CN114853880A - WT1 antigen-specific T cell receptor and anti-tumor application thereof - Google Patents

WT1 antigen-specific T cell receptor and anti-tumor application thereof Download PDF

Info

Publication number
CN114853880A
CN114853880A CN202210435441.9A CN202210435441A CN114853880A CN 114853880 A CN114853880 A CN 114853880A CN 202210435441 A CN202210435441 A CN 202210435441A CN 114853880 A CN114853880 A CN 114853880A
Authority
CN
China
Prior art keywords
tcr
antigen
seq
cell
binding fragment
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.)
Granted
Application number
CN202210435441.9A
Other languages
Chinese (zh)
Other versions
CN114853880B (en
Inventor
高福
蒋敏
谭曙光
孙文桥
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.)
Institute of Microbiology of CAS
Original Assignee
Institute of Microbiology of CAS
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 Institute of Microbiology of CAS filed Critical Institute of Microbiology of CAS
Priority to CN202210435441.9A priority Critical patent/CN114853880B/en
Publication of CN114853880A publication Critical patent/CN114853880A/en
Application granted granted Critical
Publication of CN114853880B publication Critical patent/CN114853880B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • 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
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001111Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer 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
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/7051T-cell receptor (TcR)-CD3 complex

Landscapes

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

Abstract

The invention provides T cell epitope specific T cell receptor murine TCR of tumor associated antigen WT1 and antigen binding fragment thereof, wherein the T cell receptor is composed of alpha and beta peptide chains respectively, nucleic acid for encoding the T cell receptor and the T cell epitope specific T cell receptor murine TCR, and the nucleic acid, vector containing the nucleic acid, host cell containing the vector and anti-tumor application of the host cell and the host cell; also provides a preparation method of the T cell epitope specific T cell receptor murine TCR of the WT1 protein. The specific T cell receptor and the antigen binding fragment thereof can be used as an immune effect activator to stimulate the immune response of an organism, thereby having the effect of resisting diseases such as tumor and the like.

