[go: up one dir, main page]

WO2025214269A1 - Chimeric antigen receptor targeting gcc and use thereof - Google Patents

Chimeric antigen receptor targeting gcc and use thereof

Info

Publication number
WO2025214269A1
WO2025214269A1 PCT/CN2025/087368 CN2025087368W WO2025214269A1 WO 2025214269 A1 WO2025214269 A1 WO 2025214269A1 CN 2025087368 W CN2025087368 W CN 2025087368W WO 2025214269 A1 WO2025214269 A1 WO 2025214269A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
acid sequence
amino acid
seq
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2025/087368
Other languages
French (fr)
Chinese (zh)
Inventor
何霆
刘栋群
丁艳萍
鲁薪安
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Imunopharm Technology Co Ltd
Original Assignee
Imunopharm Technology Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Imunopharm Technology Co Ltd filed Critical Imunopharm Technology Co Ltd
Publication of WO2025214269A1 publication Critical patent/WO2025214269A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the present application relates to a chimeric antigen receptor targeting GCC, a recombinant vector comprising the chimeric antigen receptor, an engineered cell, and uses thereof in drug preparation.
  • CRC colorectal cancer
  • the 5-year survival rate for colorectal cancer can drop from approximately 90% in stage I to less than 10% in stage IV [Reference 2].
  • a small number of patients without KRAS mutations can be treated with a combination of chemotherapy and the targeted drugs Avastin and Erbitux, but there are no clinically effective treatment options for patients with KRAS or BRAF mutations.
  • the immune checkpoint inhibitor PD-1 antibody has become a first-line treatment for patients with metastatic colorectal cancer, but it is limited to patients with microsatellite instability (MSI), which accounts for approximately 15% of all colorectal cancer patients. This proportion further decreases as the degree of CRC progression increases, and the proportion of clinical stage IV patients can drop to 4%.
  • MSI microsatellite instability
  • MSS microsatellite stable
  • Guanylyl cyclase 2C (GUCY2C or GCC) is a single-pass transmembrane protein expressed in intestinal epithelial cells. It converts GTP to cyclic GMP in response to guanylin, uroguanylin, or other ligands.
  • GCC expression in normal tissues and organs is highly specific, primarily concentrated in the intestinal tract and expressed primarily at the apical level [Reference 5]. Therefore, GCC's expression within the intestinal epithelium has the potential to reduce the toxic side effects of targeted drugs on normal tissues.
  • GCC is expressed in tubular adenomas, inflammatory bowel disease, colorectal precancerous lesions, primary and metastatic colorectal cancers, and metastatic lesions of these diseases in the lymph nodes and liver. Furthermore, GCC is expressed in all dysplasias and adenocarcinomas arising from esophageal and gastrointestinal metaplasia [Reference 6]. In contrast, tumors unrelated to intestinal metaplasia, including gastric signet ring carcinoma, do not express GCC. In one study, researchers evaluated GCC protein expression in 627 gastrointestinal tumors and found that 98% of colorectal cancers expressed GCC, the highest positive rate [Reference 7]. Among other gastrointestinal malignancies, 59% of esophageal cancers, 68% of gastric cancers, and 64% of pancreatic cancers expressed GCC. Therefore, GCC could be a potential drug target for the targeted treatment of these cancers.
  • Document 4 D.T.Le, J.N.Uram, H.Wang, B.R.Bartlett, H.Kemberling, A.D.Eyring, A.D.Skora, B.S.Luber, N.S.Azad, D.Laheru, B.Biedrzycki, R.C .Donehower,A.Zaheer,G.A.Fisher,T.S.Crocenzi,J.J.Lee,S.M.Duffy,R.M.Goldberg,A.de la Chapelle,M.Koshiji,F.Bhaijee,T.Huebn er,R.H.Hruban,L.D.Wood,N.Cuka,D.M.Pardoll,N.Papadopoulos,K.W.Kinzler,S.Zhou,T.C.Cornish,J.M.Taube,R.A.Anders,J.R.Eshle
  • Document 5 D. Mathur, A. R. Root, B. Bugaj-Gaweda, S. Bisulco, X. Tan, W. Fang, J. C. Kearney, J. Lucas, M. Guffroy, J. Golas, C. M. Rohde, C.
  • the present invention aims to provide a new chimeric antigen receptor that specifically targets GCC, which has a strong and sustained killing ability against tumor cells with low antigen density, can enhance the persistence of CAR-T in the body, and enhance the anti-tumor effect.
  • the present invention relates to a chimeric antigen receptor that specifically targets GCC, which includes: an scFv that specifically recognizes GCC, a CD8 hinge region or a CD28 hinge region, a CD8 transmembrane region or a CD28 transmembrane region, a CD28 co-stimulatory signal domain or a 4-1BB co-stimulatory signal domain, and a CD3 ⁇ signal domain; wherein the scFv that specifically recognizes GCC comprises a heavy chain variable region VH and a light chain variable region VL, the VH comprises an HC CDR1 with the amino acid sequence shown in SEQ ID NO: 1, an HC CDR2 with the amino acid sequence shown in SEQ ID NO: 2, and an HC CDR3 with the amino acid sequence shown in SEQ ID NO: 3, and the VL comprises an LC CDR1 with the amino acid sequence shown in SEQ ID NO: 4, an LC CDR2 with the amino acid sequence shown in SEQ ID NO: 5, and an LC CDR
  • the present invention also relates to a nucleic acid molecule comprising a nucleic acid sequence encoding the chimeric antigen receptor of the present invention, a recombinant vector comprising the nucleic acid molecule, and an engineered cell comprising the chimeric antigen receptor, nucleic acid molecule or recombinant vector.
  • the present invention also relates to the use of the chimeric antigen receptor, nucleic acid molecule, recombinant vector or engineered cell in the preparation of a drug for treating GCC-positive tumors.
  • the present invention further relates to a method for treating GCC-positive tumors, which comprises administering an effective amount of the chimeric antigen receptor, nucleic acid molecule, recombinant vector or engineered cell of the present invention to a patient in need thereof.
  • Figure 1 shows the in vitro proliferation fold of resting T cells (Figure 1A) and the in vitro proliferation fold of CAR-T cells ( Figure 1B).
  • Figure 2 shows the CAR positive rate (Figure 2A) and CAR mean fluorescence intensity (Figure 2B) of cells in each group.
  • FIG3 shows the GCC expression levels of HCT116 cell lines at different antigen densities.
  • Figure 4 shows the killing curves of CAR-T in each group against HCT116-GCC low-expressing cells.
  • Figure 5 shows the residual amount of tumor cells (Figure 5A) and the number of CAR-T cells (Figure 5B) after 10 rounds of stimulation of HCT116-GCC low-expressing cells.
  • Figure 6 shows the residual amount of tumor cells (Figure 6A) and the number of CAR-T cells ( Figure 6B) after 12 rounds of stimulation of HCT116-GCC low-expressing cells.
  • Figure 7 shows the cytokine secretion levels of each group of CAR-T cells under stimulation with HCT116-GCC low-expressing cells.
  • Figure 8 shows the cytokine secretion levels of each group of CAR-T cells after culture in IL-2-free medium for 24 hours.
  • Figure 9 shows the cell proliferation (Figure 9A) and cell viability (Figure 9B) of each group of CAR-T cells in IL-2-free culture medium.
  • Figure 10 shows the in vitro proliferation folds of resting T cells (Figure 10A) and CAR-T cells ( Figure 10B) in different signal domains.
  • FIG11 shows the CAR positivity rate ( FIG11A ) and the average fluorescence intensity ( FIG11B ) of CAR-T cells with different signal domains detected by flow cytometry.
  • Figure 13 shows the residual amount of tumor cells (Figure 13A) and the number of CAR-T cells ( Figure 13B) after 7 rounds of stimulation of HCT116-GCC low-expressing cells.
  • Figure 14 shows the in vitro proliferation folds of resting T cells (Figure 14A) and CAR-T cells ( Figure 14B) with different promoters.
  • FIG15 shows the CAR positive rates of different promoters.
  • Figure 16 shows the killing curves of CAR-T combined with different promoters against HCT116-GCC low-expressing cells.
  • Figure 17 shows the residual amount of tumor cells (Figure 17A) and the number of CAR-T cells ( Figure 17B) after 6 rounds of stimulation of HCT116-GCC low-expressing cells.
  • Figure 18 shows the residual amount of tumor cells (Figure 18A) and the number of CAR-T cells ( Figure 18B) after 10 rounds of stimulation of HCT116-GCC low-expressing cells.
  • Figure 19 shows the cytokine secretion levels of CAR-T with different promoters after stimulation of HCT116-GCC low-expressing cells for 24 hours.
  • Figure 20 shows the cytokine secretion levels of CAR-T cells with different promoters after culture in IL-2-free medium for 24 hours.
  • Figure 21 shows the cell proliferation (Figure 21A) and cell viability (Figure 21B) of CAR-T cells with different promoters in IL-2-free culture medium.
  • Figure 22 shows the tumor inhibition ability of truncated EF1 ⁇ -CAR-T-1 in NSG tumor-bearing mice (Figure 22A) and the CAR-T expansion ability in peripheral blood (Figure 22B).
  • FIG23 shows the CT images of patient 01.
  • FIG24 shows the number of peripheral blood CAR-T cells and plasma cytokine levels of patient 01.
  • FIG25 shows tumor marker levels of patient 01.
  • FIG26 shows a CT image of Patient 02.
  • FIG27 shows the number of peripheral blood CAR-T cells and plasma cytokine levels of patient 02.
  • FIG28 shows tumor marker levels of patient 02.
  • FIG29 shows the CAR positivity rate and CAR mean fluorescence intensity of CAR-T-1 and CAR-T-7.
  • Figure 30 shows the real-time killing effects of CAR-T-1 and CAR-T-7.
  • Figure 31 shows the repeated stimulation and killing effects of CAR-T-1 and CAR-T-7.
  • the scFv sequence of the GCC CAR molecule of the present invention is derived from a monoclonal antibody that specifically recognizes the GCC target (reference patents WO2011050242A1 and WO2019178580A1).
  • the CDRs of the antibodies and antigen-binding fragments disclosed in the present invention are defined or identified by Kabat numbering.
  • the antibody or antigen-binding fragment comprises a heavy chain variable region VH and a light chain variable region VL, wherein the VH comprises an HC CDR1 having an amino acid sequence as shown in SEQ ID NO: 1, an HC CDR2 having an amino acid sequence as shown in SEQ ID NO: 2, and an HC CDR3 having an amino acid sequence as shown in SEQ ID NO: 3, and the VL comprises an LC CDR1 having an amino acid sequence as shown in SEQ ID NO: 4, an LC CDR2 having an amino acid sequence as shown in SEQ ID NO: 5, and an LC CDR3 having an amino acid sequence as shown in SEQ ID NO: 6.
  • VH is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 7
  • VL is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence of SEQ ID NO: 8.
  • the nucleotide sequence encoding the VH is shown in SEQ ID NO: 27, and the nucleotide sequence encoding the VL is shown in SEQ ID NO: 28.
  • the scFv specifically recognizing GCC used in the present invention is at least 85% identical to the amino acid sequence of SEQ ID NO: 9, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical.
  • the hinge region used in the GCC CAR molecule of the present invention is a CD8 hinge region or a CD28 hinge region, wherein the amino acid sequence of the CD8 hinge region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 10, and the amino acid sequence of the CD28 hinge region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 11.
  • the transmembrane region used in the GCC CAR molecule of the present invention is the CD8 transmembrane region or the CD28 transmembrane region, wherein the amino acid sequence of the CD8 transmembrane region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 13, and the amino acid sequence of the CD28 transmembrane region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 14.
  • the costimulatory signal domain used in the GCC CAR molecule of the present invention is a CD28 costimulatory signal domain or a 4-1BB costimulatory signal domain, wherein the amino acid sequence of the CD28 costimulatory signal domain is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 15, and the amino acid sequence of the 4-1BB costimulatory signal domain is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 16.
  • the amino acid sequence of the CD3 ⁇ signaling domain used in the GCC CAR molecule of the present invention is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 17.
  • the chimeric antigen receptor of the present invention may or may not contain other genes such as tags, cytokines, or regulatory genes as needed.
  • Tags include, for example, Strep tag II tags.
  • the present invention provides the following six chimeric antigen receptors:
  • CAR-1 contains GCC scFv, CD8 hinge region, CD8 transmembrane region, CD28 costimulatory signaling domain, and CD3 ⁇ intracellular signaling domain;
  • CAR-2 contains GCC scFv, CD28 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3 ⁇ intracellular signaling domain;
  • CAR-3 contains GCC scFv, CD8 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3 ⁇ intracellular signaling domain;
  • CAR-4 contains GCC scFv, IgG4 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3 ⁇ intracellular signaling domain;
  • CAR-5 contains GCC scFv, IgG4 hinge region, CD8 transmembrane region, CD28 costimulatory signaling domain, and CD3 ⁇ intracellular signaling domain;
  • CAR-6 contains GCC scFv, CD8 hinge region, CD8 transmembrane region, 4-1BB co-stimulatory signaling domain, and CD3 ⁇ intracellular signaling domain.
  • the six chimeric antigen receptors described above further comprise a protective peptide.
  • the protective peptide is the amino acid sequence shown in SEQ ID NO: 24.
  • the present invention further provides the following seventh chimeric antigen receptor:
  • CAR-7 contains the protective peptide shown in SEQ ID NO: 24, GCC scFv, CD8 hinge region, CD8 transmembrane region, CD28 co-stimulatory signal domain, and CD3 ⁇ intracellular signal domain.
  • CAR-1 to CAR-7 of the present invention are as follows:
  • the amino acid sequence of CAR-1 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 18;
  • the amino acid sequence of CAR-2 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 19;
  • the amino acid sequence of CAR-3 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 20;
  • the amino acid sequence of CAR-4 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 21;
  • the amino acid sequence of CAR-5 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 22;
  • the amino acid sequence of CAR-6 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 23.
  • the amino acid sequence of CAR-7 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 25.
  • the present invention also relates to a nucleic acid molecule comprising a nucleic acid sequence encoding the chimeric antigen receptor of the present invention as described above.
  • the present invention also relates to a recombinant vector comprising the nucleic acid molecule described above, including but not limited to lentivirus, adenovirus, adeno-associated virus, retrovirus, transposon, and the like.
  • the recombinant vector of the present invention may optionally further comprise a promoter, such as a truncated EF1 ⁇ promoter, a full-length EF1 ⁇ promoter, a CMV promoter, an MND promoter, a truncated PGK promoter (PGK300 promoter), or a full-length PGK promoter (PGK400 promoter), preferably a truncated EF1 ⁇ promoter, a full-length EF1 ⁇ promoter, or a CMV promoter.
  • a promoter such as a truncated EF1 ⁇ promoter, a full-length EF1 ⁇ promoter, a CMV promoter, an MND promoter, a truncated PGK promoter (PGK300 promoter), or a full-length PGK promoter (PGK400 promoter), preferably a truncated EF1 ⁇ promoter, a full-length EF1 ⁇ promoter, or a CMV promoter
  • the present invention also relates to engineered cells comprising the chimeric antigen receptor, nucleic acid molecule, or recombinant vector of the present invention as described above.
  • the cells are preferably immune cells, including, for example, T cells, B cells, ⁇ T cells, NK cells, NKT cells, monocytes, macrophages, and the like.
  • the present invention further relates to the use of the chimeric antigen receptor, nucleic acid molecule, recombinant vector, and engineered cell described above in the preparation of a medicament for treating GCC-positive tumors.
  • tumors include, but are not limited to, digestive tract tumors, such as colorectal cancer, esophageal cancer, gastric cancer, or pancreatic cancer.
  • the present invention further relates to a method for treating GCC-positive tumors, comprising administering an effective amount of a chimeric antigen receptor, nucleic acid molecule, recombinant vector, or engineered cell of the present invention to a patient in need thereof.
  • the term "treat” encompasses both alleviation and cure.
  • Specific examples of GCC-positive tumors include, but are not limited to, digestive tract tumors, such as colorectal cancer, esophageal cancer, gastric cancer, or pancreatic cancer.
  • amino acid sequences and nucleotide sequences described in this application are shown in Tables 1 and 2 below, respectively.
  • Example 1 Construction of GCC CAR molecules with different hinge and transmembrane regions and differences in in vitro proliferation
  • the gene fragments and plasmid fragments after double digestion were purified using a DNA gel recovery kit and then ligated with T4 DNA ligase to obtain the pLenti6.3/V5 plasmid carrying the CAR-1, CAR-2, CAR-3, CAR-4, or CAR-5 gene.
  • the lentiviral packaging plasmids pLP/VSVG, pLP1/MDK, and pLP2/RSK (Thermo Fisher, Waltham, MA, USA) and the lentiviral transfer plasmid obtained in step (3) were transfected into HEK293T cells using Lipofectamine 3000 (Thermo Fisher, Waltham, MA, USA). After 48 hours, the culture medium was collected and centrifuged at 300 g to remove cell debris. The cells were then ultracentrifuged at 25,000 rpm for 3 hours. The precipitate was dissolved in 1 mL of saline to obtain the desired lentiviral vector.
  • T cells Collect cells that have been attached for 2 hours, centrifuge at 400g for 5 minutes, count, and take 2 ⁇ 10 5 cells to stain with CD3APC (Biolegend) antibody to detect the proportion of T cells.
  • complete culture medium X-VIVO 15 culture medium containing 500IU/mL IL-2
  • CAR-T cells were expanded and cultured in complete culture medium for 12 days, and passaged and counted every 2 days.
  • FIG. 1A The results of resting T cell proliferation in vitro are shown in Figure 1A.
  • the T cell proliferation rates of the five groups were similar.
  • the T cell proliferation rate in the CAR-T-5 group was significantly faster, while the proliferation rate in the CAR-T-2 group was slightly lower.
  • CAR positivity was detected using GCC protein conjugated to a PE fluorescent molecule (prepared by Beijing Yimi Shenzhou Pharmaceutical Technology Co., Ltd. after expressing and purifying GCC recombinant protein in CHO cells and then conjugating it to a PE fluorescent molecule).
  • the proliferation fold of CAR-T cells in each group was calculated, as shown in Figure 1B.
  • the CAR-T cell proliferation rates of the five groups were similar, but by Day 12, the proliferation rate of CAR-T-5 was significantly increased, while there were no significant differences in the other four groups.
  • Example 3 In vitro evaluation of the cytotoxicity of GCC CAR-T cells with different hinge and transmembrane regions
  • HCT116-GCC low expression cells were made by infecting HCT-116 wild-type tumor cells with lentivirus containing the human full-length GCC gene, and then detecting and sorting the cell line with low GCC expression intensity by flow cytometry.
  • HCT-116 wild-type tumor cells (not expressing human GCC) were purchased from the Cell Resource Center of the Institute of Basic Medicine, Chinese Academy of Medical Sciences.
  • the full-length human GCC sequence (NCBI sequence number: NM_004963.3) was synthesized by gene synthesis, and the DNA sequence containing the full-length human GCC was cloned into the lentiviral vector pLenti6.3/V5.
  • the lentivirus containing the full-length human GCC was prepared according to the method of step 1.1 (2) of Example 1. The lentivirus was used to infect HCT-116 wild-type cells. After 48 hours, the cells were inoculated with Alexa Fluor. The cells were incubated with 488-labeled GCC antibody (R&D Systems) at room temperature for 20 minutes.
  • the GCC expression of the cells was detected by flow cytometry (BD Arial II), and the "HCT-116-GCC low expression” cells with low GCC expression intensity and the "HCT-116-GCC high expression” cells with high GCC expression intensity were sorted. The sorted cells were expanded and cultured to a sufficient number.
  • HCT-116 wild-type tumor cells HCT116-GCC low-expressing cells and HCT116-GCC high-expressing cells were respectively After incubation with 488-labeled GCC antibody (R&D Systems) at room temperature for 20 minutes, GCC expression was detected by flow cytometry (NovoCyte 2060R, ACEA Biosciences, San Diego, CA, USA) to verify the GCC expression intensity of the two sorted cells.
  • flow cytometry NovoCyte 2060R, ACEA Biosciences, San Diego, CA, USA
  • Tumor cell plating 50 ⁇ L of IMDM complete medium containing 10% FBS was added to each well of a 96-well plate (E-plate 96) used in conjunction with a real-time killing instrument (Agilent xCELLigence RTCA SP), and the plate was placed on the RTCA Station to detect the baseline.
  • HCT-116-GCC tumor cells were digested with trypsin (Hyclone), counted, and then the cell density was adjusted to 2 ⁇ 10 5 cells/mL with IMDM complete medium containing 10% FBS.
