As a further improvement of the above T cell construction method, CAR-T cells include CD19 CART, CD20 CART, BMSACART, MSLN CART, EGFRVIIICART, Her2CART, GD2CART, CEACART.
一种治疗肿瘤的方法,包括如下步骤:A method for treating a tumor includes the following steps:
1)分离肿瘤病人的T细胞;1) Isolate T cells from tumor patients;
2)对T细胞进行CAR修饰并内源性沉默T细胞中的SHP-1基因;2) CAR modification of T cells and endogenous silencing of SHP-1 gene in T cells;
3)将改造后的T细胞输入病人体内。3) Transfuse the transformed T cells into the patient.
作为上述方法的进一步改进,T细胞的SHP-1基因或其调控基因被至少部分敲除。As a further improvement of the above method, the SHP-1 gene of T cells or its regulatory gene is at least partially knocked out.
作为上述方法的进一步改进,T细胞的SHP-1基因的第3个外显子被敲除。As a further improvement of the above method, the third exon of the SHP-1 gene of T cells was knocked out.
作为上述方法的进一步改进,CAR修饰选自CD133、CD19、CD20、BMSA、MSLN、EGFRVIII、Her2、GD2、CEA修饰。As a further improvement of the above method, the CAR modification is selected from the group consisting of CD133, CD19, CD20, BMSA, MSLN, EGFRVIII, Her2, GD2, and CEA modifications.
本发明的有益效果是:The beneficial effects of the present invention are:
本发明突破现有技术的局限,创造性地将T细胞中的SHP-1内源性阻断,有效提高了T细胞对肿瘤的杀伤作用。特别的,将CAR-T细胞,如CD133 CAR-T、CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T细胞中的SHP-1敲除,可以有效提高CAR-T细胞杀伤肿瘤细胞的能力,为肿瘤免疫疗法提供了新的靶点。The invention breaks through the limitations of the prior art, creatively blocks SHP-1 in T cells endogenously, and effectively improves the killing effect of T cells on tumors. In particular, CAR-T cells, such as CD133 CAR-T, CD19 CAR T, CD20 CAR T, BMSA CAR T, MSLN CAR T, EGFRVIII CAR T, Her2 CAR T, GD2 CAR T, CEA SHP in CAR T cells -1 knockout can effectively improve the ability of CAR-T cells to kill tumor cells and provide a new target for tumor immunotherapy.
通过借助质粒DNA电转,将表达Cas9和sgRNA的质粒电转到T细胞中,实现了高效的SHP-1基因的敲除,与之前报道的借助Cas9蛋白或者Cas9 mRNA与体外转录的sgRNA进行电转相比,节约了制备的时间和成本。By electroporation of plasmid DNA, Cas9 and sgRNA-expressing plasmids were transferred to T cells to achieve efficient knockout of the SHP-1 gene. Compared with the previously reported use of Cas9 protein or Cas9 mRNA and electrotransduction of sgRNA transcribed in vitro , Saving time and cost of preparation.
附图说明BRIEF DESCRIPTION OF THE DRAWINGS
图1为本发明采用的三代CAR的结构,包括CSF2RA嵌合受体信号肽、胞膜外抗原结合区(scFv)、c-Myc标签肽段、CD8铰链区、胞内信号传导区;FIG. 1 is a structure of a third-generation CAR used in the present invention, including a CSF2RA chimeric receptor signal peptide, an extracellular antigen-binding region (scFv), a c-Myc tag peptide, a CD8 hinge region, and an intracellular signaling region;
图2为本发明实施例1中通过表达Cas9和sgRNA的质粒电转T细胞后,T细胞中SHP-1基因的敲除情况;FIG. 2 shows the knockout of the SHP-1 gene in T cells after T cells were transfected by the plasmid expressing Cas9 and sgRNA in Example 1 of the present invention; FIG.
图3为本发明中通过质粒电转在T细胞中实现过表达CD133 CAR和敲除SHP-1基因的结果,分别检测了T细胞中CAR基因的表达情况和SHP-1基因的敲除情况;3 is the result of overexpression of CD133 CAR and knockout of the SHP-1 gene in T cells by plasmid electrotransformation in the present invention, and the expression of the CAR gene and the knockout of the SHP-1 gene in the T cells were detected, respectively;
图4是不同sgRNA的效率比较;Figure 4 is a comparison of the efficiency of different sgRNAs;
图5为本发明实施例3中制得的SHP-1敲除的CD133 CAR-T细胞杀伤肿瘤细胞能力检测和细胞因子分泌情况检测;FIG. 5 is a detection of the ability to kill tumor cells and detection of cytokine secretion by SHP-1 knockout CD133 CAR-T cells prepared in Example 3 of the present invention; FIG.
图6是CRISPR/Cas9介导的体外细胞SHP-1基因敲除的安全性实验结果。Figure 6 shows the results of the safety experiment of SHP-1 gene knockout in vitro mediated by CRISPR / Cas9.
