CN109251243B - T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor - Google Patents

T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor Download PDF

Info

Publication number
CN109251243B
CN109251243B CN201710570782.6A CN201710570782A CN109251243B CN 109251243 B CN109251243 B CN 109251243B CN 201710570782 A CN201710570782 A CN 201710570782A CN 109251243 B CN109251243 B CN 109251243B
Authority
CN
China
Prior art keywords
tcr
chain
seq
ser
variable domain
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.)
Active
Application number
CN201710570782.6A
Other languages
Chinese (zh)
Other versions
CN109251243A (en
Inventor
李懿
陈安安
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangzhou Institute of Biomedicine and Health of CAS
Original Assignee
Guangzhou Institute of Biomedicine and Health 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 Guangzhou Institute of Biomedicine and Health of CAS filed Critical Guangzhou Institute of Biomedicine and Health of CAS
Priority to CN201710570782.6A priority Critical patent/CN109251243B/en
Publication of CN109251243A publication Critical patent/CN109251243A/en
Application granted granted Critical
Publication of CN109251243B publication Critical patent/CN109251243B/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
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • A61K38/00Medicinal preparations containing peptides
    • 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)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Plant Pathology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention provides a T Cell Receptor (TCR) capable of specifically binding to short peptide VFSTVPPAFI derived from SAGE1 antigen, which antigen short peptide VFSTVPPAFI can form a complex with HLA a2402 and be presented together on the cell surface. The invention also provides nucleic acid molecules encoding the TCRs and vectors comprising the nucleic acid molecules. In addition, the invention provides cells that transduce a TCR of the invention.