  • the E-plate 96 was removed and 100 ⁇ L (2 ⁇ 10 4 cells/well) of tumor cell suspension was added to each well, and the plate was placed at room temperature for 30 min.
  • the E-plate 96 was placed on the RTCA Station in the incubator to monitor and record the cell proliferation curve in real time.
  • CAR-T cells were collected, centrifuged at 400 g for 5 min, resuspended in X-VIVO 15 medium, and the cell density was adjusted to 1 ⁇ 10 6 cells/mL. 1 ⁇ 10 6 T cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes.
  • APC-labeled CD3 antibody Biolegend
  • GCC protein coupled with PE fluorescent molecule prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.
  • the CAR positive rate was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA); the E-plate 96-well plate was removed and 50 ⁇ L of supernatant was aspirated from each well.
  • T cells were supplemented to each group until the total number of T cells in each group was the same, and finally X-VIVO 15 was added to 100 ⁇ L. After preparation, each group of CAR-T cells was added to each well. The number of cells in the untransfected T cell group was kept consistent with the total T cells in each group of CAR-T, and 15 to 100 ⁇ L of X-VIVO was added.
  • Tumor cell plating HCT116-GCC low-expressing tumor cells were digested with trypsin (Hyclone), counted, and then the cell density was adjusted to 1 ⁇ 10 5 cells/mL using IMDM complete medium containing 10% FBS. 500 ⁇ L of tumor cell suspension was added to each well of a 48-well plate and cultured overnight.
  • CAR-T cells Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1 ⁇ 10 6 cells/mL. Take 1 ⁇ 10 6 T cells and incubate with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. Use full spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect CAR positive rate.
  • APC-labeled CD3 antibody Biolegend
  • GCC protein coupled with PE fluorescent molecule prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.
  • Example 4 Cytokine secretion levels of GCC CAR-T cells with different hinge and transmembrane regions upon target cell stimulation
  • HCT116-GCC low-expressing tumor cells were digested with trypsin (Hyclone) and counted. The cell density was then adjusted to 1 ⁇ 10 5 cells/mL using IMDM complete medium containing 10% FBS. 500 ⁇ L of tumor cell suspension was added to each well of a 48-well plate and cultured overnight to allow the tumor cells to adhere.
  • CAR-T cells were harvested, centrifuged at 400 g for 5 min, and resuspended in X-VIVO 15 medium. The cell density was adjusted to 1 ⁇ 10 6 cells/mL. CAR positivity was determined using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA). The 48-well plate was removed and the required volume of CAR-T cells for each group was calculated based on the measured CAR positivity, using an E:T ratio of 1:1. T cells were supplemented to maintain the same total T cell count for each group, and finally, X-VIVO 15 was added to 500 ⁇ L. After preparation, CAR-T cells from each group were added to each well.
  • Example 5 Tonic signaling levels of GCC CAR-T cells with different hinge and transmembrane regions during in vitro culture
  • Basal signaling is a measure of the ability of CAR-T cells to initiate downstream signaling, promoting cell proliferation and differentiation, in the absence of exogenous antigen stimulation, relying on their own CAR molecules. Moderate basal signaling is crucial for maintaining CAR-T effector function and survival.
  • Day 8 CAR-T cells in each group were cultured in X-VIVO 15 (without exogenous IL-2). Cytokine levels were measured 24 hours later, and cell counts were performed every 2-3 days to comprehensively analyze the basal signal strength of CAR-T cells.
  • Example 6 CAR-T cell proliferation assay containing GCC CAR molecules with different co-stimulatory signaling domains
  • CAR-T-1 Based on the CAR-T-1 with the best killing function in vitro, the functions of CAR-T with two co-stimulatory signaling domains (CD28 and 4-1BB) were compared.
  • CAR-6 The amino acid sequence of CAR-6 is shown in Table 1.
  • CAR-T-1 and CAR-T-6 cells containing CAR-6 were prepared according to the method in Example 1. On the 5th and 11th days of culture, the cells were collected, centrifuged and counted. 2 ⁇ 10 5 cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein conjugated with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The expression of CAR in each group of cells was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA).
  • CAR-T cells were expanded and cultured in X-VIVO 15 medium containing 500 IU/mL for 11 days, with passages counted every three days. CAR-T cells were harvested and centrifuged, then resuspended in 1-2 ml of X-VIVO 15 medium. Ten microliters of cell culture medium were diluted to a certain multiple and stained with trypan blue at a 1:1 volume ratio. Cell viability and viable cell concentration were recorded using a cell counter, and the total cell count in each group was calculated for statistical analysis of cell proliferation.
  • the cell proliferation results are shown in Figure 10.
  • the T cell proliferation rates and CAR-T cell proliferation rates of the CAR-T-1 and CAR-T-6 groups were similar, with no significant difference.
  • CAR expression in each CAR-T cell group was detected on days 5 and 11.
  • the CAR positivity rate of CAR-T-1 was slightly higher than that of CAR-T-6, but the mean fluorescence intensity of CAR expression was slightly lower than that of CAR-T-6.
  • Example 7 Evaluation of the in vitro killing function of GCC CAR-T cells with different costimulatory signaling domains
  • Tumor cell plating Add 50 ⁇ L of IMDM complete medium containing 10% FBS to the wells of a 96-well plate (E-plate 96) used with a real-time killing instrument (Agilent xCELLigence RTCA SP), place it on the RTCA Station, and detect the baseline; digest the HCT116-GCC low-expressing tumor cells with trypsin, count them, and then adjust the cell density to 2 ⁇ 10 5 cells/mL with IMDM complete medium containing 10% FBS; remove the E-plate 96, add 100 ⁇ L (2 ⁇ 10 4 cells/well) of tumor cell suspension to the wells, and place them at room temperature for 30 minutes; place the E-plate 96 on the RTCA Station in the incubator, and detect the cell proliferation curve after 24 hours.
  • E-plate 96 96-well plate
  • RTCA SP real-time killing instrument
  • CAR-T cells On Day 11 were collected, centrifuged at 400 g for 5 min, resuspended in X-VIVO 15 medium, and the cell density was adjusted to 1 ⁇ 10 6 cells/mL. 1 ⁇ 10 6 T cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein conjugated with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The CAR positive rate was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA).
  • the E-plate 96-well plate was removed and 50 ⁇ L of supernatant was aspirated from each well.
  • each group needs to be supplemented with T cells, and finally, 15 to 100 ⁇ L of X-VIVO is added.
  • each group of CAR-T cells is added to each well. The number of cells in the untransfected T cell group is consistent with the total T cell count in each CAR-T group, and 15 to 100 ⁇ L of X-VIVO is added.
  • the killing results are shown in Figure 12.
  • the tumor cell killing ability of CAR-T-1 is significantly better than that of CAR-T-6, indicating that the killing efficiency of CAR-T containing the CD28 costimulatory signal domain is better than that of CAR-T containing the 4-1BB costimulatory signal domain.
  • Tumor cell plating HCT116-GCC low-expressing tumor cells were digested with trypsin (Hyclone), counted, and then resuspended in IMDM complete medium containing 10% FBS. The cell density was adjusted to 1 ⁇ 10 5 cells/mL. 500 ⁇ L of tumor cell suspension was added to each well of a 48-well plate and cultured overnight.
  • CAR-T cell co-incubation Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1 ⁇ 10 6 cells/mL. Take 1 ⁇ 10 6 T cells and incubate with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. Then, use a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect the CAR positive rate.
  • APC-labeled CD3 antibody Biolegend
  • GCC protein coupled with PE fluorescent molecule prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.
  • CAR-T-1 has a stronger ability to continuously kill tumor cells than CAR-T-6, and the number of CAR-T cell proliferation is greater, indicating that CAR-T containing the CD28 co-stimulatory signal domain has better long-term killing ability and persistence than CAR-T containing the 4-1BB co-stimulatory signal domain, that is, it has a better ability to resist tumor recurrence.
  • the promoter of the lentiviral vector is designed and screened.
  • CAR-T cells were prepared in the same manner as in Example 1. CAR-T cells were expanded and cultured in complete medium for 12 days, and passaged and counted every 2 days. The in vitro proliferation of T cells and CAR-T cells is shown in Figure 14. As the culture time increases, the proliferation rate of full-length EF1 ⁇ -CAR-T-1 is the fastest, and that of PGK300-CAR-T-1 is the slowest. From day 6 to day 12 of culture, the CAR positive rate of each group of CAR-T was detected. The results are shown in Figure 15. The CAR positive rate of truncated EF1 ⁇ -CAR-T-1 and MND-CAR-T-1 is the highest.
  • Tumor cell plating Add 50 ⁇ L of IMDM complete medium containing 10% FBS to the wells of a 96-well plate (E-plate 96) used with a real-time killing instrument (Agilent xCELLigence RTCA SP), place it on the RTCA Station, and detect the baseline; digest the HCT116-GCC low-expressing tumor cells with trypsin, count them, and then adjust the cell density to 2 ⁇ 105 cells/mL with IMDM complete medium containing 10% FBS; remove the E-plate 96, add 100 ⁇ L (2 ⁇ 104 cells/well) of tumor cell suspension to each well, and place it at room temperature for 30 minutes; place the E-plate 96 on the RTCA Station in the incubator to monitor the cell proliferation curve in real time.
  • E-plate 96 96-well plate
  • RTCA SP real-time killing instrument
  • CAR-T cells were collected, centrifuged at 400 g for 5 min, resuspended in X-VIVO 15 medium, and the cell density was adjusted to 1 ⁇ 10 6 cells/mL. 1 ⁇ 10 6 T cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The CAR positive rate was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA).
  • the E-plate 96-well plate was removed and 50 ⁇ L of supernatant was aspirated from each well.
  • T cells were supplemented to each group until the total number of T cells in each group was consistent, and finally X-VIVO 15 was added to 100 ⁇ L.
  • each group of CAR-T cells was added to each well. The number of cells in the untransfected T cell group was kept consistent with the total T cells in each CAR-T group, and X-VIVO 15 to 100 ⁇ L was added.
  • Tumor cell plating HCT116-GCC tumor cells were digested with trypsin (Hyclone), counted, and then the cell density was adjusted to 1 ⁇ 10 5 cells/mL. 500 ⁇ L of tumor cell suspension was added to each well of a 48-well plate and cultured overnight.
  • CAR-T cell co-incubation Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1 ⁇ 10 6 cells/mL. Take 1 ⁇ 10 6 T cells and incubate with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. Use full spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect CAR positivity.
  • Truncated EF1 ⁇ -CAR-T-1, full-length EF1 ⁇ -CAR-T-1, and CMV-CAR-T-1 demonstrated superior continuous tumor killing abilities, while MND-CAR-T-1 demonstrated reduced tumor cell killing and a low number of expanded CAR-T cells.
  • the truncated EF1 ⁇ -CAR-T-1, full-length EF1 ⁇ -CAR-T-1 and CMV-CAR-T-1 have stronger abilities to continuously kill tumor cells, with larger numbers of CAR-T amplified and better persistence, further proving that the three promoters, namely truncated EF1 ⁇ , full-length EF1 ⁇ and CMV, can effectively regulate CAR-T-1 gene expression and anti-tumor function.
  • Tumor cell plating HCT116-GCC low-expressing tumor cells were digested with trypsin and counted. The cell density was then adjusted to 1 ⁇ 10 5 cells/mL. 500 ⁇ L of tumor cell suspension was added to each well of a 48-well plate and cultured overnight to allow the tumor cells to adhere to the wall.
  • CAR-T cell co-incubation Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1 ⁇ 10 6 cells/mL.
  • a full-spectrum flow cytometer Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA
  • T cells were added until the total T cells in each experimental group were consistent, and finally X-VIVO 15 was added to 500 ⁇ L.
  • each group of CAR-T cells was added to each well, and the untransfected T cell group was added with an equal amount of total T cells as each group of CAR-T, and X-VIVO 15 was added to 500 ⁇ L.
  • the test results are shown in Figure 19.
  • the truncated EF1 ⁇ -CAR-T-1, full-length EF1 ⁇ -CAR-T-1 and CMV-CAR-T-1 can all secrete more cytokines TNF- ⁇ , IFN- ⁇ , and IL-2, indicating that the three groups of CAR-T cells can effectively recognize GCC target antigens and activate CAR-T cells.
  • Example 11 Basal signaling levels of GCC CAR-T cells with different promoters during in vitro culture
  • Each group of CAR-T cells was cultured in X-VIVO 15 (without exogenous IL-2 supplementation). Cytokines and proliferation rates were measured after 24 hours. Cell counts and statistical analysis were performed every 2-3 days thereafter to reflect the basal signal strength of the CAR-T cells. The results are shown in Figure 20.
  • the truncated EF1 ⁇ -CAR-T-1, full-length EF1 ⁇ -CAR-T-1, and CMV-CAR-T-1 groups secreted higher levels of cytokines, including TNF- ⁇ , IFN- ⁇ , and IL-2, with CMV-CAR-T-1 producing the highest secretion level.
  • In vitro cell proliferation results are shown in Figure 21.
  • Example 12 Tumor Suppression and In Vivo Expansion Ability of CAR-T-1 Cells in a Subcutaneous Colorectal Cancer PDX Mouse Model
  • This example takes the detection of the tumor inhibition ability and in vivo expansion ability of CAR-T (truncated EF1 ⁇ -CAR-T-1) containing the above-mentioned scFv (promoter is truncated EF1 ⁇ ), CD8 hinge region, CD8 transmembrane region, CD28 co-stimulatory signal domain, and CD3 ⁇ intracellular signal domain in PDX tumor-bearing mice as an example.
  • CAR-T-1 cells were prepared according to the above-mentioned CAR-T cell preparation method. When sufficient cells were cultured, they were resuspended in cryopreservative solution and stored in liquid nitrogen until use. Tumor tissue from colorectal cancer patients with high GCC expression detected by immunohistochemistry was obtained and cut into 2-3 mm3 fragments. The cells were inoculated into 6-8 week-old NSG mice. A small incision was made in the lower back of each mouse, and the tumor tissue sample was then subcutaneously transplanted into the surgical area. After 7 days, the mice were analyzed by luciferase live imaging (Lumina II Small Animal Live Imaging System, PerkinElmer, USA) to verify the successful establishment of the mouse xenograft model.
  • luciferase live imaging Lumina II Small Animal Live Imaging System, PerkinElmer, USA
  • each group of mice was injected with CAR-T-1 cells (5 ⁇ 106 CAR-T cells/mouse) through the tail vein.
  • the other two groups of mice were injected with the same volume of Dulbecco's phosphate buffered saline (DPBS) and the same number of T cells but without lentivirus transduction of control T cells.
  • DPBS Dulbecco's phosphate buffered saline
  • the tumor size of mice was measured 1 day before CAR-T cell injection and on days 3, 7, 12, 15, 18, 22, and 26 after injection.
  • the number of CAR-T cells in peripheral blood was detected on days 2, 6, 14, 21, and 28 after CAR-T cell injection.
  • FIG 22A Compared with the tumor burden of mice in the DPBS group and the control T cell group, CAR-T-1 cells slowed tumor growth 3 days after infusion and effectively reduced tumor burden within 26 days of detection, showing significant anti-tumor activity.
  • Figure 22B shows the number of CAR-T-1 cells in the peripheral blood of mice at different times. A high level of CAR-T cells was detected 10 days after infusion, and then decreased and maintained at a certain level.
  • Example 13 CAR-T cell expansion and anti-tumor activity in patients
  • the clinical study was designed to evaluate the safety and efficacy of infusing autologous truncated EF1- ⁇ -CAR-T-1 cells into patients with GCC tumors.
  • PBMCs Human peripheral blood mononuclear cells
  • CD3/CD28 Dynabeads (Thermo) were added at a ratio of 1.5:1 (bead:T cell ratio).
  • the cells were placed on a sample sorter and gently shaken for 30 minutes.
  • the beads were then adsorbed to the beads using a magnetic stand (Invitrogen) to isolate CD3-positive T cells.
  • To fully activate the T cells they were resuspended in complete culture medium (X-VIVO 15 + 500 IU/mL IL-2) and expanded at a density of 1.5 ⁇ 106 cells/mL. After 24 hours of magnetic bead activation, the cells were harvested and counted.
  • T cells were infected with lentivirus at an MOI of 2. 24 hours after infection, the cells were centrifuged at 400 g for 5 minutes and replaced with fresh complete culture medium for continued culture. On day 8 of culture, cells were harvested, Dynabeads removed using a magnetic rack, and centrifuged at 400 g for 5 minutes. The CAR-T cells were washed and cryopreserved using transfusion-safe freezing medium. The cell suspension was placed in cryopreservation bags, which were then cooled to -90°C and transferred to a vapor phase liquid nitrogen tank for storage. Before patient transfusion, the frozen CAR-T cells were transported to the hospital and transfused within 30 minutes after resuscitation. The transfusion dose was 1.2 ⁇ 10 9 CAR-T cells.
  • Patient 01 was diagnosed with advanced metastatic colorectal cancer, with metastases to the abdominal aorta, right iliac lymph nodes, pelvic cavity, peritoneum, anterior uterine wall, and multiple nodular metastases in both lungs. 28 days after infusion of truncated EF1- ⁇ -CAR-T-1 cells, CT scan images were obtained, as shown in Figure 23 . Lung lesions 1 and 2, as well as peritoneal metastases, were significantly reduced. Changes in tumor diameter are detailed in Table 3 . CAR-T cells expanded significantly in vivo, reaching a maximum of 3.2 ⁇ 10 7 CAR-Ts/L.
  • IL-6 levels were significantly elevated, reaching a maximum of 2666.7 pg/mL (Figure 24 ).
  • Carcinoembryonic antigen and cancer antigen levels decreased significantly during treatment ( Figure 25 ).
  • This patient was assessed as having a partial response (PR) at month 1 and a complete response (CR) at month 7. No severe CRS (e.g., no more than grade 2 CRS) was observed in Patient 01 during treatment.
  • PR partial response
  • CR complete response
  • Patient 02 was diagnosed with advanced metastatic colorectal cancer with mesenteric lymph node metastasis, retroperitoneal lymph node metastasis, lung metastasis and liver metastasis.
  • the lung lesions and liver metastases were significantly reduced, and the total tumor diameter was reduced by more than 30%.
  • the changes in tumor diameter are shown in Table 4.
  • CAR-T was significantly amplified in vivo, reaching a maximum of 1.37 ⁇ 10 8 CAR-T/L.
  • the cytokine IL-6 was significantly increased, reaching a maximum of 3500pg/mL.
  • the results are shown in Figure 27.
  • Carcinoembryonic antigen and cancer antigen decreased significantly during treatment, and the results are shown in Figure 28.
  • the patient was rated as PR (partial remission) at month 3 and maintained PR (partial remission) for more than 8 months.
  • PR partial remission
  • PR partial remission
  • no severe CRS e.g., no more than grade 2 CRS
  • Example 14 Construction of GCC CAR molecules with protective peptides and preparation of CAR-T cells
  • a chimeric gene encoding CAR-T-7 with a Strep tag II protective peptide at the N-terminus and restriction enzyme sites XbaI and SalI at both ends was synthesized by gene synthesis (Beijing Bomade Gene Technology Co., Ltd.). The specific amino acid sequence of the protective peptide is shown in Table 1.
  • the lentiviral transfer plasmid was constructed and the lentivirus was prepared according to the same method as in Example 1.
  • CAR-T cells were prepared using the same method as in Example 1. CAR-T cells were expanded and cultured in complete medium for 8 days, with passages and counts every 2 days. On day 8 of culture, the CAR positivity and mean fluorescence intensity of each CAR-T group were measured. The results are shown in Figure 29. The CAR positivity rate of CAR-T-7 was lower than that of CAR-T-1, but the CAR mean fluorescence intensity was slightly higher.
  • Example 15 In vitro evaluation of the cytotoxicity of GCC CAR-T cells with protective peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Provided is a chimeric antigen receptor targeting GCC, comprising: an scFv that specifically recognizes GCC, a CD8 hinge region or a CD28 hinge region, a CD8 transmembrane region or a CD28 transmembrane region, a CD28 co-stimulatory signal domain or a 4-1BB co-stimulatory signal domain, and a CD3ζ signal domain; the scFv that specifically recognizes GCC comprises a heavy chain variable region VH and a light chain variable region VL, the VH comprising an HC CDR1 having the amino acid sequence shown in SEQ ID NO: 1, an HC CDR2 having the amino acid sequence shown in SEQ ID NO: 2, and an HC CDR3 having the amino acid sequence shown in SEQ ID NO: 3, and the VL comprising an LC CDR1 having the amino acid sequence shown in SEQ ID NO: 4, an LC CDR2 having the amino acid sequence shown in SEQ ID NO: 5, and an LC CDR3 having the amino acid sequence shown in SEQ ID NO: 6.