具体实施方式detailed description
高效杀伤肿瘤细胞的T细胞,T细胞的SHP-1基因被内源性沉默。The T cells of tumor cells are efficiently killed, and the SHP-1 gene of T cells is endogenously silenced.
基因沉默,指基因的表达量下降或基本不表达。内源性沉默指基因本身的表达量下降或基本不表达。Gene silencing refers to a decrease in the expression level of a gene or a lack of expression. Endogenous silencing means that the expression of the gene itself is reduced or is not expressed.
作为上述T细胞的进一步改进,T细胞的SHP-1基因或其调控基因被至少部分敲除。使得SHP-1无法正常表达,或表达之后也无法得到具有活性的SHP-1。As a further improvement of the above T cells, the SHP-1 gene of T cells or its regulatory gene is at least partially knocked out. So that SHP-1 cannot be expressed normally, or active SHP-1 cannot be obtained after expression.
作为上述T细胞的进一步改进,T细胞的SHP-1基因的第3个外显子被敲除。实验数据表明,针对这一外显子进行敲除,具有更高的效率,靶向性更好,相应地安全性也更佳。As a further improvement of the aforementioned T cells, the third exon of the SHP-1 gene of the T cells was knocked out. Experimental data show that knockout of this exon has higher efficiency, better targeting, and correspondingly better safety.
CAR-T细胞对特定的细胞具有特异杀伤力。作为上述T细胞的进一步改进,T细胞为 CAR-T细胞。CAR-T cells have specific lethality to specific cells. As a further improvement of the above T cells, the T cells are CAR-T cells.
作为上述T细胞的进一步改进,CAR-T细胞包括但不限于CD133 CAR T,CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T等。As a further improvement of the above T cells, CAR-T cells include but are not limited to CD133 CAR T, CD19 CAR T, CD20 CAR T, BMSA CAR T, MSLN CAR T, EGFRVIII CAR T, Her2 CAR T, GD2 CAR T, CEA CAR T etc.
上述T细胞的构建方法,包括向T细胞导入表达Cas9和sgRNA的质粒DNA、Cas9蛋白或sgRNA-Cas9蛋白复合体,敲除SHP-1的至少部分序列或其至少部分表达调控序列使SHP-1基因内源性沉默。The method for constructing the T cell includes introducing into the T cell a plasmid DNA expressing Cas9 and sgRNA, a Cas9 protein or a sgRNA-Cas9 protein complex, and knocking out at least a part of the sequence of SHP-1 or at least a part of its expression control sequence to make SHP-1 Gene endogenous silencing.
作为上述T细胞的构建方法的进一步改进,sgRNA序列靶向SHP-1基因的第3个外显子。As a further improvement of the above T-cell construction method, the sgRNA sequence targets the third exon of the SHP-1 gene.
作为上述T细胞的构建方法的进一步改进,CAR-T细胞包括CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T。As a further improvement of the above T cell construction method, CAR-T cells include CD19 CART, CD20 CART, BMSACART, MSLN CART, EGFRVIIICART, Her2CART, GD2CART, CEACART.
下面结合实施例及实验,进一步示例性说明本发明的技术方案。其他CAR T细胞的敲除可以参照下述的方法进行。The technical solutions of the present invention will be further exemplified below in combination with examples and experiments. Knockout of other CAR T cells can be performed with reference to the method described below.
实施例1Example 1
通过Cas9蛋白和sgRNA电转在T细胞中实现SHP-1基因的敲除Knockout of SHP-1 gene in T cells by Cas9 protein and sgRNA electroporation
1)实验用PBMC需要先经由human anti-CD3/CD28 Dynabeads激活,培养在含10%FBS和300U/ml IL-2的AIM-V培养基中;1) Experimental PBMCs need to be activated by human anti-CD3 / CD28 and Dynabeads, and cultured in AIM-V medium containing 10% FBS and 300U / ml IL-2;
2)两天后去除Dynabeads并进行电转:2) After two days, remove Dynabeads and perform electric transfer:
3)取4μg体外转录好的sgRNA与16μg Cas9蛋白混匀,室温孵育20分钟;3) Take 4μg of sgRNA transcribed in vitro and mix with 16μg Cas9 protein, and incubate at room temperature for 20 minutes;
4)向上述体系中加入82μl电转buffer(Lonza,V4XP-3024),18μl supplement 1,混匀;4) Add 82 μl electroporation buffer (Lonza, V4XP-3024), 18 μl supplementation 1 to the above system, and mix well;
