CN112063640A - Chimeric antigen receptor targeting humanized CEA and uses thereof - Google Patents

Chimeric antigen receptor targeting humanized CEA and uses thereof Download PDF

Info

Publication number
CN112063640A
CN112063640A CN201910501250.6A CN201910501250A CN112063640A CN 112063640 A CN112063640 A CN 112063640A CN 201910501250 A CN201910501250 A CN 201910501250A CN 112063640 A CN112063640 A CN 112063640A
Authority
CN
China
Prior art keywords
nucleic acid
acid sequence
cancer
cea
sequence
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
CN201910501250.6A
Other languages
Chinese (zh)
Inventor
王恩秀
汪晨
张海
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.)
Nanjing Aide Institute Of Immunotherapy Co ltd
Original Assignee
Nanjing Aide Institute Of Immunotherapy 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 Nanjing Aide Institute Of Immunotherapy Co ltd filed Critical Nanjing Aide Institute Of Immunotherapy Co ltd
Priority to CN201910501250.6A priority Critical patent/CN112063640A/en
Publication of CN112063640A publication Critical patent/CN112063640A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001182Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • 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
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • Virology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Mycology (AREA)
  • Reproductive Health (AREA)
  • Pregnancy & Childbirth (AREA)

Abstract

The invention relates to a chimeric antigen receptor of targeting humanized CEA and application thereof. In particular, the invention provides a nucleic acid sequence selected from the group consisting of: contains the coding sequence of human DAP12, the coding sequence of T2A, the coding sequence of humanized CEA single-chain antibody, and the coding sequence of human TREM1 transmembrane and intracellular region which are connected in sequence. The invention also provides a related fusion protein, a vector containing the coding sequence, and applications of the fusion protein, the coding sequence and the vector.