Description

靶向GCC的嵌合抗原受体及其用途Chimeric antigen receptor targeting GCC and its use

本申请要求申请日为2024年04月08日、申请号为2024104170823、发明名称为“靶向GCC的嵌合抗原受体及其用途”的中国申请的优先权。该优先权申请的全部内容构成本申请的一部分。本申请中的全部引用文献内容均构成本申请的一部分。This application claims priority to a Chinese application filed on April 8, 2024, with application number 2024104170823, entitled “Chimeric Antigen Receptor Targeting GCC and Uses Thereof.” The entire contents of that priority application are incorporated herein by reference. The contents of all references cited in this application are incorporated herein by reference.

技术领域Technical Field

本申请涉及靶向GCC的嵌合抗原受体、包含该嵌合抗原受体的重组载体、工程细胞、以及它们在药物制备中的用途。The present application relates to a chimeric antigen receptor targeting GCC, a recombinant vector comprising the chimeric antigen receptor, an engineered cell, and uses thereof in drug preparation.

背景技术Background Art

最新数据显示,在全世界范围内,结直肠癌(colorectal cancer,CRC)在男性和女性中都具有很高的发生率和致死率,均位于第三位。在美国,每10万人中结直肠癌的发病率为38.7,死亡率则为13.9。在中国,2018年的结直肠癌新发病例约为31万,在所有新发癌症中占比12.2%;发病率为23.7/10万,死亡率为10.9/10万[文献1]。Recent data show that colorectal cancer (CRC) has a high incidence and mortality rate in both men and women worldwide, ranking third in both categories. In the United States, the incidence rate for colorectal cancer is 38.7 per 100,000 people, while the mortality rate is 13.9. In China, there were approximately 310,000 new cases of colorectal cancer in 2018, accounting for 12.2% of all new cancers; the incidence rate was 23.7 per 100,000 people, and the mortality rate was 10.9 per 100,000 people [Reference 1].

根据诊断阶段不同,结直肠癌的5年存活率从I期的90%左右可降至IV期的不足10%[文献2]。少数KRAS没有发生突变的患者,可采用化疗药和靶向药安维汀(Avastin)、爱必妥(Erbitux)的联合治疗,但对于KRAS、BRAF有突变的患者,临床上尚无有效治疗方案。免疫检查点抑制剂PD-1抗体已上升为转移性结直肠癌患者的一线治疗药物,但限于微卫星不稳定型(microsatellite instability,MSI)患者,这部分患者占所有结直肠癌患者约为15%,且该比例随着CRC恶化程度的增加会进一步降低,临床IV期的比例可降至4%,大部分患者是MMR基因没有突变的微卫星稳定型(microsatellite stable,MSS),占比达85%[文献3]。PD-1药物在MSI患者上具有较好的免疫应答、显著改善生存率,但对于MSS患者则几乎不起作用[文献4],因而极大限制了该药物的应用范围。Depending on the stage of diagnosis, the 5-year survival rate for colorectal cancer can drop from approximately 90% in stage I to less than 10% in stage IV [Reference 2]. A small number of patients without KRAS mutations can be treated with a combination of chemotherapy and the targeted drugs Avastin and Erbitux, but there are no clinically effective treatment options for patients with KRAS or BRAF mutations. The immune checkpoint inhibitor PD-1 antibody has become a first-line treatment for patients with metastatic colorectal cancer, but it is limited to patients with microsatellite instability (MSI), which accounts for approximately 15% of all colorectal cancer patients. This proportion further decreases as the degree of CRC progression increases, and the proportion of clinical stage IV patients can drop to 4%. The majority of patients are microsatellite stable (MSS) with no mutations in the MMR gene, accounting for 85% [Reference 3]. PD-1 drugs have a good immune response in MSI patients and significantly improve survival rates, but they are almost ineffective in MSS patients [Reference 4], which greatly limits the scope of application of this drug.

鸟苷酸环化酶(guanylyl cyclase 2C,简称为GUCY2C或GCC)是表达在肠道表皮细胞中的单次跨膜蛋白,能够响应鸟苷蛋白、尿鸟苷素或其它配体将三磷酸鸟苷转换为环磷酸鸟苷。从组织表达分布上看,GCC在正常组织或器官中的表达谱高度特异,主要集中于肠道系统,且主要在细胞顶部表达[文献5],因此GCC在肠上皮管腔表面的表达特点将有望减少靶向药对正常组织的毒副作用。Guanylyl cyclase 2C (GUCY2C or GCC) is a single-pass transmembrane protein expressed in intestinal epithelial cells. It converts GTP to cyclic GMP in response to guanylin, uroguanylin, or other ligands. GCC expression in normal tissues and organs is highly specific, primarily concentrated in the intestinal tract and expressed primarily at the apical level [Reference 5]. Therefore, GCC's expression within the intestinal epithelium has the potential to reduce the toxic side effects of targeted drugs on normal tissues.

在疾病情况下,GCC在管状腺瘤、炎性肠道疾病、结直肠癌前病变、原发和转移性结直肠癌、以及这些疾病在淋巴结和肝脏的转移灶均有表达。此外,GCC在所有食管和胃肠化生引起的异型增生和腺癌中也表达[文献6]。相反,与肠化生无关的肿瘤,包括胃印戒癌,不表达GCC。在一项研究中,研究者评估了627例胃肠道肿瘤中GCC蛋白的表达,结果显示,98%的结直肠癌表达GCC,阳性率最高[文献7]。在其他胃肠道恶性肿瘤中,59%的食管癌、68%的胃癌和64%的胰腺癌均表达GCC。因此可将GCC作为上述癌种靶向治疗的药物靶点。In the context of disease, GCC is expressed in tubular adenomas, inflammatory bowel disease, colorectal precancerous lesions, primary and metastatic colorectal cancers, and metastatic lesions of these diseases in the lymph nodes and liver. Furthermore, GCC is expressed in all dysplasias and adenocarcinomas arising from esophageal and gastrointestinal metaplasia [Reference 6]. In contrast, tumors unrelated to intestinal metaplasia, including gastric signet ring carcinoma, do not express GCC. In one study, researchers evaluated GCC protein expression in 627 gastrointestinal tumors and found that 98% of colorectal cancers expressed GCC, the highest positive rate [Reference 7]. Among other gastrointestinal malignancies, 59% of esophageal cancers, 68% of gastric cancers, and 64% of pancreatic cancers expressed GCC. Therefore, GCC could be a potential drug target for the targeted treatment of these cancers.

针对晚期结直肠癌患者,上海斯丹赛生物技术有限公司在2023年ESMO(European Society for Medical Oncology)年会上报道了一项来自中国5家临床中心2个剂量爬坡试验组21例患者的GUCY2C-CD19CAR-T产品的安全性和疗效的数据。其中1级剂量(1×106个细胞/kg)组有13名患者,2级剂量组(2×106个细胞/kg)有8名患者。临床结果显示,1级剂量组客观缓解率(ORR)为15.4%(2/13),2级剂量组客观缓解率为50%(4/8)。受试患者最常见的不良反应事件是细胞因子释放综合征(CRS)和腹泻,2例患者出现神经中毒,经皮质类固醇治疗后得到缓解。因此开发靶向GUCY2C的CAR-T有望长期缓解晚期结直肠癌患者的病情。At the 2023 ESMO (European Society for Medical Oncology) Annual Meeting, Shanghai Stansay Biotechnology Co., Ltd. reported safety and efficacy data from a GUCY2C-CD19 CAR-T product in 21 patients across two dose-escalation groups at five clinical centers in China for patients with advanced colorectal cancer. Thirteen patients were enrolled in the 1st dose group (1×10 6 cells/kg) and eight patients were enrolled in the 2nd dose group (2×10 6 cells/kg). Clinical results showed an objective response rate (ORR) of 15.4% (2/13) in the 1st dose group and 50% (4/8) in the 2nd dose group. The most common adverse events in the trial patients were cytokine release syndrome (CRS) and diarrhea. Two patients experienced neurotoxicity, which resolved after treatment with corticosteroids. Therefore, the development of CAR-T targeting GUCY2C has the potential to provide long-term relief for patients with advanced colorectal cancer.

虽然斯丹赛公司的GCC19CAR-T取得了一定的疗效,但该方法会杀伤人体正常的B淋巴细胞,引起B淋巴细胞缺失,从而出现体液免疫受损,导致人体易被病原体等侵染,增加了护理的成本。因此开发靶向GCC更加安全有效的CAR-T细胞药物的需求仍然很紧迫。While Stansay's GCC19 CAR-T has achieved some efficacy, this approach kills the body's normal B lymphocytes, causing B lymphocyte depletion and thus impairing humoral immunity, making the body susceptible to infection by pathogens and increasing the cost of care. Therefore, the need to develop safer and more effective CAR-T cell therapies targeting GCC remains urgent.

文献1:R.L.Siegel,K.D.Miller,A.Jemal,Cancer statistics,2020,CA Cancer J Clin 70(1)(2020)7-30.Document 1: R.L.Siegel, K.D.Miller, A.Jemal, Cancer statistics, 2020, CA Cancer J Clin 70(1)(2020)7-30.

文献2:R.L.Siegel,K.D.Miller,A.Goding Sauer,S.A.Fedewa,L.F.Butterly,J.C.Anderson,A.Cercek,R.A.Smith,A.Jemal,Colorectal cancer statistics,2020,CA Cancer J Clin 70(3)(2020)145-164.Document 2: R.L.Siegel, K.D.Miller, A.Goding Sauer, S.A.Fedewa, L.F.Butterly, J.C.Anderson, A.Cercek, R.A.Smith, A.Jemal, Colorectal cancer statistics, 2020, CA Cancer J Clin 70(3)(2020)145-164.

文献3:R.Gupta,S.Sinha,R.N.Paul,The impact of microsatellite stability status in colorectal cancer,Curr Probl Cancer 42(6)(2018)548-559.Document 3: R.Gupta, S.Sinha, R.N.Paul, The impact of microsatellite stability status in colorectal cancer, Curr Probl Cancer 42(6)(2018)548-559.

文献4:D.T.Le,J.N.Uram,H.Wang,B.R.Bartlett,H.Kemberling,A.D.Eyring,A.D.Skora,B.S.Luber,N.S.Azad,D.Laheru,B.Biedrzycki,R.C.Donehower,A.Zaheer,G.A.Fisher,T.S.Crocenzi,J.J.Lee,S.M.Duffy,R.M.Goldberg,A.de la Chapelle,M.Koshiji,F.Bhaijee,T.Huebner,R.H.Hruban,L.D.Wood,N.Cuka,D.M.Pardoll,N.Papadopoulos,K.W.Kinzler,S.Zhou,T.C.Cornish,J.M.Taube,R.A.Anders,J.R.Eshleman,B.Vogelstein,L.A.Diaz,Jr.,PD-1Blockade in Tumors with Mismatch-Repair Deficiency,N Engl J Med 372(26)(2015)2509-20.Document 4: D.T.Le, J.N.Uram, H.Wang, B.R.Bartlett, H.Kemberling, A.D.Eyring, A.D.Skora, B.S.Luber, N.S.Azad, D.Laheru, B.Biedrzycki, R.C .Donehower,A.Zaheer,G.A.Fisher,T.S.Crocenzi,J.J.Lee,S.M.Duffy,R.M.Goldberg,A.de la Chapelle,M.Koshiji,F.Bhaijee,T.Huebn er,R.H.Hruban,L.D.Wood,N.Cuka,D.M.Pardoll,N.Papadopoulos,K.W.Kinzler,S.Zhou,T.C.Cornish,J.M.Taube,R.A.Anders,J.R.Eshle man,B.Vogelstein,L.A.Diaz,Jr.,PD-1Blockade in Tumors with Mismatch-Repair Deficiency,N Engl J Med 372(26)(2015)2509-20.

文献5:D.Mathur,A.R.Root,B.Bugaj-Gaweda,S.Bisulco,X.Tan,W.Fang,J.C.Kearney,J.Lucas,M.Guffroy,J.Golas,C.M.Rohde,C.Stevens,C.Kamperschroer,K.Kelleher,R.F.Lawrence-Henderson,E.Upeslacis,J.Yao,J.Narula,E.R.LaVallie,D.R.Fernandez,B.S.Buetow,E.Rosfjord,L.Bloom,L.E.King,L.Tchistiakova,A.Nguyen,P.Sapra,A Novel GUCY2C-CD3T-Cell Engaging Bispecific Construct(PF-07062119)for the Treatment of Gastrointestinal Cancers,Clin Cancer Res 26(9)(2020)2188-2202.Document 5: D. Mathur, A. R. Root, B. Bugaj-Gaweda, S. Bisulco, X. Tan, W. Fang, J. C. Kearney, J. Lucas, M. Guffroy, J. Golas, C. M. Rohde, C. Stevens,C.Kamperschroer,K.Kelleher,R.F.Lawrence-Henderson,E.Upeslacis,J.Yao,J.Narula,E.R.LaVallie,D.R.Fernandez ,B.S.Buetow,E.Rosfjord,L.Bloom,L.E.King,L.Tchistiakova,A.Nguyen,P.Sapra,A Novel GUCY2C-CD3T-Cell Engaging Bispe cific Construct(PF-07062119) for the Treatment of Gastrointestinal Cancers, Clin Cancer Res 26(9)(2020)2188-2202.

文献6:H.Sung,J.Ferlay,R.L.Siegel,M.Laversanne,I.Soerjomataram,A.Jemal,F.Bray,Global Cancer Statistics 2020:GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36Cancers in 185Countries,CA Cancer J Clin 71(3)(2021)209-249.Document 6: H.Sung, J.Ferlay, R.L.Siegel, M.Laversanne, I.Soerjomataram, A.Jemal, F.Bray, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36Cancers in 185Countries,CA Cancer J Clin 71(3)(2021)209-249.

文献7:H.Danaee,T.Kalebic,T.Wyant,M.Fassan,C.Mescoli,F.Gao,W.L.Trepicchio,M.Rugge,Consistent expression of guanylyl cyclase-C in primary and metastatic gastrointestinal cancers,PLoS One 12(12)(2017)e0189953.Document 7: H. Danaee, T. Kalebic, T. Wyant, M. Fassan, C. Mescoli, F. Gao, W. L. Trepicchio, M. Rugge, Consistent expression o f guanylyl cyclase-C in primary and metastatic gastrointestinal cancers,PLoS One 12(12)(2017)e0189953.

文献8:Gross G,Waks T,Eshhar Z.Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity.Proc Natl Acad Sci U S A.1989Dec;86(24):10024-8.Document 8: Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A.1989Dec; 86(24):10024-8.

文献9:Khalil DN,Smith EL,Brentjens RJ,Wolchok JD.The future of cancer treatment:immunomodulation,CARs and combination immunotherapy.Nat Rev Clin Oncol2016;13(6):394.Document 9: Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol2016; 13(6):394.

发明内容Summary of the Invention

本发明旨在提供新的特异性靶向GCC的嵌合抗原受体,其对低抗原密度的肿瘤细胞具有较强的持续杀伤能力,能够增强CAR-T在体内的持续性,增强抗肿瘤效果。The present invention aims to provide a new chimeric antigen receptor that specifically targets GCC, which has a strong and sustained killing ability against tumor cells with low antigen density, can enhance the persistence of CAR-T in the body, and enhance the anti-tumor effect.

本发明一方面涉及特异性靶向GCC的嵌合抗原受体,其包括:特异性识别GCC的scFv、CD8铰链区或CD28铰链区、CD8跨膜区或CD28跨膜区、CD28共刺激信号域或4-1BB共刺激信号域、CD3ζ信号域;其中所述特异性识别GCC的scFv包含重链可变区VH和轻链可变区VL,所述VH包含SEQ ID NO:1所示氨基酸序列的HC CDR1、SEQ ID NO:2所示氨基酸序列的HC CDR2和SEQ ID NO:3所示氨基酸序列的HC CDR3,所述VL包含SEQ ID NO:4所示氨基酸序列的LC CDR1、SEQ ID NO:5所示氨基酸序列的LC CDR2和SEQ ID NO:6所示氨基酸序列的LC CDR3。On the one hand, the present invention relates to a chimeric antigen receptor that specifically targets GCC, which includes: an scFv that specifically recognizes GCC, a CD8 hinge region or a CD28 hinge region, a CD8 transmembrane region or a CD28 transmembrane region, a CD28 co-stimulatory signal domain or a 4-1BB co-stimulatory signal domain, and a CD3ζ signal domain; wherein the scFv that specifically recognizes GCC comprises a heavy chain variable region VH and a light chain variable region VL, the VH comprises an HC CDR1 with the amino acid sequence shown in SEQ ID NO: 1, an HC CDR2 with the amino acid sequence shown in SEQ ID NO: 2, and an HC CDR3 with the amino acid sequence shown in SEQ ID NO: 3, and the VL comprises an LC CDR1 with the amino acid sequence shown in SEQ ID NO: 4, an LC CDR2 with the amino acid sequence shown in SEQ ID NO: 5, and an LC CDR3 with the amino acid sequence shown in SEQ ID NO: 6.

本发明还涉及包含编码本发明嵌合抗原受体的核酸序列的核酸分子、包含所述核酸分子的重组载体、包含所述嵌合抗原受体、核酸分子或重组载体的工程细胞。The present invention also relates to a nucleic acid molecule comprising a nucleic acid sequence encoding the chimeric antigen receptor of the present invention, a recombinant vector comprising the nucleic acid molecule, and an engineered cell comprising the chimeric antigen receptor, nucleic acid molecule or recombinant vector.

本发明还涉及上述嵌合抗原受体、核酸分子、重组载体或工程细胞在制备治疗GCC阳性肿瘤的药物中的用途。The present invention also relates to the use of the chimeric antigen receptor, nucleic acid molecule, recombinant vector or engineered cell in the preparation of a drug for treating GCC-positive tumors.

本发明进一步涉及治疗GCC阳性肿瘤的方法,该方法包括给有需要的患者施用有效量的本发明嵌合抗原受体、核酸分子、重组载体或工程细胞。The present invention further relates to a method for treating GCC-positive tumors, which comprises administering an effective amount of the chimeric antigen receptor, nucleic acid molecule, recombinant vector or engineered cell of the present invention to a patient in need thereof.

本申请中所列文献的全部内容均以引用方式并入本文中。The entire contents of the documents listed in this application are incorporated herein by reference.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

图1显示静息态T细胞体外增殖倍数(图1A)、CAR-T细胞体外增殖倍数(图1B)。Figure 1 shows the in vitro proliferation fold of resting T cells (Figure 1A) and the in vitro proliferation fold of CAR-T cells (Figure 1B).

图2显示各组细胞的CAR阳性率(图2A)、CAR平均荧光强度(图2B)。Figure 2 shows the CAR positive rate (Figure 2A) and CAR mean fluorescence intensity (Figure 2B) of cells in each group.