5)取1million PBMC细胞,PBS清洗两遍后,用上述体系重悬,并转移到电转杯中,利用Lonza-4D电转仪EO-115程序电转;5) Take 1 million PBMC cells, wash them twice with PBS, resuspend them in the above system, transfer them to an electric rotor, and use the Lonza-4D electrorotator EO-115 program to electrotransform;
6)电转结束立即将细胞转移至提前预热的培养基中培养。6) Immediately after the electroporation, transfer the cells to a pre-warmed medium for cultivation.
7)电转后10天计数,并进行T7EN1酶切鉴定和Sanger测序确定敲除效率。7) Count 10 days after electroporation, and perform T7EN1 digestion identification and Sanger sequencing to determine the knockout efficiency.
结果如图1所示。图1a展示的是sgRNA靶向的序列在SHP1基因上的位置。T7EN1酶切显示(图1b),在sgRNA靶向位点发生了DNA的切割。Sanger测序结果显示(图1c),SHP-1的基因突变率为87.5%(八分之七)。The results are shown in Figure 1. Figure 1a shows the position of the sgRNA-targeted sequence on the SHP1 gene. T7EN1 digestion showed (Figure 1b) that cleavage of DNA occurred at the sgRNA targeting site. Sanger sequencing results showed (Figure 1c) that the mutation rate of SHP-1 gene was 87.5% (sevenths of eight).
实施例2:Example 2:
通过质粒电转在T细胞中实现过表达CD133 CAR和敲除SHP-1基因。Overexpression of CD133CAR and knockout of SHP-1 gene in T cells were achieved by plasmid electroporation.
1)本实施例采用的CAR靶向的抗原为CD133,CAR的结构如图2所示,依次包括 CSF2RA嵌合受体信号肽(SEQ ID NO:1)、胞膜外抗原结合区(scFv,SEQ ID NO:1) The CAR-targeted antigen used in this example is CD133, and the structure of the CAR is shown in Figure 2. In this order, it includes the CSF2RA chimeric receptor signal peptide (SEQ ID NO: 1), the extracellular antigen-binding region (scFv, SEQ ID:
2)、c-Myc标签肽段(SEQ ID NO:3)、CD8铰链区、胞内信号传导区(SEQ ID NO:2), c-Myc tag peptide (SEQ ID NO: 3), CD8 hinge region, intracellular signaling region (SEQ ID NO:
4)和T2A短肽连接的抗性基因puromycin。整个CAR基因被装载于PiggyBac质粒中,构成PiggyBac-CD133 CAR。CAR基因在EF1α启动子的驱动下起始基因的表达。CAR基因整合到基因组中依赖的是转座子的转座作用;4) Puromycin, a resistance gene linked to a T2A short peptide. The entire CAR gene is loaded into the PiggyBac plasmid and constitutes PiggyBac-CD133 CAR. The CAR gene initiates gene expression driven by the EF1α promoter. The integration of the CAR gene into the genome is dependent on the transposition of the transposon;
2)将PiggyBac-CD133 CAR质粒、transposase质粒、Cas9质粒(Addgene plasmid#44758)和敲除SHP-1基因的sgRNA质粒(sgRNA的序列如SEQ ID NO:5所示)各5μg与Amaxa电转试剂盒中的电转试剂混合,并以此混合液重悬2×10 7个PBMC细胞并转移到电转杯中,利用Lonza-2B电转仪U-014程序电转; 2) 5 μg of PiggyBac-CD133 CAR plasmid, transposase plasmid, Cas9 plasmid (Addgene plasmid # 44758) and SHP-1 gene knockout (sgRNA sequence shown in SEQ ID NO: 5) and Amaxa electroporation kit The electrotransformation reagents in the solution were mixed, and 2 × 10 7 PBMC cells were resuspended in the mixed solution and transferred to an electrorotation cup, and electrotransformed using the Lonza-2B electrorotator U-014 program;
3)电转结束立即将细胞转移至提前预热的培养基中培养;3) Immediately transfer the cells to the pre-warmed medium for cultivation immediately;
4)一天后,使用human anti-CD3/CD28 Dynabeads激活PBMC,培养在含10%FBS和300U/ml IL-2的AIM-V培养基中,细胞激活5天后,去掉Dynabeads继续培养,并以0.5μg/ml的puromycin药筛,富集CAR阳性的T细胞;4) One day later, PBMCs were activated with human anti-CD3 / CD28 and Dynabeads, and cultured in AIM-V medium containing 10% FBS and 300U / ml IL-2. After 5 days of cell activation, Dynabeads were removed and cultured, and 0.5 μg / ml puromycin drug sieve to enrich CAR positive T cells;
5)第十四天时,通过T7EN1酶切鉴定和Sanger测序确定敲除效率;通过流式分析检测CAR的表达情况。5) On the fourteenth day, knockout efficiency was determined by T7EN1 digestion identification and Sanger sequencing; CAR expression was detected by flow cytometry.