Description

Chimeric antigen receptor targeting humanized CEA and uses thereof
Technical Field
The invention belongs to the field of chimeric antigen receptors, and particularly relates to a CEA-targeted chimeric antigen receptor and application thereof.
Background
CEA was first discovered from fetal colon cancer tissues and is an embryonic carcinogenic antigen, and is therefore termed "carcinoembryonic antigen". The CEA coding gene is located on chromosome 19, and the expression product is glycoprotein with relative molecular mass of 150 kDa-300 kDa. Research shows that the CEA is not single in component, but is a protein complex rich in polysaccharide, and the protein content of the protein is 45%, and the protein complex contains fucose, mannose, galactose, sialic acid and the like. The early digestive tract and some tissues of the fetus have the capability of synthesizing CEA, but the CEA content is gradually reduced after six months of pregnancy, the postnatal content is extremely low, but the content can be found to be abnormally increased in the serum of some malignant tumor patients. Because CEA produced by cell secretion enters local body fluid and blood, the phenomenon of abnormal increase of CEA content can occur in blood serum, chest, ascites and digestive juice of tumor patients with rectal cancer, colon cancer, liver cancer, kidney cancer, breast cancer, esophageal cancer and the like, and the CEA content of the chest water of the lung cancer patients is often higher than that of the blood serum. Mild increases in CEA are also seen in certain benign digestive tract diseases such as intestinal obstruction, biliary obstruction, pancreatitis, cirrhosis, colonic polyps, ulcerative colitis, as well as in smokers and the elderly. CEA is a common tumor-associated antigen and internationally recognized tumor marker that is expressed in most human tumors of germ layer origin. Carcinoembryonic antigen (CEA) mainly exists in tissues of straight and colon cancers, and also exists in tumor tissues of liver cancer, pancreatic cancer, kidney cancer, breast cancer, esophageal cancer, ovarian cancer and the like, and a small amount of CEA also exists in normal colon tissues. CEA indexes are obviously increased in intestinal mucosa of fetus, colon cancer, gastric cancer, lung cancer, bile duct cancer and the like.
With the development of tumor immunology theory and clinical technology, Chimeric antigen receptor T-cell therapy (CAR-T) is one of the most promising tumor immunotherapy at present [ Schmitz M, et al. 10.1155/2010/956304.]. Chimeric Antigen Receptors (CARs) are a core component of CAR-T, CARs can redirect their specificity and reactivity towards selected immune cells, thus conferring on T cells the ability to recognize tumor antigens in an HLA-independent manner, which makes CAR-engineered T cells capable of activating and proliferating, and thus capable of efficiently killing tumor cells, unlike normal T Cell Receptor (TCR) responses, independent of MHC restriction.
Chimeric Antigen Receptors (CARs) express synthetic proteins on T cells that fuse antigen recognition fragments of antibodies (e.g., antibody single chain variable fragments) to intracellular signaling domains. The research finds that the single-chain variable region (scFv) of the tumor-specific monoclonal antibody is used for replacing the alpha and beta variable regions of the TCR, the scFv is directly connected with the T cell signaling domain to form a Chimeric Antigen Receptor (CAR) which is expressed on the surface of a T cell, the tumor-specific antigen can be recognized by the scFv, the activation signal of the T cell is directly generated, the activation and proliferation of the T cell are promoted, and the tumor cell is specifically killed. The process mainly depends on the specific recognition of scFv on the surface of the CAR-T cell to tumor antigen, and the specificity and the killing property of immune response are stronger.
The invention adopts humanized CEA (huM5A) single-chain antibody, huM5A has high affinity to the constructed novel CAR targeting CEA. huM5A is a humanized form of a monoclonal antibody targeting CEA and has therapeutic and diagnostic effects on tumors. The invention modifies CAR-T cells and plays a CAR-T cell anti-tumor role aiming at CEA targets. The invention lays a good foundation for clinical experiments and clinical treatment.
With the accumulation and continued sophistication of the experience of CAR-T cell therapy, there is increasing interest in its use in malignancies. Under the environment, the pace is to be accelerated, and the CAR-T cell therapy is promoted to rapidly advance on the way of treating malignant tumors by applying and developing clinical tests by utilizing the existing work foundation, research and development teams and medical teams.
Disclosure of Invention
In a first aspect, the present invention provides a nucleic acid sequence selected from the group consisting of:
(1) contains the coding sequence of human natural killer activated receptor related protein (DAP12), the coding sequence of T2A, the coding sequence of anti-CEA single-chain antibody, and the coding sequence of transmembrane and intracellular regions of human myeloid triggering receptor (TREM1) which are connected in sequence.
In one or more embodiments, the coding sequence of the signal peptide preceding the coding sequence of the anti-CEA single chain antibody is as shown in nucleotide sequence 412-474 of SEQ ID NO: 1. In one or more embodiments, the coding sequence of the anti-CEA single-chain antibody is shown in the 481-1221 th nucleotide sequence of SEQ ID NO. 1. In one or more embodiments, the nucleic acid sequence of human DAP12 is set forth in nucleic acids SEQ ID NO. 1, positions 1-339. In one or more embodiments, the nucleic acid sequence of T2A is as shown in SEQ ID NO 1 at position 355-405. In one or more embodiments, the nucleic acid sequence of the transmembrane and intracellular region of human TREM1 is as set forth in SEQ ID NO 1 at nucleic acid positions 1258-1395. In a second aspect, the invention provides a fusion protein selected from the group consisting of:
(2) the fusion protein comprises human DAP12, T2A, anti-CEA single-chain antibody, human TREM1 transmembrane and intracellular region which are connected in sequence, and fusion protein which is formed by substituting, deleting or adding one or more amino acids in a limited amino acid sequence and retains the activity of activated T cells;
preferably, the anti-CEA monoclonal antibody huM 5A.
In one or more embodiments, the nucleic acid sequence further comprises a coding sequence for a signal peptide prior to the coding sequence for the anti-CEA single chain antibody. In one or more embodiments, the amino acid sequence of the signal peptide is as shown in SEQ ID NO:2 amino acids 138-158. In one or more embodiments, the amino acid sequence of the anti-CEA single chain antibody is shown as amino acids 161-407 of SEQ ID NO 2. In one or more embodiments, the amino acid sequence of human DAP12 is set forth in SEQ ID NO. 2 amino acids 1-113. In one or more embodiments, the amino acid sequence of T2A is depicted as amino acids 119-135 of SEQ ID NO 2. In one or more embodiments, the amino acid sequence of the transmembrane and intracellular region of human TREM1 is depicted as amino acids 420-464 of SEQ ID NO 2.