图3显示不同抗原密度的HCT116细胞系的GCC表达水平。FIG3 shows the GCC expression levels of HCT116 cell lines at different antigen densities.

图4显示各组CAR-T对HCT116-GCC低表达细胞的杀伤曲线。Figure 4 shows the killing curves of CAR-T in each group against HCT116-GCC low-expressing cells.

图5显示HCT116-GCC低表达细胞10轮刺激后的肿瘤细胞残余量(图5A)、CAR-T细胞数量(图5B)。Figure 5 shows the residual amount of tumor cells (Figure 5A) and the number of CAR-T cells (Figure 5B) after 10 rounds of stimulation of HCT116-GCC low-expressing cells.

图6显示HCT116-GCC低表达细胞12轮刺激后的肿瘤细胞残余量(图6A)、CAR-T细胞数量(图6B)。Figure 6 shows the residual amount of tumor cells (Figure 6A) and the number of CAR-T cells (Figure 6B) after 12 rounds of stimulation of HCT116-GCC low-expressing cells.

图7显示各组CAR-T在HCT116-GCC低表达细胞刺激下的细胞因子分泌水平。Figure 7 shows the cytokine secretion levels of each group of CAR-T cells under stimulation with HCT116-GCC low-expressing cells.

图8显示各组CAR-T在无IL-2的培养基中培养24小时的细胞因子分泌水平。Figure 8 shows the cytokine secretion levels of each group of CAR-T cells after culture in IL-2-free medium for 24 hours.

图9显示各组CAR-T在无IL-2的培养基中的细胞增殖情况(图9A)、细胞活率(图9B)。Figure 9 shows the cell proliferation (Figure 9A) and cell viability (Figure 9B) of each group of CAR-T cells in IL-2-free culture medium.

图10显示不同信号域的静息态T细胞体外增殖倍数(图10A)、CAR-T细胞体外增殖倍数(图10B)。Figure 10 shows the in vitro proliferation folds of resting T cells (Figure 10A) and CAR-T cells (Figure 10B) in different signal domains.

图11显示流式检测不同信号域CAR-T的CAR阳性率(图11A)、CAR平均荧光强度(图11B)。FIG11 shows the CAR positivity rate ( FIG11A ) and the average fluorescence intensity ( FIG11B ) of CAR-T cells with different signal domains detected by flow cytometry.

图12显示不同信号域CAR-T对HCT116-GCC低表达细胞的杀伤曲线(E:T=1:3)。Figure 12 shows the killing curves of CAR-T with different signal domains against HCT116-GCC low-expressing cells (E:T=1:3).

图13显示HCT116-GCC低表达细胞7轮刺激后的肿瘤细胞残余量(图13A)、CAR-T细胞数量(图13B)。Figure 13 shows the residual amount of tumor cells (Figure 13A) and the number of CAR-T cells (Figure 13B) after 7 rounds of stimulation of HCT116-GCC low-expressing cells.

图14显示不同启动子的静息态T细胞体外增殖倍数(图14A)、CAR-T细胞体外增殖倍数(图14B)。Figure 14 shows the in vitro proliferation folds of resting T cells (Figure 14A) and CAR-T cells (Figure 14B) with different promoters.

图15显示不同启动子的CAR阳性率。FIG15 shows the CAR positive rates of different promoters.

图16显示与不同启动子组合的CAR-T对HCT116-GCC低表达细胞的杀伤曲线。Figure 16 shows the killing curves of CAR-T combined with different promoters against HCT116-GCC low-expressing cells.

图17显示HCT116-GCC低表达细胞6轮刺激后的肿瘤细胞残余量(图17A)、CAR-T细胞数量(图17B)。Figure 17 shows the residual amount of tumor cells (Figure 17A) and the number of CAR-T cells (Figure 17B) after 6 rounds of stimulation of HCT116-GCC low-expressing cells.

图18显示HCT116-GCC低表达细胞10轮刺激后的肿瘤细胞残余量(图18A)、CAR-T细胞数量(图18B)。Figure 18 shows the residual amount of tumor cells (Figure 18A) and the number of CAR-T cells (Figure 18B) after 10 rounds of stimulation of HCT116-GCC low-expressing cells.

图19显示不同启动子CAR-T在HCT116-GCC低表达细胞刺激24小时后的细胞因子分泌水平。Figure 19 shows the cytokine secretion levels of CAR-T with different promoters after stimulation of HCT116-GCC low-expressing cells for 24 hours.

图20显示不同启动子CAR-T在无IL-2的培养基中培养24小时的细胞因子分泌水平。Figure 20 shows the cytokine secretion levels of CAR-T cells with different promoters after culture in IL-2-free medium for 24 hours.

图21显示不同启动子CAR-T在无IL-2的培养基中的细胞增殖情况(图21A)、细胞活率(图21B)。Figure 21 shows the cell proliferation (Figure 21A) and cell viability (Figure 21B) of CAR-T cells with different promoters in IL-2-free culture medium.

图22显示截短EF1α-CAR-T-1在NSG荷瘤小鼠中的抑瘤能力(图22A)、外周血中CAR-T扩增能力(图22B)。Figure 22 shows the tumor inhibition ability of truncated EF1α-CAR-T-1 in NSG tumor-bearing mice (Figure 22A) and the CAR-T expansion ability in peripheral blood (Figure 22B).

图23显示患者01的CT影像。FIG23 shows the CT images of patient 01.

图24显示患者01的外周血CAR-T细胞数量和血浆中细胞因子水平。FIG24 shows the number of peripheral blood CAR-T cells and plasma cytokine levels of patient 01.

图25显示患者01的肿瘤标志物水平。FIG25 shows tumor marker levels of patient 01.

图26显示患者02的CT影像。FIG26 shows a CT image of Patient 02.

图27显示患者02的外周血CAR-T细胞数量和血浆中细胞因子水平。FIG27 shows the number of peripheral blood CAR-T cells and plasma cytokine levels of patient 02.

图28显示患者02的肿瘤标志物水平。FIG28 shows tumor marker levels of patient 02.

图29显示CAR-T-1和CAR-T-7的CAR阳性率和CAR平均荧光强度。FIG29 shows the CAR positivity rate and CAR mean fluorescence intensity of CAR-T-1 and CAR-T-7.

图30显示CAR-T-1和CAR-T-7的实时杀伤效果。Figure 30 shows the real-time killing effects of CAR-T-1 and CAR-T-7.

图31显示CAR-T-1和CAR-T-7的重复刺激杀伤效果。Figure 31 shows the repeated stimulation and killing effects of CAR-T-1 and CAR-T-7.

具体实施方式DETAILED DESCRIPTION

1.GCC CAR分子的设计1. Design of GCC CAR Molecules

本发明GCC CAR分子的scFv序列来源于特异识别GCC靶点的单克隆抗体(参考专利WO2011050242A1和WO2019178580A1)。本发明公开的抗体和抗原结合片段的CDR由Kabat编号所定义或识别。所述抗体或抗原结合片段包含重链可变区VH和轻链可变区VL,所述VH包含SEQ ID NO:1所示氨基酸序列的HC CDR1、SEQ ID NO:2所示氨基酸序列的HC CDR2和SEQ ID NO:3所示氨基酸序列的HC CDR3,所述VL包含SEQ ID NO:4所示氨基酸序列的LC CDR1、SEQ ID NO:5的LC CDR2和SEQ ID NO:6所示氨基酸序列的LC CDR3。The scFv sequence of the GCC CAR molecule of the present invention is derived from a monoclonal antibody that specifically recognizes the GCC target (reference patents WO2011050242A1 and WO2019178580A1). The CDRs of the antibodies and antigen-binding fragments disclosed in the present invention are defined or identified by Kabat numbering. The antibody or antigen-binding fragment comprises a heavy chain variable region VH and a light chain variable region VL, wherein the VH comprises an HC CDR1 having an amino acid sequence as shown in SEQ ID NO: 1, an HC CDR2 having an amino acid sequence as shown in SEQ ID NO: 2, and an HC CDR3 having an amino acid sequence as shown in SEQ ID NO: 3, and the VL comprises an LC CDR1 having an amino acid sequence as shown in SEQ ID NO: 4, an LC CDR2 having an amino acid sequence as shown in SEQ ID NO: 5, and an LC CDR3 having an amino acid sequence as shown in SEQ ID NO: 6.

在具体实施方式中,本发明GCC CAR分子的scFv中,VH与SEQ ID NO:7所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同,VL与SEQ ID NO:8的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。In a specific embodiment, in the scFv of the GCC CAR molecule of the present invention, VH is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 7, and VL is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence of SEQ ID NO: 8.

在更具体的实施方式中,编码上述VH的核苷酸序列如SEQ ID NO:27所示,编码上述VL的核苷酸序列如SEQ ID NO:28所示。在更具体的实施方式中,本发明所用的特异性识别GCC的scFv与SEQ ID NO:9的氨基酸序列至少85%相同、优选至少90%、更优选至少95%、进一步优选100%相同。In a more specific embodiment, the nucleotide sequence encoding the VH is shown in SEQ ID NO: 27, and the nucleotide sequence encoding the VL is shown in SEQ ID NO: 28. In a more specific embodiment, the scFv specifically recognizing GCC used in the present invention is at least 85% identical to the amino acid sequence of SEQ ID NO: 9, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical.

本发明GCC CAR分子中使用的铰链区为CD8铰链区或CD28铰链区,其中CD8铰链区的氨基酸序列与SEQ ID NO:10所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同,CD28铰链区的氨基酸序列与SEQ ID NO:11所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。The hinge region used in the GCC CAR molecule of the present invention is a CD8 hinge region or a CD28 hinge region, wherein the amino acid sequence of the CD8 hinge region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 10, and the amino acid sequence of the CD28 hinge region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 11.

本发明GCC CAR分子中使用的跨膜区为CD8跨膜区或CD28跨膜区,其中CD8跨膜区的氨基酸序列与SEQ ID NO:13所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同,CD28跨膜区的氨基酸序列与SEQ ID NO:14所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。The transmembrane region used in the GCC CAR molecule of the present invention is the CD8 transmembrane region or the CD28 transmembrane region, wherein the amino acid sequence of the CD8 transmembrane region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 13, and the amino acid sequence of the CD28 transmembrane region is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 14.

本发明GCC CAR分子中使用的共刺激信号域为CD28共刺激信号域或4-1BB共刺激信号域,其中CD28共刺激信号域的氨基酸序列与SEQ ID NO:15所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同,4-1BB共刺激信号域的氨基酸序列与SEQ ID NO:16所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。The costimulatory signal domain used in the GCC CAR molecule of the present invention is a CD28 costimulatory signal domain or a 4-1BB costimulatory signal domain, wherein the amino acid sequence of the CD28 costimulatory signal domain is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 15, and the amino acid sequence of the 4-1BB costimulatory signal domain is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 16.

本发明GCC CAR分子中使用的CD3ζ信号域的氨基酸序列与SEQ ID NO:17所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。The amino acid sequence of the CD3ζ signaling domain used in the GCC CAR molecule of the present invention is at least 85%, preferably at least 90%, more preferably at least 95%, and further preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 17.

本发明的嵌合抗原受体中,可以根据需要包含或不包含标签、细胞因子或调控基因等其它基因。标签可以列举例如Strep tag II标签等。The chimeric antigen receptor of the present invention may or may not contain other genes such as tags, cytokines, or regulatory genes as needed. Tags include, for example, Strep tag II tags.

在更具体的实施方案中,本发明提供如下六种嵌合抗原受体:In more specific embodiments, the present invention provides the following six chimeric antigen receptors:

CAR-1包含GCC scFv、CD8铰链区、CD8跨膜区、CD28共刺激信号域、CD3ζ胞内信号域;CAR-1 contains GCC scFv, CD8 hinge region, CD8 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain;

CAR-2包含GCC scFv、CD28铰链区、CD28跨膜区、CD28共刺激信号域、CD3ζ胞内信号域;CAR-2 contains GCC scFv, CD28 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain;

CAR-3包含GCC scFv、CD8铰链区、CD28跨膜区、CD28共刺激信号域、CD3ζ胞内信号域;CAR-3 contains GCC scFv, CD8 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain;

CAR-4包含GCC scFv、IgG4铰链区、CD28跨膜区、CD28共刺激信号域、CD3ζ胞内信号域;CAR-4 contains GCC scFv, IgG4 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain;

CAR-5包含GCC scFv、IgG4铰链区、CD8跨膜区、CD28共刺激信号域、CD3ζ胞内信号域;CAR-5 contains GCC scFv, IgG4 hinge region, CD8 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain;

CAR-6包含GCC scFv、CD8铰链区、CD8跨膜区、4-1BB共刺激信号域、CD3ζ胞内信号域。CAR-6 contains GCC scFv, CD8 hinge region, CD8 transmembrane region, 4-1BB co-stimulatory signaling domain, and CD3ζ intracellular signaling domain.

在其它实施方案中,上述六种嵌合抗原受体进一步包含保护肽。In other embodiments, the six chimeric antigen receptors described above further comprise a protective peptide.

在更具体的实施方案中,保护肽为SEQ ID NO:24所示的氨基酸序列。In a more specific embodiment, the protective peptide is the amino acid sequence shown in SEQ ID NO: 24.

在更具体的实施方案中,本发明进一步提供如下的第七种嵌合抗原受体:In a more specific embodiment, the present invention further provides the following seventh chimeric antigen receptor:

CAR-7包含SEQ ID NO:24所示的保护肽、GCC scFv、CD8铰链区、CD8跨膜区、CD28共刺激信号域、CD3ζ胞内信号域。CAR-7 contains the protective peptide shown in SEQ ID NO: 24, GCC scFv, CD8 hinge region, CD8 transmembrane region, CD28 co-stimulatory signal domain, and CD3ζ intracellular signal domain.

在更具体的实施方案中,本发明的CAR-1~CAR-7分别如下:In a more specific embodiment, CAR-1 to CAR-7 of the present invention are as follows:

CAR-1的氨基酸序列与SEQ ID NO:18所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同;The amino acid sequence of CAR-1 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 18;

CAR-2的氨基酸序列与SEQ ID NO:19所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同;The amino acid sequence of CAR-2 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 19;

CAR-3的氨基酸序列与SEQ ID NO:20所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同;The amino acid sequence of CAR-3 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 20;

CAR-4的氨基酸序列与SEQ ID NO:21所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同;The amino acid sequence of CAR-4 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 21;

CAR-5的氨基酸序列与SEQ ID NO:22所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同;The amino acid sequence of CAR-5 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence of SEQ ID NO: 22;

CAR-6的氨基酸序列与SEQ ID NO:23所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。The amino acid sequence of CAR-6 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 23.

CAR-7的氨基酸序列与SEQ ID NO:25所示的氨基酸序列至少85%、优选至少90%、更优选至少95%、进一步优选100%相同。The amino acid sequence of CAR-7 is at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably 100% identical to the amino acid sequence shown in SEQ ID NO: 25.

本发明还涉及核酸分子,其包含编码本发明如上所述嵌合抗原受体的核酸序列。The present invention also relates to a nucleic acid molecule comprising a nucleic acid sequence encoding the chimeric antigen receptor of the present invention as described above.

本发明还涉及重组载体,其包含如上所述的核酸分子。所述载体包括但不限于慢病毒、腺病毒、腺相关病毒、逆转录病毒、转座子等。The present invention also relates to a recombinant vector comprising the nucleic acid molecule described above, including but not limited to lentivirus, adenovirus, adeno-associated virus, retrovirus, transposon, and the like.

本发明的重组载体还可以任选进一步包含启动子,例如截短EF1α启动子、全长EF1α启动子、CMV启动子、MND启动子、截短PGK启动子PGK300启动子、全长PGK启动子PGK400启动子,优选截短EF1α启动子、全长EF1α启动子、CMV启动子。以上启动子的核苷酸序列分别对应SEQ ID NO:29~34。The recombinant vector of the present invention may optionally further comprise a promoter, such as a truncated EF1α promoter, a full-length EF1α promoter, a CMV promoter, an MND promoter, a truncated PGK promoter (PGK300 promoter), or a full-length PGK promoter (PGK400 promoter), preferably a truncated EF1α promoter, a full-length EF1α promoter, or a CMV promoter. The nucleotide sequences of the above promoters correspond to SEQ ID NOs: 29 to 34, respectively.

本发明还涉及工程细胞,其包含本发明如上所述的嵌合抗原受体、核酸分子或重组载体。所述细胞优选为免疫细胞,包括例如T细胞、B细胞、γδT细胞、NK细胞、NKT细胞、单核细胞、巨噬细胞等。The present invention also relates to engineered cells comprising the chimeric antigen receptor, nucleic acid molecule, or recombinant vector of the present invention as described above. The cells are preferably immune cells, including, for example, T cells, B cells, γδT cells, NK cells, NKT cells, monocytes, macrophages, and the like.

本发明进一步涉及如上所述的嵌合抗原受体、核酸分子、重组载体和工程细胞在制备治疗GCC阳性的肿瘤的药物中的用途。所述肿瘤的具体例子包括但不限于消化道肿瘤,例如结直肠癌、食管癌、胃癌或胰腺癌等。The present invention further relates to the use of the chimeric antigen receptor, nucleic acid molecule, recombinant vector, and engineered cell described above in the preparation of a medicament for treating GCC-positive tumors. Specific examples of such tumors include, but are not limited to, digestive tract tumors, such as colorectal cancer, esophageal cancer, gastric cancer, or pancreatic cancer.

本发明进一步涉及治疗GCC阳性肿瘤的方法,该方法包括给有需要的患者施用有效量的本发明嵌合抗原受体、核酸分子、重组载体或工程细胞。术语“治疗”包括缓解和治愈。GCC阳性肿瘤的具体例子包括但不限于消化道肿瘤,例如结直肠癌、食管癌、胃癌或胰腺癌等。The present invention further relates to a method for treating GCC-positive tumors, comprising administering an effective amount of a chimeric antigen receptor, nucleic acid molecule, recombinant vector, or engineered cell of the present invention to a patient in need thereof. The term "treat" encompasses both alleviation and cure. Specific examples of GCC-positive tumors include, but are not limited to, digestive tract tumors, such as colorectal cancer, esophageal cancer, gastric cancer, or pancreatic cancer.

本申请所记载的各氨基酸序列和核苷酸序列分别如下表1和表2所示。The amino acid sequences and nucleotide sequences described in this application are shown in Tables 1 and 2 below, respectively.

表1氨基酸序列



Table 1 Amino acid sequence



表2核苷酸序列



Table 2 Nucleotide sequences



实施例Example

实施例1:具有不同铰链区和跨膜区的GCC CAR分子的构建及体外增殖差异Example 1: Construction of GCC CAR molecules with different hinge and transmembrane regions and differences in in vitro proliferation

1.1慢病毒转移质粒的构建和慢病毒制备1.1 Construction of lentiviral transfer plasmids and lentivirus preparation

(1)CAR-1、CAR-2、CAR-3、CAR-4、CAR-5的氨基酸序列如表1中所示。将CAR-1、CAR-2、CAR-3、CAR-4、CAR-5的基因的5’端和3’端分别添加XbaI和SalI酶切位点,并通过基因合成方法合成这些基因(北京博迈德基因技术有限公司)。将以上5个基因质粒用XbaI和SalI双酶切,将含有截短的EF1α启动子的载体pLenti6.3/V5(Thermo Fisher,Waltham,MA,USA)也同样用XbaI和SalI双酶切,将双酶切后的基因片段和质粒片段分别用DNA凝胶回收试剂盒纯化后,再用T4DNA连接酶连接,获得带有CAR-1、CAR-2、CAR-3、CAR-4或CAR-5基因的pLenti6.3/V5质粒。(1) The amino acid sequences of CAR-1, CAR-2, CAR-3, CAR-4, and CAR-5 are shown in Table 1. XbaI and SalI restriction sites were added to the 5' and 3' ends of the genes of CAR-1, CAR-2, CAR-3, CAR-4, and CAR-5, respectively, and these genes were synthesized by gene synthesis (Beijing Bomade Gene Technology Co., Ltd.). The above five gene plasmids were double-digested with XbaI and SalI, and the vector pLenti6.3/V5 (Thermo Fisher, Waltham, MA, USA) containing the truncated EF1α promoter was also double-digested with XbaI and SalI. The gene fragments and plasmid fragments after double digestion were purified using a DNA gel recovery kit and then ligated with T4 DNA ligase to obtain the pLenti6.3/V5 plasmid carrying the CAR-1, CAR-2, CAR-3, CAR-4, or CAR-5 gene.