结果如图3所示。用于SHP-1敲除的sgRNA序列与图1a一致。T7EN1酶切显示(图3a),在sgRNA靶向位点发生了DNA的切割。Sanger测序结果显示(图3b),SHP-1的基因突变率为87.5%(八分之七)。CAR的表达在敲除组与未敲除组中均在95%以上(图3c)。The results are shown in Figure 3. The sgRNA sequence used for SHP-1 knockout is consistent with Figure 1a. T7EN1 digestion showed (Figure 3a) that DNA cleavage occurred at the sgRNA targeting site. Sanger sequencing results showed (Figure 3b) that the mutation rate of SHP-1 gene was 87.5% (sevenths of eight). CAR expression was above 95% in both the knockout group and the non-knockout group (Figure 3c).
与上述所用sgRNA相对的,发明人同时选用了一些其他位点靶向的sgRNA进行对照(图4a和图4b)。在使用T7EN1酶切检测并比较不同sgRNA的切割效果时,可以发现上述所用的sg1效果最好,而其他靶向第2个外显子,或者同样是第3个外显子的sgRNA(sg2-4),编辑效率均比sg1要低(编辑效率与产生的较小条带的亮度成正比,图4b)。实验数据表明,本发明中经过选择、优化后的序列,具有更好的靶向性和剪切效率,具有意料之外的效果。In contrast to the sgRNA used above, the inventors also selected sgRNAs targeted at other sites for comparison (Figure 4a and Figure 4b). When using T7EN1 to detect and compare the cleavage effect of different sgRNAs, it can be found that the sg1 used above has the best effect, while the other sgRNA (sg2- 4), the editing efficiency is lower than sg1 (editing efficiency is proportional to the brightness of the smaller bands generated, Figure 4b). The experimental data show that the sequence selected and optimized in the present invention has better targeting and cutting efficiency, and has unexpected effects.
实施例3:Example 3:
体外实验证明SHP-1敲除的CD133 CAR-T细胞对肿瘤的杀伤效果。In vitro experiments demonstrated the killing effect of CD133 CAR-T cells knocked out by SHP-1 on tumors.
通过体外杀伤实验、细胞因子分泌实验证明SHP-1敲除对CD133 CAR-T细胞抗肿瘤能力的影响。In vitro killing experiments and cytokine secretion experiments demonstrated the effect of SHP-1 knockout on the antitumor ability of CD133 CAR-T cells.
细胞杀伤检测采用的方法是Luciferase-Luciferin化学发光检测法,杀伤用靶细胞中过表达Luciferase,T细胞杀伤后,靶细胞数目减少,诱导化学发光检测值降低。通过比较杀伤前与杀伤后靶细胞的化学发光检测值,预估T细胞的杀伤能力。本发明采用的靶细胞为过表达 CD133抗原和Luciferase的U251细胞(U251-CD133-luc),非靶细胞为过表达luciferase的U251细胞(U251-CD133)。具体操作如下:The method used for cell killing detection is the Luciferase-Luciferin chemiluminescence detection method. Luciferase is overexpressed in the target cells for killing. After T cells are killed, the number of target cells is reduced and the chemiluminescence detection value is reduced. By comparing the chemiluminescence detection values of target cells before and after killing, the killing ability of T cells is estimated. The target cells used in the present invention are U251 cells (U251-CD133-luc) that overexpress CD133 antigen and Luciferase, and the non-target cells are U251 cells (U251-CD133) that overexpress luciferase. The specific operations are as follows:
1)取2×10 5个靶细胞或非靶细胞铺于48孔板中; 1) Take 2 × 10 5 target cells or non-target cells and spread them in a 48-well plate;
2)按照效靶比1∶1,4∶1,16∶1的比例,分别加入未电转过的T细胞(T cell),CAR T细胞(CD133 CAR),以及基因编辑过的CAR T细胞(SHP-1 KO CD133 CAR),其中对照组不加T细胞培养,总体积300μl/孔,每种效靶比设置三个复孔,放于37℃,5%CO 2细胞培养箱内培养16-18小时;显微镜拍照检测T细胞杀伤能力; 2) According to the effect target ratio of 1: 1, 4: 1, 16: 1, add un-transduced T cells (T cells), CAR T cells (CD133 CAR), and gene-edited CAR T cells ( SHP-1 KO CD133 CAR), in which the control group was cultured without T cells, with a total volume of 300 μl / well, three replicates were set for each effect target ratio, and the cells were cultured in a 5% CO 2 cell incubator at 37 ° C for 16- 18 hours; Photomicrograph to detect T cell killing ability;
3)收集培养的细胞,PBS清洗一次后,以100μl PBS重悬。并将细胞转移至不透明的96孔板中,加入100μl浓度为150μg/ml的D-luciferin,酶标仪检测Luminal 560nm发光值;3) The cultured cells were collected, washed once with PBS, and resuspended in 100 μl of PBS. The cells were transferred to an opaque 96-well plate, 100 μl of D-luciferin at a concentration of 150 μg / ml was added, and the luminal at 560 nm was detected by a microplate reader;