In a third aspect, the invention provides a nucleic acid vector comprising a nucleic acid sequence as described herein.
In one or more embodiments, the nucleic acid construct is a lentiviral vector comprising a nucleic acid sequence as described herein, and optionally a selectable marker.
In a fourth aspect, the invention provides a lentivirus comprising a nucleic acid vector as described herein, preferably comprising said lentivirus vector.
In a fifth aspect, the invention provides a genetically modified T cell comprising a nucleic acid sequence as described herein, or comprising a nucleic acid vector as described herein, or infected with a lentivirus as described herein, or stably expressing a fusion protein as described herein.
In a sixth aspect, the invention provides a pharmaceutical composition comprising a genetically modified T cell as described herein.
In a seventh aspect, the invention provides the use of a nucleic acid sequence, fusion protein, nucleic acid vector or lentivirus as described herein in the preparation of an activated T cell.
In an eighth aspect, the invention provides the use of a nucleic acid sequence, fusion protein, nucleic acid vector, lentivirus, or genetically modified T cell, or a pharmaceutical composition thereof, as described herein, in the preparation of a medicament for the treatment of a CEA-mediated disease.
In one or more embodiments, the CEA-mediated disease is brain cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, liver cancer, kidney cancer, lymphoma, leukemia, lung cancer, melanoma, metastatic melanoma, mesothelioma, neuroblastoma, ovarian cancer, prostate cancer, pancreatic cancer, renal cancer, skin cancer, thymoma, sarcoma, non-hodgkin's lymphoma, uterine cancer. In one or more embodiments, the CEA-mediated disease is colorectal cancer, liver cancer, breast cancer, melanoma, pancreatic cancer.
Drawings
FIG. 1: schematic representation of pELNS-CEA CAR lentiviral expression vector.
FIG. 2: structural schematic representation of CEA-CAR
Detailed Description
The present invention provides a Chimeric Antigen Receptor (CAR) targeting humanized CEA. The CAR contains sequentially linked human DAP12, T2A, anti-CEA single chain antibody, human TREM1 transmembrane and intracellular domains.
The anti-CEA single chain antibody suitable for use in the present invention may be derived from a variety of anti-CEA monoclonal antibodies known in the art.
Thus, in certain embodiments, an anti-CEA chain antibody suitable for use in the present invention contains a peptide that specifically recognizes human CEA. Optionally, the light chain variable region and the heavy chain variable region may be linked together by a linker sequence. Such single chain antibodies that may be exemplified include, but are not limited to, 10E1, M5A. In certain embodiments, the monoclonal antibody is huM 5A.
The fusion proteins of the present invention, such as human DAP12, T2A, anti-CEA single chain antibody, human TREM1 transmembrane and intracellular domains, etc., can be directly linked to each other or can be linked through a linker sequence. The linker sequence may be one known in the art to be suitable for use with antibodies, for example, a G and S containing linker sequence. Typically, the linker contains one or more motifs which repeat back and forth. For example, the motif may be GGGS, GGGGS, SSSSG, GSGSA and GGSGG. Preferably, the motifs are adjacent in the linker sequence with no intervening amino acid residues between the repeats. The linker sequence may comprise 1, 2, 3, 4 or 5 repeat motifs. The linker may be 3 to 25 amino acid residues in length, for example 3 to 15, 5 to 15, 10 to 20 amino acid residues. In certain embodiments, the linker sequence is a polyglycine linker sequence. The number of glycines in the linker sequence is not particularly limited, and is usually 2 to 20, such as 2 to 15, 2 to 10, 2 to 8. In addition to glycine and serine, other known amino acid residues may be contained in the linker, such as alanine (a), leucine (L), threonine (T), glutamic acid (E), phenylalanine (F), arginine (R), glutamine (Q), and the like.
It will be appreciated that in gene cloning procedures it is often necessary to design appropriate cleavage sites which will introduce one or more irrelevant residues at the end of the expressed amino acid sequence without affecting the activity of the sequence of interest. In order to construct a fusion protein, facilitate expression of a recombinant protein, obtain a recombinant protein that is automatically secreted outside of a host cell, or facilitate purification of a recombinant protein, it is often necessary to add some amino acids to the N-terminus, C-terminus, or other suitable regions within the recombinant protein, for example, including, but not limited to, suitable linker peptides, signal peptides, leader peptides, terminal extensions, and the like. Thus, the amino-terminus or the carboxy-terminus of the fusion protein of the invention (i.e., the CAR) may also contain one or more polypeptide fragments as protein tags. Any suitable label may be used herein. For example, the tag may be FLAG, HA, HA1, c-Myc, Poly-His, Poly-Arg, Strep-TagII, AU1, EE, T7, 4A6, B, gE and Ty 1. These tags can be used to purify proteins.
The invention also includes a CAR represented by the amino acid sequence at positions 1-464 or a mutant of the CAR represented by SEQ ID NO. 2. These mutants include: an amino acid sequence that has at least 80%, preferably at least 85%, preferably at least 90%, preferably at least 95%, preferably at least 97% sequence identity to the CAR and retains the biological activity (e.g., activating T cells) of the CAR. Sequence identity between two aligned sequences can be calculated using, for example, BLASTp from NCBI.
Mutants also include: an amino acid sequence having one or several mutations (insertions, deletions or substitutions) in the amino acid sequence as depicted in positions 1-464 of SEQ ID NO:2 or in the amino acid sequence as depicted in SEQ ID NO:2, while still retaining the biological activity of the CAR. The number of mutations usually means within 1-10, such as 1-8, 1-5 or 1-3. The substitution is preferably a conservative substitution. For example, conservative substitutions with amino acids of similar or similar properties are not typically used in the art to alter the function of a protein or polypeptide. "amino acids with similar or analogous properties" include, for example, families of amino acid residues with analogous side chains, including amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine tryptophan, histidine). Thus, substitution of one or more sites with another amino acid residue from the same side chain species in the polypeptide of the invention will not substantially affect its activity.
The invention includes nucleic acid sequences encoding the fusion proteins of the invention. The nucleic acid sequences of the invention may be in the form of DNA or in the form of RNA. The form of DNA includes cDNA, genomic DNA or artificially synthesized DNA. The DNA may be single-stranded or double-stranded. The DNA may be the coding strand or the non-coding strand. The invention also includes degenerate variants of the nucleic acid sequences encoding the fusion proteins, i.e., nucleotide sequences which encode the same amino acid sequence but differ in nucleotide sequence.