(2)将慢病毒包装质粒pLP/VSVG,pLP1/MDK,pLP2/RSK(Thermo Fisher,Waltham,MA,USA)与步骤(3)获得的慢病毒转移质粒,用Lipofectamine 3000(Thermo Fisher,Waltham,MA,USA)转染至HEK293T细胞,48小时后收集培养基,300g离心去除细胞碎片后,用超速离心机25000rpm离心3小时。将沉淀用1mL生理盐水溶解,即为所需的慢病毒载体。(2) The lentiviral packaging plasmids pLP/VSVG, pLP1/MDK, and pLP2/RSK (Thermo Fisher, Waltham, MA, USA) and the lentiviral transfer plasmid obtained in step (3) were transfected into HEK293T cells using Lipofectamine 3000 (Thermo Fisher, Waltham, MA, USA). After 48 hours, the culture medium was collected and centrifuged at 300 g to remove cell debris. The cells were then ultracentrifuged at 25,000 rpm for 3 hours. The precipitate was dissolved in 1 mL of saline to obtain the desired lentiviral vector.

1.2 CAR-T细胞制备及增殖检测1.2 CAR-T cell preparation and proliferation detection

(1)单采血淘洗:使用生理盐水将健康志愿者的单采血清洗两遍,然后用X-VIVO 15(Lonza)培养基重悬,计数。根据计数结果,按照1×108个细胞/30mL的密度将细胞贴壁,时间2小时。(1) Single-sample blood washing: Single-sample blood samples from healthy volunteers were washed twice with physiological saline, then resuspended in X-VIVO 15 (Lonza) medium and counted. Based on the count results, cells were allowed to adhere to the wall at a density of 1 × 10 8 cells/30 mL for 2 hours.

(2)T细胞的分选和激活:收集贴壁2小时的细胞,400g离心5min,计数,取2×105个细胞使用CD3APC(Biolegend)抗体染色,以检测T细胞比例。根据细胞计数结果和T细胞比例,按照磁珠:细胞=1.5:1(数量比)加入CD3/CD28Dynabeads(Thermo),放在样品分选仪(Thermo,pR5-5/12)上轻轻震荡30min。之后利用磁力架(Invitrogen)对磁珠的吸附作用,分选得到CD3阳性的T细胞。为使T细胞被充分激活,使用完全培养基(含有500IU/mL IL-2的X-VIVO 15培养基)重悬,按照1.5×106个细胞/mL的密度进行扩增培养。(2) Sorting and activation of T cells: Collect cells that have been attached for 2 hours, centrifuge at 400g for 5 minutes, count, and take 2×10 5 cells to stain with CD3APC (Biolegend) antibody to detect the proportion of T cells. According to the cell counting results and T cell ratio, CD3/CD28Dynabeads (Thermo) were added at a ratio of magnetic beads: cells = 1.5:1 (quantity ratio), and placed on a sample sorter (Thermo, pR5-5/12) and gently shaken for 30 minutes. Then, CD3-positive T cells were sorted by adsorption of magnetic beads using a magnetic stand (Invitrogen). In order to fully activate the T cells, they were resuspended in complete culture medium (X-VIVO 15 culture medium containing 500IU/mL IL-2) and expanded and cultured at a density of 1.5×10 6 cells/mL.

(3)磁珠激活24小时后,收集细胞,离心并计数。根据计数结果,平均分组,分别使用CAR-1、CAR-2、CAR-3、CAR-4和CAR-5慢病毒按MOI=2感染T细胞,得到CAR-T-1、CAR-T-2、CAR-T-3、CAR-T-4和CAR-T-5。病毒感染细胞24小时后,400g离心5min,更换新鲜的完全培养基继续培养。(3) After 24 hours of magnetic bead activation, cells were collected, centrifuged, and counted. Based on the counting results, the cells were grouped and infected with CAR-1, CAR-2, CAR-3, CAR-4, and CAR-5 lentivirus at an MOI of 2 to obtain CAR-T-1, CAR-T-2, CAR-T-3, CAR-T-4, and CAR-T-5. After 24 hours of viral infection, the cells were centrifuged at 400 g for 5 minutes and replaced with fresh complete medium for continued culture.

(4)培养4天后,收集细胞,使用磁力架去除Dynabeads,400g离心5min,使用完全培养基重悬,然后计数,按照3×105个细胞/mL的密度继续培养。(4) After 4 days of culture, the cells were collected, Dynabeads were removed using a magnetic rack, centrifuged at 400 g for 5 min, resuspended in complete medium, counted, and cultured at a density of 3 × 10 5 cells/mL.

(5)细胞扩增:将CAR-T细胞用完全培养基扩增培养12天,每隔2天传代计数。(5) Cell expansion: CAR-T cells were expanded and cultured in complete culture medium for 12 days, and passaged and counted every 2 days.

静息态T细胞体外增殖结果如图1A所示,Day 10之前,五组的T细胞增殖速率相差不大;到Day 12,CAR-T-5组的T细胞增殖速率明显更快,CAR-T-2组增殖速率略低。用偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司在CHO细胞中表达和纯化GCC重组蛋白后,偶联PE荧光分子制备而成)检测CAR阳性率,经计算得到各组CAR-T细胞的增殖倍数,结果如图1B所示,Day 10之前,五组的CAR-T细胞增殖速率水平相近,但到Day 12,CAR-T-5增殖速率明显提高,其他四组无显著差异。The results of resting T cell proliferation in vitro are shown in Figure 1A. Before Day 10, the T cell proliferation rates of the five groups were similar. By Day 12, the T cell proliferation rate in the CAR-T-5 group was significantly faster, while the proliferation rate in the CAR-T-2 group was slightly lower. CAR positivity was detected using GCC protein conjugated to a PE fluorescent molecule (prepared by Beijing Yimi Shenzhou Pharmaceutical Technology Co., Ltd. after expressing and purifying GCC recombinant protein in CHO cells and then conjugating it to a PE fluorescent molecule). The proliferation fold of CAR-T cells in each group was calculated, as shown in Figure 1B. Before Day 10, the CAR-T cell proliferation rates of the five groups were similar, but by Day 12, the proliferation rate of CAR-T-5 was significantly increased, while there were no significant differences in the other four groups.

实施例2:具有不同铰链区和跨膜区的GCC CAR-T细胞CAR表达率检测Example 2: Detection of CAR expression rates in GCC CAR-T cells with different hinge and transmembrane regions

取实施例1中不同培养时间点(Day 6、Day 8、Day 10、Day 12)的不同组CAR-T细胞,每组取2×105个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测各组细胞CAR表达情况。Different groups of CAR-T cells at different culture time points (Day 6, Day 8, Day 10, Day 12) in Example 1 were taken, and 2×10 5 T cells were taken from each group. After incubation with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes, the CAR expression of each group of cells was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA).

结果如图2A所示,各组CAR-T细胞的CAR阳性率均随着时间延长呈现出上升的趋势,其中CAR-T-3和CAR-T-4略低,但仍在50%以上。如2B所示,各组CAR-T的CAR平均荧光强度看,在Day 12之前,CAR-T-1的CAR平均荧光强度最高,说明CAR-T-1细胞表面具有最多的CAR分子水平。The results, as shown in Figure 2A, show an increasing trend in the CAR-T cell positivity rate across all groups over time, with rates slightly lower for CAR-T-3 and CAR-T-4, but still above 50%. As shown in Figure 2B, the mean fluorescence intensity of CAR in each group was highest before Day 12 for CAR-T-1, indicating that CAR-T-1 cells had the highest levels of CAR molecules on their surface.

实施例3:不同铰链区和跨膜区GCC CAR-T细胞的体外杀伤功能评价Example 3: In vitro evaluation of the cytotoxicity of GCC CAR-T cells with different hinge and transmembrane regions

为了筛选出对肿瘤GCC抗原灵敏度高的CAR-T,本实验使用“HCT116-GCC低表达”细胞作为靶细胞,该细胞是使HCT-116野生型肿瘤细胞感染人全长GCC基因的慢病毒后,通过流式细胞仪检测并分选出低GCC表达强度的细胞系。具体地,HCT-116野生型肿瘤细胞(不表达人GCC)购自中国医学科学院基础医学研究所细胞资源中心。通过基因合成方法合成全长人GCC序列(NCBI序列号:NM_004963.3),将含有全长人GCC的DNA序列克隆至慢病毒载体pLenti6.3/V5中,按照实施例1步骤1.1(2)的方法制备包含全长人GCC的慢病毒,用该慢病毒感染HCT-116野生型细胞,48小时后将细胞与Alexa488标记的GCC抗体(R&D Systems)室温孵育20分钟,通过流式细胞仪(BD Arial II)检测细胞的GCC表达,并分选出低GCC表达强度的“HCT-116-GCC低表达”的细胞和高GCC表达强度的“HCT-116-GCC高表达”的细胞;将分选获得的细胞分别扩增培养至足够数量。In order to screen out CAR-T with high sensitivity to tumor GCC antigen, this experiment used "HCT116-GCC low expression" cells as target cells. The cells were made by infecting HCT-116 wild-type tumor cells with lentivirus containing the human full-length GCC gene, and then detecting and sorting the cell line with low GCC expression intensity by flow cytometry. Specifically, HCT-116 wild-type tumor cells (not expressing human GCC) were purchased from the Cell Resource Center of the Institute of Basic Medicine, Chinese Academy of Medical Sciences. The full-length human GCC sequence (NCBI sequence number: NM_004963.3) was synthesized by gene synthesis, and the DNA sequence containing the full-length human GCC was cloned into the lentiviral vector pLenti6.3/V5. The lentivirus containing the full-length human GCC was prepared according to the method of step 1.1 (2) of Example 1. The lentivirus was used to infect HCT-116 wild-type cells. After 48 hours, the cells were inoculated with Alexa Fluor. The cells were incubated with 488-labeled GCC antibody (R&D Systems) at room temperature for 20 minutes. The GCC expression of the cells was detected by flow cytometry (BD Arial II), and the "HCT-116-GCC low expression" cells with low GCC expression intensity and the "HCT-116-GCC high expression" cells with high GCC expression intensity were sorted. The sorted cells were expanded and cultured to a sufficient number.

将HCT-116野生型肿瘤细胞、HCT116-GCC低表达细胞和HCT116-GCC高表达细胞分别与Alexa488标记的GCC抗体(R&D Systems)室温孵育20分钟后,用流式细胞仪(NovoCyte 2060R,ACEA Biosciences,San Diego,CA,USA)检测GCC表达,以验证分选出的两种细胞的GCC表达强度,结果如图3所示,HCT-116野生型肿瘤细胞无GCC阳性信号,“HCT-116-GCC低表达”的细胞有中度的GCC阳性信号,“HCT-116-GCC高表达”的细胞显示出最强的GCC阳性信号。HCT-116 wild-type tumor cells, HCT116-GCC low-expressing cells and HCT116-GCC high-expressing cells were respectively After incubation with 488-labeled GCC antibody (R&D Systems) at room temperature for 20 minutes, GCC expression was detected by flow cytometry (NovoCyte 2060R, ACEA Biosciences, San Diego, CA, USA) to verify the GCC expression intensity of the two sorted cells. The results are shown in Figure 3. HCT-116 wild-type tumor cells had no GCC-positive signal, "HCT-116-GCC low expression" cells had moderate GCC-positive signals, and "HCT-116-GCC high expression" cells showed the strongest GCC-positive signal.

3.1实时杀伤实验3.1 Real-time killing experiment

(1)肿瘤细胞铺板:在实时杀伤仪器(Agilent xCELLigence RTCA SP)配套使用的96孔板(E-plate 96)孔中,每孔加入50μL含10% FBS的IMDM完全培养基,放置于RTCA Station上,检测基线;用胰酶(Hyclone)消化HCT-116-GCC肿瘤细胞,计数,然后用含10% FBS的IMDM完全培养基将细胞密度调为2×105个细胞/mL;取出E-plate 96,在每孔中加入100μL(2×104个细胞/孔)肿瘤细胞悬液,室温放置30min;将E-plate 96放到培养箱中的RTCA Station上,实时监测和记录细胞增殖曲线。(1) Tumor cell plating: 50 μL of IMDM complete medium containing 10% FBS was added to each well of a 96-well plate (E-plate 96) used in conjunction with a real-time killing instrument (Agilent xCELLigence RTCA SP), and the plate was placed on the RTCA Station to detect the baseline. HCT-116-GCC tumor cells were digested with trypsin (Hyclone), counted, and then the cell density was adjusted to 2×10 5 cells/mL with IMDM complete medium containing 10% FBS. The E-plate 96 was removed and 100 μL (2×10 4 cells/well) of tumor cell suspension was added to each well, and the plate was placed at room temperature for 30 min. The E-plate 96 was placed on the RTCA Station in the incubator to monitor and record the cell proliferation curve in real time.

(2)CAR-T细胞加入:收集Day 8的CAR-T细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,取1×106个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率;取出E-plate 96孔板,每孔吸出50μL上清液。根据测得的CAR阳性率,按E:T=1:3计算每组CAR-T细胞所需细胞量。对各组补充T细胞至各组T细胞总数一致,最后补加X-VIVO 15至100μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组的细胞数目与各组CAR-T的总T细胞保持一致,并补加X-VIVO 15至100μL。(2) Addition of CAR-T cells: CAR-T cells on Day 8 were collected, centrifuged at 400 g for 5 min, resuspended in X-VIVO 15 medium, and the cell density was adjusted to 1×10 6 cells/mL. 1×10 6 T cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The CAR positive rate was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA); the E-plate 96-well plate was removed and 50 μL of supernatant was aspirated from each well. According to the measured CAR positive rate, the required cell amount of CAR-T cells for each group was calculated according to E:T=1:3. T cells were supplemented to each group until the total number of T cells in each group was the same, and finally X-VIVO 15 was added to 100 μL. After preparation, each group of CAR-T cells was added to each well. The number of cells in the untransfected T cell group was kept consistent with the total T cells in each group of CAR-T, and 15 to 100 μL of X-VIVO was added.

(3)将E-Plate 96孔板放回实时杀伤仪器,实时监测CAR-T对肿瘤细胞的杀伤作用,纵轴“归一化的肿瘤细胞指数”代表杀伤效率,值越低,杀伤效率越高。结果如图4A和4B所示,在E:T=1:3时,CAR-T-4和CAR-T-5对靶细胞没有杀伤作用,CAR-T-1、CAR-T-2、CAR-T-3均能较好地杀完肿瘤细胞。图4C所示,选取CAR-T和肿瘤细胞共孵育20小时的实验数据进行统计学分析,CAR-T-1、CAR-T-2和CAR-T-3彼此之间无显著性差异,但与CAR-T-4和CAR-T-5相比,均具有显著性差异。(3) The E-Plate 96-well plate was placed back into the real-time killing instrument to monitor the killing effect of CAR-T on tumor cells in real time. The vertical axis "normalized tumor cell index" represents the killing efficiency. The lower the value, the higher the killing efficiency. The results are shown in Figures 4A and 4B. When E:T = 1:3, CAR-T-4 and CAR-T-5 had no killing effect on target cells, while CAR-T-1, CAR-T-2, and CAR-T-3 were able to kill tumor cells well. As shown in Figure 4C, the experimental data of CAR-T and tumor cells co-incubated for 20 hours were selected for statistical analysis. There was no significant difference between CAR-T-1, CAR-T-2, and CAR-T-3, but there were significant differences compared with CAR-T-4 and CAR-T-5.

3.2重复刺激杀伤实验3.2 Repeated stimulation killing experiment

(1)肿瘤细胞铺板:用胰酶(Hyclone)消化HCT116-GCC低表达肿瘤细胞,计数,然后将细胞密度用含10% FBS的IMDM完全培养基调为1×105个细胞/mL,在48孔板中每孔加入500μL肿瘤细胞悬液,过夜培养。(1) Tumor cell plating: HCT116-GCC low-expressing tumor cells were digested with trypsin (Hyclone), counted, and then the cell density was adjusted to 1×10 5 cells/mL using IMDM complete medium containing 10% FBS. 500 μL of tumor cell suspension was added to each well of a 48-well plate and cultured overnight.

(2)CAR-T细胞加入:收集细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,取1×106个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率。取出48孔板,根据测得的CAR阳性率,按E:T=1:1计算每组CAR-T细胞所需细胞量,对各组补充T细胞至各组总T细胞保持一致,最后补加X-VIVO 15至500μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组加入与各组CAR-T等量的总T细胞,并补加X-VIVO 15至500μL。(2) Addition of CAR-T cells: Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1×10 6 cells/mL. Take 1×10 6 T cells and incubate with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. Use full spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect CAR positive rate. Take out the 48-well plate, calculate the required amount of CAR-T cells for each group according to the measured CAR positive rate, and calculate the required amount of CAR-T cells for each group according to E:T=1:1. Supplement T cells to each group until the total number of T cells in each group is consistent. Finally, add X-VIVO 15 to 500μL. After preparation, each group of CAR-T cells was added to each well. The untransfected T cell group was added with an equal amount of total T cells as that of each group of CAR-T, and X-VIVO 15 to 500 μL was added.

(3)每隔2~3天,在显微镜下观察细胞,当上一轮肿瘤细胞被全部杀完后,将孔中的CAR-T细胞收集并加至提前一天铺好的肿瘤细胞中进行重复刺激实验。当CAR-T细胞不能完全杀死肿瘤细胞时,使用胰酶(Hyclone)消化,收集所有细胞,使用APC标记的CD3抗体(Biolegend)和PE标记的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟,进行染色。之后通过流式细胞仪(NovoCyte 2060R,ACEA Biosciences,San Diego,CA,USA)统计在多轮靶细胞刺激后,肿瘤细胞的残余数量和CAR-T细胞的扩增数量。(3) Every 2 to 3 days, the cells were observed under a microscope. When all the tumor cells in the previous round were killed, the CAR-T cells in the wells were collected and added to the tumor cells that were plated one day in advance for repeated stimulation experiments. When the CAR-T cells could not completely kill the tumor cells, they were digested with trypsin (Hyclone), all the cells were collected, and incubated with APC-labeled CD3 antibody (Biolegend) and PE-labeled GCC protein (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes for staining. Afterwards, the number of residual tumor cells and the number of expanded CAR-T cells after multiple rounds of target cell stimulation were counted by flow cytometry (NovoCyte 2060R, ACEA Biosciences, San Diego, CA, USA).

重复杀伤结果如图5所示,经过10轮靶细胞重复刺激后,CAR-T-1、CAR-T-2和CAR-T-3杀伤能力较好,CAR-T扩增能力也较好,均显著优于CAR-T-4和CAR-T-5。进一步经过12轮靶细胞重复刺激,结果如图6所示,CAR-T-1保持最强的连续杀伤肿瘤的能力,且CAR-T细胞扩增数量最多,持久性最好,显著优于CAR-T-2和CAR-T-3。The results of repeated killing are shown in Figure 5. After 10 rounds of repeated target cell stimulation, CAR-T-1, CAR-T-2, and CAR-T-3 showed excellent killing and CAR-T expansion capabilities, all significantly superior to CAR-T-4 and CAR-T-5. After 12 rounds of repeated target cell stimulation, as shown in Figure 6, CAR-T-1 maintained the strongest ability to continuously kill tumors, with the largest number of expanded CAR-T cells and the best persistence, significantly superior to CAR-T-2 and CAR-T-3.