Description

WT1 antigen-specific T cell receptor and anti-tumor application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a T Cell Receptor (TCR) or an antigen binding fragment thereof capable of specifically recognizing HLA-A11 restricted T cell epitope of tumor associated antigen WT 1.
Background
In 2011, cancer surpasses heart disease, and becomes the first leading cause of death worldwide. WHO published 12 months in 2013, the number of newly added cancer patients worldwide has exceeded 1400 million every year, which is greatly increased compared to 1270 million patients as a statistical result in 2008. By 2020, cancer causes 960 million deaths per year, with about 1 trillion dollars worldwide each year in cancer diagnosis.
The immune cell therapy modified by genetic engineering technology is gradually becoming an important means for treating solid tumors and hematologic malignancies in recent years [1,2] Among them, the engineered T cell therapies represented by chimeric antigen receptor engineered T cell (CAR-T) and T cell receptor engineered T cell (TCR-T) therapies are expected to become a new strategy for clinical treatment of tumors. CD 19-targeted CAR-T cells have been shown to have significant therapeutic efficacy in the clinical treatment of a variety of B cell malignancies and there are a number of products approved for marketing, but more than half of patients with malignant lymphoma relapse after treatment due to poor CAR-T cell persistence or antigen loss [3,4] . TCRs recognize intracellular or extracellular antigens presented by the Major Histocompatibility Complex (MHC), and have the characteristics of a broader antigen recognition range and higher immune response sensitivity than CAR, and TCR-T therapy has been demonstrated to have certain clinical therapeutic value for a variety of tumors including melanoma and multiple sarcoma [5]
The selection of safe and effective targets is critical to TCR-T cell therapy, and the ideal antigen target should have the characteristic of high expression specificity in tumor cells in the first place. The Wilms tumor 1(WT1) protein was found to be overexpressed in various tumor cells such as acute myeloid leukemia and breast cancer, but not expressed in normal hematopoietic cells and paraneoplastic cells [6,7] It is considered to be a safe and therapeutically promising T cell target. WT1 gene was found in Wilms tumors in 1990 [8] Which encodes a zinc finger protein transcription factor in an individualDuring development, WT1 gene is mainly expressed in kidney, testis and ovary of fetus [9] . Since the mutation of WT1 gene was found to be involved in the development of genitourinary diseases and renal tumors in early studies, it was defined as a cancer suppressor gene [10,11] . However, in the research of various tumor cells such as leukemia, breast cancer and the like, the wild type full-length WT1 gene is found to show high expression [12,13] Most of the studies at present support that WT1 is a protooncogene, and its high expression in tumor cells can promote the proliferation of tumor cells [14-16]
WT1 protein is found to have strong immunogenicity, and is defined as a preferred target antigen of a cancer vaccine by the national cancer research center in 2009 [17] Only 2 of the WT 1-specific CD8+ T cell epitopes identified so far, were HLA-A2-restricted epitopes WT1126-134(RMFPNAPYL) [18] And its derived epitope YMFPNAPYL [19,20] HLA-A24 restricted epitope WT1235-243(CMTWNQMNL) [21] And its derived epitope CYTWNQMNL [22] . The existing clinical research data show that the tumor polypeptide vaccine based on the WT1 antigen not only has good safety, but also can induce continuous immune response in patients with Acute Myelogenous Leukemia (AML) and the like [23,24] . In addition, a study of TCR-T cell therapy of AML bone marrow transplant patients has shown that adoptive delivery of TCR-T cells targeting the HLA-A2-restricted WT1 epitope to patients is effective in preventing AML recurrence following bone marrow transplant [25,26]
Although TCR-T cell therapy specific for the WT1 antigen has shown good therapeutic promise in hematological tumors, therapeutic strategies developed against the only epitopes currently available do not benefit the broader population of patients due to their HLA-restriction. HLA-A11 is one of the most common HLA class I genotypes in the world, with a phenotype frequency of about 20-30% in the Chinese population. However, there is no HLA-A11 restricted WT1 epitope, and therefore, there is a need to identify HLA-A11 restricted WT1 epitope and screen specific TCR to provide a basis for clinical treatment of tumor patients with HLA-A11 genetic background.
Disclosure of Invention
Specifically, tumor-associated antigen WT1, when used as an antigen, can cause the body to produce CD8 + CTL (cytotoxic T lymphocyte) responses. Some of the polypeptides in the WT1 protein can be presented on the cell surface by HLA molecules and recognized by T cells. The identification of T cell epitopes of HLA-A11-restricted WT1 protein and the discovery of TCRs are of great significance for immunotherapy targeting WT 1.
The inventors screened for a specific T Cell Receptor (TCR) that specifically targets the above epitope of the tumor associated antigen WT1 by a TCR single cell screening technique.
One embodiment of the present invention includes specific T cell receptors and antigen binding fragments thereof that provide targeting to an epitope of the tumor associated antigen WT 1. Another embodiment of the present invention encompasses the use of the above-described T cell receptor and antigen binding fragments thereof for the preparation of a medicament for the treatment of tumors carrying the tumor associated antigen WT1 gene.
The invention is based on the principle, the specific TCR of the tumor associated antigen WT1 (shown as SEQ ID NO: 17) polypeptide or the antigen binding fragment thereof is specifically bound with the compound molecule of the tumor associated antigen WT1 (shown as SEQ ID NO: 17) polypeptide and HLA-A11, thereby stimulating the activation of T cells, inducing the T cells to secrete cytokines such as IFN-gamma and the like, and further killing tumor cells expressing the tumor associated antigen WT1 gene, in particular expressing the tumor associated antigen WT1(414-423) polypeptide (shown as SEQ ID NO: 17) and HLA-A11.
In the present invention, the expression "tumor associated antigen WT1 specific TCR" or "murine tumor associated antigen WT1 specific TCR" is a murine TCR against the HLA-A11 restricted CTL epitope polypeptide of tumor associated antigen WT1 (the sequence of which is shown in SEQ ID NO: 17), and in the present embodiment is referred to as WT1-9E TCR or 9E.
The present application includes TCRs or derivatives that specifically bind to a complex molecule of the tumor associated antigen WT1 polypeptide (shown in SEQ ID NO: 17) and HLA-A11, as well as TCR fragments that exhibit substantially the same functional antigen specificity as the original TCR.
In the present invention, "fragments of a TCR" or "antigen-binding fragments" refer to antigen-binding fragments and TCR analogs of TCRs, which typically include at least a portion of the antigen-binding or variable region of the parent TCR, such as one or more (e.g., six) CDRs. Such antigen-binding fragments or fragments of TCRs retain at least some of the binding specificity of the parent TCR, and their use is also encompassed by the invention.
"specific" binding, when referring to ligand/receptor, antibody/antigen or other binding pairs, refers to determining the presence or absence of a binding reaction of a protein, e.g., a polypeptide (as shown in SEQ ID NO: 17), to a molecule of HLA-A11 complex in a heterogeneous population of proteins and/or other biological agents. Thus, under the conditions specified, a particular ligand/antigen binds to a particular receptor/antibody and does not bind in significant amounts to other proteins present in the sample.
The invention also provides a pharmaceutical composition comprising a TCR specific for a polypeptide of the tumor associated antigen WT1 (shown in SEQ ID NO: 17) or an antigen binding fragment thereof. To prepare the pharmaceutical composition, the tumor associated antigen WT1 (shown in SEQ ID NO: 17) polypeptide-specific TCR or antigen-binding fragment thereof can be prepared into various desired dosage forms by mixing with a pharmaceutically acceptable carrier or excipient. Examples of the dosage form of the pharmaceutical composition of the present invention include tablets, powders, pills, powders, granules, fine granules, soft/hard capsules, film-coated preparations, pellets, sublingual tablets, and ointments, which are oral preparations, and examples of non-oral preparations include injections, suppositories, transdermal preparations, ointments, plasters, and external liquid preparations, and those skilled in the art can select an appropriate dosage form according to the administration route, the administration target, and the like.
The dose of the active ingredient of the pharmaceutical composition of the present invention varies depending on the subject, the target organ, the symptom, the administration method, and the like, and can be determined by the judgment of the doctor in consideration of the type of the formulation, the administration method, the age and weight of the patient, the symptom of the patient, and the like.
The pharmaceutical compositions of the invention may also contain other agents, including but not limited to cytotoxic, cytostatic, antiangiogenic or antimetabolic agents, tumor-targeting agents, immunostimulants or immunomodulators or TCRs in combination with cytotoxic, cytostatic or other toxic agents.
Specifically, the present invention provides the following.
A T Cell Receptor (TCR) or an antigen binding fragment thereof, which TCR or antigen binding fragment thereof is capable of binding to a T cell epitope of the tumour associated antigen WT1 and to the HLA-A11 complex, and which TCR comprises an alpha chain variable region and a beta chain variable region,
the TCR, or antigen-binding fragment thereof, comprises the following alpha chain Complementarity Determining Regions (CDRs) and beta chain Complementarity Determining Regions (CDRs):
an alpha chain complementarity determining region CDR1 shown in SEQ ID NO. 2;
an alpha chain complementarity determining region CDR2 shown in SEQ ID NO. 3;
an alpha chain complementarity determining region CDR3 shown in SEQ ID NO. 4; and
a beta strand complementarity determining region CDR1 shown in SEQ ID NO. 6;
the beta strand complementarity determining region CDR2 shown in SEQ ID NO. 7;
the beta-strand complementarity determining region CDR3 shown in SEQ ID NO. 8,
wherein the amino acid sequence of the T cell epitope is shown as SEQ ID NO. 17.