4)计算T细胞的杀伤能力:将只含有PBS的孔作为空白对照,读数为N0,只含有肿瘤细胞的孔内读数设为N,实验组读数设为N1,则细胞杀伤效率为1-(N1-N0)/(N-N0)×100%。4) Calculate the killing capacity of T cells: Take the well containing only PBS as a blank control, the reading is N0, the reading in the well containing only tumor cells is set to N, and the reading in the experimental group is set to N1, then the cell killing efficiency is 1- ( N1-N0) / (N-N0) x 100%.
细胞因子释放实验采用AlphaLISA系列试剂盒检测:Cytokine release experiments were tested using AlphaLISA series kits:
1)将2×10 5个靶细胞U251-CD133-luc铺于48孔板中,按照效靶比为16∶1的比例,向每孔中加入一定量的CAR-T细胞或SHP-1敲除的CAR-T细胞,每种T细胞分为三个复孔,共加入300μl AIM-V培养液于37℃培养箱里培养; 1) Spread 2 × 10 5 target cells U251-CD133-luc in a 48-well plate, and add a certain amount of CAR-T cells or SHP-1 to each well according to a 16: 1 target ratio. In addition to CAR-T cells, each T cell was divided into three duplicate wells, and a total of 300 μl of AIM-V medium was added and cultured in a 37 ° C incubator;
2)16个小时后,吸取100μl上清,500g离心5分钟,收集上清液用于后续检测。使用PerkinElmer公司的AlphaLISA系列试剂盒检测IFN-γ和TNF-α细胞因子释放情况。2) After 16 hours, pipette 100 μl of the supernatant, centrifuge at 500 g for 5 minutes, and collect the supernatant for subsequent detection. PerkinElmer's AlphaLISA series kits were used to detect the release of IFN-γ and TNF-α cytokines.
结果如图5所示。SHP-1敲除后显著增强了CART细胞杀伤肿瘤细胞的能力(图5a,5b),增强了CAR T细胞分泌细胞因子TNF-α和IFN-γ的能力(图5c)。The results are shown in Figure 5. SHP-1 knockout significantly enhanced the ability of CART cells to kill tumor cells (Figures 5a, 5b), and enhanced the ability of CAR T cells to secrete cytokines TNF-α and IFN-γ (Figure 5c).
实施例4:Example 4:
体外细胞实验证明CRISPR/Cas9介导的SHP-1基因敲除的安全性。In vitro cell experiments demonstrated the safety of CRISPR / Cas9-mediated SHP-1 gene knockout.
T细胞电转采用质粒电转的形式,可能会造成基因组的随机插入,Cas9和sgRNA的同时插入会造成基因组的多次切割,提高脱靶效应。因为Cas9蛋白融合有GFP荧光蛋白,通过检测是否有GFP信号可以大体判断Cas9的整合情况。通过流式检测了电转后一天和电转后24天的Cas9的插入情况(图6a)。结果表明并未检测到有Cas9基因整合到基因组中。T cell electroporation uses plasmid electrotransformation, which may cause random insertion of the genome. Simultaneous insertion of Cas9 and sgRNA will cause multiple cuts of the genome and improve off-target effects. Because the Cas9 protein is fused with a GFP fluorescent protein, the integration of Cas9 can be roughly judged by detecting whether there is a GFP signal. The insertion of Cas9 was detected by flow cytometry one day and 24 days after electric rotation (Figure 6a). The results showed that no Cas9 gene was integrated into the genome.
CRISPR/Cas9系统进行基因编辑的另外一个安全性即拖把效应。根据靶向SHP-1的sgRNA序列在www.crispr.mit.edu网站上寻找打分最高的脱靶位点。最终选取前十个打分最高 的位点作为潜在的脱靶位点(图6b)。进一步通过PCR和T7EN1对前四个潜在的脱靶位点进行了检测。T7EN1酶切分析显示(图6c),在相应的位点并没有出现脱靶效应。实验数据表明本发明的体系具有很好的安全性。Another safety feature of the CRISPR / Cas9 system for gene editing is the mop effect. Find the highest scoring off-target site based on the sgRNA sequence targeting SHP-1 on www.crispr.mit.edu. Finally, the top ten sites with the highest scores were selected as potential off-target sites (Figure 6b). The first four potential off-target sites were further detected by PCR and T7EN1. T7EN1 digestion analysis (Figure 6c) showed no off-target effect at the corresponding site. Experimental data show that the system of the present invention has good security.
其他CAR T细胞也可以采用类似的方法,在不付出创造性劳动的情况下构建得到。Other CAR T cells can be constructed in a similar way without creative effort.