The nucleic acid sequences described herein can generally be obtained by PCR amplification. Specifically, primers can be designed based on the nucleic acid sequences disclosed herein, particularly open reading frame sequences, and the relevant sequences can be amplified using commercially available cDNA libraries or cDNA libraries prepared by conventional methods known to those skilled in the art as templates. When the sequence is long, two or more PCR amplifications are often required, and then the amplified fragments are spliced together in the correct order. For example, in certain embodiments, the nucleic acid sequence encoding the fusion protein described herein is as set forth in nucleic acid SEQ ID NO. 1, nucleic acids 1-1395.
The invention also relates to nucleic acid vectors comprising the nucleic acid sequences described herein, and one or more regulatory sequences operatively linked to these sequences. The nucleic acid sequences of the invention can be manipulated in a variety of ways to ensure expression of the fusion protein (CAR). The nucleic acid vector may be manipulated prior to its insertion into the vector depending on the identity or requirements of the expression vector. Techniques for altering nucleic acid sequences using recombinant DNA methods are known in the art.
The control sequence may be an appropriate promoter sequence. The promoter sequence is typically operably linked to the coding sequence of the protein to be expressed. The promoter may be any nucleotide sequence which shows transcriptional activity in the host cell of choice including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell. The control sequence may also be a suitable transcription terminator sequence, a sequence recognized by a host cell to terminate transcription. The terminator sequence is operably linked to the 3' terminus of the nucleotide sequence encoding the polypeptide. Any terminator which is functional in the host cell of choice may be used in the present invention. The control sequence may also be a suitable leader sequence, a nontranslated region of an mRNA which is important for translation by the host cell. The leader sequence is operably linked to the 5' terminus of the nucleotide sequence encoding the polypeptide. Any terminator which is functional in the host cell of choice may be used in the present invention.
The nucleic acid sequences of the present invention can be cloned into many types of vectors. For example, it can be cloned into plasmids, phagemids, phage derivatives, animal viruses and cosmids. Further, the vector is an expression vector. The expression vector may be provided to the cell in the form of a viral vector. Viral vector technology is well known in the art and is described, for example, in Sambrook et al (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York) and other virology and Molecular biology manuals. Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses.
Generally, suitable vectors comprise an origin of replication, a promoter sequence, a convenient restriction enzyme site, and one or more selectable markers that function in at least one organism (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).
For example, in certain embodiments, the invention employs a lentiviral vector comprising a nucleic acid sequence as described herein, and optionally a selectable marker.
An example of a suitable promoter is the elongation growth factor-1 alpha (EF-1 alpha) promoter sequence. Another example of a suitable promoter is immediate early Cytomegalovirus (CMV). However, other constitutive promoter sequences may also be used, including, but not limited to, the simian virus 40(SV40) early promoter, the mouse mammary cancer virus (MMTV), the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia virus promoter, the EB virus immediate early promoter, the rous sarcoma virus promoter, and human gene promoters such as, but not limited to, the actin promoter, myosin promoter, heme promoter, and creatine kinase promoter. Further, inducible promoters are also contemplated. The use of an inducible promoter provides a molecular switch that is capable of turning on expression of a nucleic acid sequence operably linked to the inducible promoter during periods of expression and turning off expression when expression is undesirable. Examples of inducible promoters include, but are not limited to, the metallothionein promoter, the glucocorticoid promoter, the progesterone promoter, and the tetracycline promoter.
To assess the expression of the CAR polypeptide or portion thereof, the expression vector introduced into the cells can also comprise either or both of a selectable marker gene or a reporter gene to facilitate identification and selection of expressing cells from a population of cells sought to be transfected or infected by the viral vector. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both the selectable marker and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in a host cell. Useful selectable markers include, for example, antibiotic resistance genes, such as neo and the like.
Reporter genes are used to identify potentially transfected cells and to evaluate the functionality of regulatory sequences. After the DNA has been introduced into the recipient cell, the expression of the reporter gene is assayed at an appropriate time. Suitable reporter genes may include genes encoding luciferase, β -galactosidase, chloramphenicol acetyltransferase, secreted alkaline phosphatase, or green fluorescent protein. Suitable expression systems are well known and can be prepared using known techniques or obtained commercially.
Methods for introducing and expressing genes into cells are known in the art. The vector may be readily introduced into a host cell by any method known in the art, for example, mammalian, bacterial, yeast or insect cells. For example, the expression vector may be transferred into a host cell by physical, chemical or biological means.
Physical methods for introducing nucleic acids into host cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Biological methods for introducing a nucleic acid of interest into a host cell include the use of DNA and RNA vectors. Chemical means of introducing nucleic acids into host cells include colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
Biological methods for introducing nucleic acids into host cells include the use of viral vectors, particularly lentiviral vectors, which have become the most widely used method for inserting genes into mammalian, e.g., human, cells. Other viral vectors may be derived from retroviruses, poxviruses, herpes simplex virus I, adenoviruses, adeno-associated viruses, and the like. Many virus-based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. The selected gene can be inserted into a vector and packaged into a retroviral particle using techniques known in the art. The recombinant virus can then be isolated and delivered to the subject cells in vivo or ex vivo. Many retroviral systems are known in the art. In some embodiments, an adenoviral vector is used. Many adenoviral vectors are known in the art.
Thus, in certain embodiments, the invention also provides a lentivirus for activating T cells, the virus comprising a lentiviral vector as described herein and a corresponding packaging gene, such as gag, pol, and vsvg.