Description

T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor
Technical Field
The invention relates to a TCR capable of recognizing short peptide derived from SAGE1 antigen, SAGE1 specific T cells obtained by transducing the TCR, and application of the T cells in preventing and treating SAGE1 related diseases.
Background
SAGE1, an endogenous tumor antigen, was degraded into small polypeptides after intracellular production and was presented on the cell surface as a complex with MHC (major histocompatibility Complex) molecules. Studies showed VFSTVPPAFI to be a short peptide derived from SAGE 1. SAGE1 antigen was expressed in tumor tissues such as melanoma, bladder Cancer, liver Cancer, epidermoid carcinoma, non-small cell lung Cancer and squamous cell carcinoma, but not in most normal tissues except testis (Martelange V1, De Smet C, De Plaen E, Lurqin C, Boon T. Cancer Res.2000; 60(14): 3848-55; Atanofovic D, et., Cancer biol. Ther.2006; 5(9): 1218-25). For the treatment of the above diseases, chemotherapy, radiotherapy and the like can be used, but both of them cause damages to normal cells themselves.
T cell adoptive immunotherapy is the transfer of reactive T cells specific for a target cell antigen into a patient to act on the target cell. The T Cell Receptor (TCR) is a membrane protein on the surface of T cells that recognizes a corresponding short peptide antigen on the surface of a target cell. In the immune system, the direct physical contact between T cells and Antigen Presenting Cells (APC) is initiated by the binding of antigen short peptide specific TCR and short peptide-major histocompatibility complex (pMHC complex), and then other cell membrane surface molecules of the T cells and APC interact to cause a series of subsequent cell signaling and other physiological reactions, so that T cells with different antigen specificities exert immune effects on their target cells. Therefore, those skilled in the art have focused on isolating a TCR specific for the SAGE1 antigen short peptide to be effective, or transducing T cells with the TCR to obtain T cells specific for the SAGE1 antigen short peptide, thereby making them effective in cellular immunotherapy.
Disclosure of Invention
The invention aims to provide a T cell receptor for recognizing SAGE1 antigen short peptide.
In a first aspect of the invention, there is provided a T Cell Receptor (TCR) capable of binding to the VFSTVPPAFI-HLA a2402 complex.
In another preferred embodiment, the TCR comprises a TCR alpha chain variable domain and a TCR beta chain variable domain, the amino acid sequence of CDR3 of the TCR alpha chain variable domain is CAVSERARLMF (SEQ ID NO: 12); and/or the amino acid sequence of CDR3 of the variable domain of the TCR beta chain is CASSLVGFTEAFF (SEQ ID NO: 15).
In another preferred embodiment, the 3 Complementarity Determining Regions (CDRs) of the TCR α chain variable domain are:
αCDR1-SSVPPY(SEQ ID NO:10)
αCDR2-YTSAATLV(SEQ ID NO:11)
alpha CDR3-CAVSERARLMF (SEQ ID NO: 12); and/or
The 3 complementarity determining regions of the TCR β chain variable domain are:
βCDR1-SGHDT(SEQ ID NO:13)
βCDR2-YYEEEE(SEQ ID NO:14)
βCDR3-CASSLVGFTEAFF(SEQ ID NO:15)。
in another preferred embodiment, the TCR comprises a TCR alpha chain variable domain which is an amino acid sequence having at least 90% sequence identity to SEQ ID No. 1, and a TCR beta chain variable domain; and/or the TCR β chain variable domain is identical to SEQ ID NO:5 an amino acid sequence having at least 90% sequence identity.
In another preferred embodiment, the TCR comprises the alpha chain variable domain amino acid sequence SEQ ID NO 1.
In another preferred embodiment, the TCR comprises the beta chain variable domain amino acid sequence SEQ ID NO 5.
In another preferred embodiment, the TCR is an α β heterodimer comprising a TCR α chain constant region TRAC 01 and a TCR β chain constant region TRBC1 01 or TRBC2 01.
In another preferred embodiment, the α chain amino acid sequence of the TCR is SEQ ID NO:3 and/or the beta chain amino acid sequence of the TCR is SEQ ID NO 7.
In another preferred embodiment, the TCR is soluble.
In another preferred embodiment, the TCR is single chain.
In another preferred embodiment, the TCR is formed by linking an α chain variable domain to a β chain variable domain via a peptide linker.
In another preferred embodiment, the TCR has one or more mutations in amino acid 11, 13, 19, 21, 53, 76, 89, 91, or 94 of the α chain variable region, and/or in the penultimate 3-, 5-, or 7-position of the short peptide amino acid of the α chain J gene; and/or the TCR has one or more mutations in beta chain variable region amino acid 11, 13, 19, 21, 53, 76, 89, 91, or 94 th, and/or beta chain J gene short peptide amino acid penultimate 2,4 or 6 th, wherein the amino acid position numbering is according to the position numbering listed in IMGT (international immunogenetic information system).
In another preferred embodiment, the TCR comprises (a) all or part of a TCR α chain, excluding the transmembrane domain; and (b) all or part of a TCR β chain, excluding the transmembrane domain;
and (a) and (b) each comprise a functional variable domain, or comprise a functional variable domain and at least a portion of the TCR chain constant domain.
In another preferred embodiment, the cysteine residues form an artificial disulfide bond between the alpha and beta chain constant domains of the TCR.
In another preferred embodiment, the cysteine residues forming the artificial disulfide bond in the TCR are substituted at one or more groups of sites selected from the group consisting of:
thr48 and TRBC1 × 01 of TRAC × 01 exon 1 or Ser57 of TRBC2 × 01 exon 1;
thr45 and TRBC1 × 01 of TRAC × 01 exon 1 or Ser77 of TRBC2 × 01 exon 1;
tyr10 and TRBC1 x 01 of exon 1 of TRAC x 01 or Ser17 of exon 1 of TRBC2 x 01;
thr45 and TRBC1 × 01 of TRAC × 01 exon 1 or Asp59 of TRBC2 × 01 exon 1;
ser15 and TRBC1 × 01 of TRAC × 01 exon 1 or Glu15 of TRBC2 × 01 exon 1;
arg53 and TRBC1 × 01 of TRAC × 01 exon 1 or Ser54 of TRBC2 × 01 exon 1;
pro89 and TRBC1 and 01 of exon 1 of TRAC 01 or Ala19 of exon 1 of TRBC2 and 01; and
tyr10 and TRBC1 × 01 of exon 1 of TRAC × 01 or Glu20 of exon 1 of TRBC2 × 01.
In another preferred embodiment, the α chain amino acid sequence of the TCR is SEQ ID NO 26 and/or the β chain amino acid sequence of the TCR is SEQ ID NO 28.
In another preferred embodiment, the TCR comprises an artificial interchain disulfide bond between the α chain variable region and the β chain constant region.
In another preferred embodiment, the cysteine residues that form the artificial interchain disulfide bond in the TCR replace one or more groups of sites selected from the group consisting of:
amino acid 46 of TRAV and amino acid 60 of exon 1 of TRBC1 x 01 or TRBC2 x 01;
amino acid 47 of TRAV and amino acid 61 of exon 1 of TRBC1 x 01 or TRBC2 x 01;
amino acid 46 of TRAV and amino acid 61 of TRBC1 x 01 or TRBC2 x 01 exon 1; or
Amino acid 47 of TRAV and amino acid 60 of exon 1 of TRBC1 x 01 or TRBC2 x 01.
In another preferred embodiment, the TCR comprises an alpha chain variable domain and a beta chain variable domain and all or part of the beta chain constant domain, excluding the transmembrane domain, but which does not comprise an alpha chain constant domain, the alpha chain variable domain of the TCR forming a heterodimer with the beta chain.
In another preferred embodiment, the TCR has a conjugate attached to the C-or N-terminus of the alpha and/or beta chain.
In another preferred embodiment, the conjugate that binds to the T cell receptor is a detectable label, a therapeutic agent, a PK modifying moiety or a combination of any of these. Preferably, the therapeutic agent is an anti-CD 3 antibody.
In a second aspect of the invention, there is provided a multivalent TCR complex comprising at least two TCR molecules, and wherein at least one of the TCR molecules is a TCR according to the first aspect of the invention.
In a third aspect of the invention, there is provided a nucleic acid molecule comprising a nucleic acid sequence encoding a TCR molecule according to the first aspect of the invention, or the complement thereof.
In another preferred embodiment, the nucleic acid molecule comprises the nucleotide sequence encoding the variable domain of the TCR α chain SEQ ID NO: 2.
in another preferred embodiment, the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 6.
in another preferred embodiment, the nucleic acid molecule comprises the nucleotide sequence encoding the TCR α chain SEQ ID NO:4 and/or comprises the nucleotide sequence encoding the TCR β chain SEQ ID NO: 8.
in a fourth aspect of the invention, there is provided a vector comprising a nucleic acid molecule according to the third aspect of the invention; preferably, the vector is a viral vector; more preferably, the vector is a lentiviral vector.
In a fifth aspect of the invention, there is provided an isolated host cell comprising a vector according to the fourth aspect of the invention or a genome into which has been integrated an exogenous nucleic acid molecule according to the third aspect of the invention.
In a sixth aspect of the invention, there is provided a cell which transduces a nucleic acid molecule according to the third aspect of the invention or a vector according to the fourth aspect of the invention; preferably, the cell is a T cell or a stem cell.
In a seventh aspect of the invention, there is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a TCR according to the first aspect of the invention, a TCR complex according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, a vector according to the fourth aspect of the invention, or a cell according to the sixth aspect of the invention.
In an eighth aspect, the invention provides the use of a T cell receptor according to the first aspect of the invention, or a TCR complex according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, a vector according to the fourth aspect of the invention, or a cell according to the sixth aspect of the invention, for the manufacture of a medicament for the treatment of a tumour or an autoimmune disease.
In a ninth aspect of the invention, there is provided a method of treating a disease comprising administering to a subject in need thereof an amount of a T cell receptor according to the first aspect of the invention, or a TCR complex according to the second aspect of the invention, a nucleic acid molecule according to the third aspect of the invention, a vector according to the fourth aspect of the invention, or a cell according to the sixth aspect of the invention, or a pharmaceutical composition according to the seventh aspect of the invention;
preferably, the disease is a tumor, preferably the tumor includes melanoma, and other solid tumors such as gastric cancer, lung cancer, esophageal cancer, bladder cancer, head and neck squamous cell carcinoma, prostate cancer, breast cancer, colon cancer, ovarian cancer, liver cancer, epidermoid carcinoma, non-small cell lung cancer, squamous cell carcinoma, and the like.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1a, FIG. 1b, FIG. 1c, FIG. 1d, FIG. 1e and FIG. 1f are the amino acid sequence of the TCR α chain variable domain, the nucleotide sequence of the TCR α chain variable domain, the amino acid sequence of the TCR α chain, the nucleotide sequence of the TCR α chain, the amino acid sequence of the TCR α chain with leader sequence and the nucleotide sequence of the TCR α chain with leader sequence, respectively.
Fig. 2a, fig. 2b, fig. 2c, fig. 2d, fig. 2e and fig. 2f are a TCR β chain variable domain amino acid sequence, a TCR β chain variable domain nucleotide sequence, a TCR β chain amino acid sequence, a TCR β chain nucleotide sequence, a TCR β chain amino acid sequence with a leader sequence and a TCR β chain nucleotide sequence with a leader sequence, respectively.
FIG. 3 is CD8 of monoclonal cells+And tetramer-PE double positive staining results.
Fig. 4a and 4b are the amino acid and nucleotide sequences, respectively, of a soluble TCR α chain.
Fig. 5a and 5b are the amino acid and nucleotide sequences, respectively, of a soluble TCR β chain.
Figure 6 is a gel diagram of the soluble TCR obtained after purification. The leftmost lane is the molecular weight marker (marker), the middle lane is the non-reducing gel, and the rightmost lane is the reducing gel.
FIG. 7 is a BIAcore kinetic profile of binding of soluble TCRs of the invention to the VFSTVPPAFI-HLA A2402 complex.
FIG. 8 shows that T cells transduced with the TCR of the invention were very responsive to activation of target cells loaded with their specific short peptides, but not to target cells not loaded with the corresponding short peptides and target cells loaded with non-specific short peptides.
FIG. 9 shows that T cells transduced by the TCR of the invention have a significant killing effect on the specific target cell 95D; has no killing effect on non-specific target cells NCI-H1299 and IM 9.
Detailed Description
The present inventors have made extensive and intensive studies to find a TCR capable of specifically binding to SAGE1 antigen short peptide VFSTVPPAFI (SEQ ID NO:9), which antigen short peptide VFSTVPPAFI can form a complex with HLA A2402 and be presented together on the cell surface. The invention also provides nucleic acid molecules encoding the TCRs and vectors comprising the nucleic acid molecules. In addition, the invention provides cells that transduce a TCR of the invention.
Term(s) for
MHC molecules are proteins of the immunoglobulin superfamily, which may be MHC class I or class II molecules. Therefore, it is specific for antigen presentation, different individuals have different MHC, and different short peptides in one protein antigen can be presented on the cell surface of respective APC. Human MHC is commonly referred to as an HLA gene or HLA complex.
The T Cell Receptor (TCR), is the only receptor for a specific antigenic peptide presented on the Major Histocompatibility Complex (MHC). In the immune system, direct physical contact between T cells and Antigen Presenting Cells (APCs) is initiated by the binding of antigen-specific TCRs to pMHC complexes, and then other cell membrane surface molecules of both T cells and APCs interact, which causes a series of subsequent cell signaling and other physiological reactions, thereby allowing T cells of different antigen specificities to exert immune effects on their target cells.
TCRs are cell membrane surface glycoproteins that exist as heterodimers from either the α chain/β chain or the γ chain/δ chain. In 95% of T cells the TCR heterodimer consists of α and β chains, while 5% of T cells have TCRs consisting of γ and δ chains. Native α β heterodimeric TCRs have an α chain and a β chain, which constitute subunits of an α β heterodimeric TCR. Broadly, each of the α and β chains comprises a variable region, a linker region and a constant region, and the β chain also typically contains a short diversity region between the variable region and the linker region, but the diversity region is often considered to be part of the linker region. Each variable region comprises 3 CDRs (complementarity determining regions) CDR1, CDR2 and CDR3, which are chimeric in framework structures (framework regions). The CDR regions determine the binding of the TCR to the pMHC complex, where CDR3 is recombined from variable and connecting regions, referred to as hypervariable regions. The α and β chains of a TCR are generally regarded as having two "domains" each, namely a variable domain and a constant domain, the variable domain being made up of linked variable regions and linking regions. The sequences of TCR constant domains can be found in public databases of the international immunogenetic information system (IMGT), such as the constant domain sequence of the α chain of the TCR molecule is "TRAC 01", the constant domain sequence of the β chain of the TCR molecule is "TRBC 1 01" or "TRBC 2 01". In addition, the α and β chains of the TCR also comprise a transmembrane region and a cytoplasmic region, the cytoplasmic region being very short.
In the present invention, the terms "polypeptide of the invention", "TCR of the invention", "T cell receptor of the invention" are used interchangeably.
Natural interchain disulfide bond and artificial interchain disulfide bond
A set of disulfide bonds, referred to herein as "native interchain disulfide bonds," exist between the C α and C β chains of the membrane proximal region of native TCRs. In the present invention, the artificially introduced interchain covalent disulfide bond whose position is different from that of the natural interchain disulfide bond is referred to as an "artificial interchain disulfide bond".
For convenience of description of the positions of disulfide bonds, the positions of the amino acid sequences of TRAC 01 and TRBC1 × 01 or TRBC2 × 01 are numbered in the order from the N-terminus to the C-terminus, such as in TRBC1 × 01 or TRBC2 × 01, and the 60 th amino acid in the order from the N-terminus to the C-terminus is P (proline), and thus in the present invention it can be described as Pro60 of TRBC1 × 01 or TRBC2 × 01 exon 1, and also as the 60 th amino acid of TRBC1 × 01 or TRBC2 × 01 exon 1, and as in 737bc 3 × 01 or TRBC2 × 01, and the 61 th amino acid in the order from the N-terminus to the C-terminus is Q (glutamine), and thus in the present invention it can be described as TRBC1 × 01 or TRBC 6301 × 01, or TRBC 8501, and similarly as TRBC 8261 or glbc 891. In the present invention, the position numbering of the amino acid sequences of the variable regions TRAV and TRBV follows the position numbering listed in IMGT. If an amino acid in TRAV, the position listed in IMGT is numbered 46, it is described herein as the 46 th amino acid of TRAV, and so on. In the present invention, the sequence position numbers of other amino acids are specifically described.