2. The T Cell Receptor (TCR) or an antigen-binding fragment thereof of item 1 comprising:
1, and
the beta chain variable region as shown in the sequence of SEQ ID NO. 5.
3. The TCR of items 1 or 2 or an antigen binding fragment thereof, wherein the TCR is a murine TCR, a human murine chimeric TCR or a humanized TCR, preferably having the sequence shown in SEQ ID NO 9 and SEQ ID NO 15.
4. A polynucleotide encoding a TCR, or an antigen-binding fragment thereof, according to any one of items 1-3.
5. An expression vector comprising the polynucleotide of item 4, said expression vector being a lentiviral vector.
6. A host cell comprising the expression vector of item 5.
7. A method of making a TCR, or an antigen-binding fragment thereof, according to any one of items 1-3, comprising:
1) culturing the host cell of item 6;
2) recovering the TCR of any of items 1-3 or the antigen-binding fragment thereof from the host cell or culture medium thereof.
8. A pharmaceutical composition comprising a TCR, or antigen-binding fragment thereof, according to any one of items 1-3, and a pharmaceutically acceptable carrier.
9. Use of a TCR, or an antigen-binding fragment thereof, according to any one of items 1-3 in the manufacture of a medicament, such as a proteinaceous agent, a protein agent conjugate, or a TCR in combination with an antigen, for increasing the level of a cytokine that secretes IFN- γ from a T cell.
10. Use of a TCR, or an antigen-binding fragment thereof, according to any one of items 1 to 3 in the preparation of a reagent for the detection of a tumor cell expressing a T cell epitope of the tumor-associated antigen WT1, or for the preparation of a reagent for the detection or diagnosis of a tumor, wherein the TCR, or the antigen-binding fragment thereof, specifically binds to a T cell epitope of the tumor-associated antigen WT1 and HLA-a11, and the sequence of the T cell epitope is set forth in SEQ ID NO: 17.
11. Use of a TCR, or an antigen-binding fragment thereof, according to any one of items 1 to 3 in the preparation of an anti-tumour medicament for the treatment of a patient having a tumour bearing the T cell epitope of the tumour-associated antigen WT1, as set out in SEQ ID No. 17, preferably ovarian cancer.
THE ADVANTAGES OF THE PRESENT INVENTION
In this study, we aimed to identify CD8+ T cell epitopes of WT1 protein from HLA-a x 11:01 transgenic mice and to screen for functional TCRs that specifically recognize the WT1 epitope. We successfully identified an HLA-A11-restricted epitope WT1(414-423) (FSCRWPSCQK) from transgenic mice and screened for an immunodominant TCR specific for WT1 (414-423). We verified the binding ability of the TCR to WT1(414-423)/HLA-A11 at the cellular level and protein level, and prepared TCR-T cells from primary T cells, and the results of in vitro cell function tests show that the TCR-T cells can specifically secrete Interferon (IFN) -gamma under the stimulation of WT1(414-423) polypeptide. The functional TCR is expected to become a potential candidate drug for future tumor TCR-T cell therapy.
The specific T cell receptor of the WT1(414-423) (FSCRWPSCQK) polypeptide epitope targeting WT1 and the T cell expressing the same have high binding property, so that the specific T cell receptor can be used for carrying out preliminary study on medicaments and developing medicaments targeting mutation related to tumor growth and development in experimental models and the like. Therefore, the compounds can be used for preparing medicaments for diagnosing and treating various tumors expressing WT1 at various stages, in particular for treating solid tumors of people with HLA-A11 genetic background.
Drawings
FIG. 1 Single cell sorting of WT1(414-423) -specific T cells from HLA-A11: 01 transgenic mice.
(a) A process of polypeptide immunization of HLA-A11: 01 transgenic mice; (b) an ELISPOT experiment detects that WT1(414-423) polypeptide causes IFN-gamma response pattern secretion of mouse splenocytes; (c) flow-detecting the ratio of Mus-2 mouse splenocytes CD3+/CD8 +/tetramer + cells and performing single cell sorting.
FIG. 2 shows the molecular sieve chromatography and biotinylation levels of WT1(414-423) and HLA-A11 complex. a is a molecular sieve chromatogram; panel b shows SDS-PAGE of the molecular sieve chromatography of biotinylated WT1(414-423) and HLA-A11 complexes, and the corresponding biotinylation level detection.
FIG. 3 293T cells co-transfected with WT1-9E TCR and CD3-CD8 and tested for binding specificity of TCRs using WT1(414-423)/HLA-A11 tetramer or control HLA-A11 tetramer.
FIG. 4 specific secretion of IFN-. gamma.cytokines by interaction of WT1-9E TCR-T cells with PANC1 target cells loaded with WT1(414-423) polypeptide. (a) Interaction of WT1-9E TCR-T with polypeptide-loaded PANC1 cells. (b, c) detecting TCR-T cells that produce IFN- γ following stimulation with polypeptide loaded PANC1 using an ELISPOT assay. Primary T cells from 3 volunteers D1, D2, and D3 were used to make WT1-9E TCR-T cells in this experiment. Wherein (b) shows positive reaction spot-forming cells (SFCs) in which the number of 9E TCR-T per well is 1X10 5 . (c) Based on (b) inStatistical analysis of SFCs. blank shown on ELISPOT plates: only 9E TCR-T; mock T cells + PANC1 cells, without added polypeptide; NC T cells + PANC1 cells + negative polypeptide control. P < 0.01, p < 0.001.
FIG. 5 WT1(414-423)/HLA-A11 pMHC and 9E TCR protein interactions. (a, b) in vitro inclusion body renaturation and purification of WT1(414-423)/HLA-A11 pMHC protein (a) and 9E TCR protein (b), the panels show the purity of the proteins as identified by SDS-PAGE. (c, d) Surface Plasmon Resonance (SPR) measurement of binding kinetics (c) and affinity (d) for WT1(414-423)/HLA-A11 pMHC and 9E TCR.
Detailed Description
The technical scheme of the invention is further illustrated by the detailed description and the attached drawings, but the technical scheme can be understood by those skilled in the art: the following detailed description and examples are intended to illustrate the invention and should not be construed as limiting the invention in any way. It will be apparent to those skilled in the art that many modifications can be made to the present invention without departing from the spirit thereof, and such modifications are intended to be within the scope of the invention.
The following experimental methods are all conventional experimental methods in the art unless otherwise specified, and the experimental materials used are all experimental materials that can be easily obtained from commercial companies unless otherwise specified.
Example 1 prediction and identification of WT1 epitope
We predicted 6 WT1 polypeptides with higher affinity to HLA-A11: 01 (Table 1) by NetMHC-4.0 online prediction tool, and immunized 6 HLA-A11: 01 transgenic mice after mixing these 6 polypeptides (Poisson diagram). After three polypeptide immunizations, the existence of T cells activated by a specific polypeptide in spleen cells of mice is detected by an IFN-gamma-ELSPOT experiment, so that the epitope of the WT1 polypeptide with immunogenicity is preliminarily identified.
TABLE 1 HLA-A11-restricted WT1 polypeptide sequence and affinity prediction
Polypeptide name Sequence of Predicted affinity (nM)
WT1 167-177 HAAQFPNHSFK 149.31
WT1 218-227 RTPYSSDNLY 613.41
WT1 323-332 FMCAYPGCNK 249.20
WT1 391-399 KFSRSDHLK 1073.82
WT1(414-423) FSCRWPSCQK 161.20
WT1 436-444 NMHQRNMTK 814.58
Data from NetMHC-4.0 online predictions. .
Transgenic C57/B6 mice expressing the human HLA-a 11:01 gene were obtained from beijing baiosaccae and housed in SPF animal houses. 100. mu.g of each of the 6 synthesized WT1 polypeptides were co-dissolved in 50. mu.L of sterile PBS and mixed with 50. mu.L of Quick CTL cell immunization adjuvant (Biodragon), and HLA-A11: 01 transgenic mice were immunized 4 to 6 weeks by inguinal subcutaneous injection, and 100. mu.L of each mouse was injected. The immunization was boosted in the same manner on day 7 and 14, respectively, and mice were euthanized and spleen cells were collected for culture and detection on day 21.
Mouse spleen cells (100. mu.L, 2.5X 10) were added to 96-well ELISPOT plates pre-coated with anti-mouse-IFN-. gamma.antibody (BD, Biosciences) 5 Cell number/well), adding 1 WT1 polypeptide (100 μ L, 40 μ g/mL) to be detected into each well, taking RPMI 1640 culture medium and PMA/ION (Beijing Dake) as negative control and positive control stimulators respectively, mixing well, and placing ELISPOT plate in 37 deg.C incubator. After 18 hours of incubation, the cells were discarded, Biotinylated antibody (Biotinylated anti-mouse-IFN-. gamma.) (BD, Biosciences) and enzyme conjugate (SA-HRP) (BD, Biosciences) were incubated in this order according to the instructions, developed with horseradish peroxidase substrate (AEC) (BD, Biosciences), and finally the development was stopped with water. Spots were captured and counted using an automatic ELISPOT reader and image analysis software (Cellular Technology Limited).
The results show that the WT1(414-423) polypeptide can activate mouse T cells to secrete IFN-gamma and present a positive reaction spot on an ELISPOT plate (FIGS. 1a and b), the ELISPOT detection finds that the WT1(414-423) polypeptide induces a specific T cell reaction in 4/6 mice, and further specific TCR screening and function research are carried out on the epitope of the WT1(414-423) (FSCRWPSCQK) polypeptide.
Example 2 WT1(414-423) (FSCRWPSCQK) polypeptide-specific T cell sorting and TCR Gene cloning
Subsequently, tetramers of WT1(414-423) polypeptide and HLA-A11 (FIG. 2) were prepared therefrom, and CD3 was selected from splenocytes of immunized mice by staining with CD3, CD8 antibodies together + CD8 + T cells, and sorting to obtain WT1(414-423) epitope specific T cells. The details are as follows.
Preparation of WT1(414-423)/HLA-A11 tetramer
The binding properties and specific T cell responses of the polypeptide WT1(414-423) and HLA-A11, as well as specific TCRs were evaluated and screened.
According to the conventional method, respectively aligning beta 2 m (beta 2-microglobulin, HLA-A11 light chain gene) (Uniprot: P61769) and HLA-A11 heavy chain gene (Uniprot: Q5S3G3) are subjected to prokaryotic codon optimization to obtain beta 2 The nucleic acid sequence of m is shown as SEQ ID NO. 14, and the coded amino acid sequence thereof is shown as SEQ ID NO. 13. The obtained HLA-A11 heavy chain gene with biotin label for prokaryotic expression is shown as SEQ ID NO. 12, and the coded amino acid sequence thereof is shown as SEQ ID NO. 11.
The heavy chain gene of HLA-A11 with biotin label is a sequence which carries expression biotin specific binding polypeptide at the C end of the heavy chain gene. The amino acid sequence of the biotin specific binding polypeptide is shown as SEQ ID NO. 22.