Claims (12)

  1. 高效杀伤肿瘤细胞的T细胞,其特征在于:T细胞的SHP-1基因被内源性沉默。T cells that efficiently kill tumor cells are characterized in that the SHP-1 gene of the T cells is endogenously silenced.
  2. 根据权利要求1所述的T细胞,其特征在于:T细胞的SHP-1基因或其调控基因被至少部分敲除。The T cell according to claim 1, wherein the SHP-1 gene of the T cell or a regulatory gene thereof is at least partially knocked out.
  3. 根据权利要求1所述的T细胞,其特征在于:T细胞的SHP-1基因的第3个外显子被敲除。The T cell according to claim 1, wherein the third exon of the SHP-1 gene of the T cell is knocked out.
  4. 根据权利要求1~3任一项所述的T细胞,其特征在于:T细胞为CAR-T细胞。The T cell according to any one of claims 1 to 3, wherein the T cell is a CAR-T cell.
  5. 根据权利要求4所述的T细胞,其特征在于:CAR-T细胞包括CD133 CAR T,CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T。The T cell according to claim 4, characterized in that: CAR-T cells include CD133 CAR T, CD19 CAR T, CD20 CAR T, BMSA CAR T, MSLN CAR T, EGFRVIII CAR T, Her2 CAR T, GD2 CAR T , CEA CAR T.
  6. 权利要求1所述T细胞的构建方法,包括向T细胞导入表达Cas9和sgRNA的质粒DNA、Cas9蛋白或sgRNA-Cas9蛋白复合体,敲除SHP-1的至少部分序列或其至少部分表达调控序列使SHP-1基因内源性沉默。The method for constructing T cells according to claim 1, comprising introducing into the T cells plasmid DNA expressing Cas9 and sgRNA, Cas9 protein or sgRNA-Cas9 protein complex, and knocking out at least part of the sequence of SHP-1 or at least part of the expression control sequence Endogenous silencing of the SHP-1 gene.
  7. 根据权利要求6所述的方法,其特征在于:sgRNA序列靶向SHP-1基因的第3个外显子。The method according to claim 6, characterized in that the sgRNA sequence targets the third exon of the SHP-1 gene.
  8. 根据权利要求6或7所述的方法,其特征在于:CAR-T细胞包括CD19 CAR T,CD20 CAR T,BMSA CAR T,MSLN CAR T,EGFRVIII CAR T,Her2 CAR T,GD2 CAR T,CEA CAR T。The method according to claim 6 or 7, characterized in that the CAR-T cells include CD19 CART, CD20 CART, BMSA CART, MSLN CART, EGFRVIIICART, Her2 CART, GD2CART, CEACAR T.
  9. 一种治疗肿瘤的方法,包括如下步骤:A method for treating a tumor includes the following steps:
    1)分离肿瘤病人的T细胞;1) Isolate T cells from tumor patients;
    2)对T细胞进行CAR修饰并内源性沉默T细胞中的SHP-1基因;2) CAR modification of T cells and endogenous silencing of SHP-1 gene in T cells;
    3)将改造后的T细胞输入病人体内。3) Transfuse the transformed T cells into the patient.
  10. 根据权利要求9所述的方法,其特征在于:T细胞的SHP-1基因或其调控基因被至少部分敲除。The method according to claim 9, wherein the SHP-1 gene of T cells or a regulatory gene thereof is at least partially knocked out.
  11. 根据权利要求9所述的方法,其特征在于:T细胞的SHP-1基因的第3个外显子被敲除。The method according to claim 9, wherein the third exon of the SHP-1 gene of the T cell is knocked out.
  12. 根据权利要求9~11任一项所述的方法,其特征在于:CAR修饰选自CD133、CD19、CD20、BMSA、MSLN、EGFRVIII、Her2、GD2、CEA修饰。The method according to any one of claims 9 to 11, wherein the CAR modification is selected from the group consisting of CD133, CD19, CD20, BMSA, MSLN, EGFRVIII, Her2, GD2, and CEA modifications.
PCT/CN2018/095635 2018-06-26 2018-07-13 Shp-1-knockout t-cell and construction method therefor WO2020000526A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810665548.6 2018-06-26
CN201810665548.6A CN108866004A (en) 2018-06-26 2018-06-26 The T cell and its construction method that SHP-1 is knocked out