实施例4:具有不同铰链区和跨膜区的GCC CAR-T细胞在靶细胞刺激下的细胞因子分泌水平Example 4: Cytokine secretion levels of GCC CAR-T cells with different hinge and transmembrane regions upon target cell stimulation

4.1肿瘤细胞铺板:4.1 Tumor cell plating:

用胰酶(Hyclone)消化HCT116-GCC低表达肿瘤细胞,计数,然后将细胞密度用含10% FBS的IMDM完全培养基调为1×105个细胞/mL,在48孔板中每孔加入500μL肿瘤细胞悬液,过夜培养,使肿瘤细胞贴壁。HCT116-GCC low-expressing tumor cells were digested with trypsin (Hyclone) and counted. The cell density was then adjusted to 1×10 5 cells/mL using IMDM complete medium containing 10% FBS. 500 μL of tumor cell suspension was added to each well of a 48-well plate and cultured overnight to allow the tumor cells to adhere.

4.2 CAR-T细胞共孵育:4.2 CAR-T cell co-incubation:

收集细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,同时使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率;取出48孔板,根据测得的CAR阳性率,按E:T=1:1计算每组CAR-T细胞所需添加体积。对各组补充T细胞至各组总T细胞保持一致,最后补加X-VIVO 15至500μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组加入与各组CAR-T等量的总T细胞,并补加X-VIVO 15至500μL。24小时后,轻柔吹打孔中细胞(尽量不要吹起肿瘤细胞),然后孔板400g离心5min。取50μL上清,使用Cytometric Bead Array(CBA)kits(BD Biosciences)试剂盒检测各组中TNF、IFN-γ、IL-2三种细胞因子的水平。Cells were harvested, centrifuged at 400 g for 5 min, and resuspended in X-VIVO 15 medium. The cell density was adjusted to 1 × 10 6 cells/mL. CAR positivity was determined using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA). The 48-well plate was removed and the required volume of CAR-T cells for each group was calculated based on the measured CAR positivity, using an E:T ratio of 1:1. T cells were supplemented to maintain the same total T cell count for each group, and finally, X-VIVO 15 was added to 500 μL. After preparation, CAR-T cells from each group were added to each well. An equal number of total T cells was added to the untransfected T cell group, and X-VIVO 15 was added to 500 μL. After 24 hours, the cells were gently pipetted in the wells (try not to agitate tumor cells), and the plate was centrifuged at 400 g for 5 min. 50 μL of supernatant was collected and the levels of TNF, IFN-γ, and IL-2 in each group were detected using Cytometric Bead Array (CBA) kits (BD Biosciences).

结果如图7所示,相比于CAR-T-4和CAR-T-5,CAR-T-1、CAR-T-2、CAR-T-3三组显著分泌更多的TNF-α、IFN-γ和IL-2细胞因子,说明这三组CAR-T细胞均能有效识别GCC靶抗原,激活CAR-T细胞行使杀伤功能,而CAR-T-4和CAR-T-5不能有效识别GCC靶抗原。The results are shown in Figure 7. Compared with CAR-T-4 and CAR-T-5, the CAR-T-1, CAR-T-2, and CAR-T-3 groups secreted significantly more TNF-α, IFN-γ, and IL-2 cytokines, indicating that these three groups of CAR-T cells can effectively recognize GCC target antigens and activate CAR-T cells to perform killing functions, while CAR-T-4 and CAR-T-5 cannot effectively recognize GCC target antigens.

实施例5:具有不同铰链区和跨膜区的GCC CAR-T细胞在体外培养过程中的基底信号(tonic signaling)传导水平Example 5: Tonic signaling levels of GCC CAR-T cells with different hinge and transmembrane regions during in vitro culture

基底信号是衡量在没有外源抗原刺激的情况下,CAR-T细胞能够依赖自身CAR分子启动下游信号,促进细胞增殖和分化的能力的指标。适度的基底信号对于维持CAR-T的效应功能和存活至关重要。将各组Day 8的CAR-T在X-VIVO 15(无外源添加IL-2)中培养,24小时后检测细胞因子的水平,以及每2-3天进行细胞计数,综合分析CAR-T的基底信号强度。Basal signaling is a measure of the ability of CAR-T cells to initiate downstream signaling, promoting cell proliferation and differentiation, in the absence of exogenous antigen stimulation, relying on their own CAR molecules. Moderate basal signaling is crucial for maintaining CAR-T effector function and survival. Day 8 CAR-T cells in each group were cultured in X-VIVO 15 (without exogenous IL-2). Cytokine levels were measured 24 hours later, and cell counts were performed every 2-3 days to comprehensively analyze the basal signal strength of CAR-T cells.

结果如图8所示,CAR-T-1、CAR-T-2、CAR-T-3三组的细胞因子TNF-α、IFN-γ、IL-2分泌水平较高,其中CAR-T-1最高,具有最高的基底信号强度。静息态细胞增殖结果如图9所示,CAR-T-1、CAR-T-2、CAR-T-3三组均出现明显的细胞扩增,而CAR-T-4和CAR-T-5的细胞基本无扩增,后期细胞死亡。这说明CAR-T-1、CAR-T-2、CAR-T-3存在明显的基底信号,其中CAR-T-1基底信号最强,而CAR-T-4和CAR-T-5不存在基底信号。此结果预示着,当CAR-T回输到体内,CAR-T-1具有更快的肿瘤抗原反应能力和较强的存活能力,有利于提升治疗效果。The results, as shown in Figure 8, showed high secretion levels of TNF-α, IFN-γ, and IL-2 in the CAR-T-1, CAR-T-2, and CAR-T-3 groups, with CAR-T-1 producing the highest secretion and possessing the highest basal signal intensity. The results of resting cell proliferation, shown in Figure 9, showed significant cell proliferation in all three groups, while CAR-T-4 and CAR-T-5 showed little proliferation and later cell death. This indicates that CAR-T-1, CAR-T-2, and CAR-T-3 exhibit significant basal signals, with CAR-T-1 exhibiting the strongest basal signal, while CAR-T-4 and CAR-T-5 exhibit no basal signal. This suggests that when CAR-T cells are infused back into the body, CAR-T-1 exhibits a more rapid tumor antigen response and enhanced survival, potentially improving therapeutic efficacy.

实施例6:包含具有不同共刺激信号域的GCC CAR分子的CAR-T细胞增殖检测Example 6: CAR-T cell proliferation assay containing GCC CAR molecules with different co-stimulatory signaling domains

在体外杀伤功能最优的CAR-T-1的基础上,进行两种共刺激信号域(CD28和4-1BB)的CAR-T的功能比较。Based on the CAR-T-1 with the best killing function in vitro, the functions of CAR-T with two co-stimulatory signaling domains (CD28 and 4-1BB) were compared.

CAR-6的氨基酸序列如表1中所示。按照实施例1中的方法制备CAR-T-1和包含CAR-6的CAR-T-6细胞。在培养第5天及第11天,收集细胞,离心并计数。取2×105个细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测各组细胞CAR表达情况。The amino acid sequence of CAR-6 is shown in Table 1. CAR-T-1 and CAR-T-6 cells containing CAR-6 were prepared according to the method in Example 1. On the 5th and 11th days of culture, the cells were collected, centrifuged and counted. 2×10 5 cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein conjugated with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The expression of CAR in each group of cells was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA).

将CAR-T细胞用含有500IU/mL的X-VIVO 15培养基扩增培养11天,每隔3天传代计数。收集CAR-T细胞并离心,加1-2毫升的X-VIVO 15培养基重悬,取10微升的细胞培养液稀释一定倍数后,用台盼蓝按照体积比一比一染色,通过细胞计数仪记录细胞活率和活细胞浓度,计算各组的总细胞数,以统计分析各组细胞的增殖情况。CAR-T cells were expanded and cultured in X-VIVO 15 medium containing 500 IU/mL for 11 days, with passages counted every three days. CAR-T cells were harvested and centrifuged, then resuspended in 1-2 ml of X-VIVO 15 medium. Ten microliters of cell culture medium were diluted to a certain multiple and stained with trypan blue at a 1:1 volume ratio. Cell viability and viable cell concentration were recorded using a cell counter, and the total cell count in each group was calculated for statistical analysis of cell proliferation.

细胞增殖结果如图10所示,CAR-T-1和CAR-T-6两组细胞的T细胞增殖速率和CAR-T细胞增殖速率均比较接近,无明显差异。如图11所示,在第5天和第11天检测各组CAR-T的CAR表达情况,CAR-T-1的CAR阳性率略高于CAR-T-6,但CAR表达的平均荧光强度略低于CAR-T-6。The cell proliferation results are shown in Figure 10. The T cell proliferation rates and CAR-T cell proliferation rates of the CAR-T-1 and CAR-T-6 groups were similar, with no significant difference. As shown in Figure 11, CAR expression in each CAR-T cell group was detected on days 5 and 11. The CAR positivity rate of CAR-T-1 was slightly higher than that of CAR-T-6, but the mean fluorescence intensity of CAR expression was slightly lower than that of CAR-T-6.

实施例7:具有不同共刺激信号域的GCC CAR-T细胞的体外杀伤功能评价Example 7: Evaluation of the in vitro killing function of GCC CAR-T cells with different costimulatory signaling domains

7.1实时杀伤7.1 Real-time Killing

(1)肿瘤细胞铺板:在实时杀伤仪器(Agilent xCELLigence RTCA SP)配套使用的96孔板(E-plate 96)孔中加入50μL含10% FBS的IMDM完全培养基,放置于RTCA Station上,检测基线;用胰酶消化HCT116-GCC低表达肿瘤细胞,计数,然后用含10% FBS的IMDM完全培养基将细胞密度调为2×105个细胞/mL;取出E-plate 96,在孔中加入100μL(2×104个细胞/孔)肿瘤细胞悬液,室温放置30min;将E-plate 96放到培养箱中的RTCA Station上,24小时后检测细胞增殖曲线。(1) Tumor cell plating: Add 50 μL of IMDM complete medium containing 10% FBS to the wells of a 96-well plate (E-plate 96) used with a real-time killing instrument (Agilent xCELLigence RTCA SP), place it on the RTCA Station, and detect the baseline; digest the HCT116-GCC low-expressing tumor cells with trypsin, count them, and then adjust the cell density to 2×10 5 cells/mL with IMDM complete medium containing 10% FBS; remove the E-plate 96, add 100 μL (2×10 4 cells/well) of tumor cell suspension to the wells, and place them at room temperature for 30 minutes; place the E-plate 96 on the RTCA Station in the incubator, and detect the cell proliferation curve after 24 hours.

(2)CAR-T细胞加入:收集Day 11的CAR-T细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,取1×106个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率;取出E-plate 96孔板,每孔吸出50μL上清液。根据测得的CAR阳性率,按E:T=1:3、E:T=1:6分别计算每组CAR-T细胞所需添加体积。为使每组总T细胞保持一致,需要对各组补充T细胞,最后补加X-VIVO 15至100μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组的细胞数目与各组CAR-T的总T细胞保持一致,并补加X-VIVO 15至100μL。(2) Addition of CAR-T cells: CAR-T cells on Day 11 were collected, centrifuged at 400 g for 5 min, resuspended in X-VIVO 15 medium, and the cell density was adjusted to 1×10 6 cells/mL. 1×10 6 T cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein conjugated with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The CAR positive rate was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA). The E-plate 96-well plate was removed and 50 μL of supernatant was aspirated from each well. According to the measured CAR positive rate, the required volume of CAR-T cells for each group was calculated according to E:T = 1:3 and E:T = 1:6. To maintain a consistent total T cell count in each group, each group needs to be supplemented with T cells, and finally, 15 to 100 μL of X-VIVO is added. After preparation, each group of CAR-T cells is added to each well. The number of cells in the untransfected T cell group is consistent with the total T cell count in each CAR-T group, and 15 to 100 μL of X-VIVO is added.

(3)将E-Plate 96孔板放回实时杀伤仪器,监测CAR-T对肿瘤细胞的杀伤作用,纵轴“归一化的肿瘤细胞指数”代表杀伤效率,值越低,杀伤效率越高。(3) Place the E-Plate 96-well plate back into the real-time killing instrument to monitor the killing effect of CAR-T on tumor cells. The vertical axis "normalized tumor cell index" represents the killing efficiency. The lower the value, the higher the killing efficiency.

杀伤结果如图12所示,CAR-T-1的肿瘤细胞杀伤能力显著优于CAR-T-6,说明含有CD28共刺激信号域的CAR-T杀伤效率优于含有4-1BB共刺激信号域的CAR-T。The killing results are shown in Figure 12. The tumor cell killing ability of CAR-T-1 is significantly better than that of CAR-T-6, indicating that the killing efficiency of CAR-T containing the CD28 costimulatory signal domain is better than that of CAR-T containing the 4-1BB costimulatory signal domain.

7.2重复刺激杀伤7.2 Repeated Stimulation Killing

(1)肿瘤细胞铺板:用胰酶(Hyclone)消化HCT116-GCC低表达肿瘤细胞,计数,然后用含10% FBS的IMDM完全培养基将细胞重悬,并调整细胞密度为1×105个细胞/mL,在48孔板中每孔加入500μL肿瘤细胞悬液,过夜培养。(1) Tumor cell plating: HCT116-GCC low-expressing tumor cells were digested with trypsin (Hyclone), counted, and then resuspended in IMDM complete medium containing 10% FBS. The cell density was adjusted to 1×10 5 cells/mL. 500 μL of tumor cell suspension was added to each well of a 48-well plate and cultured overnight.

(2)CAR-T细胞共孵育:收集细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,取1×106个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率。取出48孔板,根据测得的CAR阳性率,按E:T=1:3计算每组CAR-T细胞所需添加体积。为使每组总T细胞保持一致,需要对各组补充T细胞,最后补加X-VIVO 15至500μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组加入与CAR-T组等量的总T细胞,并补加X-VIVO 15至500μL。(2) CAR-T cell co-incubation: Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1×10 6 cells/mL. Take 1×10 6 T cells and incubate with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. Then, use a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect the CAR positive rate. Take out the 48-well plate and calculate the required volume of CAR-T cells for each group according to the measured CAR positive rate at E:T=1:3. In order to keep the total number of T cells in each group consistent, it is necessary to supplement T cells to each group and finally add X-VIVO 15 to 500μL. After preparation, each group of CAR-T cells was added to each well. The untransfected T cell group was added with an equal amount of total T cells as the CAR-T group, and X-VIVO 15 to 500 μL was added.

(3)每隔2~3天,当上一轮肿瘤细胞被全部杀完后,将孔中的CAR-T细胞收集并加至提前一天铺好的肿瘤细胞中进行重复刺激实验。当CAR-T细胞不能完全杀死肿瘤细胞时,使用胰酶(Hyclone)消化,收集所有细胞,使用APC标记的CD3抗体(Biolegend)和PE标记的CAR抗体(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟,进行染色。之后通过流式细胞仪(NovoCyte 2060R,ACEA Biosciences,San Diego,CA,USA)统计在多轮靶细胞刺激后,每孔中肿瘤细胞的残余数量和CAR-T细胞的数量。(3) Every 2 to 3 days, when all tumor cells in the previous round were killed, the CAR-T cells in the wells were collected and added to the tumor cells that were plated one day in advance for repeated stimulation experiments. When CAR-T cells could not completely kill the tumor cells, they were digested with trypsin (Hyclone), all cells were collected, and APC-labeled CD3 antibody (Biolegend) and PE-labeled CAR antibody (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) were incubated at room temperature in the dark for 15 minutes for staining. Afterwards, the number of residual tumor cells and the number of CAR-T cells in each well after multiple rounds of target cell stimulation were counted by flow cytometry (NovoCyte 2060R, ACEA Biosciences, San Diego, CA, USA).

重复刺激杀伤的结果如图13所示,经过7轮靶细胞刺激后,CAR-T-1比CAR-T-6具有更强的连续杀伤肿瘤细胞的能力,且CAR-T细胞扩增数量更多,说明含有CD28共刺激信号域的CAR-T比含有4-1BB共刺激信号域的CAR-T的长期杀伤能力和持久性更好,即具有更优的抵抗肿瘤复发的能力。The results of repeated stimulation and killing are shown in Figure 13. After 7 rounds of target cell stimulation, CAR-T-1 has a stronger ability to continuously kill tumor cells than CAR-T-6, and the number of CAR-T cell proliferation is greater, indicating that CAR-T containing the CD28 co-stimulatory signal domain has better long-term killing ability and persistence than CAR-T containing the 4-1BB co-stimulatory signal domain, that is, it has a better ability to resist tumor recurrence.

实施例8:具有不同启动子的GCC CAR分子构建和CAR-T细胞制备Example 8: Construction of GCC CAR molecules with different promoters and preparation of CAR-T cells

在CAR-T-1分子的基础上,对慢病毒载体进行启动子的设计和筛选。Based on the CAR-T-1 molecule, the promoter of the lentiviral vector is designed and screened.

8.1慢病毒转移质粒的构建和慢病毒制备8.1 Construction of Lentiviral Transfer Plasmid and Lentivirus Preparation

(1)将两端分别带有限制性酶切位点XbaI和SalI的全长EF1α-CAR-T-1、截短EF1α-CAR-T-1、CMV-CAR-T-1、MND-CAR-T-1、PGK300-CAR-T-1和PGK400-CAR-T-1的GCC CAR的嵌合基因通过基因合成的方法合成(北京博迈德基因技术有限公司),启动子的具体序列如表2中所示。按照与实施例1相同的方法构建慢病毒转移质粒和制备慢病毒。(1) Chimeric genes of GCC CAR of full-length EF1α-CAR-T-1, truncated EF1α-CAR-T-1, CMV-CAR-T-1, MND-CAR-T-1, PGK300-CAR-T-1, and PGK400-CAR-T-1 with restriction enzyme sites XbaI and SalI at both ends were synthesized by gene synthesis (Beijing Bomade Gene Technology Co., Ltd.). The specific sequence of the promoter is shown in Table 2. Lentivirus transfer plasmids were constructed and lentivirus was prepared according to the same method as in Example 1.

8.2 CAR-T细胞制备及增殖检测8.2 CAR-T Cell Preparation and Proliferation Detection

按照与实施例1中相同的方法制备CAR-T细胞。将CAR-T细胞用完全培养基扩增培养12天,每隔2天传代计数。T细胞、CAR-T细胞的体外增殖情况如图14所示,随着培养时间的延长,全长EF1α-CAR-T-1的增殖速率最快,PGK300-CAR-T-1最慢。在培养第6天到第12天,检测各组CAR-T的CAR的阳性率,结果如图15所示,截短EF1α-CAR-T-1和MND-CAR-T-1的CAR阳性率最高。CAR-T cells were prepared in the same manner as in Example 1. CAR-T cells were expanded and cultured in complete medium for 12 days, and passaged and counted every 2 days. The in vitro proliferation of T cells and CAR-T cells is shown in Figure 14. As the culture time increases, the proliferation rate of full-length EF1α-CAR-T-1 is the fastest, and that of PGK300-CAR-T-1 is the slowest. From day 6 to day 12 of culture, the CAR positive rate of each group of CAR-T was detected. The results are shown in Figure 15. The CAR positive rate of truncated EF1α-CAR-T-1 and MND-CAR-T-1 is the highest.

实施例9:具有不同启动子的GCC CAR-T细胞的体外杀伤功能评价Example 9: Evaluation of the in vitro killing function of GCC CAR-T cells with different promoters

9.1实时杀伤9.1 Real-time Killing

(1)肿瘤细胞铺板:在实时杀伤仪器(Agilent xCELLigence RTCA SP)配套使用的96孔板(E-plate 96)孔中加入50μL含10% FBS的IMDM完全培养基,放置于RTCA Station上,检测基线;用胰酶消化HCT116-GCC低表达肿瘤细胞,计数,然后用含10% FBS的IMDM完全培养基将细胞密度调为2×105个细胞/mL;取出E-plate 96,在每孔中加入100μL(2×104个细胞/孔)肿瘤细胞悬液,室温放置30min;将E-plate 96放到培养箱中的RTCA Station上,实时监测细胞增殖曲线。(1) Tumor cell plating: Add 50 μL of IMDM complete medium containing 10% FBS to the wells of a 96-well plate (E-plate 96) used with a real-time killing instrument (Agilent xCELLigence RTCA SP), place it on the RTCA Station, and detect the baseline; digest the HCT116-GCC low-expressing tumor cells with trypsin, count them, and then adjust the cell density to 2× 105 cells/mL with IMDM complete medium containing 10% FBS; remove the E-plate 96, add 100 μL (2× 104 cells/well) of tumor cell suspension to each well, and place it at room temperature for 30 minutes; place the E-plate 96 on the RTCA Station in the incubator to monitor the cell proliferation curve in real time.