Thus, in certain embodiments, the invention provides a genetically modified T cell comprising a nucleic acid sequence as described herein, or comprising a lentiviral vector as described herein, or infected with a lentivirus as described herein, or prepared by a method as described herein, or stably expressing a fusion protein as described herein.
The CAR-T cells of the invention can undergo robust in vivo T cell expansion and sustained at high levels in the blood and bone marrow for extended amounts of time, and form specific memory T cells. Without wishing to be bound by any particular theory, the CAR-T cells of the invention can differentiate into a central memory-like state in vivo upon encountering and subsequently depleting target cells expressing a surrogate antigen.
The invention also includes a class of cell therapies in which T cells are genetically modified to express a CAR described herein, and the CAR-T cells are injected into a recipient in need thereof. The injected cells are capable of killing tumor cells of the recipient. Unlike antibody therapy, CAR-T cells are able to replicate in vivo, resulting in long-term persistence that can lead to sustained tumor control.
The anti-tumor immune response elicited by the CAR-T cells can be an active or passive immune response. Additionally, the CAR-mediated immune response can be part of an adoptive immunotherapy step, in which the CAR-T cells induce an immune response specific for the antigen-binding portion in the CAR.
Thus, the diseases that can be treated with the CARs, their coding sequences, nucleic acid vectors, expression vectors, viruses, and CAR-T cells of the invention are preferably CEA-mediated diseases.
In particular, herein, "CEA-mediated disease" includes various types of brain cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, liver cancer, kidney cancer, lymphoma, leukemia, lung cancer, melanoma, metastatic melanoma, mesothelioma, neuroblastoma, ovarian cancer, prostate cancer, pancreatic cancer, renal cancer, skin cancer, thymoma, sarcoma, non-hodgkin's lymphoma, uterine cancer, among others. In certain embodiments, the CEA-mediated disease is colorectal cancer, liver cancer, breast cancer, melanoma, pancreatic cancer.
The CAR-modified T cells of the invention can be administered alone or as a pharmaceutical composition in combination with diluents and/or with other components such as relevant cytokines or cell populations. Briefly, a pharmaceutical composition of the invention may comprise CAR-T cells as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients. Such compositions may include buffers such as neutral buffered saline, sulfate buffered saline, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; a protein; polypeptides or amino acids such as glycine; an antioxidant; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative.
The pharmaceutical compositions of the present invention may be administered in a manner suitable for the disease to be treated (or prevented). The amount and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease.
When referring to an "immunologically effective amount", "an anti-tumor effective amount", "a tumor-inhibiting effective amount", or a "therapeutic amount", the precise amount of the composition of the invention to be administered can be determined by a physician, taking into account the age, weight, tumor size, extent of infection or metastasis, and individual differences in the condition of the patient (subject). It can be generally pointed out that: pharmaceutical compositions comprising T cells described herein can be in the range of 104To 109Dosage of individual cells/kg body weight, preferably 105To 106Dosage of individual cells/kg body weight. The T cell composition may also be administered multiple times at these doses. Cells can be administered by using infusion techniques well known in immunotherapy (see, e.g., Rosenberg et al, New Eng.J.of Med.319:1676, 1988). Optimal dosages and treatment regimens for a particular patient can be readily determined by those skilled in the medical arts by monitoring the patient for signs of disease and adjusting the treatment accordingly.
Administration of the subject composition may be carried out in any convenient manner, including by spraying, injection, swallowing, infusion, implantation or transplantation. The compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodal, intraspinally, intramuscularly, by intravenous injection, or intraperitoneally. In one embodiment, the T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection. In another embodiment, the T cell composition of the invention is preferably administered by intravenous injection. The composition of T cells can be injected directly into the tumor, lymph node or site of infection.
In some embodiments of the invention, the CAR-T cells of the invention or compositions thereof can be combined with other therapies known in the art. Such therapies include, but are not limited to, chemotherapy, radiation therapy, and immunosuppressive agents. For example, the treatment may be in conjunction with radiotherapeutic or chemotherapeutic agents known in the art for the treatment of CEA-mediated diseases.
The present invention is described in further detail by referring to the following experimental examples. These examples are provided for illustrative purposes only and are not intended to be limiting unless otherwise specified. Accordingly, the present invention should in no way be construed as limited to the following examples, but rather should be construed to include any and all variations which become apparent in light of the teachings provided herein. The methods and reagents used in the examples are, unless otherwise indicated, conventional in the art.
Example 1 chimeric antigen receptor preparation
The invention provides a targeting humanized CEA chimeric antigen receptor. The design sequentially comprises DAP12, T2A, a humanized CEA single-chain variable region and TREM1, and the structure of the design is shown in figure 1.
pELNS-DAP12-T2A plasmid was stored by Nanjing Katsui medicine science, Inc., or was constructed according to the method disclosed in the literature (Enxiu Wang et al, Generation of patent T-cell Immunology for Cancer Using DAP12-Based, Multichain, Chimeric Immunoreppers.2015, Cancer Immunology Research,3(7):815), CEA scFv-TREM1 gene synthesis was synthesized and provided by Biotech corporation, Yoshigawa, Inc., plasmid pELNS-DAP12-T2A and the synthesized gene fragments were separately digested by BamHI and I double (purchased from Saltakara), the digestion was performed as described, and they were ligated after digestion to obtain a lentiviral vector expressing Chimeric antigen receptor:
5 mu.L of lentiviral vector was transformed into E.coli TOP10 competent cells (purchased from Nanjing Ande high Biotech Co., Ltd.), and after culturing at 37 ℃ for 16h, monoclonals were picked up, and after culturing at 37 ℃ for 12h, plasmids were extracted with a plasmid extraction kit (purchased from Takara Co., Ltd.), and the specific method is described in the specification.
Example 2 Lentiviral packaging
The slow virus is packaged by a calcium phosphate method, and the method comprises the following specific steps:
293T cells were passaged every other day, 5X 10 cells were seeded per T150 cell flask6And (4) cells. After 48 hours, the cell number should reach 20-25 million/vial. In the case of 1T 150 cell culture flask, the cells were gently washed twice with about 15ml of 1 XPBS, 3ml of 0.25% pancreatin-2.21 mM EDTA was added until the cells were exfoliated, 12ml of 10% (wt) FBS (from Gibico) DMEM medium (from corning) was added to the exfoliated cells, the cells were collected and transferred to a sterile centrifuge tube, 1000rpm was applied, the cell was centrifuged for 10 minutes, the supernatant was aspirated, and the pellet was resuspended in 10ml of 10% (wt) FBS DMEM medium. Cell count, 12X 10 from cell concentration6The required volume of each cell, cells and 25ml of 10% (wt) FBS DMEM medium were combined, placed in a T150 cell flask, and shaken gently to distribute the cells evenly to the bottom of the cell flask at 37 deg.C and cultured overnight in a 5% CO2 incubator.
Cells were observed to reach a cell density of approximately 80% -90% at which time transfection was initiated by gently aspirating the medium 30-60 minutes prior to transfection. Plasmid DNA and calcium chloride solution were mixed, taking a T150 vial as an example, requiring 28ug pRSV.rev (available from Invitrogen), 28ug pGAG-Pol (available from Invitrogen), 11ug pVSVG (available from Invitrogen), 23ug recombinant lentivirus expression plasmid mesoCAR-1/mesoCAR-2/mesoCAR-3/mesoCAR-4, added to 1.5ml of calcium chloride solution and mixed well. Adding 1.5ml of BBS solution into a 15ml sterile centrifuge tube, uniformly mixing the DNA-calcium chloride solution with a 1ml gun head, dropwise adding the mixture into the BBS solution, rapidly mixing the mixture uniformly for 15-20 ℃, and incubating the mixture for 25-30 minutes at room temperature. The DNA-calcium chloride-BBS mixture (available from Shanghai Bintian Biotechnology Co., Ltd.) was added dropwise to the T150 bottle uniformly using a 5ml pipette. Culturing in a cell culture box containing 5% carbon dioxide at 37 deg.C for 6 hr. And changing the liquid after 6 h. The plate was gently shaken several times to suspend some calcium phosphate precipitate sufficiently, the culture solution containing calcium phosphate precipitate was aspirated, 20ml of fresh 5% (wt) FBS DMEM culture solution was added, and the culture was continued.
The 293T cell culture supernatant transfected the previous day was collected into a centrifuge tube, centrifuged at 1000rpm for 5 minutes, labeled, and stored in a 4 ℃ freezer. 20ml of 5% (wt) FBS DMEM medium previously preheated was added to the cell flask and the cell culture was continued overnight in the cell incubator at 37 ℃. The viral supernatant was collected a second time (48 h/day). The supernatants from both collections were pooled together and filtered through a 0.45 μm filter to remove cellular debris. Centrifuging at 12000-24000rpm at 4 ℃ overnight, pouring out all supernatant after centrifugation, adding fresh 5% (wt) FBS DMEM culture medium for resuspension, performing virus split charging, and rapidly storing in a refrigerator at-80 ℃.
Sequence listing
<110> Nanjing Aide immunotherapy research institute Co., Ltd
<120> chimeric antigen receptor targeting humanized CEA and use thereof
<130> 2019
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 1395
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 1
atggggggac ttgaaccctg cagcaggttc ctgctcctgc ctctcctgct ggctgtaagt 60
ggtctccgtc ctgtccaggt ccaggcccag agcgattgca gttgctctac ggtgagcccg 120
ggcgtgctgg cagggatcgt gatgggagac ctggtgctga cagtgctcat tgccctggcc 180
gtgtacttcc tgggccggct ggtccctcgg gggcgagggg ctgcggaggc agcgacccgg 240
aaacagcgta tcactgagac cgagtcgcct tatcaggagc tccagggtca gaggtcggat 300
gtctacagcg acctcaacac acagaggccg tattacaaag tcgagggcgg cggagagggc 360
agaggaagtc ttctaacatg cggtgacgtg gaggagaatc ccggccctag gatggcctta 420
ccagtgaccg ccttgctcct gccgctggcc ttgctgctcc acgccgccag gccgggatcc 480
gatattcagt tgacacagag ccctagcagc ctgagcgcca gtgtggggga tagagtgaca 540
attacttgta gagccgggga gagcgtggat atcttcgggg tgggcttcct gcactggtac 600
cagcagaagc cagggaaggc ccctaagctg ctgatctaca gggcgagcaa cctggagtcc 660
ggcgtgccca gccggttctc cgggagcggg tccaggaccg acttcacact gacaatcagc 720
agcctgcagc ccgaggattt tgccacctac tactgccagc agacaaacga ggacccatac 780
accttcgggc aggggacaaa ggtcgagatc aagggcgggg gggggagcgg ggggggcggc 840
tccggggggg gcgggagcga ggtgcagctg gtggagagcg gggggggcct ggtgcagccc 900
ggcggcagcc tgaggctgag ctgcgccgcc agcggcttca acattaagga cacatacatg 960
cactgggtga ggcaggcccc cggcaagggg ctggagtggg tggcccggat cgatcccgcc 1020
aacggcaaca gtaagtacgc tgatagcgtg aagggaaggt ttacaattag cgctgataca 1080
agtaaaaaca cagcttacct gcagatgaac agtcttaggg ccgaggatac agccgtgtac 1140
tattgcgccc catttggata ctacgtgtct gattacgcta tggcctactg gggacagggg 1200
acactggtga cagtgagcag cgctagcggt ggcggaggtt ctggaggtgg gggttccatc 1260
aaccttacaa atgtgacaga tatcatcagg gttccggtgt tcaacattgt cattctcctg 1320
gctggtggat tcctgagtaa gagcctggtc ttctctgtcc tgtttgctgt cacgctgagg 1380
tcatttgtac cctag 1395
<210> 2
<211> 464
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 2
Met Gly Gly Leu Glu Pro Cys Ser Arg Phe Leu Leu Leu Pro Leu Leu
1 5 10 15
Leu Ala Val Ser Gly Leu Arg Pro Val Gln Val Gln Ala Gln Ser Asp
20 25 30
Cys Ser Cys Ser Thr Val Ser Pro Gly Val Leu Ala Gly Ile Val Met
35 40 45
Gly Asp Leu Val Leu Thr Val Leu Ile Ala Leu Ala Val Tyr Phe Leu
50 55 60
Gly Arg Leu Val Pro Arg Gly Arg Gly Ala Ala Glu Ala Ala Thr Arg
65 70 75 80
Lys Gln Arg Ile Thr Glu Thr Glu Ser Pro Tyr Gln Glu Leu Gln Gly
85 90 95
Gln Arg Ser Asp Val Tyr Ser Asp Leu Asn Thr Gln Arg Pro Tyr Tyr
100 105 110
Lys Val Glu Gly Gly Gly Glu Gly Arg Gly Ser Leu Leu Thr Cys Gly
115 120 125
Asp Val Glu Glu Asn Pro Gly Pro Arg Met Ala Leu Pro Val Thr Ala
130 135 140
Leu Leu Leu Pro Leu Ala Leu Leu Leu His Ala Ala Arg Pro Gly Ser
145 150 155 160
Asp Ile Gln Leu Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
165 170 175
Asp Arg Val Thr Ile Thr Cys Arg Ala Gly Glu Ser Val Asp Ile Phe
180 185 190
Gly Val Gly Phe Leu His Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro
195 200 205
Lys Leu Leu Ile Tyr Arg Ala Ser Asn Leu Glu Ser Gly Val Pro Ser
210 215 220
Arg Phe Ser Gly Ser Gly Ser Arg Thr Asp Phe Thr Leu Thr Ile Ser
225 230 235 240
Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Thr Asn
245 250 255
Glu Asp Pro Tyr Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Gly
260 265 270
Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Glu Val
275 280 285
Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly Ser Leu
290 295 300
Arg Leu Ser Cys Ala Ala Ser Gly Phe Asn Ile Lys Asp Thr Tyr Met
305 310 315 320
His Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Arg
325 330 335
Ile Asp Pro Ala Asn Gly Asn Ser Lys Tyr Ala Asp Ser Val Lys Gly
340 345 350
Arg Phe Thr Ile Ser Ala Asp Thr Ser Lys Asn Thr Ala Tyr Leu Gln
355 360 365
Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys Ala Pro
370 375 380
Phe Gly Tyr Tyr Val Ser Asp Tyr Ala Met Ala Tyr Trp Gly Gln Gly
385 390 395 400
Thr Leu Val Thr Val Ser Ser Ala Ser Gly Gly Gly Gly Ser Gly Gly
405 410 415
Gly Gly Ser Ile Asn Leu Thr Asn Val Thr Asp Ile Ile Arg Val Pro
420 425 430
Val Phe Asn Ile Val Ile Leu Leu Ala Gly Gly Phe Leu Ser Lys Ser
435 440 445
Leu Val Phe Ser Val Leu Phe Ala Val Thr Leu Arg Ser Phe Val Pro
450 455 460