Detailed Description
TCR molecules
During antigen processing, antigens are degraded within cells and then carried to the cell surface by MHC molecules. T cell receptors are capable of recognizing peptide-MHC complexes on the surface of antigen presenting cells. Accordingly, a first aspect of the invention provides a TCR molecule capable of binding VFSTVPPAFI-HLA a2402 complex. Preferably, the TCR molecule is isolated or purified. The α and β chains of the TCR each have 3 Complementarity Determining Regions (CDRs).
In a preferred embodiment of the invention, the α chain of the TCR comprises CDRs having the amino acid sequence:
αCDR1-SSVPPY(SEQ ID NO:10)
αCDR2-YTSAATLV(SEQ ID NO:11)
alpha CDR3-CAVSERARLMF (SEQ ID NO: 12); and/or
The 3 complementarity determining regions of the TCR β chain variable domain are:
βCDR1-SGHDT(SEQ ID NO:13)
βCDR2-YYEEEE(SEQ ID NO:14)
βCDR3-CASSLVGFTEAFF(SEQ ID NO:15)。
chimeric TCRs can be prepared by embedding the above-described amino acid sequences of the CDR regions of the invention into any suitable framework. One skilled in the art can design or synthesize a TCR molecule with the corresponding function based on the CDR regions disclosed herein, so long as the framework structure is compatible with the CDR regions of the TCR of the invention. Thus, the TCR molecules of the invention are those which comprise the above-described α and/or β chain CDR region sequences and any suitable framework structure. The TCR α chain variable domain of the invention is an amino acid sequence having at least 90%, preferably 95%, more preferably 98% sequence identity to SEQ ID No. 1; and/or the TCR β chain variable domain of the invention is a variant of SEQ ID NO:5, having at least 90%, preferably 95%, more preferably 98% sequence identity.
In a preferred embodiment of the invention, the TCR molecules of the invention are heterodimers consisting of α and β chains. In particular, in one aspect the α chain of the heterodimeric TCR molecules comprises a variable domain and a constant domain, the α chain variable domain amino acid sequence comprising CDR1(SEQ ID NO: 10), CDR2(SEQ ID NO: 11) and CDR3(SEQ ID NO:12) of the above-described α chain. Preferably, the TCR molecule comprises the alpha chain variable domain amino acid sequence SEQ ID NO 1. More preferably, the amino acid sequence of the α chain variable domain of the TCR molecule is SEQ ID NO 1. In another aspect, the β chain of the heterodimeric TCR molecule comprises a variable domain and a constant domain, and the β chain variable domain amino acid sequence comprises CDR1(SEQ ID NO:13), CDR2(SEQ ID NO: 14), and CDR3(SEQ ID NO:15) of the above-described β chain. Preferably, the TCR molecule comprises the beta chain variable domain amino acid sequence SEQ ID NO 5. More preferably, the amino acid sequence of the β chain variable domain of the TCR molecule is SEQ ID NO 5.
In a preferred embodiment of the invention, the TCR molecules of the invention are single chain TCR molecules consisting of part or all of the α chain and/or part or all of the β chain. Single chain TCR molecules are described in Chung et al (1994) Proc. Natl. Acad. Sci. USA 91, 12654-. From the literature, those skilled in the art are readily able to construct single chain TCR molecules comprising the CDRs regions of the invention. In particular, the single chain TCR molecule comprises V α, V β and C β, preferably linked in order from N-terminus to C-terminus.
The alpha chain variable domain amino acid sequence of the single chain TCR molecule comprises the CDR1(SEQ ID NO: 10), CDR2(SEQ ID NO: 11) and CDR3(SEQ ID NO:12) of the alpha chain described above. Preferably, the single chain TCR molecule comprises the alpha chain variable domain amino acid sequence SEQ ID NO 1. More preferably, the α chain variable domain amino acid sequence of the single chain TCR molecule is SEQ ID NO 1. The amino acid sequence of the beta chain variable domain of the single chain TCR molecule comprises the CDR1(SEQ ID NO:13), CDR2(SEQ ID NO: 14) and CDR3(SEQ ID NO:15) of the above-described beta chain. Preferably, the single chain TCR molecule comprises the beta chain variable domain amino acid sequence SEQ ID NO 5. More preferably, the amino acid sequence of the β chain variable domain of the single chain TCR molecule is SEQ ID NO 5.
In a preferred embodiment of the invention, the constant domain of the TCR molecules of the invention is a human constant domain. The human constant domain amino acid sequences are known to those skilled in the art or can be obtained by consulting published databases of relevant books or IMGT (international immunogenetic information system). For example, the constant domain sequence of the α chain of the TCR molecules of the invention can be "TRAC 01", and the constant domain sequence of the β chain of the TCR molecules can be "TRBC 1 01" or "TRBC 2 01". Arg at position 53 of the amino acid sequence given in TRAC 01 of IMGT, here denoted: TRAC × 01 Arg53 of exon 1, and so on. Preferably, the amino acid sequence of the α chain of the TCR molecule of the invention is SEQ ID NO 3 and/or the amino acid sequence of the β chain is SEQ ID NO 7.
Naturally occurring TCRs are membrane proteins that are stabilized by their transmembrane regions. Like immunoglobulins (antibodies) as antigen recognition molecules, TCRs can also be developed for diagnostic and therapeutic applications, where soluble TCR molecules are required. Soluble TCR molecules do not include their transmembrane regions. Soluble TCRs have a wide range of uses, not only for studying the interaction of TCRs with pmhcs, but also as diagnostic tools for detecting infections or as markers for autoimmune diseases. Similarly, soluble TCRs can be used to deliver therapeutic agents (e.g., cytotoxic or immunostimulatory compounds) to cells presenting a specific antigen, and in addition, soluble TCRs can be conjugated to other molecules (e.g., anti-CD 3 antibodies) to redirect T cells to target them to cells presenting a particular antigen. The invention also obtained a soluble TCR specific for the SAGE1 antigen short peptide.
To obtain a soluble TCR, in one aspect, the inventive TCR may be one in which an artificial disulfide bond is introduced between residues of the constant domains of its alpha and beta chains. Cysteine residues form an artificial interchain disulfide bond between the alpha and beta chain constant domains of the TCR. Cysteine residues may be substituted for other amino acid residues at appropriate positions in native TCRs to form artificial interchain disulfide bonds. For example, a disulfide bond is formed by substituting Thr48 of exon 1 of TRAC × 01 and a cysteine residue of Ser57 of exon 1 of TRBC1 × 01 or TRBC2 × 01. Other sites for introducing cysteine residues to form disulfide bonds may also be: thr45 and TRBC1 × 01 of TRAC × 01 exon 1 or Ser77 of TRBC2 × 01 exon 1; tyr10 and TRBC1 x 01 of exon 1 of TRAC x 01 or Ser17 of exon 1 of TRBC2 x 01; thr45 and TRBC1 × 01 of TRAC × 01 exon 1 or Asp59 of TRBC2 × 01 exon 1; ser15 and TRBC1 × 01 of TRAC × 01 exon 1 or Glu15 of TRBC2 × 01 exon 1; arg53 and TRBC1 × 01 of TRAC × 01 exon 1 or Ser54 of TRBC2 × 01 exon 1; pro89 and TRBC1 and 01 of exon 1 of TRAC 01 or Ala19 of exon 1 of TRBC2 and 01; or Tyr10 and TRBC1 and 01 of TRAC 01 exon 1 or Glu20 of TRBC2 and 01 exon 1. I.e., a cysteine residue, in place of any of the above-described alpha and beta chain constant domains. The TCR constant domains of the invention may be truncated at one or more of their C-termini by up to 50, or up to 30, or up to 15, or up to 10, or up to 8 or fewer amino acids, so as not to include a cysteine residue for the purpose of deleting the native disulphide bond, or by mutating the cysteine residue forming the native disulphide bond to another amino acid.
As described above, the TCRs of the invention may comprise artificial disulfide bonds introduced between residues of the constant domains of their alpha and beta chains. It should be noted that the TCRs of the invention may each contain both TRAC constant domain sequences and TRBC1 or TRBC2 constant domain sequences, with or without the artificial disulfide bonds introduced as described above between the constant domains. The TRAC constant domain sequence and TRBC1 or TRBC2 constant domain sequences of the TCR may be linked by the native disulfide bond present in the TCR.
To obtain a soluble TCR, on the other hand, the inventive TCR also comprises a TCR having a mutation in its hydrophobic core region, preferably a mutation that enables an improved stability of the inventive soluble TCR, as described in the patent publication WO 2014/206304. Such TCRs may be mutated at the following variable domain hydrophobic core positions: (alpha and/or beta chain) variable region amino acid positions 11, 13, 19, 21, 53, 76, 89, 91, 94, and/or positions 3,5,7 of the reciprocal amino acid position of the short peptide of the alpha chain J gene (TRAJ), and/or positions 2,4,6 of the reciprocal amino acid position of the short peptide of the beta chain J gene (TRBJ), wherein the position numbering of the amino acid sequence is according to the position numbering listed in the International immunogenetic information System (IMGT). The above-mentioned international system of immunogenetics information is known to the skilled person and the position numbering of the amino acid residues of the different TCRs in IMGT can be derived from this database.
The TCR with the mutated hydrophobic core region of the invention can be a stable soluble single chain TCR formed by connecting the variable domains of the alpha and beta chains of the TCR by a flexible peptide chain. It should be noted that the flexible peptide chain of the present invention can be any peptide chain suitable for linking the TCR α and β chain variable domains.
In addition, for stability, patent document PCT/CN2016/077680 also discloses that the introduction of an artificial interchain disulfide bond between the α chain variable region and the β chain constant region of the TCR can significantly improve the stability of the TCR. Thus, the high affinity TCRs of the invention may also contain an artificial interchain disulfide bond between the α chain variable region and the β chain constant region. Specifically, the cysteine residues that form the artificial interchain disulfide bond between the α chain variable region and the β chain constant region of the TCR are substituted for: amino acid 46 of TRAV and amino acid 60 of exon 1 of TRBC1 x 01 or TRBC2 x 01; amino acid 47 of TRAV and amino acid 61 of exon 1 of TRBC1 x 01 or TRBC2 x 01; amino acid 46 of TRAV and amino acid 61 of TRBC1 x 01 or TRBC2 x 01 exon 1; or amino acid 47 of TRAV and amino acid 60 of exon 1 of TRBC1 x 01 or TRBC2 x 01. Preferably, such a TCR may comprise (i) all or part of a TCR α chain, excluding its transmembrane domain, and (ii) all or part of a TCR β chain, excluding its transmembrane domain, wherein (i) and (ii) both comprise the variable domain and at least part of the constant domain of the TCR chain, the α chain forming a heterodimer with the β chain. More preferably, such a TCR may comprise the a chain variable domain and the β chain variable domain and all or part of the β chain constant domain, excluding the transmembrane domain, but which does not comprise the a chain constant domain, the a chain variable domain of the TCR forming a heterodimer with the β chain.
The TCRs of the invention may also be provided in the form of multivalent complexes. Multivalent TCR complexes of the invention comprise polymers formed by association of two, three, four or more TCRs of the invention, such as might be produced as a tetramer using the tetrameric domain of p53, or a complex formed by association of a plurality of TCRs of the invention with another molecule. The TCR complexes of the invention can be used to track or target cells presenting a particular antigen in vitro or in vivo, and can also be used to generate intermediates for other multivalent TCR complexes having such applications.
The TCRs of the invention may be used alone or in covalent or other association, preferably covalently, with a conjugate. The conjugates include a detectable label (for diagnostic purposes, where the TCR is used to detect the presence of cells presenting the VFSTVPPAFI-HLA a2402 complex), a therapeutic agent, a PK (protein kinase) modifying moiety, or a combination of any of the above.
Detectable labels for diagnostic purposes include, but are not limited to: a fluorescent or luminescent label, a radioactive label, an MRI (magnetic resonance imaging) or CT (computed tomography) contrast agent, or an enzyme capable of producing a detectable product.
Therapeutic agents that may be associated or conjugated with the TCRs of the invention include, but are not limited to: 1. radionuclides (Koppe et al, 2005, Cancer metastasis reviews (Cancer metastasis) 24, 539); 2. biotoxicity (Chaudhary et al, 1989, Nature 339, 394; Epel et al, 2002, Cancer Immunology and Immunotherapy 51, 565); 3. cytokines such as IL-2 and the like (Gillies et al, 1992, Proc. Natl. Acad. Sci. USA (PNAS)89, 1428; Card et al, 2004, Cancer Immunology and Immunotherapy)53, 345; Halin et al, 2003, Cancer Research 63, 3202); 4. antibody Fc fragment (Mosquera et al, 2005, Journal Of Immunology 174, 4381); 5. antibody scFv fragments (Zhu et al, 1995, International Journal of Cancer 62,319); 6. gold nanoparticles/nanorods (Lapotko et al, 2005, Cancer letters 239, 36; Huang et al, 2006, Journal of the American Chemical Society 128, 2115); 7. viral particles (Peng et al, 2004, Gene therapy 11, 1234); 8. liposomes (Mamot et al, 2005, Cancer research 65, 11631); 9. nano magnetic particles; 10. prodrug activating enzymes (e.g., DT-diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)); 11. chemotherapeutic agents (e.g., cisplatin) or nanoparticles in any form, and the like.
In addition, the TCRs of the invention may also be hybrid TCRs comprising sequences derived from more than one species. For example, studies have shown that murine TCRs are more efficiently expressed in human T cells than human TCRs. Thus, the inventive TCR may comprise a human variable domain and a murine constant domain. The drawback of this approach is the possibility of eliciting an immune response. Thus, there should be a regulatory regimen to immunosuppresse when it is used for adoptive T cell therapy to allow for the engraftment of murine expressing T cells.
It should be understood that the amino acid names herein are expressed in terms of international single-letter or three-letter english letters, and the single-letter english letter and three-letter english letters of the amino acid names correspond to the following relationships: ala (A), Arg (R), Asn (N), Asp (D), Cys (C), Gln (Q), Glu (E), Gly (G), His (H), Ile (I), Leu (L), Lys (K), Met (M), Phe (F), Pro (P), Ser (S), Thr (T), Trp (W), Tyr (Y) and Val (V).
Nucleic acid molecules
A second aspect of the invention provides a nucleic acid molecule encoding a TCR molecule of the first aspect of the invention or a portion thereof, which may be one or more CDRs, variable domains of the alpha and/or beta chains, and the alpha and/or beta chains.
The nucleotide sequence encoding the α chain CDR regions of the TCR molecules of the first aspect of the invention is as follows:
αCDR1-
Figure BDA0001349595040000091
(SEQ ID NO:16)
αCDR2-
Figure BDA0001349595040000092
(SEQ ID NO:17)
αCDR3-
Figure BDA0001349595040000093
(SEQ ID NO:18)
the nucleotide sequence encoding the β chain CDR regions of the TCR molecules of the first aspect of the invention is as follows:
βCDR1-
Figure BDA0001349595040000094
(SEQ ID NO:19)
βCDR2-
Figure BDA0001349595040000095
(SEQ ID NO:20)
βCDR3-
Figure BDA0001349595040000096
(SEQ ID NO:21)
thus, the nucleotide sequence of the nucleic acid molecule of the invention encoding the TCR alpha chain of the invention comprises SEQ ID NO 16, 17 and 18 and/or the nucleotide sequence of the nucleic acid molecule of the invention encoding the TCR beta chain of the invention comprises SEQ ID NO 19, 20 and 21.
The nucleotide sequence of the nucleic acid molecule of the invention may be single-stranded or double-stranded, the nucleic acid molecule may be RNA or DNA, and may or may not comprise an intron. Preferably, the nucleotide sequence of the nucleic acid molecule of the invention does not comprise an intron but is capable of encoding a polypeptide of the invention, e.g. the nucleotide sequence of the nucleic acid molecule of the invention encoding a TCR alpha chain variable domain of the invention comprises SEQ ID NO 2 and/or the nucleotide sequence of the nucleic acid molecule of the invention encoding a TCR beta chain variable domain of the invention comprises SEQ ID NO 6. More preferably, the nucleotide sequence of the nucleic acid molecule of the invention comprises SEQ ID NO. 4 and/or SEQ ID NO. 8.