(2)CAR-T细胞加入:收集Day 8的CAR-T细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,取1×106个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率;取出E-plate 96孔板,每孔吸出50μL上清液。根据测得的CAR阳性率,按E:T=1:3计算每组CAR-T细胞所需添加体积。对各组补充T细胞至每组总T细胞保持一致,最后补加X-VIVO 15至100μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组的细胞数目与各CAR-T组的总T细胞保持一致,并补加X-VIVO 15至100μL。(2) Addition of CAR-T cells: CAR-T cells on Day 8 were collected, centrifuged at 400 g for 5 min, resuspended in X-VIVO 15 medium, and the cell density was adjusted to 1×10 6 cells/mL. 1×10 6 T cells were taken and incubated with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. The CAR positive rate was detected using a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA). The E-plate 96-well plate was removed and 50 μL of supernatant was aspirated from each well. According to the measured CAR positive rate, the required volume of CAR-T cells for each group was calculated according to E:T=1:3. T cells were supplemented to each group until the total number of T cells in each group was consistent, and finally X-VIVO 15 was added to 100 μL. After preparation, each group of CAR-T cells was added to each well. The number of cells in the untransfected T cell group was kept consistent with the total T cells in each CAR-T group, and X-VIVO 15 to 100 μL was added.

(3)将E-Plate 96孔板放回实时杀伤仪器,监测CAR-T对肿瘤细胞的杀伤作用,纵轴“归一化的肿瘤细胞指数”代表杀伤效率,值越低,杀伤效率越高。(3) Place the E-Plate 96-well plate back into the real-time killing instrument to monitor the killing effect of CAR-T on tumor cells. The vertical axis "normalized tumor cell index" represents the killing efficiency. The lower the value, the higher the killing efficiency.

实时杀伤结果如图16所示,在E:T=1:3下,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1和CMV-CAR-T-1比其他组CAR-T的杀伤速率更快,但三组之间没有显著性差异,说明截短EF1α、全长EF1α和CMV这三种启动子均能有效调控CAR-T-1基因表达及抗肿瘤功能,而PGK300、PGK400启动子均无法使CAR-T-1实现有效的抗肿瘤功能。The real-time killing results are shown in Figure 16. At E:T = 1:3, the killing rates of truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1 and CMV-CAR-T-1 were faster than those of other groups of CAR-T, but there was no significant difference among the three groups, indicating that the three promoters, truncated EF1α, full-length EF1α and CMV, can effectively regulate the gene expression and anti-tumor function of CAR-T-1, while the PGK300 and PGK400 promoters cannot enable CAR-T-1 to achieve effective anti-tumor function.

9.2重复刺激杀伤9.2 Repeated Stimulation Killing

(1)肿瘤细胞铺板:用胰酶(Hyclone)消化HCT116-GCC肿瘤细胞,计数,然后将细胞密度调为1×105个细胞/mL,在48孔板中每孔加入500μL肿瘤细胞悬液,过夜培养。(1) Tumor cell plating: HCT116-GCC tumor cells were digested with trypsin (Hyclone), counted, and then the cell density was adjusted to 1×10 5 cells/mL. 500 μL of tumor cell suspension was added to each well of a 48-well plate and cultured overnight.

(2)CAR-T细胞共孵育:收集细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,取1×106个T细胞,与APC标记的CD3抗体(Biolegend)和偶联PE荧光分子的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟后,使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率。取出48孔板,根据测得的CAR阳性率,按E:T=1:1计算每组CAR-T细胞所需添加体积。对各组补充T细胞至每组总T细胞保持一致,最后补加X-VIVO 15至500μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组加入与各组CAR-T等量的总T细胞,并补加X-VIVO 15至500μL。(2) CAR-T cell co-incubation: Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1×10 6 cells/mL. Take 1×10 6 T cells and incubate with APC-labeled CD3 antibody (Biolegend) and GCC protein coupled with PE fluorescent molecule (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) at room temperature in the dark for 15 minutes. Use full spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect CAR positivity. Take out the 48-well plate and calculate the required volume of CAR-T cells for each group according to the measured CAR positivity rate at E:T=1:1. Supplement T cells to each group until the total number of T cells in each group is consistent, and finally add X-VIVO 15 to 500μL. After preparation, each group of CAR-T cells was added to each well. The untransfected T cell group was added with an equal amount of total T cells as that of each group of CAR-T, and X-VIVO 15 to 500 μL was added.

(3)每隔2~3天,当上一轮肿瘤细胞被全部杀完后,将孔中的CAR-T细胞收集并加至提前一天铺好的肿瘤细胞中进行重复刺激实验。当CAR-T细胞不能完全杀死肿瘤细胞时,使用胰酶(Hyclone)消化,收集所有细胞,使用APC标记的CD3抗体(Biolegend)和PE标记的GCC蛋白(北京艺妙神州医药科技有限公司制备)室温避光孵育15分钟,进行染色。之后通过流式细胞仪(NovoCyte 2060R,ACEA Biosciences,San Diego,CA,USA)统计在多轮靶细胞刺激后,肿瘤细胞的残余数量和CAR-T细胞的扩增数量。(3) Every 2 to 3 days, when all tumor cells in the previous round were killed, the CAR-T cells in the wells were collected and added to the tumor cells that were plated one day in advance for repeated stimulation experiments. When CAR-T cells could not completely kill the tumor cells, they were digested with trypsin (Hyclone), all cells were collected, and APC-labeled CD3 antibody (Biolegend) and PE-labeled GCC protein (prepared by Beijing Yimiao Shenzhou Pharmaceutical Technology Co., Ltd.) were incubated at room temperature in the dark for 15 minutes for staining. Afterwards, the number of residual tumor cells and the number of expanded CAR-T cells after multiple rounds of target cell stimulation were counted by flow cytometry (NovoCyte 2060R, ACEA Biosciences, San Diego, CA, USA).

6轮靶细胞重复刺激后的结果如图17所示,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1、CMV-CAR-T-1和MND-CAR-T-1杀伤能力较好,CAR-T扩增数量较多,各组间无显著性差异,但均显著优于PGK300-CAR-T-1和PGK400-CAR-T-1。进一步经过10轮靶细胞重复刺激后,结果如图18所示,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1、CMV-CAR-T-1具有较好的肿瘤连续杀伤能力,而MND-CAR-T-1对肿瘤细胞的杀伤减弱,CAR-T扩增数量偏少。综合来看,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1和CMV-CAR-T-1的连续杀伤肿瘤细胞的能力较强,CAR-T扩增数量较多,持久性较好,进一步证明截短EF1α、全长EF1α和CMV这三种启动子均能有效调控CAR-T-1基因表达及抗肿瘤功能。The results after six rounds of repeated target cell stimulation are shown in Figure 17. Truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1, CMV-CAR-T-1, and MND-CAR-T-1 demonstrated superior cytotoxicity and a high number of expanded CAR-T cells. There were no significant differences between the groups, but all were significantly superior to PGK300-CAR-T-1 and PGK400-CAR-T-1. After a further 10 rounds of repeated target cell stimulation, the results are shown in Figure 18. Truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1, and CMV-CAR-T-1 demonstrated superior continuous tumor killing abilities, while MND-CAR-T-1 demonstrated reduced tumor cell killing and a low number of expanded CAR-T cells. Overall, the truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1 and CMV-CAR-T-1 have stronger abilities to continuously kill tumor cells, with larger numbers of CAR-T amplified and better persistence, further proving that the three promoters, namely truncated EF1α, full-length EF1α and CMV, can effectively regulate CAR-T-1 gene expression and anti-tumor function.

实施例10:具有不同启动子的GCC CAR-T细胞的细胞因子分泌水平Example 10: Cytokine secretion levels of GCC CAR-T cells with different promoters

(1)肿瘤细胞铺板:用胰酶消化HCT116-GCC低表达肿瘤细胞,计数,然后将细胞密度调为1×105个细胞/mL,在48孔板中每孔加入500μL肿瘤细胞悬液,过夜培养,使肿瘤细胞贴壁。(1) Tumor cell plating: HCT116-GCC low-expressing tumor cells were digested with trypsin and counted. The cell density was then adjusted to 1×10 5 cells/mL. 500 μL of tumor cell suspension was added to each well of a 48-well plate and cultured overnight to allow the tumor cells to adhere to the wall.

(2)CAR-T细胞共孵育:收集细胞,400g离心5min,用X-VIVO 15培养基重悬,并调整细胞密度为1×106个细胞/mL,同时使用全光谱流式细胞仪(Northern Lights,N7-00008-0A,Cytek Biosciences,Fremont,CA 94538,USA)检测CAR阳性率;取出48孔板,根据测得的CAR阳性率,按E:T=1:1计算每组CAR-T细胞所需添加体积。为使每组能进行平行比较,补加T细胞至各实验组的总T细胞保持一致,最后补加X-VIVO 15至500μL。配制完成后,将各组CAR-T细胞加至各孔中,未转染的T细胞组加入与各组CAR-T等量的总T细胞,并补加X-VIVO 15至500μL。(2) CAR-T cell co-incubation: Collect cells, centrifuge at 400g for 5 minutes, resuspend in X-VIVO 15 medium, and adjust the cell density to 1×10 6 cells/mL. At the same time, use a full-spectrum flow cytometer (Northern Lights, N7-00008-0A, Cytek Biosciences, Fremont, CA 94538, USA) to detect the CAR positive rate; take out the 48-well plate, and calculate the required volume of CAR-T cells for each group according to the measured CAR positive rate at E:T=1:1. In order to make each group able to be compared in parallel, T cells were added until the total T cells in each experimental group were consistent, and finally X-VIVO 15 was added to 500μL. After the preparation was completed, each group of CAR-T cells was added to each well, and the untransfected T cell group was added with an equal amount of total T cells as each group of CAR-T, and X-VIVO 15 was added to 500μL.

(3)24小时后,轻柔吹打孔中细胞(尽量不要吹起肿瘤细胞),然后孔板400g离心5min。取50μL上清,使用Cytometric Bead Array(CBA)kits(BD Biosciences)试剂盒检测各组中细胞因子TNF、IFN-γ、IL-2。(3) After 24 hours, gently pipette the cells in the wells (try not to blow up the tumor cells), and then centrifuge the wells at 400g for 5 minutes. Take 50μL of the supernatant and use Cytometric Bead Array (CBA) kits (BD Biosciences) to detect the cytokines TNF, IFN-γ, and IL-2 in each group.

检测结果如图19所示,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1和CMV-CAR-T-1均能分泌较多的细胞因子TNF-α、IFN-γ、IL-2,表明该三组CAR-T细胞能有效识别GCC靶抗原,进而活化CAR-T细胞。The test results are shown in Figure 19. The truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1 and CMV-CAR-T-1 can all secrete more cytokines TNF-α, IFN-γ, and IL-2, indicating that the three groups of CAR-T cells can effectively recognize GCC target antigens and activate CAR-T cells.

实施例11:具有不同启动子的GCC CAR-T细胞在体外培养过程中的基底信号(tonic signaling)传导水平Example 11: Basal signaling levels of GCC CAR-T cells with different promoters during in vitro culture

将各组CAR-T在X-VIVO 15(无外源添加IL-2)中培养,24小时后检测细胞因子和增殖率,之后每2-3天进行细胞计数和统计分析,以反映CAR-T的基底信号强度。结果如图20所示,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1和CMV-CAR-T-1三组的细胞因子TNF-α、IFN-γ、IL-2分泌水平较高,其中以CMV-CAR-T-1最高。体外细胞增殖结果如图21所示,截短EF1α-CAR-T-1、全长EF1α-CAR-T-1和CMV-CAR-T-1组出现明显的细胞扩增,而MND-CAR-T-1组的扩增水平较低,PGK300-CAR-T-1和PGK400-CAR-T-1组的细胞无扩增,后期细胞死亡。这说明截短EF1α-CAR-T-1、全长EF1α-CAR-T-1和CMV-CAR-T-1存在明显的基底信号,其中以CMV-CAR-T-1最强。Each group of CAR-T cells was cultured in X-VIVO 15 (without exogenous IL-2 supplementation). Cytokines and proliferation rates were measured after 24 hours. Cell counts and statistical analysis were performed every 2-3 days thereafter to reflect the basal signal strength of the CAR-T cells. The results are shown in Figure 20. The truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1, and CMV-CAR-T-1 groups secreted higher levels of cytokines, including TNF-α, IFN-γ, and IL-2, with CMV-CAR-T-1 producing the highest secretion level. In vitro cell proliferation results are shown in Figure 21. Significant cell proliferation was observed in the truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1, and CMV-CAR-T-1 groups, while the MND-CAR-T-1 group had a lower level of cell proliferation. Cells in the PGK300-CAR-T-1 and PGK400-CAR-T-1 groups showed no cell proliferation and subsequently underwent cell death. This indicates that there are obvious basal signals in truncated EF1α-CAR-T-1, full-length EF1α-CAR-T-1 and CMV-CAR-T-1, among which CMV-CAR-T-1 is the strongest.

实施例12:CAR-T-1细胞在结直肠癌PDX皮下小鼠模型中的抑瘤能力及体内扩增能力Example 12: Tumor Suppression and In Vivo Expansion Ability of CAR-T-1 Cells in a Subcutaneous Colorectal Cancer PDX Mouse Model

本实施例以检测包含上述scFv(启动子为截短EF1α)、CD8铰链区、CD8跨膜区、CD28共刺激信号域、CD3ζ胞内信号域的CAR-T(截短EF1α-CAR-T-1)对PDX荷瘤小鼠的抑瘤能力和体内扩增能力为例。This example takes the detection of the tumor inhibition ability and in vivo expansion ability of CAR-T (truncated EF1α-CAR-T-1) containing the above-mentioned scFv (promoter is truncated EF1α), CD8 hinge region, CD8 transmembrane region, CD28 co-stimulatory signal domain, and CD3ζ intracellular signal domain in PDX tumor-bearing mice as an example.

按照上述CAR-T细胞制备方法制备CAR-T-1细胞,培养至足够数量时,将细胞以冻存液重悬后保存至液氮中待用。取免疫组化检测GCC高表达的结直肠癌患者的肿瘤组织,切成2-3mm3碎片。接种于6-8周龄NSG小鼠,每只小鼠在下背部创建小切口,然后将肿瘤组织样本皮下移植到手术区域。7天后,对小鼠进行荧光素酶活体成像(Lumina II小动物活体成像系统,PerkinElmer,USA)分析,以验证小鼠异种移植模型是否构建成功。待肿瘤长到100mm3时,每组小鼠分别从尾静脉注射CAR-T-1细胞(5×106个CAR-T细胞/只),另外两组小鼠分别注射同体积杜氏磷酸盐缓冲液(DPBS)和相同T细胞数量的未转染慢病毒的对照T细胞。小鼠于CAR-T细胞注射前1天、以及注射后第3、7、12、15、18、22、26天统计肿瘤大小,于CAR-T细胞注射后第2、6、14、21和28天进行外周血CAR-T数量检测。CAR-T-1 cells were prepared according to the above-mentioned CAR-T cell preparation method. When sufficient cells were cultured, they were resuspended in cryopreservative solution and stored in liquid nitrogen until use. Tumor tissue from colorectal cancer patients with high GCC expression detected by immunohistochemistry was obtained and cut into 2-3 mm³ fragments. The cells were inoculated into 6-8 week-old NSG mice. A small incision was made in the lower back of each mouse, and the tumor tissue sample was then subcutaneously transplanted into the surgical area. After 7 days, the mice were analyzed by luciferase live imaging (Lumina II Small Animal Live Imaging System, PerkinElmer, USA) to verify the successful establishment of the mouse xenograft model. When the tumors grew to 100 mm³, each group of mice was injected with CAR-T-1 cells (5× 106 CAR-T cells/mouse) through the tail vein. The other two groups of mice were injected with the same volume of Dulbecco's phosphate buffered saline (DPBS) and the same number of T cells but without lentivirus transduction of control T cells. The tumor size of mice was measured 1 day before CAR-T cell injection and on days 3, 7, 12, 15, 18, 22, and 26 after injection. The number of CAR-T cells in peripheral blood was detected on days 2, 6, 14, 21, and 28 after CAR-T cell injection.

具体结果如图22A所示,与DPBS组和对照T细胞组的小鼠肿瘤负荷相比,CAR-T-1细胞回输3天后就减缓了肿瘤生长的速度,并在检测的26天内有效降低肿瘤负荷,具有显著的抗肿瘤活性。图22B显示了不同时间小鼠外周血中CAR-T-1细胞的数量,回输10天后就能检测到较高水平的CAR-T细胞,之后降低并维持在一定的水平。The specific results are shown in Figure 22A. Compared with the tumor burden of mice in the DPBS group and the control T cell group, CAR-T-1 cells slowed tumor growth 3 days after infusion and effectively reduced tumor burden within 26 days of detection, showing significant anti-tumor activity. Figure 22B shows the number of CAR-T-1 cells in the peripheral blood of mice at different times. A high level of CAR-T cells was detected 10 days after infusion, and then decreased and maintained at a certain level.

实施例13:患者中的CAR-T细胞扩增和抗肿瘤活性Example 13: CAR-T cell expansion and anti-tumor activity in patients

临床研究设计的目的在于评定将自体截短EF1-α-CAR-T-1细胞输注到患者中的安全性和疗效,肿瘤靶点为GCC。The clinical study was designed to evaluate the safety and efficacy of infusing autologous truncated EF1-α-CAR-T-1 cells into patients with GCC tumors.

从患者获得人外周血单核细胞(PBMC),根据细胞计数结果和T细胞比例,按照磁珠:T细胞=1.5:1(数量比)加入CD3/CD28Dynabeads(Thermo),放在样品分选仪上轻轻震荡30min。之后利用磁力架(Invitrogen)对磁珠的吸附作用,分选得到CD3阳性的T细胞。为使T细胞被充分激活,使用完全培养基(X-VIVO 15+500IU/mL IL-2)重悬,按照1.5×106个细胞/mL的密度进行扩增培养。磁珠激活24小时后,收集细胞,计数。根据计数结果,将慢病毒按MOI=2感染T细胞。病毒感染细胞24小时后,400g离心5min,更换新鲜的完全培养基继续培养。培养至第8天,收集细胞,使用磁力架去除Dynabeads,400g离心5min,洗涤CAR-T细胞,使用可回输冻存液进行冷冻保存。将细胞悬浮液装入冻存袋中,然后将袋子梯度冷却至-90℃并转移至用于存储的气相液氮罐中。患者回输前,将冷冻的CAR-T运输至医院,复苏后30分钟内完成患者回输。回输剂量为1.2×109个CAR-T细胞。Human peripheral blood mononuclear cells (PBMCs) were obtained from patients. Based on cell count results and the T cell ratio, CD3/CD28 Dynabeads (Thermo) were added at a ratio of 1.5:1 (bead:T cell ratio). The cells were placed on a sample sorter and gently shaken for 30 minutes. The beads were then adsorbed to the beads using a magnetic stand (Invitrogen) to isolate CD3-positive T cells. To fully activate the T cells, they were resuspended in complete culture medium (X-VIVO 15 + 500 IU/mL IL-2) and expanded at a density of 1.5 × 10⁶ cells/mL. After 24 hours of magnetic bead activation, the cells were harvested and counted. Based on the count results, T cells were infected with lentivirus at an MOI of 2. 24 hours after infection, the cells were centrifuged at 400 g for 5 minutes and replaced with fresh complete culture medium for continued culture. On day 8 of culture, cells were harvested, Dynabeads removed using a magnetic rack, and centrifuged at 400 g for 5 minutes. The CAR-T cells were washed and cryopreserved using transfusion-safe freezing medium. The cell suspension was placed in cryopreservation bags, which were then cooled to -90°C and transferred to a vapor phase liquid nitrogen tank for storage. Before patient transfusion, the frozen CAR-T cells were transported to the hospital and transfused within 30 minutes after resuscitation. The transfusion dose was 1.2 × 10 9 CAR-T cells.