Claims (9)

1. A nucleic acid sequence selected from the group consisting of: contains the coding sequence of human DAP12, the coding sequence of T2A, the coding sequence of anti-CEA single-chain antibody, and the coding sequence of human TREM1 transmembrane and intracellular region which are connected in sequence.
2. The nucleic acid sequence as claimed in claim 1, wherein the nucleic acid sequence further comprises a nucleic acid sequence of a signal peptide before the coding sequence of the anti-CEA single-chain antibody, preferably the nucleic acid sequence of the signal peptide is as shown in SEQ ID NO 1, nucleic acid 412 and 474; and/or the nucleic acid sequence of the anti-CEA antibody is shown as the 481-1221 bit polynucleic acid of SEQ ID NO. 1; and/or the nucleic acid sequence of the human DAP12 is shown as the 1 st-339 st polynucleic acid of SEQ ID NO 1; and/or the nucleic acid sequence of the T2A is shown as the 355-405 th nucleic acid of SEQ ID NO. 1; and/or the nucleic acid sequence of the transmembrane and intracellular region of the human TREM1 is shown as the nucleic acid at position 1258-1395 of SEQ ID NO. 1.
3. A fusion protein selected from the group consisting of: a fusion protein containing sequentially connected human DAP12, T2A, anti-CEA single-chain antibody, human TREM1 transmembrane and intracellular regions; and a derivative fusion protein in which one or more amino acids are substituted, deleted or added in the defined amino acid sequence and which retains the activity of activated T cells; preferably, the anti-humanized CEA monoclonal antibody is huM 5A.
4. The fusion protein of claim 3, wherein the fusion protein has one or more of the following characteristics: the fusion protein also comprises a signal peptide at the N end of the anti-CEA antibody, preferably, the amino acid sequence of the signal peptide is shown as the amino acid at the 138 th and 158 th positions of SEQ ID NO. 2; the amino acid sequence of the anti-CEA single-chain antibody is shown as the amino acid at the 161 st and 407 st positions of SEQ ID NO 2; the amino acid sequence of the human DAP12 is shown as amino acids 1-113 of SEQ ID NO. 2; the amino acid sequence of the T2A is shown as the 119 th and 135 th amino acids of SEQ ID NO 2; the amino acid sequence of the transmembrane and intracellular region of the human TREM1 is shown as the amino acids 420-464 of SEQ ID NO. 2.
5. A nucleic acid vector comprising the nucleic acid sequence of any one of claims 1-2; preferably, the nucleic acid vector is a lentiviral vector comprising the nucleic acid sequence of any one of claims 1-2.
6. A lentivirus comprising the nucleic acid vector of claim 5, preferably said lentivirus vector.
7. A genetically modified T-cell or a pharmaceutical composition comprising a genetically modified T-cell, wherein the cell comprises the nucleic acid sequence of any one of claims 1-2, or comprises the nucleic acid vector of claim 5, or is infected with the lentivirus of claim 6, or stably expresses the fusion protein of claim 4.
8. Use of the nucleic acid sequence of any one of claims 1-2, the fusion protein of any one of claims 3-4, the nucleic acid vector of claim 5, or the lentivirus of claim 7 for the preparation of activated T cells.
9. Use of the nucleic acid sequence of any one of claims 1-2, the fusion protein of any one of claims 3-4, the nucleic acid vector of claim 5, the lentivirus of claim 6, or the genetically modified T cell of claim 7, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating a CEA-mediated disease; preferably, the CEA-mediated disease is brain cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, liver cancer, kidney cancer, lymphoma, leukemia, lung cancer, melanoma, metastatic melanoma, mesothelioma, neuroblastoma, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, skin cancer, thymoma, sarcoma, non-hodgkin lymphoma, uterine cancer, preferably colorectal cancer, liver cancer, breast cancer, melanoma, pancreatic cancer.
CN201910501250.6A 2019-06-11 2019-06-11 Chimeric antigen receptor targeting humanized CEA and uses thereof Pending CN112063640A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910501250.6A CN112063640A (en) 2019-06-11 2019-06-11 Chimeric antigen receptor targeting humanized CEA and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910501250.6A CN112063640A (en) 2019-06-11 2019-06-11 Chimeric antigen receptor targeting humanized CEA and uses thereof