It will be appreciated that, due to the degeneracy of the genetic code, different nucleotide sequences may encode the same polypeptide. Thus, the nucleic acid sequence encoding the TCR of the present invention may be identical to or a degenerate variant of the nucleic acid sequences shown in the figures of the present invention. As illustrated by one of the examples herein, a "degenerate variant" refers to a nucleic acid sequence that encodes a protein sequence having SEQ ID NO. 1, but differs from the sequence of SEQ ID NO. 2.
The nucleotide sequence may be codon optimized. Different cells differ in the utilization of specific codons, and the expression level can be increased by changing the codons in the sequence according to the type of the cell. Codon usage tables for mammalian cells as well as for various other organisms are well known to those skilled in the art.
The full-length sequence of the nucleic acid molecule of the present invention or a fragment thereof can be obtained by, but not limited to, PCR amplification, recombination, or artificial synthesis. At present, DNA sequences encoding the TCRs of the invention (or fragments or derivatives thereof) have been obtained entirely by chemical synthesis. The DNA sequence may then be introduced into various existing DNA molecules (or vectors, for example) and cells known in the art. The DNA may be the coding strand or the non-coding strand.
Carrier
The invention also relates to vectors comprising the nucleic acid molecules of the invention, including expression vectors, i.e. constructs capable of expression in vivo or in vitro. Commonly used vectors include bacterial plasmids, bacteriophages and animal and plant viruses.
Viral delivery systems include, but are not limited to, adenoviral vectors, adeno-associated virus (AAV) vectors, herpes viral vectors, retroviral vectors, lentiviral vectors, baculoviral vectors.
Preferably, the vector can transfer the nucleotide of the invention into a cell, such as a T cell, such that the cell expresses a TCR specific for the SAGE1 antigen. Ideally, the vector should be capable of sustained high level expression in T cells.
Cells
The invention also relates to genetically engineered host cells that have been engineered with the vectors or coding sequences of the invention. The host cell comprises a vector of the invention or has integrated into its chromosome a nucleic acid molecule of the invention. The host cell is selected from: prokaryotic and eukaryotic cells, such as E.coli, yeast cells, CHO cells, and the like.
In addition, the invention also includes isolated cells, particularly T cells, that express the TCRs of the invention. The T cell may be derived from a T cell isolated from a subject, or may be part of a mixed population of cells isolated from a subject, such as a population of Peripheral Blood Lymphocytes (PBLs). For example, the cells may be isolated from Peripheral Blood Mononuclear Cells (PBMC), which may be CD4+Helper T cell or CD8+Cytotoxic T cells. The cell may be in CD4+Helper T cell/CD 8+A mixed population of cytotoxic T cells. Generally, the cells can be activated with an antibody (e.g., an anti-CD 3 or anti-CD 28 antibody) to render them more amenable to transfection, e.g., transfection with a vector comprising a nucleotide sequence encoding a TCR molecule of the invention.
Alternatively, the cell of the invention may also be or be derived from a stem cell, such as a Hematopoietic Stem Cell (HSC). Gene transfer to HSCs does not result in TCR expression on the cell surface, since the CD3 molecule is not expressed on the stem cell surface. However, when stem cells differentiate into lymphoid precursors (lymphoid precursors) that migrate to the thymus, expression of the CD3 molecule will initiate expression of the introduced TCR molecule on the surface of the thymocytes.
There are many methods suitable for T cell transfection using DNA or RNA encoding the TCR of the invention (e.g., Robbins et al, (2008) J.Immunol.180: 6116-. T cells expressing the TCRs of the invention may be used for adoptive immunotherapy. Those skilled in the art will be able to recognize many suitable methods for adoptive therapy (e.g., Rosenberg et al, (2008) Nat Rev Cancer8 (4): 299-308).
SAGE1 antigen-related diseases
The present invention also relates to a method for treating and/or preventing a disease associated with SAGE1 in a subject, comprising the step of adoptively transferring SAGE 1-specific T cells to the subject. The SAGE 1-specific T cells recognized the VFSTVPPAFI-HLA A2402 complex.
The SAGE1 specific T cells of the invention can be used for treating any SAGE1 related disease presenting SAGE1 antigen short peptide VFSTVPPAFI-HLA A2402 complex. Including but not limited to tumors such as melanoma, and other solid tumors such as bladder cancer, liver cancer, epidermoid carcinoma, non-small cell lung cancer, squamous cell carcinoma, and the like.
Method of treatment
Treatment may be carried out by isolating T cells from patients or volunteers with a disease associated with SAGE1 antigen and introducing the TCR of the invention into such T cells, followed by reinfusion of these genetically engineered cells into the patient. Accordingly, the present invention provides a method of treating a disease associated with SAGE1 comprising infusing into a patient isolated T cells expressing a TCR according to the invention, preferably the T cells are derived from the patient themselves. Generally, this involves (1) isolating T cells from the patient, (2) transducing T cells in vitro with a nucleic acid molecule of the invention or a nucleic acid molecule capable of encoding a TCR molecule of the invention, and (3) infusing the genetically modified T cells into the patient. The number of cells isolated, transfected and transfused can be determined by a physician.
The main advantages of the invention are:
(1) the TCR disclosed by the invention can be combined with SAGE1 antigen short peptide complex VFSTVPPAFI-HLA A2402, and cells transduced with the TCR disclosed by the invention can be specifically activated and have a strong killing effect on target cells.
The following specific examples further illustrate the invention. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, for which specific conditions are not indicated in the following examples, are generally carried out according to conventional conditions, for example as described in Sambrook and Russell et al, Molecular Cloning: A Laboratory Manual (third edition) (2001) CSHL Press, or according to the conditions as recommended by the manufacturer. Unless otherwise indicated, percentages and parts are by weight. Unless otherwise indicated, percentages and parts are by weight. The test materials and reagents used in the following examples are commercially available without specific reference.
EXAMPLE 1 cloning SAGE 1-specific T cells
Peripheral Blood Lymphocytes (PBLs) from healthy volunteers of genotype HLA-A2402 were stimulated with synthetic short peptide (Beijing Baisheng Gene technology, Inc.) VFSTVPPAFI (SEQ ID NO.9, named PX149 in the present invention). The short peptide was renatured with HLA-A2402 having a biotin label to prepare a pHLA homopolymer. These monomers were combined with streptavidin labeled with PE (BD Co.) to form PE-labeled tetramers, which were then sorted for double positive cells against CD 8-APC. The sorted cells were expanded and subjected to secondary sorting as described above, followed by monoclonal culture by limiting dilution.
Because the whole experiment process takes longer time, the influence factors are extremely large, the experiment is complex, the cell performance can not be predicted at all, and the success rate of obtaining the corresponding T cell monoclonal is very low even through layer-by-layer screening and strict detection. It is generally difficult to obtain TCRs with the desired activity through only a few batches of experiments.
In the invention, through intensive research and a large number of experiments of the inventor, the double-positive monoclonal cell meeting the conditions is finally obtained. Monoclonal cells were stained with tetramer and double positive clones were selected as shown in FIG. 3.
Even if a single T cell clone is successfully selected, the resulting TCR may not be satisfactory because in many cases the resulting TCR may not be successfully renatured or may have poor affinity for the corresponding epitope after renaturation, or may not bind. Further validation of binding activity is required.
Example 2 construction of TCR Gene and vector for obtaining SAGE 1-specific T cell clone
Using Quick-RNATMTotal RNA from VFSTVPPAFI-specific, HLA-A2402-restricted T cell clones selected in example 1 were extracted by MiniPrep (ZYMO research). cDNA was synthesized using the SMART RACE cDNA amplification kit from clontech, using primers designed to be conserved at the C-terminus of the human TCR gene. The sequences were cloned into the T vector (TAKARA) and sequenced. The alpha chain and beta chain sequence structures of the TCR expressed by the double positive clone are respectively shown in figure 1 a-figure 1f and figure 2 a-figure 2f after sequencing, and figure 1a, figure 1b, figure 1c and figure 1d are respectively a TCR alpha chain variable domain amino acid sequence, a TCR alpha chain variable domain nucleotide sequence, a TCR alpha chain amino acid sequence and a TCR alpha chain nucleotide sequence; fig. 2a, fig. 2b, fig. 2c and fig. 2d are a TCR β chain variable domain amino acid sequence, a TCR β chain variable domain nucleotide sequence, a TCR β chain amino acid sequence and a TCR β chain nucleotide sequence, respectively.
The alpha chain was identified to comprise CDRs with the following amino acid sequences:
αCDR1-SSVPPY(SEQ ID NO:10)
αCDR2-YTSAATLV(SEQ ID NO:11)
αCDR3-CAVSERARLMF(SEQ ID NO:12)
the beta chain comprises CDRs having the following amino acid sequences:
βCDR1-SGHDT(SEQ ID NO:13)
βCDR2-YYEEEE(SEQ ID NO:14)
βCDR3-CASSLVGFTEAFF(SEQ ID NO:15)。
variable domains of TCR α and β chains were spliced separately to conserved domains of mouse TCR α and β chains, respectively, by overlap (overlap) PCR into full-length genes and ligated into lentiviral expression vector plenti (adddge). The method specifically comprises the following steps: v region genes of a TCR alpha chain and a TCR beta chain are respectively connected with C regions of conserved regions of a mouse TCR alpha chain and a TCR beta chain by overlap PCR to obtain a TCR alpha-2A-TCR beta fragment. And (3) carrying out enzyme digestion and connection on the lentivirus expression vector and the TCR alpha-2A-TCR beta to obtain a pLenti-SAGE1TRA-2A-TRB-IRES-NGFR plasmid. As a control, a lentiviral vector pLenti-eGFP expressing eGFP was also constructed. The pseudovirus was then packaged again at 293T/17.
Example 3 expression, refolding and purification of SAGE1 antigen short peptide specific soluble TCR
To obtain soluble TCR molecules, the α and β chains of the TCR molecules of the invention may comprise only the variable and part of the constant domains thereof, respectively, and a cysteine residue has been introduced into the constant domains of the α and β chains, respectively, to form artificial interchain disulfide bonds, at the positions Thr48 of exon 1 TRAC × 01 and Ser57 of exon 1 TRBC2 × 01, respectively; the amino acid sequence and nucleotide sequence of the alpha chain are shown in FIGS. 4a and 4b, respectively, and the amino acid sequence and nucleotide sequence of the beta chain are shown in FIGS. 5a and 5b, respectively, and the introduced cysteine residues are shown in bold and underlined letters. The desired gene sequences of the above TCR alpha and beta chains were synthesized and inserted into the expression vector pET28a + (Novagene) by standard methods described in "Molecular Cloning A Laboratory Manual" (third edition, Sambrook and Russell), upstream and downstream Cloning sites being NcoI and NotI, respectively. The insert was confirmed by sequencing without error.
The expression vectors of TCR alpha and beta chains are transformed into expression bacteria BL21(DE3) by chemical transformation method, and the bacteria are grown in LB culture solution and OD600Inclusion bodies formed after expression of the α and β chains of the TCR were extracted by BugBuster Mix (Novagene) and washed repeatedly with BugBuster solution several times at 0.6 final induction with final concentration of 0.5mM IPTG, and finally dissolved in 6M guanidine hydrochloride, 10mM Dithiothreitol (DTT),10mM ethylenediaminetetraacetic acid (EDTA),20mM Tris (pH 8.1).
The TCR α and β chains after lysis were separated by 1: 1 was rapidly mixed in 5M urea, 0.4M arginine, 20mM Tris (pH 8.1),3.7mM cystamine,6.6mM β -mercaptamine (4 ℃) to a final concentration of 60 mg/mL. After mixing, the solution was dialyzed against 10 volumes of deionized water (4 ℃ C.) and after 12 hours, the deionized water was changed to a buffer (20mM Tris, pH 8.0) and dialysis was continued at 4 ℃ for 12 hours. The solution after completion of dialysis was filtered through a 0.45. mu.M filter and then purified by an anion exchange column (HiTrap Q HP,5ml, GE Healthcare). The TCR eluted with peaks containing successfully renatured α and β dimers was confirmed by SDS-PAGE gel. The TCR was subsequently further purified by gel filtration chromatography (HiPrep 16/60, Sephacryl S-100HR, GE Healthcare). The purity of the purified TCR was greater than 90% as determined by SDS-PAGE and the concentration was determined by BCA. The SDS-PAGE gel of the soluble TCR of the invention is shown in FIG. 6.
Example 4 binding characterisation
BIAcore analysis
This example demonstrates that soluble TCR molecules of the invention are capable of specifically binding to the VFSTVPPAFI-HLA a2402 complex.
The binding activity of the TCR molecules obtained in examples 3 and 5 to the VFSTVPPAFI-HLA a2402 complex was tested using a BIAcore T200 real-time assay system. Anti-streptavidin antibody (GenScript) was added to coupling buffer (10mM sodium acetate buffer, pH 4.77), and then the antibody was passed through CM5 chip previously activated with EDC and NHS to immobilize the antibody on the chip surface, and finally the unreacted activated surface was blocked with ethanolamine hydrochloric acid solution to complete the coupling process at a coupling level of about 15,000 RU.
The low concentration of streptavidin was flowed over the antibody coated chip surface, then VFSTVPPAFI-HLA A2402 complex was flowed over the detection channel, the other channel served as a reference channel, and 0.05mM biotin was flowed over the chip at a flow rate of 10. mu.L/min for 2min to block the remaining binding sites of streptavidin.
The VFSTVPPAFI-HLA A2402 complex is prepared as follows:
a. purification of
Collecting 100ml E.coli liquid for inducing expression of heavy chain or light chain, centrifuging at 4 ℃ for 10min at 8000g, washing the thalli once with 10ml PBS, then resuspending the thalli with 5ml BugBuster Master Mix Extraction Reagents (Merck) by vigorous shaking, rotatably incubating at room temperature for 20min, centrifuging at 4 ℃ for 15min at 6000g, discarding supernatant, and collecting inclusion body.
Resuspending the inclusion bodies in 5ml of BugBuster Master Mix, and rotary incubating at room temperature for 5 min; adding 30ml of 10-fold diluted BugBuster, uniformly mixing, and centrifuging at 4 ℃ at 6000g for 15 min; discarding the supernatant, adding 30ml of 10-fold diluted BugBuster to resuspend the inclusion bodies, mixing uniformly, centrifuging at 4 ℃ for 15min at 6000g, repeating twice, adding 30ml of 20mM Tris-HCl pH 8.0 to resuspend the inclusion bodies, mixing uniformly, centrifuging at 4 ℃ for 15min at 6000g, finally dissolving the inclusion bodies by using 20mM Tris-HCl 8M urea, detecting the purity of the inclusion bodies by SDS-PAGE, and detecting the concentration by using a BCA kit.
b. Renaturation
The synthesized short peptide VFSTVPPAFI (Beijing Baisheng Gene technology Co., Ltd.) was dissolved in DMSO to a concentration of 20 mg/ml. Inclusion of light and heavy chains was solubilized with 8M Urea, 20mM Tris pH 8.0, 10mM DTT and further denatured by addition of 3M guanidine hydrochloride, 10mM sodium acetate, 10mM EDTA prior to renaturation. VFSTVPPAFI peptide was added to a renaturation buffer (0.4M L-arginine, 100mM Tris pH 8.3, 2mM EDTA, 0.5mM oxidative glutathione, 5mM reduced glutathione, 0.2mM PMSF, cooled to 4 ℃) at 25mg/L (final concentration), followed by the addition of 20mg/L of light chain and 90mg/L of heavy chain in sequence (final concentration, heavy chain was added in three portions, 8 h/time), and renaturation was carried out at 4 ℃ for at least 3 days until completion, and SDS-PAGE checked for success or failure of the renaturation.
c. Purification after renaturation
The renaturation buffer was replaced by dialysis against 10 volumes of 20mM Tris pH 8.0, at least twice to reduce the ionic strength of the solution sufficiently. After dialysis, the protein solution was filtered through a 0.45 μm cellulose acetate filter and then loaded onto a HiTrap Q HP (GE general electric) anion exchange column (5ml bed volume). The protein was eluted using an Akta purifier (GE general electric) with a 0-400mM NaCl linear gradient prepared in 20mM Tris pH 8.0, pMHC was eluted at about 250mM NaCl, the peak fractions were collected, and the purity was checked by SDS-PAGE.
d. Biotinylation of the compound