Publications (1)

Publication Number Publication Date
WO2020000526A1 true WO2020000526A1 (en) 2020-01-02

Family

ID=64294683

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/095635 WO2020000526A1 (en) 2018-06-26 2018-07-13 Shp-1-knockout t-cell and construction method therefor

Country Status (2)

Country Link
CN (1) CN108866004A (en)
WO (1) WO2020000526A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116334077B (en) * 2022-07-29 2024-02-02 江苏省人民医院(南京医科大学第一附属医院) Construction method and application of airway epithelial cell SHP-1 gene specific knockout mouse model

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016154596A1 (en) * 2015-03-25 2016-09-29 Editas Medicine, Inc. Crispr/cas-related methods, compositions and components
CN106163547A (en) * 2014-03-15 2016-11-23 诺华股份有限公司 Use Chimeric antigen receptor treatment cancer
CN107109421A (en) * 2014-10-09 2017-08-29 国立大学法人山口大学 CAR expression vectors and CAR expression T cells
WO2018096361A1 (en) * 2016-11-28 2018-05-31 Autolus Limited Signal transduction modifying protein
CN108138183A (en) * 2014-04-18 2018-06-08 爱迪塔斯医药公司 For CRISPR-CAS correlation techniques, composition and the component of immunotherapy for cancer