Publications (1)

Publication Number Publication Date
CN112063640A true CN112063640A (en) 2020-12-11

Family

ID=73658496

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910501250.6A Pending CN112063640A (en) 2019-06-11 2019-06-11 Chimeric antigen receptor targeting humanized CEA and uses thereof

Country Status (1)

Country Link
CN (1) CN112063640A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112557662A (en) * 2021-02-22 2021-03-26 北京瀚梅生物科技有限公司 Colorectal cancer detection kit

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112557662A (en) * 2021-02-22 2021-03-26 北京瀚梅生物科技有限公司 Colorectal cancer detection kit
CN112557662B (en) * 2021-02-22 2021-07-06 和卓生物科技(上海)有限公司 Colorectal cancer detection kit

Similar Documents

Publication Publication Date Title
CN108504668B (en) Chimeric antigen receptor targeting CD19 and CD22 and uses thereof
CN108018299B (en) Chimeric antigen receptor targeting BCMA and uses thereof
CN108070607B (en) Chimeric antigen receptor targeting CD19-41BB-tEGFR and application thereof
CN109320615B (en) Chimeric antigen receptor targeting novel BCMA and uses thereof
CN108004259B (en) Chimeric antigen receptor targeting B cell maturation antigen and uses thereof
CN108728459B (en) Method and use of chimeric antigen receptor targeting CD19 and co-expressing IL-15
CN107841506B (en) Mesothelin-targeted chimeric antigen receptors and uses thereof
CN109503721B (en) Chimeric antigen receptor targeting CD19 and uses thereof
CN110923255B (en) Chimeric antigen receptor targeting BCMA and CD19 and uses thereof
CN108441505B (en) Chimeric antigen receptor targeting ROR1 and application thereof
CN108866088B (en) Targeting CLL-1 chimeric antigen receptor and uses thereof
CN113896801B (en) Chimeric antigen receptor cell targeting human Claudin18.2 and NKG2DL, and preparation method and application thereof
CN112522208A (en) Transgenic tumor infiltrating lymphocyte and application thereof
CN108239623B (en) Preparation method and application of mixed CART cells
CN108624608B (en) Preparation method and application of fourth generation chimeric antigen receptor targeting mesothelin
CN109504698B (en) Method for targeting chimeric antigen receptor of fully humanized mesothelin and application
CN108624607B (en) Methods and uses of chimeric antigen receptors targeting mesothelin and dual modifications thereof
CN110714018B (en) Chimeric antigen receptor targeting EGFRVIII and application thereof
CN108707619B (en) Chimeric antigen receptor targeting ROR1 and application thereof
CN108728458B (en) Methods and uses of chimeric antigen receptors targeting mesothelin in combination with IL-15 expression
CN109134660B (en) Methods of targeting chimeric antigen receptors of Mesothelin in combination with expression of anti-PD1 antibodies and uses thereof
CN112063640A (en) Chimeric antigen receptor targeting humanized CEA and uses thereof
WO2020061796A1 (en) Bcma-and-cd19-targeting chimeric antigen receptor and uses thereof
CN110845621A (en) Chimeric antigen receptor method targeting EGFR and CD19 double targets
CN112080510A (en) Chimeric antigen receptor targeting humanized GD2 and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20201211

WD01 Invention patent application deemed withdrawn after publication