The purified pMHC molecules were concentrated using Millipore ultrafiltration tubes while displacing the buffer to 20mM Tris pH 8.0, followed by addition of biotinylation reagent 0.05M Bicine pH 8.3, 10mM ATP, 10mM MgOAc, 50. mu. M D-Biotin, 100. mu.g/ml BirA enzyme (GST-BirA), incubation of the mixture overnight at room temperature, and SDS-PAGE to determine the completion of biotinylation.
e. Purification of biotinylated complexes
The biotinylated pMHC molecules were concentrated to 1ml using Millipore ultrafiltration tubes, the biotinylated pMHC was purified by gel filtration chromatography, and HiPrep was pre-equilibrated with filtered PBS using Akta purifier (GE general electric Co., Ltd.)TM16/60S200HR column (GE general electric) was loaded with 1ml of concentrated biotinylated pMHC molecules and then eluted with PBS at a flow rate of 1 ml/min. Biotinylated pMHC molecules appeared as a unimodal elution at approximately 55 ml. The fractions containing the protein were pooled, concentrated using Millipore ultrafiltration tubes, protein concentration was determined by BCA (Thermo), and biotinylated pMHC molecules were stored in aliquots at-80 ℃ by addition of the protease inhibitor cocktail (Roche).
Kinetic parameters were calculated using BIAcore Evaluation software, and kinetic profiles of the binding of soluble TCR molecules of the invention to VFSTVPPAFI-HLA A2402 complex were obtained as shown in FIG. 7. The profile shows that the soluble TCR molecules obtained by the invention can be combined with VFSTVPPAFI-HLA A2402 complex. Meanwhile, the method is used for detecting the binding activity of the soluble TCR molecule and the short peptides of other unrelated antigens and the HLA complex, and the result shows that the TCR molecule is not bound with other unrelated antigens.
Example 5 SAGE 1-specific T cell receptor Lentiviral packaging and transfection of primary T cells with SAGE1TCR
(a) Production of lentiviruses by Rapid-mediated transient transfection of 293T/17 cells
A third generation lentiviral packaging system was used to package lentiviruses containing the gene encoding the desired TCR. 293T/17 cells were transfected with 4 plasmids (a lentiviral vector containing pLenti-SAGE1TRA-2A-TRB-IRES-NGFR as described In example 2, and 3 plasmids containing other components necessary for the construction of infectious but non-replicating lentiviral particles) using rapid-mediated transient transfection (Express-In-mediated transfection (Open Biosystems)).
For transfection, cells were seeded at day 0 on a 15 cm petri dish at 1.7X 107293T/17 cells, which were distributed evenly on the culture dish with a degree of confluence slightly higher than 50%. On day 1, plasmids were transfected, pLenti-SAGE1TRA-2A-TRB-IRES-NGFR and pLenti-eGFP pseudoviruses were packaged, and the above expression plasmids were mixed with packaging plasmids pMDLg/pRRE, pRSV-REV and pMD.2G in a 15 cm diameter plate in the following amounts: 22.5 microgram: 15 microgram: 7.5 micrograms. The ratio of the transfection reagent PEI-MAX to the plasmid was 2:1 and the amount used was 114.75. mu.g per dish. The specific operation is as follows: adding the expression plasmid and the packaging plasmid into 1800 microliters of OPTI-MEM (Gibbo, catalog No. 31985-2And (5) culturing. 5-7 hours of transfection, the transfection medium was removed and replaced with DMEM (Gibbo, Cat. C11995500bt) complete medium containing 10% fetal bovine serum at 37 deg.C/5% CO2And (5) culturing. Culture supernatants containing packaged lentiviruses were collected on days 3 and 4. To harvest the packaged lentivirus, the collected culture supernatant was centrifuged at 3000g for 15min to remove cell debris, filtered through a 0.22 micron filter (Merck Millipore, catalog # SLGP033RB) and finally concentrated using a 50KD cut-off concentrator tube (Merck Millipore), catalog # UFC905096 to remove most of the supernatant, finally concentrated to 1ml and aliquoted and frozen at-80 ℃. Pseudovirus samples were taken for virus titer determination, procedures were referenced to p24ELISA (Clontech, cat # 632200) kit instructions. For comparison, the pLenti-eGFP vector was also includedPseudoviruses.
(b) Transduction of primary T cells with lentiviruses containing SAGE 1-specific T cell receptor genes
CD8 isolated from blood of healthy volunteers+T cells, then transduced with the packaged lentivirus. These cells were counted in 48-well plates in 1X 10 medium 1640 (Gibbo, Cat. No. C11875500bt) with 10% FBS (Gibbo, Gibco, Cat. No. C10010500BT) containing 50IU/ml IL-2 and 10ng/ml IL-76Cells/ml (0.5 ml/well) were incubated with pre-washed anti-CD 3/CD28 antibody-coated beads (T cell amplicons, life technologies, cat No. 11452D) overnight for stimulation, cells: bead 3: 1.
after overnight stimulation, according to the virus titer measured by the p24ELISA kit, the concentrated slow virus of SAGE 1-specific T cell receptor gene was added at an MOI of 10 and centrifuged at 32 ℃ and 900g for 1 hour. After infection, the lentivirus infection solution was removed and the cells were resuspended in 1640 medium supplemented with 30IU/ml IL-2 containing 10% FBS at 37 ℃/5% CO2The cells were cultured for 3 days. Cells were counted 3 days after transduction and diluted to 0.5X 106Individual cells/ml. The cells were counted every two days, replaced or added with fresh medium containing 3IU/ml IL-2, maintaining the cells at 0.5X 106-1×106Individual cells/ml. Cells were analyzed by flow cytometry starting on day 3 and starting on day 5 for functional assays (e.g., ELISPOT for IFN- γ release and non-radioactive cytotoxicity assays). Freezing storage of aliquots of cells, at least 4X 10, from day 10 or as the cells slow down division and become smaller in size6Individual cell/tube (1X 10)7Individual cells/ml, 90% FBS/10% DMSO).
Example 6 cell activation functional validation
ELISPOT scheme
The following assay was performed to demonstrate activation of TCR-transduced T cells in response specifically to target cells. IFN-. gamma.production as measured by the ELISPOT assay was used as a readout for T cell activation.
Reagent
Test medium: 10% FBS (Gibbo, catalog number 16000-
Washing buffer solution: 0.01M PBS/0.05% Tween 20
PBS (Gibbo Co., catalog number C10010500BT)
PVDF ELISPOT 96-well plate (Merck Millipore, Cat. No. MSIPS4510)
Human IFN-. gamma.ELISPOT PVDF-enzyme kit (BD) contains all other reagents required (capture and detection antibody, streptavidin-alkaline phosphatase and BCIP/NBT solution)
Method
Target cell preparation
The target cells of this example were Epstein-Barr virus (EBV) transformed immortalized Lymphoblast Cell Lines (LCLs). B95-8 cells were induced to produce EBV-containing culture supernatants by phorbol myristate acetate (TPA), centrifuged at 4 deg.C/600 g for 10min to remove impurities, filtered through 0.22 μm filter, and aliquoted for-70 deg.C storage. From Peripheral Blood Lymphocytes (PBLs) of healthy volunteers of the genotype HLA-A11/A02/A24 (both homozygote and heterozygote), 10ml of 2X 10-concentration blood was taken7One ml PBL suspension in 25 cm square culture flask, adding cyclosporine at 37 deg.C/CO2Incubate for 1 hour in incubator, thaw one EBV portion quickly, add to the cells at 1/10 dilution, shake gently and place the flask upright at 37 deg.C/CO2Culturing in an incubator. After 12 days of culture, the culture was continued by adding 10ml of a medium, and after about 30 days, the culture was further expanded and subjected to flow assay, in which CD19 was present+CD23hiCD58+Is an immortalized Lymphoblastoid Cell Line (LCL). The ELISPOT test uses HLA-A24 as a specific target cell.
Effector cell preparation
The effector cells (T cells) of this assay were CD8 expressing SAGE 1-specific TCR analyzed by flow cytometry in example 3+T cells and CD8 of the same volunteer+T served as negative control effector cells. Stimulation of T cells with anti-CD 3/CD28 coated beads (T cell amplicons, Life technologies), transduction with lentiviruses carrying SAGE 1-specific TCR genes (according to the examples)3) After amplification in 1640 medium containing 10% FBS containing 30IU/ml IL-2 until 9-12 days after transduction, the cells were placed in the test medium and washed by centrifugation at 300g for 10 minutes at room temperature. The cells were then resuspended in the test medium at 2 × the desired final concentration. Negative control effector cells were treated as well.
ELISPOT
The well plate was prepared as follows according to the manufacturer's instructions: 10ml of sterile PBS per plate 1: anti-human IFN-. gamma.capture antibody was diluted at 200, and 100. mu.l of the diluted capture antibody was aliquoted into each well. The plates were incubated overnight at 4 ℃. After incubation, the well plates were washed to remove excess capture antibody. 100 μ l/well of RPMI1640 medium containing 10% FBS was added and the well plates were incubated at room temperature for 2 hours to close the well plates. The media was then washed from the well plate, and any residual wash buffer was removed by flicking and tapping the ELISPOT well plate on paper.
SAGE1CD8+T cells (TCR-transduced T cells of the invention, i.e., effector cells, also designated "VF 6CD 8" in the invention+TCELL "), CD8+ T cells (negative control effector cells) and LCL-A24/A11 (target cells) were prepared as described in example 3, and the corresponding short peptides were added to the corresponding experimental groups, wherein PX149 was SAGE1PX 149597-606 VFSTVPPAFI short peptide, and the remainder were non-SAGE 1 TCR-specific binding short peptides.
The components of the assay were then added to ELISPOT well plates in the following order:
77000 cells/ml of 130 microliters of target cells (resulting in a total of about 10000 target cells/well).
50 microliter of effector cells (1000 SAGE1TCR double positive T cells).
20 microliter 10-5Mol/l SAGE1PX 149597-606 VFSTVPPAFI/nonspecific short peptide solution (final concentration is 10)-6Moles/liter).
All wells were made in triplicate for addition.
The well plates were then incubated overnight (37 ℃/5% CO)2) The next day, the medium was discarded, the well plate was washed 2 times with double distilled water and 3 times with wash buffer, and tapped on a paper towel to remove residual wash buffer. However, the device is not suitable for use in a kitchenPrimary antibody was then detected by dilution with PBS containing 10% FBS and added to each well at 100. mu.l/well. The well plate was incubated at room temperature for 2 hours, washed 3 times with wash buffer and the well plate was tapped on a paper towel to remove excess wash buffer.
PBS containing 10% FBS was used at 1: streptavidin-alkaline phosphatase was diluted 100, 100 microliters of diluted streptavidin-alkaline phosphatase was added to each well and the wells were incubated for 1 hour at room temperature. The plate was then washed 3 times with wash buffer and 2 times with PBS, and the excess wash buffer and PBS was removed by tapping the plate on a paper towel. After washing, 100 microliter of BCIP/NBT solution provided by the kit is added for development. And covering the well plate with tinfoil paper in the developing period, keeping the well plate in the dark, and standing for 5-15 minutes. Spots on the developing plate were routinely detected during this period to determine the optimum time for terminating the reaction. The BCIP/NBT solution was removed and the well plate was rinsed with double-distilled water to stop the development reaction, spun-dried, then the bottom of the well plate was removed, the well plate was dried at room temperature until each well was completely dried, and then the spots formed in the bottom film of the well plate were counted using an immune spot plate counter (CTL, cell Technology Limited).
Results
SAGE1TCR transduced T cells were tested by ELISPOT assay (described above) for IFN- γ release in response to SAGE1PX 149597-606 VFSTVPPAFI short peptide loaded target cells and non-specific short peptide loaded target cells. The number of ELSPOT spots observed in each well was plotted using a graphipad prism 6.
The results of the experiment are shown in FIG. 8, SAGE1CD8 alone+Little IFN-. gamma.is released by T cells (effector cells) or LCL cells (target cells) when the corresponding short peptides are added.
SAGE1CD8+T cells (effector cells) were able to react with LCL-A24/A11 cells supplemented with PX149 to release more IFN- γ.
SAGE1CD8+T cells (effector cells) released very little IFN-. gamma.when added to LCL cells with nonspecific short peptides.
CD8+T cells (negative control effector cells) added LCL-A24/A11 cells of PX149 with minimal IFN- γ release.
In conclusion, T cells transduced with the TCR of the invention have a good activation response to target cells loaded with the specific short peptide, and no activation response to target cells not loaded with the corresponding short peptide and target cells loaded with non-specific short peptide.
Example 7 verification of cell killing function
This test is a colorimetric substitution test for the 51Cr release cytotoxicity test, and quantitatively determines Lactate Dehydrogenase (LDH) released after cell lysis. LDH released in the medium was detected using a 30 min coupled enzymatic reaction in which LDH converted a tetrazolium salt (INT) to red formazan (formazan). The amount of red product produced is proportional to the number of cells lysed. 490nm visible absorbance data can be collected using a standard 96-well plate reader.
Material
CytoTox
Figure BDA0001349595040000161
Non-radioactive cytotoxicity assays (Promega, G1780) contained a substrate mixture, assay buffer, lysis solution and stop buffer.
Test medium: 10% FBS (heat-inactivated, Gibbo, Cat. No. 16000-.
Microwell round bottom 96 well tissue culture plates (Nunc, Cat. No. 163320)
96-well immunoplate Maxisorb (Nunc, Cat. No. 442404)
Method
Target cell preparation
The test adopts three tumor cell lines of 95D, NCI-H1299 and IM 9 as target cells. Target cells were prepared in assay medium: the concentration of the target cells is adjusted to 3X 105One/ml, 50. mu.l/well to obtain 1.5X 104Individual cells/well.
Effector cell preparation
The effector cells (T cells) of this assay were CD8 analyzed by flow cytometry in example 3+T cells. Fine effectCell to target cell ratio 10:1,5: 1,2.5: 1,1.25:1 (diluted to 3X 10 if 10:1)6One/ml, 50. mu.l/well to obtain 1.5X 105Individual cells/well). Set up the source CD8+T cells plus target cell control (10: 1).
SAGE 1-specific TCR transduced CD8+T cell specific killing tumor cell assay
Preparation of the test
The components of the assay were added to a microwell round bottom 96 well tissue culture plate in the following order:
45ul of target cells (prepared as described above) were added to each well
45ul of effector cells (prepared as described above) were added to each well
A control group was prepared as follows:
effector cells release spontaneously: there were only 45ul of effector cells.
Target cells release: there were only 45ul of target cells.
Maximum release of target cells: there were only 45ul of target cells.
-media control: only 90ul of medium was present.
All wells were made in triplicate and the final volume was 90ul (insufficient media make up).
Incubate at 37 ℃ for 24 hours. Before collecting the supernatants from all wells, the target cells maximum release control wells were placed on the cells at-70 ℃ for approximately 30 minutes and thawed at 37 ℃ for 15 minutes to allow total lysis of the target cells.
The plate was centrifuged at 250g for 4 min. 50ul of supernatant from each well of the assay plate was transferred to the corresponding well of a 96-well immunoplate Maxisorb plate. The substrate mixture was reconstituted with assay buffer (12ml) and 50ul was added to each well of the plate. The plate was covered and incubated in the dark at room temperature for 30 minutes. 50ul of stop solution was added to each well of the plate to stop the reaction. The absorbance at 490nm was recorded counted over 1 hour after addition of the stop solution.
Calculation results
The absorbance values of the medium background were subtracted from all the absorbance values of the experimental, target cell spontaneous release and effector cell spontaneous release groups.
The corrected values obtained above were substituted into the following formula to calculate the percent cytotoxicity resulting from each effect-to-target ratio.
% cytotoxicity 100 × (experiment-effector cell spontaneous-target cell spontaneous)/(target cell maximal-target cell spontaneous)
Results
SAGE1TCR transduced T cells were tested for LDH release in response to specific target cells by a non-radioactive cytotoxicity assay (as described above). The absorbance of 490nm visible light in each well was plotted using graphpad prism 6.
The statistical results of the experimental data are shown in fig. 9, where the effective target ratio is 10:1, the T cells transduced by the TCR of the invention have obvious killing effect on specific target cells 95D; has no killing effect on non-specific target cells NCI-H1299 and IM 9. Homologous CD8+T cells kill target cells 95D only weakly.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.
Sequence listing
<110> Guangzhou biomedical and health research institute of Chinese academy of sciences
<120> T cell receptor recognizing SAGE1 antigen and nucleic acid encoding the receptor
<130> P2017-1271
<160> 29
<170> PatentIn version 3.5
<210> 1
<211> 111
<212> PRT
<213> artificial sequence
<220>
<223> TCR alpha chain variable Domain
<400> 1
Ala Gln Ser Val Thr Gln Leu Gly Ser His Val Ser Val Ser Glu Gly
1 5 10 15