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107164407A (en) * 2017-07-04 2017-09-15 王小平 Gene knockout and gene overexpression are carried out simultaneously without the eucaryote that species are limited

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106163547A (en) * 2014-03-15 2016-11-23 诺华股份有限公司 Use Chimeric antigen receptor treatment cancer
CN108138183A (en) * 2014-04-18 2018-06-08 爱迪塔斯医药公司 For CRISPR-CAS correlation techniques, composition and the component of immunotherapy for cancer
CN107109421A (en) * 2014-10-09 2017-08-29 国立大学法人山口大学 CAR expression vectors and CAR expression T cells
WO2016154596A1 (en) * 2015-03-25 2016-09-29 Editas Medicine, Inc. Crispr/cas-related methods, compositions and components
WO2018096361A1 (en) * 2016-11-28 2018-05-31 Autolus Limited Signal transduction modifying protein

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WATSON, H. A. ET AL.: "SHP-1: the next checkpoint target for cancer immunotherapy", BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 44, no. 2, 15 April 2016 (2016-04-15), pages 356 - 362, XP055469547, DOI: 10.1042/BST20150251 *

Also Published As

Publication number Publication date
CN108866004A (en) 2018-11-23

Similar Documents

Publication Publication Date Title
Mathewson et al. Inhibitory CD161 receptor identified in glioma-infiltrating T cells by single-cell analysis
Ding et al. PARP inhibition elicits STING-dependent antitumor immunity in Brca1-deficient ovarian cancer
US10034925B2 (en) Modified natural killer cells and natural killer cell lines having increased cytotoxicity
Li et al. Anti-cancer efficacy of SREBP inhibitor, alone or in combination with docetaxel, in prostate cancer harboring p53 mutations
Mandula et al. Ablation of the endoplasmic reticulum stress kinase PERK induces paraptosis and type I interferon to promote anti-tumor T cell responses
Park et al. Generation of lung cancer cell lines harboring EGFR T790M mutation by CRISPR/Cas9-mediated genome editing
US20220235380A1 (en) Immune cells having co-expressed shrnas and logic gate systems
CN107699547A (en) The targeting CD133 of the gene silencings of PD 1 CAR T cells and its application
Gjuka et al. Enzyme-mediated depletion of methylthioadenosine restores T cell function in MTAP-deficient tumors and reverses immunotherapy resistance
WO2020000526A1 (en) Shp-1-knockout t-cell and construction method therefor
Yang et al. Targeting RAD51 enhances chemosensitivity of adult T‑cell leukemia‑lymphoma cells by reducing DNA double‑strand break repair
Miyauchi et al. Reprogramming of tumor-associated macrophages via NEDD4-mediated CSF1R degradation by targeting USP18
Li et al. Aurora A kinase inhibition induces accumulation of SCLC tumor cells in mitosis with restored interferon signaling to increase response to PD-L1
Morman et al. BATF regulates the expression of Nfil3, Wnt10a and miR155hg for efficient induction of antibody class switch recombination in mice
Ho et al. The CD58: CD2 axis is co-regulated with PD-L1 via CMTM6 and governs anti-tumor immunity
CN114929853A (en) Natural killer cell immunotherapy for the treatment of glioblastoma and other cancers
CN115348870B (en) Universal chimeric antigen receptor T cells and uses thereof
Kinsella et al. Attenuation of homeostatic signaling from apoptotic thymocytes triggers a global regenerative response in the thymus
CN114949218B (en) PD-L1 modulator and application thereof
Jacob et al. Gene therapy in the treatment and prevention of oral cancer: An overview
US20220304990A1 (en) Mi-2beta Inhibitor as an Immunotherapy Agent
EP3635098B1 (en) T-cells modified to overexpress phf19
Bag et al. The immunomodulatory properties of the HDAC6 inhibitor ACY241 supports robust anti-tumor response in NSCLC when coupled with the chemotherapy drug Oxaliplatin
Mengwasser Genetic screening approaches to cancer driver characterization and synthetic lethal target discovery
Su In vivo CRISPR screens identify dual function of MEN1 in regulating tumor-microenvironment interactions

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: 18924341

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 19.05.2021)

122 Ep: pct application non-entry in european phase

Ref document number: 18924341

Country of ref document: EP

Kind code of ref document: A1