The respective DNA sequences were synthesized by the assignee Inc. (Nanjing Kingsry Co.), and the Nde I and Xho I cleavage sites were introduced, respectively, wherein the Nde I cleavage site was located at the 5 'end of the sequence and the Xho I cleavage site was located at the 3' end of the sequence. Using the cleavage sites Nde I and Xho I, the synthesized beta 2 The DNA sequences of the m and HLA-A11 heavy chain genes were cloned into expression vector pET-21a (Invitrogen corporation) to establish beta 2 Prokaryotic recombinant expression plasmids of m and HLA-A11 heavy chain protein beta 2m-pET 21a and HLA-A11-pET 21 a.
The two expression plasmids are respectively transferred into E.coli.BL21(DE3) competent cells (purchased from Tianenzze organisms) by a heat shock method, IPTG is added for induction expression, escherichia coli is crushed, and the homogenate is homogenized to extract inclusion bodies, so as to obtain the inclusion body proteins of beta 2m and HLA-A11 heavy chains in the state of the inclusion bodies.
1mL of. beta.2m inclusion bodies (30mg/mL in a solution containing 6M Gua-HCl, 50mM Tris pH8.0, 100mM NaCl, 10mM EDTA and 10mM DTT) were slowly added dropwise to 1L of a renaturation solution (20mM Tris-HCl, 400mM L-arginine, EDTA 2mM, GSH/GSSG 5mM/1mM) containing 5mg of the WT1 (414-.
After 1 hour, the reaction was carried out as β 2 m: the heavy chain inclusion body of HLA-a11 was slowly added dropwise to the above renaturation solution at a molar ratio of HLA-a11 heavy chain of 1:1, and renaturation was carried out for 8 hours or more. Concentration: passing the renatured sample through a 10kDa filter using an ultrafiltration cup to concentrate the sample and changing the buffer to 20mM Tris-Cl, 50mM NaCl, pH8.0 by concentration; liquid is changed twice: after concentrating the sample to about 20mL, add to 200mL of buffer containing 20mM Tris-Cl, 50mM NaCl, pH 8.0; then, after concentrating to about 20mL, 20mM Tris-Cl, 50mM NaCl, pH8.0 buffer solution was added again to 100mL, and finally concentrated to about 10-20mL volume, resulting in WT1 (414-.
After the sample was removed, the supernatant was concentrated to about 0.5-1mL by centrifugation at 12000rpm for 10min at 4 ℃ and the WT1(414-423)/HLA-A11 complex was purified by superdex200 molecular sieves (from GE Healthcare). The protein peak of WT1(414-423)/HLA-A11 complex (peak at about 15.8mL) was collected based on the light absorption value at 280nm for purification. The protein sample purified by the molecular sieve is collected in an ultrafiltration concentration tube, concentrated to about 500 mu L and then centrifuged at 4 ℃ to remove the precipitate, thus obtaining the WT1(414-423)/HLA-A11 complex protein sample.
2. Biotinylation reaction
Using the WT1(414-423)/HLA-A11 complex protein sample obtained in step (1), the following biotinylation reaction system (500. mu.L) was prepared.
Biotinylation reaction system (purchased from avidly corporation): a total of 500. mu.L
Protein sample 1mg/mL 200. mu.L, Buffer A (bicine Buffer) 50. mu.L, Buffer B (ATP, biotin) 50. mu.L, 200. mu.M biotin, Bir-Ase (3mg/mL) 20. mu.L, made up to 500. mu.L with 20mM Tris-Cl, 50mM NaCl, pH 8.0. After the preparation, the mixture is mixed evenly, placed on ice and incubated overnight in a refrigerator at 4 ℃.
The biotinylated reaction system was purified by Superdex200 molecular sieves to remove excess biotin, and the peak values of the biotinylated WT1 (414-.
And (3) detecting biotinylation efficiency: the biotinylated WT1(414-423)/HLA-A11 complex was concentrated to about 500. mu.L, and samples were taken for SDS-PAGE shift assay to verify the biotinylation effect.
One sample and two controls were set for three groups:
group A (MHC): biotinylated WT1(414-423)/HLA-A11 complex sample 8. mu.L + molecular sieve buffer 2. mu.L;
group B (+ SA): biotinylated WT1(414-423)/HLA-A11 complex sample 8. mu.L + streptavidin 2. mu.L (20 mg/mL);
group C (SA): streptavidin 2. mu.L + molecular sieve buffer 8. mu.L.
The three samples were incubated on ice for 30min and then identified by SDS-PAGE, the results of which are shown in FIG. 2.
The results show that: from the light absorption at 280nm and the corresponding SDS-PAGE, a band lag was observed in the SDS-PAGE of the WT1(414-423)/HLA-A11 complex after biotinylation due to its binding to streptavidin as a macromolecule, which caused the band lag.
From the SDS-PAGE band intensity, it was observed that the WT1(414-423)/HLA-A11 complex was significantly reduced in intensity after biotinylation, and it was judged that the WT1(414-423)/HLA-A11 complex could be efficiently biotinylated (as shown in FIG. 2).
Preparation of WT1(414-423)/HLA-A11 tetramer
Biotinylated WT1 (414-: the molar ratio of WT1 (414-.
WT1(414-423)/HLA-A11-PE tetramer specific T cell sorting and single cell TCR gene amplification and sequencing
Washing with PBS, resuspending, collecting the mouse splenocytes obtained after WT1(414-423) polypeptide immunization at 1X10 7 Centrifuging at 200-250g for 10 min; washed three times with PBS containing 0.5% BSA and centrifuged at 200-250g for 10 min.
WT1(414-423)/HLA-A11-PE tetramer (obtained from the above tetramer preparation), PerCP-Cy5-CD8 (from BD Co.) and FITC-CD3 fluorescent antibody (from BD Co.) were incubated with splenocytes at a molar ratio of 1:1:1 for 20 minutes at 25 ℃; washed three times with PBS containing 0.5% BSA and centrifuged for 10min at 200-250 g; cells were resuspended in PBS containing 0.5% BSA.
Then the cells are subjected to flow cytometry single cell sorting. Wherein the control group is a negative control which does not stain the tetramer, the experimental group is a WT1(414-423)/HLA-A11 tetramer staining group, the abscissa is WT1(414-423)/HLA-A11 tetramer, and the ordinate is CD8 staining. Selecting lymphocyte subset, selecting CD3 + CD8 + T cells, sorted to give WT1(414-423)/HLA-A11-PE tetramer positive CD8+ T cells, and the results are shown in FIG. 1 c.
Single positive cells were sorted into 96-well plates containing cell lysates (purchased from tiangen organisms) and rnase inhibitors (purchased from kanji century organisms). Total RNA was then extracted from WT1(414-423)/HLA-A11-PE tetramer positive T cells in each well and subjected to 5' RACE TCR gene amplification as described below.
The 5' RACE is divided into three steps: reverse transcription (RT-PCR), first round PCR amplification and second round PCR amplification. The procedure was carried out using the Takara D315-FullRACE Kit according to the instructions.
(1) RT-PCR: the downstream primer used was a TCR gene constant region specific primer GSP1 (available from Takara Co.), and the upstream primer was a target-switching primer (available from Takara Co.) with an Oligo-guanine deoxyribonucleic acid (Oligo dG) at the 3' end.
(2) First round PCR: the first PCR product of the α chain or β chain of TCR was obtained by using the cDNA of TCR obtained in (1) above as a template, the upstream Primer as outer linker Primer 1 (5' RACE outer Primer, Takara Co.), and the downstream Primer as a specific Primer for a TCR constant region upstream of the constant region GSP1 (purchased from Takara Co.).
(3) Second round PCR: the first round PCR product of α chain or β chain of TCR obtained in (2) above was used as template, the upstream Primer was inner linker Primer 2 (5' RACE inner Primer), and the downstream Primer was a section of TCR constant region specific Primer (purchased from Takara D315-FullRACE Kit) upstream of constant region GSP2 to obtain the second round PCR product of α chain or β chain of TCR, respectively.
And carrying out agarose gel electrophoresis on the amplified second round PCR amplification product containing the TCR alpha chain and the beta chain variable region gene to obtain the TCR alpha chain or beta chain variable region target gene at the position of 500 bp. The band of interest was recovered and the gene fragment of interest was ligated to a T vector (pMD18T, Takara) using T4 ligase. The ligation products were then transformed into DH 5. alpha. cells (purchased from Tiangen organisms) and subjected to monoclonal gene sequencing (Kurtoui Boxing family).
The obtained WT1(414-423) polypeptide specific T cells are subjected to single-cell TCR gene amplification sequencing through the processes, and after result analysis, alpha chains and beta chains which appear frequently in the T cells are selected to combine into a new TCR which is named as WT-1-9E TCR, and the new TCR is further combined and functionally verified. The WT-1-9E TCR of the invention has an alpha chain variable region as shown in the sequence of SEQ ID NO. 1 and a beta chain variable region as shown in the sequence of SEQ ID NO. 5.
Example 2 binding of WT1(414-423)/HLA-A11 tetramer to cells expressing WT1-9E TCR
In this example, the inventors further confirmed that the screened WT1-9E TCR had specific binding to WT1(414-423) polypeptide/HLA-A11.
Validation of WT1-9E TCR binding specificity
First, the α chain and β chain variable regions (V regions) (amino acid sequences are shown in SEQ ID NO:1 and SEQ ID NO:5, respectively) of WT1-9E TCR were ligated to the α chain and β chain constant region (C region) genes of human TCR (synthesized by Hongyingbio corporation) to obtain WT1-9E chimeric TCR α and β chain sequences, the chimeric TCR α chain having the amino acid sequence shown in SEQ ID NO:9, the chimeric TCR α chain having the nucleic acid sequence shown in SEQ ID NO:10, the chimeric TCR β chain having the amino acid sequence shown in SEQ ID NO:15 and the chimeric TCR β chain having the nucleic acid sequence shown in SEQ ID NO: 16.
And a slow virus expression vector, namely a slow virus expression vector WT1-9E-pCDH, of the chimeric WT1-9E TCR is constructed by connecting the alpha chain and the beta chain of the WT1-9E TCR by a P2A sequence (the amino acid sequence of the P2A sequence is shown as SEQ ID NO: 19) and taking a slow virus expression plasmid pCDH (purchased from Invitrogen) as a starting plasmid.
HEK-293T cells (purchased from ATCC) were co-transfected with WT1-9E-pCDH lentiviral expression vectors and CD3-CD8-pCDH plasmids (purchased from Nanjing Kingsler) expressing CD3 and CD8, respectively, in a number 1:1 ratio. 24 hours after co-transfection, cells were centrifuged at 200 and 250g for 10 min; HEK-293T cells expressing WT1-9E TCR were obtained by washing three times with PBS containing 0.5% BSA and centrifugation at 200-250g for 10 min.
Then, in order to further verify the binding of WT1-9E TCR to WT1(414-423) polypeptide/HLA-A11, the tetramer of WT1(414-423) polypeptide/HLA-A11 prepared above was stained with 293T cells expressing WT1-9E TCR to evaluate the binding specificity.