Ala Leu Val Leu Leu Arg Cys Asn Tyr Ser Ser Ser Val Pro Pro Tyr
20 25 30
Leu Phe Trp Tyr Val Gln Tyr Pro Asn Gln Gly Leu Gln Leu Leu Leu
35 40 45
Lys Tyr Thr Ser Ala Ala Thr Leu Val Lys Gly Ile Asn Gly Phe Glu
50 55 60
Ala Glu Phe Lys Lys Ser Glu Thr Ser Phe His Leu Thr Lys Pro Ser
65 70 75 80
Ala His Met Ser Asp Ala Ala Glu Tyr Phe Cys Ala Val Ser Glu Arg
85 90 95
Ala Arg Leu Met Phe Gly Asp Gly Thr Gln Leu Val Val Lys Pro
100 105 110
<210> 2
<211> 333
<212> DNA
<213> artificial sequence
<220>
<223> TCR alpha chain variable Domain
<400> 2
gcccagtcgg tgacccagct tggcagccac gtctctgtct ctgagggagc cctggttctg 60
ctgaggtgca actactcatc gtctgttcca ccatatctct tctggtatgt gcaatacccc 120
aaccaaggac tccagcttct cctgaagtac acatcagcgg ccaccctggt taaaggcatc 180
aacggttttg aggctgaatt taagaagagt gaaacctcct tccacctgac gaaaccctca 240
gcccatatga gcgacgcggc tgagtacttc tgtgctgtga gtgaaagagc cagactcatg 300
tttggagatg gaactcagct ggtggtgaag ccc 333
<210> 3
<211> 252
<212> PRT
<213> artificial sequence
<220>
<223> TCR alpha chain
<400> 3
Ala Gln Ser Val Thr Gln Leu Gly Ser His Val Ser Val Ser Glu Gly
1 5 10 15
Ala Leu Val Leu Leu Arg Cys Asn Tyr Ser Ser Ser Val Pro Pro Tyr
20 25 30
Leu Phe Trp Tyr Val Gln Tyr Pro Asn Gln Gly Leu Gln Leu Leu Leu
35 40 45
Lys Tyr Thr Ser Ala Ala Thr Leu Val Lys Gly Ile Asn Gly Phe Glu
50 55 60
Ala Glu Phe Lys Lys Ser Glu Thr Ser Phe His Leu Thr Lys Pro Ser
65 70 75 80
Ala His Met Ser Asp Ala Ala Glu Tyr Phe Cys Ala Val Ser Glu Arg
85 90 95
Ala Arg Leu Met Phe Gly Asp Gly Thr Gln Leu Val Val Lys Pro Asn
100 105 110
Ile Gln Asn Pro Asp Pro Ala Val Tyr Gln Leu Arg Asp Ser Lys Ser
115 120 125
Ser Asp Lys Ser Val Cys Leu Phe Thr Asp Phe Asp Ser Gln Thr Asn
130 135 140
Val Ser Gln Ser Lys Asp Ser Asp Val Tyr Ile Thr Asp Lys Thr Val
145 150 155 160
Leu Asp Met Arg Ser Met Asp Phe Lys Ser Asn Ser Ala Val Ala Trp
165 170 175
Ser Asn Lys Ser Asp Phe Ala Cys Ala Asn Ala Phe Asn Asn Ser Ile
180 185 190
Ile Pro Glu Asp Thr Phe Phe Pro Ser Pro Glu Ser Ser Cys Asp Val
195 200 205
Lys Leu Val Glu Lys Ser Phe Glu Thr Asp Thr Asn Leu Asn Phe Gln
210 215 220
Asn Leu Ser Val Ile Gly Phe Arg Ile Leu Leu Leu Lys Val Ala Gly
225 230 235 240
Phe Asn Leu Leu Met Thr Leu Arg Leu Trp Ser Ser
245 250
<210> 4
<211> 756
<212> DNA
<213> artificial sequence
<220>
<223> TCR alpha chain
<400> 4
gcccagtcgg tgacccagct tggcagccac gtctctgtct ctgagggagc cctggttctg 60
ctgaggtgca actactcatc gtctgttcca ccatatctct tctggtatgt gcaatacccc 120
aaccaaggac tccagcttct cctgaagtac acatcagcgg ccaccctggt taaaggcatc 180
aacggttttg aggctgaatt taagaagagt gaaacctcct tccacctgac gaaaccctca 240
gcccatatga gcgacgcggc tgagtacttc tgtgctgtga gtgaaagagc cagactcatg 300
tttggagatg gaactcagct ggtggtgaag cccaacatcc agaaccctga ccctgccgtg 360
taccagctga gagactctaa atccagtgac aagtctgtct gcctattcac cgattttgat 420
tctcaaacaa atgtgtcaca aagtaaggat tctgatgtgt atatcacaga caaaactgtg 480
ctagacatga ggtctatgga cttcaagagc aacagtgctg tggcctggag caacaaatct 540
gactttgcat gtgcaaacgc cttcaacaac agcattattc cagaagacac cttcttcccc 600
agcccagaaa gttcctgtga tgtcaagctg gtcgagaaaa gctttgaaac agatacgaac 660
ctaaactttc aaaacctgtc agtgattggg ttccgaatcc tcctcctgaa agtggccggg 720
tttaatctgc tcatgacgct gcggctgtgg tccagc 756
<210> 5
<211> 112
<212> PRT
<213> artificial sequence
<220>
<223> TCR beta chain variable domain
<400> 5
Asp Ala Gly Val Thr Gln Ser Pro Thr His Leu Ile Lys Thr Arg Gly
1 5 10 15
Gln Gln Val Thr Leu Arg Cys Ser Pro Lys Ser Gly His Asp Thr Val
20 25 30
Ser Trp Tyr Gln Gln Ala Leu Gly Gln Gly Pro Gln Phe Ile Phe Gln
35 40 45
Tyr Tyr Glu Glu Glu Glu Arg Gln Arg Gly Asn Phe Pro Asp Arg Phe
50 55 60
Ser Gly His Gln Phe Pro Asn Tyr Ser Ser Glu Leu Asn Val Asn Ala
65 70 75 80
Leu Leu Leu Gly Asp Ser Ala Leu Tyr Leu Cys Ala Ser Ser Leu Val
85 90 95
Gly Phe Thr Glu Ala Phe Phe Gly Gln Gly Thr Arg Leu Thr Val Val
100 105 110
<210> 6
<211> 336
<212> DNA
<213> artificial sequence
<220>
<223> TCR beta chain variable domain
<400> 6
gacgctggag tcacccaaag tcccacacac ctgatcaaaa cgagaggaca gcaagtgact 60
ctgagatgct ctcctaagtc tgggcatgac actgtgtcct ggtaccaaca ggccctgggt 120
caggggcccc agtttatctt tcagtattat gaggaggaag agagacagag aggcaacttc 180
cctgatcgat tctcaggtca ccagttccct aactatagct ctgagctgaa tgtgaacgcc 240
ttgttgctgg gggactcggc cctctatctc tgtgccagca gcttggtagg gttcactgaa 300
gctttctttg gacaaggcac cagactcaca gttgta 336
<210> 7
<211> 289
<212> PRT
<213> artificial sequence
<220>
<223> TCR beta chain
<400> 7
Asp Ala Gly Val Thr Gln Ser Pro Thr His Leu Ile Lys Thr Arg Gly
1 5 10 15
Gln Gln Val Thr Leu Arg Cys Ser Pro Lys Ser Gly His Asp Thr Val
20 25 30
Ser Trp Tyr Gln Gln Ala Leu Gly Gln Gly Pro Gln Phe Ile Phe Gln
35 40 45
Tyr Tyr Glu Glu Glu Glu Arg Gln Arg Gly Asn Phe Pro Asp Arg Phe
50 55 60
Ser Gly His Gln Phe Pro Asn Tyr Ser Ser Glu Leu Asn Val Asn Ala
65 70 75 80
Leu Leu Leu Gly Asp Ser Ala Leu Tyr Leu Cys Ala Ser Ser Leu Val
85 90 95
Gly Phe Thr Glu Ala Phe Phe Gly Gln Gly Thr Arg Leu Thr Val Val
100 105 110
Glu Asp Leu Asn Lys Val Phe Pro Pro Glu Val Ala Val Phe Glu Pro
115 120 125
Ser Glu Ala Glu Ile Ser His Thr Gln Lys Ala Thr Leu Val Cys Leu
130 135 140
Ala Thr Gly Phe Phe Pro Asp His Val Glu Leu Ser Trp Trp Val Asn
145 150 155 160
Gly Lys Glu Val His Ser Gly Val Ser Thr Asp Pro Gln Pro Leu Lys
165 170 175
Glu Gln Pro Ala Leu Asn Asp Ser Arg Tyr Cys Leu Ser Ser Arg Leu
180 185 190
Arg Val Ser Ala Thr Phe Trp Gln Asn Pro Arg Asn His Phe Arg Cys
195 200 205
Gln Val Gln Phe Tyr Gly Leu Ser Glu Asn Asp Glu Trp Thr Gln Asp
210 215 220
Arg Ala Lys Pro Val Thr Gln Ile Val Ser Ala Glu Ala Trp Gly Arg
225 230 235 240
Ala Asp Cys Gly Phe Thr Ser Val Ser Tyr Gln Gln Gly Val Leu Ser
245 250 255
Ala Thr Ile Leu Tyr Glu Ile Leu Leu Gly Lys Ala Thr Leu Tyr Ala
260 265 270
Val Leu Val Ser Ala Leu Val Leu Met Ala Met Val Lys Arg Lys Asp
275 280 285
Phe
<210> 8
<211> 867
<212> DNA
<213> artificial sequence
<220>
<223> TCR beta chain
<400> 8
gacgctggag tcacccaaag tcccacacac ctgatcaaaa cgagaggaca gcaagtgact 60
ctgagatgct ctcctaagtc tgggcatgac actgtgtcct ggtaccaaca ggccctgggt 120
caggggcccc agtttatctt tcagtattat gaggaggaag agagacagag aggcaacttc 180
cctgatcgat tctcaggtca ccagttccct aactatagct ctgagctgaa tgtgaacgcc 240
ttgttgctgg gggactcggc cctctatctc tgtgccagca gcttggtagg gttcactgaa 300
gctttctttg gacaaggcac cagactcaca gttgtagagg acctgaacaa ggtgttccca 360
cccgaggtcg ctgtgtttga gccatcagaa gcagagatct cccacaccca aaaggccaca 420
ctggtgtgcc tggccacagg cttcttcccc gaccacgtgg agctgagctg gtgggtgaat 480
gggaaggagg tgcacagtgg ggtcagcacg gacccgcagc ccctcaagga gcagcccgcc 540
ctcaatgact ccagatactg cctgagcagc cgcctgaggg tctcggccac cttctggcag 600
aacccccgca accacttccg ctgtcaagtc cagttctacg ggctctcgga gaatgacgag 660
tggacccagg atagggccaa acccgtcacc cagatcgtca gcgccgaggc ctggggtaga 720
gcagactgtg gctttacctc ggtgtcctac cagcaagggg tcctgtctgc caccatcctc 780
tatgagatcc tgctagggaa ggccaccctg tatgctgtgc tggtcagcgc ccttgtgttg 840
atggccatgg tcaagagaaa ggatttc 867
<210> 9
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> antigen short peptide
<400> 9
Val Phe Ser Thr Val Pro Pro Ala Phe Ile
1 5 10
<210> 10
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> α CDR1
<400> 10
Ser Ser Val Pro Pro Tyr
1 5
<210> 11
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> α CDR2
<400> 11
Tyr Thr Ser Ala Ala Thr Leu Val
1 5
<210> 12
<211> 11
<212> PRT
<213> artificial sequence
<220>
<223> α CDR3
<400> 12
Cys Ala Val Ser Glu Arg Ala Arg Leu Met Phe
1 5 10
<210> 13
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> β CDR1
<400> 13
Ser Gly His Asp Thr
1 5
<210> 14
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> β CDR2
<400> 14
Tyr Tyr Glu Glu Glu Glu
1 5
<210> 15
<211> 13
<212> PRT
<213> artificial sequence
<220>
<223> β CDR3
<400> 15
Cys Ala Ser Ser Leu Val Gly Phe Thr Glu Ala Phe Phe
1 5 10
<210> 16
<211> 18
<212> DNA
<213> artificial sequence
<220>
<223> α CDR1
<400> 16
tcgtctgttc caccatat 18
<210> 17
<211> 24
<212> DNA
<213> artificial sequence
<220>
<223> α CDR2
<400> 17
tacacatcag cggccaccct ggtt 24
<210> 18
<211> 33
<212> DNA
<213> artificial sequence
<220>
<223> α CDR3
<400> 18
tgtgctgtga gtgaaagagc cagactcatg ttt 33
<210> 19
<211> 15
<212> DNA
<213> artificial sequence
<220>
<223> β CDR1
<400> 19
tctgggcatg acact 15
<210> 20
<211> 18
<212> DNA
<213> artificial sequence
<220>
<223> β CDR2
<400> 20
tattatgagg aggaagag 18
<210> 21
<211> 39
<212> DNA
<213> artificial sequence
<220>
<223> β CDR3
<400> 21
tgtgccagca gcttggtagg gttcactgaa gctttcttt 39
<210> 22
<211> 271
<212> PRT
<213> artificial sequence
<220>
<223> TCR alpha chain having leader sequence
<400> 22
Met Leu Leu Leu Leu Val Pro Val Leu Glu Val Ile Phe Thr Leu Gly
1 5 10 15
Gly Thr Arg Ala Gln Ser Val Thr Gln Leu Gly Ser His Val Ser Val
20 25 30
Ser Glu Gly Ala Leu Val Leu Leu Arg Cys Asn Tyr Ser Ser Ser Val
35 40 45
Pro Pro Tyr Leu Phe Trp Tyr Val Gln Tyr Pro Asn Gln Gly Leu Gln
50 55 60
Leu Leu Leu Lys Tyr Thr Ser Ala Ala Thr Leu Val Lys Gly Ile Asn
65 70 75 80
Gly Phe Glu Ala Glu Phe Lys Lys Ser Glu Thr Ser Phe His Leu Thr
85 90 95
Lys Pro Ser Ala His Met Ser Asp Ala Ala Glu Tyr Phe Cys Ala Val
100 105 110
Ser Glu Arg Ala Arg Leu Met Phe Gly Asp Gly Thr Gln Leu Val Val
115 120 125
Lys Pro Asn Ile Gln Asn Pro Asp Pro Ala Val Tyr Gln Leu Arg Asp
130 135 140
Ser Lys Ser Ser Asp Lys Ser Val Cys Leu Phe Thr Asp Phe Asp Ser
145 150 155 160
Gln Thr Asn Val Ser Gln Ser Lys Asp Ser Asp Val Tyr Ile Thr Asp
165 170 175
Lys Thr Val Leu Asp Met Arg Ser Met Asp Phe Lys Ser Asn Ser Ala
180 185 190
Val Ala Trp Ser Asn Lys Ser Asp Phe Ala Cys Ala Asn Ala Phe Asn
195 200 205
Asn Ser Ile Ile Pro Glu Asp Thr Phe Phe Pro Ser Pro Glu Ser Ser
210 215 220
Cys Asp Val Lys Leu Val Glu Lys Ser Phe Glu Thr Asp Thr Asn Leu
225 230 235 240
Asn Phe Gln Asn Leu Ser Val Ile Gly Phe Arg Ile Leu Leu Leu Lys
245 250 255
Val Ala Gly Phe Asn Leu Leu Met Thr Leu Arg Leu Trp Ser Ser
260 265 270
<210> 23
<211> 813
<212> DNA
<213> artificial sequence
<220>
<223> TCR alpha chain having leader sequence
<400> 23
atgctcctgc tgctcgtccc agtgctcgag gtgattttta ccctgggagg aaccagagcc 60
cagtcggtga cccagcttgg cagccacgtc tctgtctctg agggagccct ggttctgctg 120
aggtgcaact actcatcgtc tgttccacca tatctcttct ggtatgtgca ataccccaac 180
caaggactcc agcttctcct gaagtacaca tcagcggcca ccctggttaa aggcatcaac 240
ggttttgagg ctgaatttaa gaagagtgaa acctccttcc acctgacgaa accctcagcc 300
catatgagcg acgcggctga gtacttctgt gctgtgagtg aaagagccag actcatgttt 360
ggagatggaa ctcagctggt ggtgaagccc aacatccaga accctgaccc tgccgtgtac 420
cagctgagag actctaaatc cagtgacaag tctgtctgcc tattcaccga ttttgattct 480
caaacaaatg tgtcacaaag taaggattct gatgtgtata tcacagacaa aactgtgcta 540
gacatgaggt ctatggactt caagagcaac agtgctgtgg cctggagcaa caaatctgac 600
tttgcatgtg caaacgcctt caacaacagc attattccag aagacacctt cttccccagc 660
ccagaaagtt cctgtgatgt caagctggtc gagaaaagct ttgaaacaga tacgaaccta 720
aactttcaaa acctgtcagt gattgggttc cgaatcctcc tcctgaaagt ggccgggttt 780
aatctgctca tgacgctgcg gctgtggtcc agc 813
<210> 24
<211> 308
<212> PRT
<213> artificial sequence
<220>
<223> TCR beta chain having leader sequence
<400> 24
Met Gly Pro Gly Leu Leu Cys Trp Ala Leu Leu Cys Leu Leu Gly Ala
1 5 10 15
Gly Leu Val Asp Ala Gly Val Thr Gln Ser Pro Thr His Leu Ile Lys
20 25 30
Thr Arg Gly Gln Gln Val Thr Leu Arg Cys Ser Pro Lys Ser Gly His
35 40 45
Asp Thr Val Ser Trp Tyr Gln Gln Ala Leu Gly Gln Gly Pro Gln Phe
50 55 60
Ile Phe Gln Tyr Tyr Glu Glu Glu Glu Arg Gln Arg Gly Asn Phe Pro
65 70 75 80
Asp Arg Phe Ser Gly His Gln Phe Pro Asn Tyr Ser Ser Glu Leu Asn
85 90 95
Val Asn Ala Leu Leu Leu Gly Asp Ser Ala Leu Tyr Leu Cys Ala Ser
100 105 110
Ser Leu Val Gly Phe Thr Glu Ala Phe Phe Gly Gln Gly Thr Arg Leu
115 120 125
Thr Val Val Glu Asp Leu Asn Lys Val Phe Pro Pro Glu Val Ala Val
130 135 140
Phe Glu Pro Ser Glu Ala Glu Ile Ser His Thr Gln Lys Ala Thr Leu
145 150 155 160
Val Cys Leu Ala Thr Gly Phe Phe Pro Asp His Val Glu Leu Ser Trp
165 170 175
Trp Val Asn Gly Lys Glu Val His Ser Gly Val Ser Thr Asp Pro Gln
180 185 190
Pro Leu Lys Glu Gln Pro Ala Leu Asn Asp Ser Arg Tyr Cys Leu Ser
195 200 205
Ser Arg Leu Arg Val Ser Ala Thr Phe Trp Gln Asn Pro Arg Asn His
210 215 220
Phe Arg Cys Gln Val Gln Phe Tyr Gly Leu Ser Glu Asn Asp Glu Trp
225 230 235 240
Thr Gln Asp Arg Ala Lys Pro Val Thr Gln Ile Val Ser Ala Glu Ala
245 250 255
Trp Gly Arg Ala Asp Cys Gly Phe Thr Ser Val Ser Tyr Gln Gln Gly
260 265 270
Val Leu Ser Ala Thr Ile Leu Tyr Glu Ile Leu Leu Gly Lys Ala Thr
275 280 285
Leu Tyr Ala Val Leu Val Ser Ala Leu Val Leu Met Ala Met Val Lys
290 295 300
Arg Lys Asp Phe
305
<210> 25
<211> 924
<212> DNA
<213> artificial sequence
<220>
<223> TCR beta chain having leader sequence
<400> 25
atgggccccg ggctcctctg ctgggcactg ctttgtctcc tgggagcagg cttagtggac 60
gctggagtca cccaaagtcc cacacacctg atcaaaacga gaggacagca agtgactctg 120
agatgctctc ctaagtctgg gcatgacact gtgtcctggt accaacaggc cctgggtcag 180
gggccccagt ttatctttca gtattatgag gaggaagaga gacagagagg caacttccct 240
gatcgattct caggtcacca gttccctaac tatagctctg agctgaatgt gaacgccttg 300
ttgctggggg actcggccct ctatctctgt gccagcagct tggtagggtt cactgaagct 360
ttctttggac aaggcaccag actcacagtt gtagaggacc tgaacaaggt gttcccaccc 420
gaggtcgctg tgtttgagcc atcagaagca gagatctccc acacccaaaa ggccacactg 480
gtgtgcctgg ccacaggctt cttccccgac cacgtggagc tgagctggtg ggtgaatggg 540
aaggaggtgc acagtggggt cagcacggac ccgcagcccc tcaaggagca gcccgccctc 600
aatgactcca gatactgcct gagcagccgc ctgagggtct cggccacctt ctggcagaac 660
ccccgcaacc acttccgctg tcaagtccag ttctacgggc tctcggagaa tgacgagtgg 720
acccaggata gggccaaacc cgtcacccag atcgtcagcg ccgaggcctg gggtagagca 780
gactgtggct ttacctcggt gtcctaccag caaggggtcc tgtctgccac catcctctat 840
gagatcctgc tagggaaggc caccctgtat gctgtgctgg tcagcgccct tgtgttgatg 900
gccatggtca agagaaagga tttc 924
<210> 26
<211> 197
<212> PRT
<213> artificial sequence
<220>
<223> soluble TCR alpha chain
<400> 26
Ala Gln Ser Val Thr Gln Leu Gly Ser His Val Ser Val Ser Glu Gly
1 5 10 15
Ala Leu Val Leu Leu Arg Cys Asn Tyr Ser Ser Ser Val Pro Pro Tyr
20 25 30
Leu Phe Trp Tyr Val Gln Tyr Pro Asn Gln Gly Leu Gln Leu Leu Leu
35 40 45
Lys Tyr Thr Ser Ala Ala Thr Leu Val Lys Gly Ile Asn Gly Phe Glu
50 55 60
Ala Glu Phe Lys Lys Ser Glu Thr Ser Phe His Leu Thr Lys Pro Ser
65 70 75 80
Ala His Met Ser Asp Ala Ala Glu Tyr Phe Cys Ala Val Ser Glu Arg
85 90 95
Ala Arg Leu Met Phe Gly Asp Gly Thr Gln Leu Val Val Lys Pro Tyr
100 105 110
Ile Gln Asn Pro Asp Pro Ala Val Tyr Gln Leu Arg Asp Ser Lys Ser
115 120 125
Ser Asp Lys Ser Val Cys Leu Phe Thr Asp Phe Asp Ser Gln Thr Asn
130 135 140
Val Ser Gln Ser Lys Asp Ser Asp Val Tyr Ile Thr Asp Lys Cys Val
145 150 155 160
Leu Asp Met Arg Ser Met Asp Phe Lys Ser Asn Ser Ala Val Ala Trp
165 170 175
Ser Asn Lys Ser Asp Phe Ala Cys Ala Asn Ala Phe Asn Asn Ser Ile
180 185 190
Ile Pro Glu Asp Thr
195
<210> 27
<211> 591
<212> DNA
<213> artificial sequence
<220>
<223> soluble TCR alpha chain
<400> 27
gcacagtctg tgacccagct tggcagccac gtctctgtct ctgagggagc cctggttctg 60
ctgaggtgca actactcatc gtctgttcca ccatatctct tctggtatgt gcaatacccc 120
aaccaaggac tccagcttct cctgaagtac acatcagcgg ccaccctggt taaaggcatc 180
aacggttttg aggctgaatt taagaagagt gaaacctcct tccacctgac gaaaccctca 240
gcccatatga gcgacgcggc tgagtacttc tgtgctgtga gtgaaagagc cagactcatg 300
tttggagatg gaactcagct ggtggtgaag ccctatatcc agaatccgga cccggccgtt 360
tatcagctgc gtgatagcaa aagcagcgat aaaagcgtgt gcctgttcac cgattttgat 420
agccagacca acgtgagcca gagcaaagat agcgatgtgt acatcaccga taaatgcgtg 480
ctggatatgc gcagcatgga tttcaaaagc aatagcgcgg ttgcgtggag caacaaaagc 540
gattttgcgt gcgcgaacgc gtttaacaac agcatcatcc cggaagatac g 591
<210> 28
<211> 242
<212> PRT
<213> artificial sequence
<220>
<223> soluble TCR beta chain
<400> 28
Asp Ala Gly Val Thr Gln Ser Pro Thr His Leu Ile Lys Thr Arg Gly
1 5 10 15
Gln Gln Val Thr Leu Arg Cys Ser Pro Lys Ser Gly His Asp Thr Val
20 25 30
Ser Trp Tyr Gln Gln Ala Leu Gly Gln Gly Pro Gln Phe Ile Phe Gln
35 40 45
Tyr Tyr Glu Glu Glu Glu Arg Gln Arg Gly Asn Phe Pro Asp Arg Phe
50 55 60
Ser Gly His Gln Phe Pro Asn Tyr Ser Ser Glu Leu Asn Val Asn Ala
65 70 75 80
Leu Leu Leu Gly Asp Ser Ala Leu Tyr Leu Cys Ala Ser Ser Leu Val
85 90 95
Gly Phe Thr Glu Ala Phe Phe Gly Gln Gly Thr Arg Leu Thr Val Val
100 105 110
Glu Asp Leu Lys Asn Val Phe Pro Pro Glu Val Ala Val Phe Glu Pro
115 120 125
Ser Glu Ala Glu Ile Ser His Thr Gln Lys Ala Thr Leu Val Cys Leu
130 135 140
Ala Thr Gly Phe Tyr Pro Asp His Val Glu Leu Ser Trp Trp Val Asn
145 150 155 160
Gly Lys Glu Val His Ser Gly Val Cys Thr Asp Pro Gln Pro Leu Lys
165 170 175
Glu Gln Pro Ala Leu Asn Asp Ser Arg Tyr Ala Leu Ser Ser Arg Leu
180 185 190
Arg Val Ser Ala Thr Phe Trp Gln Asp Pro Arg Asn His Phe Arg Cys
195 200 205
Gln Val Gln Phe Tyr Gly Leu Ser Glu Asn Asp Glu Trp Thr Gln Asp
210 215 220
Arg Ala Lys Pro Val Thr Gln Ile Val Ser Ala Glu Ala Trp Gly Arg
225 230 235 240
Ala Asp
<210> 29
<211> 726
<212> DNA
<213> artificial sequence
<220>
<223> soluble TCR beta chain
<400> 29
gatgcaggtg ttacccaaag tcccacacac ctgatcaaaa cgagaggaca gcaagtgact 60
ctgagatgct ctcctaagtc tgggcatgac actgtgtcct ggtaccaaca ggccctgggt 120
caggggcccc agtttatctt tcagtattat gaggaggaag agagacagag aggcaacttc 180
cctgatcgat tctcaggtca ccagttccct aactatagct ctgagctgaa tgtgaacgcc 240
ttgttgctgg gggactcggc cctctatctc tgtgccagca gcttggtagg gttcactgaa 300
gctttctttg gacaaggcac cagactcaca gttgtagaag atctgaaaaa tgtgtttccg 360
ccggaagtcg cggtgttcga accgtcggaa gccgaaatta gccataccca gaaagcaacg 420
ctggtgtgcc tggctaccgg cttttatccg gatcatgtgg aactgtcctg gtgggttaac 480
ggcaaagaag tgcactcagg tgtttgtacg gatccgcagc cgctgaaaga acaaccggca 540
ctgaatgact cgcgttatgc tctgagttcc cgtctgcgcg ttagcgccac cttctggcag 600
gatccgcgta accactttcg ctgtcaggtc caattctacg gcctgtccga aaatgatgaa 660
tggacccagg accgtgcaaa accggtcacg caaatcgtgt cagcagaagc ttggggtcgt 720
gcagat 726