患者回输CAR-T前均接受基于氟达拉滨和环磷酰胺的调理治疗,患者回输CAR-T后,密切观察患者至少2小时。定期收集患者外周血,通过流式细胞术监测CAR-T扩增情况,使用BDTMCytometric Bead Array(CBA)Human Th1/Th2/Th17CBA Kit试剂盒监测血浆中的细胞因子水平。所述方案由进行试验的医院批准。向所有患者提供书面知情同意书。All patients received fludarabine and cyclophosphamide-based conditioning therapy before CAR-T cell infusion. After CAR-T cell infusion, patients were closely observed for at least 2 hours. Peripheral blood was collected regularly to monitor CAR-T cell expansion by flow cytometry, and plasma cytokine levels were monitored using the BD Cytometric Bead Array (CBA) Human Th1/Th2/Th17 CBA Kit. The protocol was approved by the hospital conducting the trial. All patients provided written informed consent.

患者01被诊断为晚期转移性结直肠癌,伴有腹主动脉、右侧髂血管旁淋巴结转移、盆腔、腹膜、子宫前壁转移、双肺多发结节转移灶。输注截短EF1-α-CAR-T-1细胞28天后,CT扫描图像的如图23所示,肺部病灶1和2、腹膜转移灶显著减少,肿瘤直径变化具体见表3。CAR-T在体内扩增明显,最高达到3.2×107个CAR-T/L,细胞因子中IL-6有明显提升,最高达到2666.7pg/mL,结果示于图24中。癌胚抗原和癌抗原在治疗期间明显下降,结果示于图25中。该患者,在第一个月被评为PR(部分缓解),在第7个月被评为CR(完全缓解)。在治疗期间,患者01中未观察到严重的CRS(例如,不大于2级CRS)。Patient 01 was diagnosed with advanced metastatic colorectal cancer, with metastases to the abdominal aorta, right iliac lymph nodes, pelvic cavity, peritoneum, anterior uterine wall, and multiple nodular metastases in both lungs. 28 days after infusion of truncated EF1-α-CAR-T-1 cells, CT scan images were obtained, as shown in Figure 23 . Lung lesions 1 and 2, as well as peritoneal metastases, were significantly reduced. Changes in tumor diameter are detailed in Table 3 . CAR-T cells expanded significantly in vivo, reaching a maximum of 3.2×10 7 CAR-Ts/L. Among cytokines, IL-6 levels were significantly elevated, reaching a maximum of 2666.7 pg/mL (Figure 24 ). Carcinoembryonic antigen and cancer antigen levels decreased significantly during treatment (Figure 25 ). This patient was assessed as having a partial response (PR) at month 1 and a complete response (CR) at month 7. No severe CRS (e.g., no more than grade 2 CRS) was observed in Patient 01 during treatment.

表3患者01的肿瘤大小变化
Table 3 Changes in tumor size of patient 01

患者02被诊断为晚期转移性结直肠癌,伴有肠系膜淋巴结转移、腹膜后淋巴结转移、肺转移和肝转移转移灶。输注截短EF1-α-CAR-T-1细胞3个月后,CT扫描图像的如图26所示,肺部病灶和肝部转移灶显著减少,肿瘤总直径减少超过30%,肿瘤直径变化具体见表4。CAR-T在体内明显扩增,最高达到1.37×108个CAR-T/L,细胞因子中IL-6有明显提升,最高达到3500pg/mL,结果示于图27中。癌胚抗原和癌抗原在治疗期间明显下降,结果示于图28中。该患者在第3个月评为PR(部分缓解),并维持了超过8个月PR(部分缓解)。在治疗期间,患者02中未观察到严重的CRS(例如,不大于2级CRS)。Patient 02 was diagnosed with advanced metastatic colorectal cancer with mesenteric lymph node metastasis, retroperitoneal lymph node metastasis, lung metastasis and liver metastasis. Three months after the infusion of truncated EF1-α-CAR-T-1 cells, the CT scan image is shown in Figure 26. The lung lesions and liver metastases were significantly reduced, and the total tumor diameter was reduced by more than 30%. The changes in tumor diameter are shown in Table 4. CAR-T was significantly amplified in vivo, reaching a maximum of 1.37×10 8 CAR-T/L. The cytokine IL-6 was significantly increased, reaching a maximum of 3500pg/mL. The results are shown in Figure 27. Carcinoembryonic antigen and cancer antigen decreased significantly during treatment, and the results are shown in Figure 28. The patient was rated as PR (partial remission) at month 3 and maintained PR (partial remission) for more than 8 months. During treatment, no severe CRS (e.g., no more than grade 2 CRS) was observed in patient 02.

表4患者02的肿瘤大小变化
Table 4 Changes in tumor size of patient 02

实施例14:具有保护肽的GCC CAR分子构建和CAR-T细胞制备Example 14: Construction of GCC CAR molecules with protective peptides and preparation of CAR-T cells

在上述CAR-T-1分子的基础上,对慢病毒载体进行保护肽的设计。Based on the above-mentioned CAR-T-1 molecule, a protective peptide was designed for the lentiviral vector.

14.1慢病毒转移质粒的构建和慢病毒制备14.1 Construction of Lentiviral Transfer Plasmids and Lentivirus Preparation

将两端分别带有限制性酶切位点XbaI和SalI的编码N末端带有Strep tag II保护肽的CAR-T-7的嵌合基因通过基因合成的方法合成(北京博迈德基因技术有限公司),保护肽的具体氨基酸序列如表1中所示。按照与实施例1相同的方法构建慢病毒转移质粒和制备慢病毒。A chimeric gene encoding CAR-T-7 with a Strep tag II protective peptide at the N-terminus and restriction enzyme sites XbaI and SalI at both ends was synthesized by gene synthesis (Beijing Bomade Gene Technology Co., Ltd.). The specific amino acid sequence of the protective peptide is shown in Table 1. The lentiviral transfer plasmid was constructed and the lentivirus was prepared according to the same method as in Example 1.

14.2 CAR-T细胞制备和CAR表达率检测14.2 CAR-T Cell Preparation and CAR Expression Rate Detection

按照与实施例1中相同的方法制备CAR-T细胞。将CAR-T细胞用完全培养基扩增培养8天,每隔2天传代计数。培养到第8天,检测各组CAR-T的CAR的阳性率和平均荧光强度,结果如图29所示,CAR-T-7的CAR阳性率与CAR-T-1相比较低,但CAR平均荧光强度略高。CAR-T cells were prepared using the same method as in Example 1. CAR-T cells were expanded and cultured in complete medium for 8 days, with passages and counts every 2 days. On day 8 of culture, the CAR positivity and mean fluorescence intensity of each CAR-T group were measured. The results are shown in Figure 29. The CAR positivity rate of CAR-T-7 was lower than that of CAR-T-1, but the CAR mean fluorescence intensity was slightly higher.

实施例15:具有保护肽的GCC CAR-T细胞的体外杀伤功能评价Example 15: In vitro evaluation of the cytotoxicity of GCC CAR-T cells with protective peptides

15.1实时杀伤15.1 Real-time Kill

按照与实施例9中相同的实验步骤进行实时杀伤实验。实时杀伤结果如图30所示,在E:T=1:3下,72小时内,CAR-T-7的杀伤速率较快,说明CAR-T-7在短期内的抗肿瘤效果优于CAR-T-1。A real-time killing experiment was performed according to the same experimental procedures as in Example 9. The real-time killing results are shown in Figure 30. At E:T = 1:3, the killing rate of CAR-T-7 was faster within 72 hours, indicating that CAR-T-7 has a better anti-tumor effect than CAR-T-1 in the short term.

15.2重复刺激杀伤15.2 Repeated Stimulation Killing

按照与实施例9中相同的实验步骤进行重复刺激杀伤实验。结果如图31所示,经过2轮靶细胞重复刺激后,CAR-T-7的杀伤能力较好,显著优于CAR-T-1,但在CAR-T扩增数量上未见明显优势。综合来看,CAR-T-7的持久性较好,具有更强的抗肿瘤功能。Repeated stimulation and killing experiments were conducted using the same experimental procedures as in Example 9. The results, as shown in Figure 31, show that after two rounds of repeated target cell stimulation, CAR-T-7 demonstrated superior killing ability, significantly outperforming CAR-T-1, but showed no significant advantage in the number of CAR-T cell expansion. Overall, CAR-T-7 demonstrated superior persistence and stronger anti-tumor activity.

Claims (12)

靶向GCC的嵌合抗原受体,其包括:特异性识别GCC的scFv、CD8铰链区或CD28铰链区、CD8跨膜区或CD28跨膜区、CD28共刺激信号域或4-1BB共刺激信号域、CD3ζ信号域;A chimeric antigen receptor targeting GCC, comprising: a scFv that specifically recognizes GCC, a CD8 hinge region or a CD28 hinge region, a CD8 transmembrane region or a CD28 transmembrane region, a CD28 costimulatory signaling domain or a 4-1BB costimulatory signaling domain, and a CD3ζ signaling domain; 所述特异性识别GCC的scFv包含重链可变区VH和轻链可变区VL,所述VH包含SEQ ID NO:1所示氨基酸序列的HC CDR1、SEQ ID NO:2所示氨基酸序列的HC CDR2和SEQ ID NO:3所示氨基酸序列的HC CDR3,所述VL包含SEQ ID NO:4所示氨基酸序列的LC CDR1、SEQ ID NO:5所示氨基酸序列的LC CDR2和SEQ ID NO:6所示氨基酸序列的LC CDR3。The scFv that specifically recognizes GCC comprises a heavy chain variable region VH and a light chain variable region VL, wherein the VH comprises HC CDR1 with the amino acid sequence shown in SEQ ID NO: 1, HC CDR2 with the amino acid sequence shown in SEQ ID NO: 2, and HC CDR3 with the amino acid sequence shown in SEQ ID NO: 3, and the VL comprises LC CDR1 with the amino acid sequence shown in SEQ ID NO: 4, LC CDR2 with the amino acid sequence shown in SEQ ID NO: 5, and LC CDR3 with the amino acid sequence shown in SEQ ID NO: 6. 根据权利要求1所述的嵌合抗原受体,其中所述VH与SEQ ID NO:7的氨基酸序列至少85%相同,所述VL与SEQ ID NO:8的氨基酸序列至少85%相同;The chimeric antigen receptor of claim 1, wherein the VH is at least 85% identical to the amino acid sequence of SEQ ID NO: 7, and the VL is at least 85% identical to the amino acid sequence of SEQ ID NO: 8; 优选地,所述VH包含SEQ ID NO:7的氨基酸序列,VL包含SEQ ID NO:8的氨基酸序列。Preferably, the VH comprises the amino acid sequence of SEQ ID NO: 7, and the VL comprises the amino acid sequence of SEQ ID NO: 8. 根据权利要求1或2所述的嵌合抗原受体,其中所述特异性识别GCC的scFv与SEQ ID NO:9的氨基酸序列至少85%相同。A chimeric antigen receptor according to claim 1 or 2, wherein the scFv that specifically recognizes GCC is at least 85% identical to the amino acid sequence of SEQ ID NO: 9. 根据权利要求1-3中任意一项所述的嵌合抗原受体,其包括选自以下(1)-(4)中的任意一项:The chimeric antigen receptor according to any one of claims 1 to 3, comprising any one selected from the following (1) to (4): (1)特异性识别GCC的scFv、CD8铰链区、CD8跨膜区、CD28共刺激信号域、CD3ζ胞内信号域,(1) scFv that specifically recognizes GCC, CD8 hinge region, CD8 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain. (2)特异性识别GCC的scFv、CD28铰链区、CD28跨膜区、CD28共刺激信号域、CD3ζ胞内信号域,(2) scFv that specifically recognizes GCC, CD28 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain, (3)特异性识别GCC的scFv、CD8铰链区、CD28跨膜区、CD28共刺激信号域、CD3ζ胞内信号域,(3) scFv that specifically recognizes GCC, CD8 hinge region, CD28 transmembrane region, CD28 costimulatory signaling domain, and CD3ζ intracellular signaling domain, (4)特异性识别GCC的scFv、CD8铰链区、CD8跨膜区、4-1BB共刺激信号域、CD3ζ胞内信号域;(4) scFv that specifically recognizes GCC, CD8 hinge region, CD8 transmembrane region, 4-1BB costimulatory signaling domain, and CD3ζ intracellular signaling domain; 其中,in, CD8铰链区的氨基酸序列与SEQ ID NO:10所示的氨基酸序列至少85%相同、CD28铰链区的氨基酸序列与SEQ ID NO:11所示的氨基酸序列至少85%相同,The amino acid sequence of the CD8 hinge region is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 10, and the amino acid sequence of the CD28 hinge region is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 11. CD8跨膜区的氨基酸序列与SEQ ID NO:13所示的氨基酸序列至少85%相同、CD28跨膜区的氨基酸序列与SEQ ID NO:14所示的氨基酸序列至少85%相同,The amino acid sequence of the CD8 transmembrane region is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 13, and the amino acid sequence of the CD28 transmembrane region is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 14. CD28共刺激信号域的氨基酸序列与SEQ ID NO:15所示的氨基酸序列至少85%相同、4-1BB共刺激信号域的氨基酸序列与SEQ ID NO:16所示的氨基酸序列至少85%相同,The amino acid sequence of the CD28 costimulatory signaling domain is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 15, and the amino acid sequence of the 4-1BB costimulatory signaling domain is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 16. CD3ζ胞内信号域的氨基酸序列与SEQ ID NO:17所示的氨基酸序列至少85%相同。The amino acid sequence of the CD3ζ intracellular signaling domain is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 17. 根据权利要求1-4中任意一项所述的嵌合抗原受体,其进一步包括保护肽,所述保护肽优选具有SEQ ID NO:24所示的氨基酸序列。The chimeric antigen receptor according to any one of claims 1-4 further comprises a protective peptide, which preferably has an amino acid sequence shown in SEQ ID NO: 24. 根据权利要求1-5中任意一项所述的嵌合抗原受体,其与SEQ ID NO:18、SEQ ID NO:19、SEQ ID NO:20、SEQ ID NO:23或SEQ ID NO:25所示的氨基酸序列至少85%相同。The chimeric antigen receptor according to any one of claims 1-5, which is at least 85% identical to the amino acid sequence shown in SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 23 or SEQ ID NO: 25. 一种核酸分子,其包含编码根据权利要求1-6中任意一项所述的嵌合抗原受体的核酸序列。A nucleic acid molecule comprising a nucleic acid sequence encoding the chimeric antigen receptor according to any one of claims 1 to 6. 一种重组载体,其包含根据权利要求7所述的核酸分子,任选进一步包含截短EF1α启动子、全长EF1α启动子、CMV启动子;优选地,A recombinant vector comprising the nucleic acid molecule according to claim 7, optionally further comprising a truncated EF1α promoter, a full-length EF1α promoter, and a CMV promoter; preferably, 截短EF1α启动子的核苷酸序列如SEQ ID NO:29所示,The nucleotide sequence of the truncated EF1α promoter is shown in SEQ ID NO: 29. 全长EF1α启动子的核苷酸序列如SEQ ID NO:30所示,The nucleotide sequence of the full-length EF1α promoter is shown in SEQ ID NO: 30. CMV启动子的核苷酸序列如SEQ ID NO:31所示;The nucleotide sequence of the CMV promoter is shown in SEQ ID NO: 31; 更优选地,所述重组载体为重组慢病毒载体。More preferably, the recombinant vector is a recombinant lentiviral vector. 一种工程细胞,其包含根据权利要求1-6中任意一项所述的嵌合抗原受体、根据权利要求7所述的核酸分子、或根据权利要求8所述的重组载体。An engineered cell comprising the chimeric antigen receptor according to any one of claims 1 to 6, the nucleic acid molecule according to claim 7, or the recombinant vector according to claim 8. 根据权利要求1-6中任意一项所述的嵌合抗原受体、权利要求7所述的核酸分子、权利要求8所述的重组载体、权利要求9所述的工程细胞在制备治疗GCC阳性肿瘤的药物中的用途。Use of the chimeric antigen receptor according to any one of claims 1 to 6, the nucleic acid molecule according to claim 7, the recombinant vector according to claim 8, or the engineered cell according to claim 9 in the preparation of a medicament for treating GCC-positive tumors. 权利要求10所述的用途,其中所述药物用于治疗消化道肿瘤。The use according to claim 10, wherein the drug is used to treat digestive tract tumors. 权利要求11所述的用途,其中所述药物用于治疗结直肠癌、食管癌、胃癌或胰腺癌。The use according to claim 11, wherein the drug is used to treat colorectal cancer, esophageal cancer, gastric cancer or pancreatic cancer.
PCT/CN2025/087368 2024-04-08 2025-04-04 Chimeric antigen receptor targeting gcc and use thereof Pending WO2025214269A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202410417082.3A CN120818065A (en) 2024-04-08 2024-04-08 Chimeric antigen receptor targeting GCC and its use
CN202410417082.3 2024-04-08

Publications (1)

Publication Number Publication Date
WO2025214269A1 true WO2025214269A1 (en) 2025-10-16

Family

ID=97349435

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2025/087368 Pending WO2025214269A1 (en) 2024-04-08 2025-04-04 Chimeric antigen receptor targeting gcc and use thereof

Country Status (2)

Country Link
CN (1) CN120818065A (en)
WO (1) WO2025214269A1 (en)

Also Published As

Publication number Publication date
CN120818065A (en) 2025-10-21

Similar Documents

Publication Publication Date Title
WO2020108645A1 (en) Cd19-and bcma-based combined car-t immunotherapy
CN110248645A (en) Novel exosome-based anticancer agents
CN110317822B (en) TROP2 chimeric antigen receptor, T cell thereof, and preparation method and application thereof
CN113214408B (en) Chimeric antigen receptor macrophage and preparation method and application thereof
WO2023151346A1 (en) Construction and application of novel bispecific chimeric antigen receptor
TWI868739B (en) T cell receptor targeting KRAS G12V mutant polypeptide and its use
CN116836282B (en) Antibodies, chimeric antigen receptors and uses thereof
WO2021197483A1 (en) Fully human anti-human cd22 chimeric antigen receptor and application thereof
CN113416260B (en) Claudin18.2-targeted specific chimeric antigen receptor cell and preparation method and application thereof
CN113621077B (en) A TIM-3/CD28 fusion protein and CAR-T cells modified by the fusion protein
WO2025056004A1 (en) Immune cell expressing fusion protein, and expression vector thereof, use thereof and preparation method therefor
CN110055269B (en) Human mesothelin chimeric antigen receptor, T cell thereof, preparation method and application thereof
WO2025108099A1 (en) Chimeric antigen receptor against glypican-3 and use thereof
WO2022222846A1 (en) Chimeric antigen receptor targeting cd19, preparation method therefor and use thereof
WO2022127401A1 (en) Chimeric antigen receptor targeting cd123 and use thereof
CN111944053B (en) anti-BCMA CAR and expression vector and application thereof
WO2025157326A1 (en) Pharmaceutical composition and use thereof
CN110669138A (en) Double-chimeric antigen receptor, T cell, construction method and application thereof
CN117402261B (en) A method for preparing CAR-NK cells based on recombinant adenovirus and its application
WO2025214269A1 (en) Chimeric antigen receptor targeting gcc and use thereof
JP2019534322A (en) Methods for treating elevated TIM-3
CN118813547A (en) A double-gene modified CAR-T cell and its application
CN110251666A (en) Composition and application thereof for cancer targeting or immunotherapy
CN118599012B (en) A CART targeting PD-1 and VEGF and its use in preparing a drug for treating tumors
WO2025139979A1 (en) Chimeric antigen receptor targeting ly6g6d and use thereof

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: 25785743

Country of ref document: EP

Kind code of ref document: A1