Specifically, the co-transfected HEK-293T cells were incubated with WT1(414-423)/HLA-A11-PE tetramer prepared above, and PerCP-Cy5-CD8 and FITC-CD3 antibodies (both BD Co.) at a molar ratio of 1:1:1 for 30 min; washing with PBS containing 0.5% BSA for three times, and centrifuging at 200-250g for 10 min; the cells were resuspended in PBS containing 0.5% BSA and used to determine the frequency of T cells positive for WT1 (414-423)/HLA-A11.
The analysis was performed using flow cytometry. Among them, mock group was HEK-293T cells not staining tetramers (negative control), WT1-9E group was HEK-293T cells staining tetramers with WT1(414-423)/HLA-A11, abscissa was WT1(414-423)/HLA-A11 tetramers, and ordinate was CD3 staining, and the results are shown in FIG. 3.
The results showed that the proportion of cells in WT1-9E group capable of binding to WT1(414-423)/HLA-A11-PE tetramer was about 40% of CD8 positive cells. Therefore, it can be seen that cell level binding experiments in HEK-293T cells indicate that WT1-9E TCR is capable of specifically binding to the WT1(414-423)/HLA-A11 complex.
Example 3 preparation of WT1-9E TCR-T cells and their immunoreaction with target cells
In this example, the WT1-9E TCR gene was introduced into T cells isolated from Peripheral Blood Mononuclear Cells (PBMC) isolated from healthy volunteers with HLA-A11 genetic background as TCR-T effector cells (FIG. 4 a). To evaluate the activity of WT1-9E TCR-T cells on target cells, the target cells were pancreatic cancer PANC-1 cell line.
The specific operation is as follows.
Preparation of TCR-T cells and detection of TCR expression efficiency
Peripheral blood lymphocytes from three healthy volunteers (D1, D2, and D3) were collected to obtain PBMCs. And a part of the PBMCs were subjected to negative selection with magnetic beads (Biolegend) to separate CD 8T cells.
WT1-9E TCR Lentiviral preparation
The expression plasmid WT1-9E-pCDH of example 2 was mixed with lentiviral packaging plasmids PLP1, PLP2 and VSVG (all available from Addgene) in the ratio PLP1: PLP2: VSVG: WT 1-9E-pCDH-1: 1:1:1, and 30. mu.L of the mixture was diluted into DMEM medium (1.25 mL).
30ul of polyetherimide (PEI, 1. mu.g/. mu.L) was added to DMEM (1.25 mL). The PEI/DMEM solution was added to the prepared DNA solution in its entirety, incubated at room temperature for 15 minutes, added to 293T cells (Shanghai cell bank) and mixed well.
After 6 hours, the culture medium was carefully aspirated, and 25mL of fresh culture medium was added to continue the culture. After 64 hours, the virus-containing supernatant was collected, which was WT1-9E TCR lentiviral supernatant.
Adding microspheres (ThermoFisher) coated with anti-CD3/anti-CD28 into T cells according to the quantity ratio of 1:1 for activation culture overnight, then adding WT1-9E TCR lentivirus into the T cells according to the volume ratio of 1:1, mixing uniformly, setting virus-free infected holes as controls, and culturing in a 5% CO2 incubator at 37 ℃. After 24 hours, the medium was changed to complete medium and the culture was continued until day 10. The microspheres of anti-CD3/anti-CD28 were removed under magnetic field conditions and washed twice with the same medium as the cell culture to obtain WT1-9E TCR-T effector cells of this example.
2. Detection of response of effector cells to target cells harboring WT1(414-423) polypeptide
Then, the level of secreted IFN-. gamma.was measured by the action of different T cell detection methods (IFN-. gamma. -ELISPOT and IFN-. gamma. -ELISA) on WT1-9E TCR-T cells and PANC1 cells (obtained from the institute of basic medicine of the Chinese medical sciences) carrying WT1 (414-.
WT1-9E TCR-T cells prepared using peripheral blood-derived T cells from three volunteers were mixed with PANC1 cells or PANC1 target cells to which WT1(414-423) polypeptide (10. mu.g/mL final concentration) was added at a ratio of 1: 1. In addition, WT1-9E TCR-T cells prepared using PBMCs from three volunteers were incubated with PBMC primary cells as Antigen Presenting Cells (APC) in a 1:1 ratio and WT1(414-423) polypeptide (10. mu.g/mL final concentration) was added.
Meanwhile, for PANC1 cells, the addition of NY-ESO-1 polypeptide (shown in SEQ ID NO: 18) was used as a negative control in place of the WT1(414-423) polypeptide.
Note that, for PANC1 cells, WT1(414-423) polypeptide and NY-ESO-1 polypeptide were added to each well at a final concentration of 10. mu.g/mL, respectively, and added to the medium of target cells before mixing them with effector cells.
The mixture was mixed in a volume of 1X10 to 100. mu.L 5 Cell number/well was added to ELISPOT plates pre-coated with anti-IFN- γ antibody for ELISPOT detection. Effector cells and target cells are mixed and then placed at 37 ℃ with 100% humidity and 5% CO 2 The cell culture chamber was continued for 18 hours.
The results show that WT1-9E TCR-T cells prepared from three volunteer-derived PBMCs all produced IFN-. gamma.upon incubation with PANC1 target cells presenting WT1(414-423) polypeptide, FIG. 4b is a photograph of ELISPOT assay results, and FIG. 4c is a statistical plot of the number of corresponding ELISPOT reaction spots in FIG. 4 b. Therefore, ELISPOT experiments show that WT1-9E TCR-T cells can specifically interact with target cells expressing WT1(414-423) to form IFN-gamma-secreting spots.
In conclusion, it was demonstrated that WT1-9E TCR-T cells are capable of specifically recognizing target cells presenting WT1(414-423)/HLA-A11 and are capable of specifically secreting the cytokine IFN-. gamma.. In view of the potential cytotoxic effects of CD8+ T cells on target cells, it was concluded that the WT1-9E TCR-T cells of the invention have potential target cell killing activity and tumor therapeutic value.
Example 4 analysis of the binding characteristics of WT1-9E TCR to WT1(414-423)/HLA-A11
In order to accurately determine the binding properties and affinity of WT1-9E TCR to WT1(414-423)/HLA-A11 complex protein, the inventors further examined the affinity at the protein level using Surface Plasmon Resonance (SPR). Since the functional domain of WT1-9E TCR is the extracellular domain and the extracellular domain, which does not contain the transmembrane region, is a soluble protein, the extracellular domain of WT1-9E TCR was synthesized. The specific operation is as follows.
TCR protein expression and purification
The extracellular region genes of WT1-9E TCR alpha chain and beta chain were optimized according to prokaryotic codons to synthesize DNA sequences of extracellular regions of WT1-9E TCR chimeric alpha chain and beta chain, respectively (WT 1-9E: alpha chain, SEQ ID NO: 20; beta chain, SEQ ID NO:21), wherein WT1-9E TCR has an alpha chain variable region as shown in the sequence of SEQ ID NO:1 and a beta chain variable region as shown in the sequence of SEQ ID NO: 5. Introducing enzyme cutting sites Nde I and Xho I respectively, wherein the enzyme cutting site Nde I is positioned at the 5 'end of the sequence, and the enzyme cutting site Xho I is positioned at the 3' end of the sequence. The DNA sequences of the extracellular regions of WT1-9E TCR alpha chain and beta chain synthesized were cloned into expression vector pET21 a (Invitrogen corporation) using restriction enzyme sites Nde I and Xho I, respectively, and prokaryotic recombinant expression plasmids of extracellular region proteins of WT1-9E TCR alpha chain and beta chain were constructed.
The expression plasmid is transferred into E.coli BL21(DE3) competent cells by a heat shock method, IPTG is added for induction expression, and the extracellular region protein of WT1-9E TCR alpha chain and beta chain in an inclusion body state is obtained.
6mL of inclusion bodies of the extracellular regions of WT1-9E TCR alpha chain and beta chain (each inclusion body dissolved at 30mg/mL in a solution containing 6M Gua-HCl, 50mM Tris pH8.0, 100mM NaCl, 10mM EDTA and 10mM DTT) were dropped in 1L of a prepared renaturation solution (5M urea; 20mM Tris-HCl; 400mM L-arginine; EDTA 2 mM; GSH/GSSG 5mM/1mM) at a mass ratio of 2:1 in two drops of 3mL each with a minimum interval of 8 hours, followed by concentration in a concentration cup (Millipore Co.).
After concentration, the mixture was placed in 4L of deionized water and 4L of 10mM Tris, pH8.0 dialysate in this order, and each dialyzed for 24 hours. Then, the protein was preliminarily purified by Source 15Q ion exchange chromatography, and the objective protein was identified by SDS-PAGE.
Specifically, the target protein was concentrated in a concentration cup (Millipore Co., Ltd.), and the solution was exchanged with 20mM Tris-HCl, 150mM NaCl, pH8.0 buffer, and after concentration, the protein was purified by Superdex200 pg molecular sieves (GE Healthcare) to obtain about 2-3mg of WT1-9E TCR protein, and the target protein was detected by reducing (containing Dithiothreitol (DTT)) and non-reducing (containing no Dithiothreitol (DTT)) SDS-PAGE (FIG. 5).
The results indicated that WT1-9E TCR molecular sieve chromatography gave a peak of protein interest at 15mL elution volume and that WT1-9E was identified by SDS-PAGE as an α β heterodimer with a band size of about 52kD on SDS-PAGE under non-reducing conditions without DTT and opened α and β inter-chain disulfide bonds on SDS-PAGE under reducing conditions with DTT, showing bands of sizes around 24kD and 28kD, respectively (FIGS. 5a and b).
SPR detection assay
WT1-9E TCR protein prepared by performing an in vitro renaturation experiment as in example 2, and biotinylated WT1(414-423)/HLA-A11 complex protein prepared in example 2 were exchanged into SPR buffer (10mM HEPES-HCl, 150mM Na-Cl, 0.005% Tween-20, pH 7.4). WT1(414-423)/HLA-A11 complex protein was diluted to 20. mu.g/mL and fixed on an SA chip (GE Health), after which gradient (0. mu.M, 0.39. mu.M, 0.78. mu.M, 1.56. mu.M, 3.125. mu.M, 6.25. mu.M, 12.5. mu.M, 25. mu.M and 50. mu.M) diluted WT1-9E TCR protein was flowed through each channel of the SA chip, respectively, and binding kinetic parameters were analyzed using BIA evaluation software and affinity constants were calculated. The affinity of WT1-9E TCR to WT1(414-423)/HLA-A11 complex protein was examined (FIG. 5 c).
The results show that WT1-9E TCR binding WT1(414-423)/HLA-A11 exhibited a fast association and dissociation pattern with a binding affinity (KD) of 11.9. mu.M (FIG. 5d), as can be seen from the curves in FIG. 5.
Therefore, WT1-9E TCR has good binding property and affinity, and it can be speculated that WT1-9E TCR can generate IFN-gamma to tumor cells carrying WT1(414-423) when used in anti-tumor therapy, thereby killing the tumor cells and achieving the effect of treating tumor.
Reference to the literature
[1].June C H,Sadelain M.Chimeric antigen receptor therapy.N Engl J Med,2018,379:64-73
[2].Thomas R,Al-Khadairi G,Roelands J,et al.NY-ESO-1based immunotherapy of cancer:current perspectives.Front Immunol,2018,9:947