Claims (27)

1. A T Cell Receptor (TCR) capable of binding to the VFSTVPPAFI-HLA a2402 complex, the TCR comprising a TCR a chain variable domain and a TCR β chain variable domain, wherein the 3 Complementarity Determining Regions (CDRs) of the TCR a chain variable domain are:
Figure FDA0003203291120000011
the 3 complementarity determining regions of the TCR β chain variable domain are:
βCDR1-SGHDT(SEQ ID NO:13)
βCDR2-YYEEEE(SEQ ID NO:14)
βCDR3-CASSLVGFTEAFF(SEQ ID NO:15)。
2. a TCR as claimed in claim 1 which comprises a TCR α chain variable domain which is an amino acid sequence having at least 90% sequence identity to SEQ ID No. 1 and a TCR β chain variable domain; and/or the TCR β chain variable domain is identical to SEQ ID NO:5 an amino acid sequence having at least 90% sequence identity.
3. A TCR as claimed in claim 1 which comprises the α chain variable domain amino acid sequence SEQ ID NO 1.
4. A TCR as claimed in claim 1 which comprises the β chain variable domain amino acid sequence SEQ ID NO 5.
5. A TCR as claimed in claim 1 which is an α β heterodimer comprising a TCR α chain constant region TRAC 01 and a TCR β chain constant region TRBC1 01 or TRBC2 01.
6. A TCR as claimed in claim 1 wherein the α chain amino acid sequence of the TCR is SEQ ID NO:3 and/or the beta chain amino acid sequence of the TCR is SEQ ID NO 7.
7. A TCR as claimed in claim 1 which is formed by the α chain variable domain linked to the β chain variable domain by a peptide linker sequence.
8. A TCR as claimed in claim 1 which comprises (a) all or part of a TCR α chain, excluding the transmembrane domain; and (b) all or part of a TCR β chain, excluding the transmembrane domain;
and (a) and (b) each comprise a functional variable domain, or,
(a) and (b) each comprises a functional variable domain and further comprises at least a portion of the TCR chain constant domain.
9. A TCR as claimed in claim 1 in which the α chain amino acid sequence of the TCR is SEQ ID No. 26 and/or the β chain amino acid sequence of the TCR is SEQ ID No. 28.
10. A TCR as claimed in claim 1 which comprises an artificial interchain disulphide bond between the α chain variable region and the β chain constant region of the TCR.
11. A TCR as claimed in claim 1 which comprises an α chain variable domain and a β chain variable domain and all or part of the β chain constant domain, excluding the transmembrane domain, but which does not comprise an α chain constant domain, the α chain variable domain of the TCR forming a heterodimer with the β chain.
12. A TCR as claimed in claim 1 wherein a conjugate is attached to the C-or N-terminus of the α and/or β chains of the TCR.
13. A TCR as claimed in claim 12 wherein the conjugate which binds to the T cell receptor is a detectable label, a therapeutic agent, a PK modifying moiety or a combination thereof.
14. A TCR as claimed in claim 13 wherein the therapeutic agent is an anti-CD 3 antibody.
15. A multivalent TCR complex comprising at least two TCR molecules, and wherein at least one TCR molecule is a TCR as claimed in any one of claims 1 to 14.
16. A nucleic acid molecule comprising a nucleic acid sequence encoding a TCR according to any one of claims 1 to 14 or the complement thereof.
17. The nucleic acid molecule of claim 16, wherein said nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 2.
18. the nucleic acid molecule of claim 16 or 17, wherein said nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 6.
19. the nucleic acid molecule of claim 16, wherein the nucleic acid molecule comprises the nucleotide sequence encoding a TCR α chain of SEQ ID NO:4 and/or comprises the nucleotide sequence encoding the TCR β chain SEQ ID NO: 8.
20. a vector comprising the nucleic acid molecule of any one of claims 16-19.
21. The vector of claim 20, wherein said vector is a viral vector.
22. The vector of claim 20, wherein said vector is a lentiviral vector.
23. An isolated host cell comprising the vector or chromosome of any one of claims 20-22 and the exogenous nucleic acid molecule of any one of claims 16-19 integrated therein.
24. A cell transduced with the nucleic acid molecule of any one of claims 16 to 19 or the vector of any one of claims 20 to 22.
25. The cell of claim 24, wherein the cell is a T cell or a stem cell.
26. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a TCR according to any one of claims 1 to 14, a TCR complex according to claim 15, a nucleic acid molecule according to any one of claims 16 to 19, a vector according to any one of claims 20 to 22, or a cell according to claim 24 or 25.
27. Use of a TCR as claimed in any one of claims 1 to 14 or a TCR complex as claimed in claim 15, a nucleic acid molecule as claimed in any one of claims 16 to 19, a vector as claimed in any one of claims 20 to 22 or a cell as claimed in claim 24 or 25 in the manufacture of a medicament for the treatment of a tumour.
CN201710570782.6A 2017-07-13 2017-07-13 T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor Active CN109251243B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201710570782.6A CN109251243B (en) 2017-07-13 2017-07-13 T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710570782.6A CN109251243B (en) 2017-07-13 2017-07-13 T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor

Publications (2)

Publication Number Publication Date
CN109251243A CN109251243A (en) 2019-01-22
CN109251243B true CN109251243B (en) 2021-10-19

Family

ID=65051172

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710570782.6A Active CN109251243B (en) 2017-07-13 2017-07-13 T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor

Country Status (1)

Country Link
CN (1) CN109251243B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110974963B (en) * 2019-12-13 2021-05-18 上海交通大学医学院附属第九人民医院 Use of a substance for modulating SAGE1-INTS3 complex expression and/or function
CN110960677B (en) * 2019-12-13 2021-07-06 上海交通大学医学院附属第九人民医院 Use of SAGE1 inhibitor in preparation of medicine or kit

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1714102A (en) * 2002-11-09 2005-12-28 阿维德克斯有限公司 T cell receptor display
CN101155829A (en) * 2005-04-01 2008-04-02 阿维德克斯有限公司 High affinity HIV T cell receptors
CN101389652A (en) * 2005-05-25 2009-03-18 麦迪金有限公司 T cell receptors which specifically bind to VYGFVRACL-HLA-A24
CN105683215A (en) * 2013-06-26 2016-06-15 广州市香雪制药股份有限公司 High-stability T-cell receptor and preparation method and application thereof
CN106519019A (en) * 2015-11-06 2017-03-22 广州市香雪制药股份有限公司 TCR for identifying PRAME antigen
CN106831978A (en) * 2015-12-04 2017-06-13 中国科学院广州生物医药与健康研究院 Recognize the φt cell receptor of DAGE

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1714102A (en) * 2002-11-09 2005-12-28 阿维德克斯有限公司 T cell receptor display
CN101155829A (en) * 2005-04-01 2008-04-02 阿维德克斯有限公司 High affinity HIV T cell receptors
CN101389652A (en) * 2005-05-25 2009-03-18 麦迪金有限公司 T cell receptors which specifically bind to VYGFVRACL-HLA-A24
CN105683215A (en) * 2013-06-26 2016-06-15 广州市香雪制药股份有限公司 High-stability T-cell receptor and preparation method and application thereof
CN106519019A (en) * 2015-11-06 2017-03-22 广州市香雪制药股份有限公司 TCR for identifying PRAME antigen
CN106831978A (en) * 2015-12-04 2017-06-13 中国科学院广州生物医药与健康研究院 Recognize the φt cell receptor of DAGE

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HLA-A2限制性survivin点突变抗原肽特异性 TCR 基因筛选及抗肿瘤功能验证;林燕梅;《中国优秀硕士学位论文全文数据库 医药卫生科技辑》;20140115(第1期);第1-77页 *
Identification on a Human Sarcoma of Two New Genes with Tumor-specific Expression;Vale´rie Martelange等;《CANCER RESEARCH》;20000715;第60卷(第14期);第3848-3855页 *
Lack of ADAM2, CALR3 and SAGE1 Cancer/Testis Antigen Expression in Lung and Breast Cancer;Emeaga Maheswaran等;《PLOS ONE 》;20150807;第10卷(第8期);第1-10页 *

Also Published As

Publication number Publication date
CN109251243A (en) 2019-01-22

Similar Documents

Publication Publication Date Title
EP3858852B1 (en) T-cell receptor recognizing ssx2 antigen
CN112646024B (en) T cell receptor for identifying KRAS mutation and coding sequence thereof
WO2017076308A1 (en) Tcr for identifying ny-eso-1 antigen oligopeptide
CN106749620B (en) T cell receptor for recognizing MAGE-A1 antigen short peptide
CN106831978B (en) T cell receptor recognizing PRAME antigen
CN110272482B (en) T cell receptor recognizing PRAME antigen short peptide
CN110343167B (en) T cell receptor recognizing SSX2 antigen short peptide
CN110776562B (en) T cell receptor for identifying AFP antigen
CN110343166B (en) T cell receptor recognizing AFP antigen short peptides
CN110577591B (en) T cell receptor for identifying AFP antigen short peptide and its coding sequence
CN113321726A (en) T cell receptor for identifying HPV
CN109575121B (en) T cell receptor recognizing AFP antigen short peptides
EP4006050A1 (en) T cell receptor for recognizing ssx2 antigen short peptide
CN109251244B (en) TCR (T cell receptor) for recognizing LMP1 antigen derived from EBV (Epstein-Barr Virus) membrane protein
WO2016177195A1 (en) T cell receptor for recognizing rhamm antigen short-chain polypeptide
CN109251243B (en) T cell receptor for recognizing SAGE1 antigen and nucleic acid for encoding receptor
CN113321727A (en) T cell receptor for identifying AFP antigen short peptide and its coding sequence
CN113321725A (en) T cell receptor for identifying AFP
CN107987156B (en) TCR for recognizing SAGE1 antigen short peptide
CN109400697B (en) TCR (T cell receptor) for identifying PRAME (platelet-activating antigen) short peptide and related composition thereof
CN109890839B (en) T cell receptor recognizing SAGE1 antigen short peptide
CN108659114B (en) TCR for recognizing PASD1 antigen short peptide
CN110577590B (en) TCR capable of recognizing AFP antigen and encoding nucleic acid thereof
CN110627894B (en) T cell receptor for recognizing NY-ESO-1 antigen short peptide and coding sequence thereof
CN108929378B (en) T cell receptor for recognizing PRAME antigen and nucleic acid for encoding receptor

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