[3].Byrne M,Oluwole O O,Savani B,et al.Understanding and managing large B cell lymphoma relapses after chimeric antigen receptor T cell therapy.Biol Blood Marrow Transplant,2019,25:e344-e351
[4].Shah N N,Fry T J.Mechanisms of resistance to CAR T cell therapy.Nat Rev Clin Oncol,2019,16:372-385
[5].Ikeda H.T-cell adoptive immunotherapy using tumor-infiltrating T cells and genetically engineered TCR-T cells.Int Immunol,2016,28:349-353
[6].Artibani M,Sims A H,Slight J,et al.WT1 expression in breast cancer disrupts the epithelial/mesenchymal balance of tumour cells and correlates with the metabolic response to docetaxel.Sci Rep,2017,7:45255
[7].Menssen H D,Renkl H J,Rodeck U,et al.Presence of Wilms'tumor gene(wt1)transcripts and the WT1 nuclear protein in the majority of human acute leukemias.Leukemia,1995,9:1060-1067
[8].Haber D A,Buckler AJ,Glaser T,et al.An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms'tumor.Cell,1990,61:1257-1269
[9].Huang A,Campbell C E,Bonetta L,et al.Tissue,developmental,and tumor-specific expression of divergent transcripts in Wilms tumor.Science,1990,250:991-994
[10].Little M,Wells C.A clinical overview of WT1 gene mutations.Hum Mutat,1997,9:209-225
[11].Iii F J R.The WT1 Wilms tumor gene product:a developmentally regulated transcription factor in the kidney that functions as a tumor suppressor.FASEB J,1993,7:896-903
[12].Nakatsuka S-i,Oji Y,Horiuchi T,et al.Immunohistochemical detection of WT1 protein in a variety of cancer cells.Mod Pathol,2006,19:804-814
[13].Sugiyama H.WT1(Wilms'tumor gene 1):biology and cancer immunotherapy.Jpn J Clin Oncol,2010,40:377-387
[14].Yang L,Han Y,Saurez Saiz F,et al.A tumor suppressor and oncogene:the WT1 story.Leukemia,2007,21:868-876
[15].Mayo M W,Wang C-Y,Drouin S S,et al.WT1 modulates apoptosis by transcriptionally upregulating the bcl-2proto-oncogene.EMBO J,1999,18:3990-4003
[16].Brett A,Pandey S,Fraizer G.The Wilms'tumor gene(WT1)regulates E-cadherin expression and migration of prostate cancer cells.Mol Cancer,2013,12:3
[17].Cheever M A,Allison J P,Ferris A S,et al.The prioritization of cancer antigens:a national cancer institute pilot project for the acceleration of translational research.Clin Cancer Res,2009,15:5323-5337
[18].
Figure BDA0003612016820000181
V,Scheibenbogen C,Thiel E,et al.Complete remission in a patient with recurrent acute myeloid leukemia induced by vaccination with WT1 peptide in the absence of hematological or renal toxicity.Leukemia,2004,18:165-166
[19].Maslak P G,Dao T,Krug L M,et al.Vaccination with synthetic analog peptides derived from WT1 oncoprotein induces T-cell responses in patients with complete remission from acute myeloid leukemia.Blood,2010,116:171-179
[20].Nguyen T H,Tan A C,Xiang S D,et al.Understanding CD8(+)T-cell responses toward the native and alternate HLA-A*02:01-restricted WT1 epitope.Clin Transl Immunology,2017,6:e134
[21].Ruggiero E,Carnevale E,Prodeus A,et al.CRISPR-based gene disruption and integration of high-avidity,WT1-specific T cell receptors improve antitumor T cell function.Sci Transl Med,2022,14:eabg8027
[22].Yasukawa M,Fujiwara H,Ochi T,et al.Clinical efficacy of WT1 peptide vaccination in patients with acute myelogenous leukemia and myelodysplastic syndrome.Am J Hematol,2009,84:314-315
[23].Tsuboi A,Oka Y,Kyo T,et al.Long-term WT1 peptide vaccination for patients with acute myeloid leukemia with minimal residual disease.Leukemia,2012,26:1410-1413
[24].Di Stasi A,Jimenez A M,Minagawa K,et al.Review of the results of WT1 peptide vaccination strategies for myelodysplastic syndromes and acute myeloid leukemia from nine different studies.Front Immunol,2015,6:36
[25].Maslak P G,Dao T,Bernal Y,et al.Phase 2 trial of a multivalent WT1 peptide vaccine(galinpepimut-S)in acute myeloid leukemia.Blood Adv,2018,2:224-234
[26].Crowther M D,Svane I M,Met
Figure BDA0003612016820000191
T-cell gene therapy in cancer immunotherapy:why it is no longer just CARs on the road.Cells,2020,9:1588
Sequence listing
1 (> SEQ ID NO:1(9E TCR. alpha. chain variable region amino acid sequence)
MKSLSVSLVVLWLQLNWVNSQQKVQQSPESLIVPEGAMTSLNCTFSDSASQYFAWYRQHSGKAPKALMSIFSNGEKEEGRFTIHLNKASLHFSLHIRDSQPSDSALYLCAVLLSGSFNKLTFGAGTRLAVCP
2(9E TCR alpha chain CDR1 amino acid sequence)
DSASQY
3(9E TCR alpha chain CDR2 amino acid sequence)
IFSNGE
4(9E TCR alpha chain CDR3 amino acid sequence)
AVLLSGSFNKLT
5(9E TCR beta variable region amino acid sequence)
MGSRLFLVLSLLCTKHMEAAVTQSPRNKVTVTGGNVTLSCCQTNSHNYMYWYRQDTGHGLRLIHYSYGAGNLQIGDVPDGYKATRTTQEDFFLLLELASPSQTSLYFCASSDGDSNYAEQFFGPGTRLTVL
6(9E TCR beta chain CDR1)
NSHNY
7(9E TCR beta chain CDR2)
SYGAGN
8(9E TCR beta chain CDR3)
ASSDGDSNYAEQF
SEQ ID NO 9(9E chimeric TCR. alpha. chain amino acid sequence)
MKSLSVSLVVLWLQLNWVNSQQKVQQSPESLIVPEGAMTSLNCTFSDSASQYFAWYRQHSGKAPKALMSIFSNGEKEEGRFTIHLNKASLHFSLHIRDSQPSDSALYLCAVLLSGSFNKLTFGAGTRLAVCPYIQNPDPAVYQLRDSKSSDKSVCLFTDFDSQTNVSQSKDSDVYITDKCVLDMRSMDFKSNSAVAWSNKSDFACANAFNNSIIPEDTFFPSPESSCDVKLVEKSFETDTNLNFQNLSVIGFRILLLKVAGFNLLMTLRLWSS
SEQ ID NO 10(9E chimeric TCR. alpha. chain nucleotide sequence)
ATGAAATCCTTGAGTGTTTCCCTAGTGGTCCTGTGGCTCCAGTTAAACTGGGTGAACAGCCAGCAGAAGGTGCAGCAGAGCCCAGAATCCCTCATTGTCCCAGAGGGAGCCATGACCTCTCTCAACTGCACTTTCAGCGACAGTGCTTCTCAGTATTTTGCATGGTACAGACAGCATTCTGGGAAAGCCCCCAAGGCACTGATGTCCATCTTCTCCAATGGTGAAAAAGAAGAAGGCAGATTCACAATTCACCTCAATAAAGCCAGTCTGCATTTCTCGCTACACATCAGAGACTCCCAGCCCAGTGACTCTGCTCTCTACCTCTGTGCAGTGTTGTTATCTGGTAGCTTCAATAAGTTGACCTTTGGAGCAGGGACCAGACTGGCTGTGTGCCCATATATCCAGAACCCTGACCCTGCCGTGTACCAGCTGAGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTCACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAGGATTCTGATGTGTATATCACAGACAAATGCGTGCTAGACATGAGGTCTATGGACTTCAAGAGCAACAGTGCTGTGGCCTGGAGCAACAAATCTGACTTTGCATGTGCAAACGCCTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCCCCAGCCCAGAAAGTTCCTGTGATGTCAAGCTGGTCGAGAAAAGCTTTGAAACAGATACGAACCTAAACTTTCAAAACCTGTCAGTGATTGGGTTCCGAATCCTCCTCCTGAAAGTGGCCGGGTTTAATCTGCTCATGACGCTGCGGCTGTGGTCCAGC
< SEQ ID NO:11(HLA-A11 amino acid sequence)
GSHSMRYFYTSVSRPGRGEPRFIAVGYVDDTQFVRFDSDAASQRMEPRAPWIEQEGPEYWDQETRNVKAQSQTDRVDLGTLRGYYNQSEDGSHTIQIMYGCDVGPDGRFLRGYRQDAYDGKDYIALNEDLRSWTAADMAAQITKRKWEAAHAAEQQRAYLEGRCVEWLRRYLENGKETLQRTDPPKTHMTHHPISDHEATLRCWALGFYPAEITLTWQRDGEDQTQDTELVETRPAGDGTFQKWAAVVVPSGQEQRYTCHVQHEGLPKPLTLRWELGSGGGLNDIFEAQKIEWH
12(HLA-A11 heavy chain prokaryotic codon optimized nucleotide sequence)
ATGGGCTCTCACTCCATGAGGTATTTCTACACCTCCGTGTCCCGGCCCGGCCGCGGGGAGCCCCGCTTCATCGCCGTGGGCTACGTGGACGACACGCAGTTCGTGCGGTTCGACAGCGACGCCGCGAGCCAGAGGATGGAGCCGCGGGCGCCGTGGATAGAGCAGGAGGGGCCGGAGTATTGGGACCAGGAGACACGGAATGTGAAGGCCCAGTCACAGACTGACCGAGTGGACCTGGGGACCCTGCGCGGCTACTACAACCAGAGCGAGGACGGTTCTCACACCATCCAGATAATGTATGGCTGCGACGTGGGGCCGGACGGGCGCTTCCTCCGCGGGTACCGGCAGGACGCCTACGACGGCAAGGATTACATCGCCCTGAACGAGGACCTGCGCTCTTGGACCGCGGCGGACATGGCAGCTCAGATCACCAAGCGCAAGTGGGAGGCGGCCCATGCGGCGGAGCAGCAGAGAGCCTACCTGGAGGGCCGGTGCGTGGAGTGGCTCCGCAGATACCTGGAGAACGGGAAGGAGACGCTGCAGCGCACGGACCCCCCCAAGACACATATGACCCACCACCCCATCTCTGACCATGAGGCCACCCTGAGGTGCTGGGCCCTGGGCTTCTACCCTGCGGAGATCACACTGACCTGGCAGCGGGATGGGGAGGACCAGACCCAGGACACGGAGCTCGTGGAGACCAGGCCTGCAGGGGATGGAACCTTCCAGAAGTGGGCGGCTGTGGTGGTGCCTTCTGGACAGGAGCAGAGATACACCTGCCATGTGCAGCATGAGGGTTTGCCCAAGCCCCTCACCCTGAGATGGGAGCTGGGATCCGGTGGTGGTCTGAACGATATTTTTGAAGCTCAGAAAATCGAATGGCATTAA
>SEQ ID NO:13(β 2 m amino acid sequence)
IQRTPKIQVYSRHPAENGKSNFLNCYVSGFHPSDIEVDLLKNGERIEKVEHSDLSFSKDWSFYLLYYTEFTPTEKDEYACRVNHVTLSQPKIVKWDRDM
>SEQ ID NO:14(β 2 m prokaryotic codon optimized nucleotide sequence)
ATGATCCAGCGTACTCCAAAGATTCAGGTTTACTCACGTCATCCAGCAGAGAATGGAAAGTCAAATTTCCTGAATTGCTATGTGTCTGGGTTTCATCCATCCGACATTGAAGTTGACTTACTGAAGAATGGAGAGAGAATTGAAAAAGTGGAGCATTCAGACTTGTCTTTCAGCAAGGACTGGTCTTTCTATCTCTTGTACTACACTGAATTCACCCCCACTGAAAAAGATGAGTATGCCTGCCGTGTGAACCATGTGACTTTGTCACAGCCCAAGATAGTTAAGTGGGATCGAGACATGTAA
SEQ ID NO 15(9E chimeric TCR. beta. chain amino acid sequence)
MGSRLFLVLSLLCTKHMEAAVTQSPRNKVTVTGGNVTLSCCQTNSHNYMYWYRQDTGHGLRLIHYSYGAGNLQIGDVPDGYKATRTTQEDFFLLLELASPSQTSLYFCASSDGDSNYAEQFFGPGTRLTVLEDLKNVFPPEVAVFEPSEAEISHTQKATLVCLATGFYPDHVELSWWVNGKEVHSGVCTDPQPLKEQPALNDSRYCLSSRLRVSATFWQNPRNHFRCQVQFYGLSENDEWTQDRAKPVTQIVSAEAWGRADCGFTSESYQQGVLSATILYEILLGKATLYAVLVSALVLMAMVKRKDSRG
SEQ ID NO 16(9E chimeric TCR beta chain nucleotide sequence)
ATGGGCTCCAGGCTCTTTCTGGTCTTGAGCCTCCTGTGTACAAAACACATGGAGGCTGCAGTCACCCAAAGCCCTAGAAACAAGGTGACAGTAACAGGAGGAAACGTGACATTGAGCTGTTGCCAGACTAATAGCCACAACTACATGTACTGGTATCGGCAGGACACTGGGCATGGGCTGAGGCTGATCCATTACTCATATGGTGCTGGCAACCTTCAAATAGGAGATGTCCCTGATGGGTACAAGGCCACCAGAACAACGCAAGAAGACTTCTTCCTCCTGCTGGAATTGGCTTCTCCCTCTCAGACATCTTTGTACTTCTGTGCCAGCAGTGATGGGGACAGCAACTATGCTGAGCAGTTCTTCGGACCAGGGACACGACTCACCGTCCTAGAGGACCTGAAAAACGTGTTCCCACCCGAGGTCGCTGTGTTTGAGCCATCAGAAGCAGAGATCTCCCACACCCAAAAGGCCACACTGGTGTGCCTGGCCACAGGCTTCTACCCCGACCACGTGGAGCTGAGCTGGTGGGTGAATGGGAAGGAGGTGCACAGTGGGGTCTGCACAGACCCGCAGCCCCTCAAGGAGCAGCCCGCCCTCAATGACTCCAGATACTGCCTGAGCAGCCGCCTGAGGGTCTCGGCCACCTTCTGGCAGAACCCCCGCAACCACTTCCGCTGTCAAGTCCAGTTCTACGGGCTCTCGGAGAATGACGAGTGGACCCAGGATAGGGCCAAACCTGTCACCCAGATCGTCAGCGCCGAGGCCTGGGGTAGAGCAGACTGTGGCTTCACCTCCGAGTCTTACCAGCAAGGGGTCCTGTCTGCCACCATCCTCTATGAGATCTTGCTAGGGAAGGCCACCTTGTATGCCGTGCTGGTCAGTGCCCTCGTGCTGATGGCCATGGTCAAGAGAAAGGATTCCAGAGGC
17 (T cell epitope amino acid sequence of WT1)
FSCRWPSCQK
18 (control NY-ESO-1 polypeptide amino acid sequence)
SLLMWITQC
19 (amino acid sequence of the P2A sequence)
ATNFSLLKQAGDVEENPGP
20(9E TCR alpha chain extracellular region prokaryotic codon optimized nucleotide sequence)
CAGCAGAAAGTGCAGCAGAGTCCGGAAAGCCTGATTGTGCCGGAAGGCGCGATGACGAGCCTGAACTGCACCTTTAGCGACAGCGCGAGTCAGTATTTTGCGTGGTATCGTCAGCATAGCGGCAAAGCGCCGAAAGCGCTGATGAGCATTTTTAGCAACGGCGAAAAAGAAGAAGGCCGCTTTACCATTCATCTGAACAAAGCGAGCCTGCATTTTAGCCTGCATATTCGCGATAGTCAGCCGAGCGATAGCGCGCTGTATCTGTGCGCGGTGCTGCTGAGCGGCAGCTTTAACAAACTGACCTTTGGCGCGGGCACCCGCCTGGCGGTGTGCCCGTATATTCAGAACCCGGATCCGGCGGTGTATCAGCTGCGCGATAGCAAAAGCAGCGATAAAAGCGTGTGCCTGTTTACCGATTTTGATAGTCAGACCAACGTGAGTCAGAGCAAAGATAGCGATGTGTATATTACCGATAAATGCGTGCTGGATATGCGCAGCATGGATTTTAAAAGCAACAGCGCGGTGGCGTGGAGCAACAAAAGCGATTTTGCGTGCGCGAACGCGTTTAACAACAGCATTATTCCGGAAGATACCTTTTTTCCGTCTCCGGAAAGCAGC
21(9E TCR beta chain extracellular region prokaryotic codon optimized nucleotide sequence)
GAAGCGGCTGTGACGCAGAGCCCGCGCAACAAAGTGACCGTGACCGGCGGCAACGTGACCCTGAGCTGCTGTCAGACCAACAGCCATAACTATATGTATTGGTATCGCCAAGATACCGGCCATGGCCTGCGCCTGATTCATTATAGCTATGGCGCGGGCAACCTGCAGATTGGCGATGTGCCGGATGGCTATAAAGCGACCCGCACCACCCAAGAAGATTTTTTTCTGCTGCTGGAACTGGCGAGCCCGAGTCAGACGAGCCTGTATTTTTGCGCGAGCAGCGATGGCGATAGCAACTATGCGGAACAGTTTTTTGGCCCGGGCACCCGCCTGACCGTGCTGGAAGATCTGAAAAACGTGTTTCCGCCGGAAGTGGCGGTGTTTGAACCGAGCGAAGCGGAAATTAGCCATACGCAGAAAGCGACCCTGGTGTGCCTGGCGACCGGCTTTTATCCGGATCATGTGGAACTGAGCTGGTGGGTGAACGGCAAAGAAGTGCATAGCGGCGTGTGCACCGATCCGCAGCCGCTGAAAGAACAGCCGGCGCTGAACGATAGCCGCTATGCGCTGAGCAGCCGCCTGCGCGTGAGCGCGACCTTTTGGCAAGATCCGCGCAACCATTTTCGCTGCCAAGTGCAGTTTTATGGCCTGAGCGAAAACGATGAATGGACCCAAGATCGCGCGAAACCGGTGACGCAGATTGTGAGCGCGGAAGCGTGGGGCCGCGCGGAT
22 (amino acid sequence of a biotin-specific binding polypeptide)
GLNDIFEAQKIEWH

Claims (11)

  1. A T Cell Receptor (TCR) or an antigen binding fragment thereof, which TCR or antigen binding fragment thereof is capable of binding to a T cell epitope of the tumour associated antigen WT1 and to the HLA-A11 complex, and which TCR comprises an alpha chain variable region and a beta chain variable region,
    the TCR, or antigen-binding fragment thereof, comprises the following alpha chain Complementarity Determining Regions (CDRs) and beta chain Complementarity Determining Regions (CDRs):
    an alpha chain complementarity determining region CDR1 shown in SEQ ID NO. 2;
    an alpha chain complementarity determining region CDR2 shown in SEQ ID NO. 3;
    an alpha chain complementarity determining region CDR3 shown in SEQ ID NO. 4; and
    a beta strand complementarity determining region CDR1 shown in SEQ ID NO. 6;
    the beta strand complementarity determining region CDR2 shown in SEQ ID NO. 7;
    the beta strand complementarity determining region CDR3 shown in SEQ ID NO. 8,
    wherein the amino acid sequence of the T cell epitope is shown as SEQ ID NO. 17.
  2. 2. The T Cell Receptor (TCR) or antigen-binding fragment thereof of claim 1, comprising:
    1, and
    the beta chain variable region as shown in the sequence of SEQ ID NO. 5.
  3. 3. A TCR or antigen-binding fragment thereof as claimed in claim 1 or 2 wherein the TCR is a murine TCR, a human murine chimeric TCR or a humanized TCR, preferably having the sequence shown in SEQ ID No. 9 and SEQ ID No. 15.
  4. 4. A polynucleotide encoding a TCR, or antigen-binding fragment thereof, according to any one of claims 1-3.
  5. 5. An expression vector comprising the polynucleotide of claim 4, said expression vector being a lentiviral vector.
  6. 6. A host cell comprising the expression vector of claim 5.
  7. 7. A method of making a TCR, or an antigen-binding fragment thereof, according to any one of claims 1-3, comprising:
    1) culturing the host cell of claim 6;
    2) recovering the TCR, or antigen-binding fragment thereof, of any one of claims 1-3 from the host cell or culture medium thereof.
  8. 8. A pharmaceutical composition comprising a TCR, or antigen-binding fragment thereof, according to any one of claims 1-3, and a pharmaceutically acceptable carrier.
  9. 9. Use of a TCR, or an antigen-binding fragment thereof, according to any one of claims 1 to 3 in the manufacture of a medicament, such as a proteinaceous drug, a protein drug conjugate or a TCR in combination with an antigen, for increasing the level of a cytokine that secretes IFN- γ from T cells.
  10. 10. Use of a TCR, or an antigen-binding fragment thereof, according to any one of claims 1 to 3 in the manufacture of a reagent for the detection of a tumour cell expressing a T cell epitope of the tumour associated antigen WT1, wherein the TCR, or antigen-binding fragment thereof, specifically binds to a T cell epitope of the tumour associated antigen WT1 and HLA-a11, and the sequence of said T cell epitope is set forth in SEQ ID No. 17, or in the manufacture of a reagent for the detection or diagnosis of a tumour.
  11. 11. Use of a TCR, or an antigen-binding fragment thereof, as claimed in any one of claims 1 to 3 in the manufacture of an anti-tumour medicament for the treatment of a patient having a tumour bearing the T cell epitope of the tumour-associated antigen WT1 as set out in SEQ ID NO 17, preferably ovarian cancer.
CN202210435441.9A 2022-04-24 2022-04-24 WT1 antigen specific T cell receptor and anti-tumor application thereof Active CN114853880B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210435441.9A CN114853880B (en) 2022-04-24 2022-04-24 WT1 antigen specific T cell receptor and anti-tumor application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210435441.9A CN114853880B (en) 2022-04-24 2022-04-24 WT1 antigen specific T cell receptor and anti-tumor application thereof

Publications (2)

Publication Number Publication Date
CN114853880A true CN114853880A (en) 2022-08-05
CN114853880B CN114853880B (en) 2023-11-10

Family

ID=82634311

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210435441.9A Active CN114853880B (en) 2022-04-24 2022-04-24 WT1 antigen specific T cell receptor and anti-tumor application thereof

Country Status (1)

Country Link
CN (1) CN114853880B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116139260A (en) * 2023-03-02 2023-05-23 广东龄值生物科技有限公司 WT1 tumor vaccine and preparation method and application thereof
WO2024088383A1 (en) * 2022-10-28 2024-05-02 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Anti-wt1/hla antibodies and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110662760A (en) * 2017-05-12 2020-01-07 奥古斯塔大学研究所公司 Human alpha-fetoprotein specific T cell receptor and uses thereof
WO2022068850A1 (en) * 2020-09-29 2022-04-07 中国科学院微生物研究所 Screening and antitumor use of kras mutation specific t cell receptor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110662760A (en) * 2017-05-12 2020-01-07 奥古斯塔大学研究所公司 Human alpha-fetoprotein specific T cell receptor and uses thereof
WO2022068850A1 (en) * 2020-09-29 2022-04-07 中国科学院微生物研究所 Screening and antitumor use of kras mutation specific t cell receptor

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024088383A1 (en) * 2022-10-28 2024-05-02 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Anti-wt1/hla antibodies and uses thereof
CN116139260A (en) * 2023-03-02 2023-05-23 广东龄值生物科技有限公司 WT1 tumor vaccine and preparation method and application thereof

Also Published As

Publication number Publication date
CN114853880B (en) 2023-11-10

Similar Documents

Publication Publication Date Title
CN112300269B (en) KRAS mutation specific T cell receptor screening and anti-tumor application
AU2017235467B2 (en) Transfected T-cells and T-cell receptors for use in immunotherapy against cancers
US11945854B2 (en) Transfected T-cells and T-cell receptors for use in immunotherapy against cancers
CN114127091B (en) Tumor neoantigen polypeptide and application thereof
KR102171794B1 (en) Method for activating helper t cell
CA3016410A1 (en) Peptides and combination of peptides for use in immunotherapy against non-small cell lung cancer and other cancers
CN114853880B (en) WT1 antigen specific T cell receptor and anti-tumor application thereof
CN112480239B (en) Human papilloma virus specific T cell receptor and anti-tumor application thereof
CN112375136B (en) NY-ESO-1 specific T cell receptor screening and anti-tumor application thereof
CN111690051B (en) Specific T cell receptor targeting NY-ESO-1(157-165) epitope and anti-tumor application
CN117552115B (en) Universal antigen peptide library for inducing tumor specific immune response and application thereof
US20220281948A1 (en) Mhc class ii molecules and methods of use thereof
CN115073584A (en) TCR for specifically recognizing PRAME antigen peptide and application thereof
EA043021B1 (en) TRANSFECTED T-CELLS AND T-CELL RECEPTORS FOR USE IN CANCER IMMUNOTHERAPY
NZ747154B2 (en) Transfected t-cells and t-cell receptors for use in immunotherapy against cancers

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant