CN113801217A - High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen - Google Patents

High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen Download PDF

Info

Publication number
CN113801217A
CN113801217A CN202010556192.XA CN202010556192A CN113801217A CN 113801217 A CN113801217 A CN 113801217A CN 202010556192 A CN202010556192 A CN 202010556192A CN 113801217 A CN113801217 A CN 113801217A
Authority
CN
China
Prior art keywords
ser
tcr
gly
leu
ala
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010556192.XA
Other languages
Chinese (zh)
Inventor
李懿
李小林
战凯
孙含丽
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.)
Xiangxue Life Science Technology Guangdong Co ltd
Original Assignee
Xiangxue Life Science Technology Guangdong Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Xiangxue Life Science Technology Guangdong Co ltd filed Critical Xiangxue Life Science Technology Guangdong Co ltd
Priority to CN202010556192.XA priority Critical patent/CN113801217A/en
Priority to PCT/CN2021/100718 priority patent/WO2021254458A1/en
Publication of CN113801217A publication Critical patent/CN113801217A/en
Pending legal-status Critical Current

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
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

The present invention provides a T Cell Receptor (TCR) having the property of binding to the YMLDLQPET-HLA a0201 complex; and the binding affinity of the TCR to the YMLDLQPET-HLA A0201 complex is at least 2-fold greater than the binding affinity of a wild-type TCR to the YMLDLQPET-HLA A0201 complex. The invention also provides fusion molecules of such TCRs with therapeutic agents. Such TCRs can be used alone or in combination with therapeutic agents to target YMLDLQPET-HLA a0201 complex presenting tumor cells.

Description

High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen
Technical Field
The present invention relates to the field of biotechnology, and more specifically to T Cell Receptors (TCRs) capable of recognizing polypeptides derived from HPV 16E 7 protein. The invention also relates to the preparation and use of said receptors.
Background
Only two types of molecules are able to recognize antigens in a specific manner. One of which is an immunoglobulin or antibody; the other is the T Cell Receptor (TCR), which is a glycoprotein on the surface of cell membranes that exists as a heterodimer from the α chain/β chain or the γ chain/δ chain. The composition of the TCR repertoire of the immune system is produced by v (d) J recombination in the thymus, followed by positive and negative selection. In the peripheral environment, TCRs mediate the specific recognition of the major histocompatibility complex-peptide complex (pMHC) by T cells, and are therefore critical for the cellular immune function of the immune system.
TCRs are the only receptors for specific antigenic peptides presented on the Major Histocompatibility Complex (MHC), and such exogenous or endogenous peptides may be the only signs of cellular abnormalities. 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 responses, thereby allowing T cells of different antigen specificities to exert immune effects on their target cells.
The MHC class I and II molecular ligands corresponding to the TCR are also proteins of the immunoglobulin superfamily but are specific for presentation of antigens, with different individuals having different MHC, and thus are capable of presenting different short peptides of a single protein antigen to the surface of respective APC cells. Human MHC is commonly referred to as an HLA gene or HLA complex.
The HPV 16E 7 gene is one of the early region genes of the Human Papilloma Virus (HPV) genome, and encodes an E7 protein which is a small acidic protein containing about 100 amino acids. The high-risk HPV 16E 7 protein is an important reason for inducing tumors such as cervical cancer, head and neck tumor, anal cancer and the like. HPV 16E 7 is degraded into small polypeptides after intracellular production and forms complexes with MHC (major histocompatibility complex) molecules, which are presented on the cell surface. YMLDLQPET is a short peptide derived from the HPV 16E 7 antigen, which is a target for the treatment of HPV 16E 7 related diseases.
Thus, the YMLDLQPET-HLA A0201 complex provides a marker for targeting of TCR to tumor cells. The TCR capable of combining the YMLDLQPET-HLA A0201 compound has high application value for treating tumors. For example, TCRs capable of targeting the tumor cell marker can be used to deliver cytotoxic or immunostimulatory agents to target cells, or transformed into T cells, enabling T cells expressing the TCR to destroy tumor cells for administration to patients in a course of treatment known as adoptive immunotherapy. For the former purpose, the ideal TCR is of higher affinity, enabling the TCR to reside on the targeted cell for a long period of time. For the latter purpose, it is preferred to use a medium affinity TCR. Accordingly, those skilled in the art are working to develop TCRs that target tumor cell markers that can be used to meet different objectives.
Disclosure of Invention
The present invention aims to provide a TCR with higher affinity for the YMLDLQPET-HLA A0201 complex.
It is a further object of the present invention to provide a method for preparing a TCR of the above type and uses thereof.
In a first aspect of the present invention, there is provided a T Cell Receptor (TCR) comprising an alpha chain variable domain and a beta chain variable domain, which has the activity of binding YMLDLQPET-HLA a0201 complex, and the amino acid sequence of the TCR alpha chain variable domain has at least 90% sequence homology with the amino acid sequence shown in SEQ ID No. 1 and the amino acid sequence of the TCR beta chain variable domain has at least 90% sequence homology with the amino acid sequence shown in SEQ ID No. 2.
In a preferred embodiment, the amino acid sequence of the TCR a chain variable domain and the amino acid sequence of the TCR β chain variable domain are not the amino acid sequence of the wild-type TCR a chain variable domain and the amino acid sequence of the wild-type TCR β chain variable domain at the same time.
In a further preferred embodiment, the amino acid sequence of the TCR alpha chain variable domain is not the amino acid sequence shown in SEQ ID NO. 1, and/or
The amino acid sequence of the variable domain of the TCR beta chain is not the amino acid sequence shown in SEQ ID NO. 2.
In another preferred embodiment, the α chain variable domain of the TCR comprises an amino acid sequence having at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence homology to the sequence set forth in SEQ ID No. 1.
In another preferred embodiment, the β chain variable domain of the TCR is an amino acid sequence having at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence homology to the sequence set forth in SEQ ID No. 2.
In another preferred embodiment, the amino acid sequence of the TCR α chain variable domain has at least 95% sequence homology with the amino acid sequence set forth in SEQ ID NO. 1.
In another preferred embodiment, the amino acid sequence of the variable domain of the TCR β chain has at least 95% sequence homology with the amino acid sequence set forth in SEQ ID NO. 2.
In another preferred embodiment, the TCR has at least 2-fold greater affinity for the YMLDLQPET-HLA a0201 complex than a wild-type TCR.
In another preferred embodiment, the reference sequence of the 3 CDR regions (complementarity determining regions) of the variable domain of the TCR alpha chain is as follows,
CDR1α:DRVSQS
CDR2α:IYSNGD
CDR3 α: AVNPRYGNKLV, and the TCR alpha chain variable domain contains at least one of the following mutations:
Figure BDA0002544355500000021
Figure BDA0002544355500000031
in another preferred embodiment, the number of amino acid mutations in the variable domain of the TCR α chain is 2 or 3 or 4.
In another preferred embodiment, the amino acid sequence of CDR3 α of the TCR α chain variable domain is: AVNPRYGNKLV are provided.
In another preferred embodiment, the 3 CDRs of the TCR α chain variable domain are: CDR1 α: DRVSQS;
CDR2 α: IYSNGD; and CDR3 α: AVNPRYGNKLV are provided.
In another preferred embodiment, the amino acid sequence of the variable domain of the TCR alpha chain is SEQ ID NO 1.
In another preferred embodiment, the 3 CDRs of the TCR β chain variable domain are:
CDR1β:KGHDR;
CDR2 β: SFDVKD; and
CDR3 β: ATSDRGQGAFGEQY, and CDR3 β contains at least one of the following mutations:
residues before mutation Post-mutation residues
Position 6G of CDR3 beta Q
Position 7Q of CDR3 beta F or W or Y
Position 11G of CDR3 beta A
Q at position 13 of CDR3 beta Y
Position 14Y of CDR3 beta F or H
In another preferred embodiment, the amino acid mutation in CDR3 β comprises:
residues before mutation Post-mutation residues
Position 6G of CDR3 beta Q
In another preferred embodiment, the number of mutations in the CDR3 β is 1 or 2 or 3.
In another preferred example, the amino acid sequence of CDR1 β in the TCR β chain variable domain is KGHDR; and CDR2 beta amino acid sequence is SFDVKD.
In another preferred embodiment, the amino acid sequence of CDR3 β in the TCR β chain variable domain is selected from: ATSDRQYGAFGEQY, ATSDRQFGAFGEQY, and ATSDRGQGAFGEQY.
In another preferred example, the TCR α chain variable domain comprises CDR1 α, CDR2 α and CDR3 α, wherein the amino acid sequence of CDR3 α is: AVNPRYGNKLV, and the amino acid sequence of CDR1 alpha is DR [1 alpha X1] [1 alpha X2] [1 alpha X3] [1 alpha X4] and the amino acid sequence of CDR2 alpha is I [2 alpha X1] [2 alpha X2] [2 alpha X3] GD, wherein [1 alpha X1] is V or L or M or H, and/or [1 alpha X2] is A or S or T or G, and/or [1 alpha X3] is Q or N or V or Y, and/or [1 alpha X4] is S or T or A or V, and/or [2 alpha X1] is Y or F, and/or [2 alpha X2] is S or N, and/or [2 alpha X3] is N or P.
In another preferred embodiment, the TCR has a mutation in the alpha chain variable domain shown in SEQ ID NO. 1 selected from one or more of V29L/M/H, S30A/T/G, Q31N/V/Y, S32A/T/V, Y51F, S52N and N53P, wherein the numbering of the amino acid residues is as shown in SEQ ID NO. 1.
In another preferred example, the TCR β chain variable domain comprises CDR1 β, CDR2 β, and CDR3 β, wherein the amino acid sequence of CDR1 β is KGHDR, the amino acid sequence of CDR2 β is SFDVKD, and the amino acid sequence of CDR3 β is: ATSDR [3 betaX 1] [3 betaX 2] GAF [3 betaX 3] E [3 betaX 4] [3 betaX 5], wherein [3 betaX 1] is Q or G, and/or [3 betaX 2] is Y or F or Q or W, and/or [3 betaX 3] is G or A, and/or [3 betaX 4] is Q or Y, and/or [3 betaX 5] is Y or H or F.
In another preferred embodiment, the TCR has a mutation in the beta chain variable domain shown in SEQ ID NO. 2 selected from one or more of G97Q, Q98Y/W/F, G102A, Q104Y, Y105F/H, wherein the amino acid residue numbering is as shown in SEQ ID NO. 2.
In another preferred embodiment, the TCR has CDRs selected from the group consisting of:
Figure BDA0002544355500000041
in another preferred embodiment, the TCR is soluble.
In another preferred embodiment, the TCR is an α β heterodimeric TCR comprising an α chain TRAC constant region sequence and a β chain TRBC1 or TRBC2 constant region sequence.
In another preferred embodiment, the TCR comprises (i) all or part of a TCR α chain, except for its transmembrane domain, and (ii) all or part of a TCR β chain, except for its transmembrane domain, wherein both (i) and (ii) comprise the variable domain and at least part of the constant domain of the TCR chain.
In another preferred embodiment, the TCR comprises an artificial interchain disulfide bond between the α chain constant region and the β chain constant region.
In another preferred embodiment, the cysteine residues forming the artificial interchain disulfide bond between the constant regions of the TCR α and β chains replace one or more groups of sites selected from:
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 TRAC × 01 exon 1 or Glu20 of TRBC2 × 01 exon 1.
In another preferred embodiment, the amino acid sequence of the α chain variable domain of the TCR is one of SEQ ID NOs 1, 13-21; and/or the amino acid sequence of the variable domain of the beta chain of the TCR is one of SEQ ID NO 2, 22-33.
In another preferred embodiment, the TCR is selected from the group consisting of:
Figure BDA0002544355500000051
Figure BDA0002544355500000061
in another preferred embodiment, the TCR is a single chain TCR.
In another preferred embodiment, the TCR is a single chain TCR consisting of an alpha chain variable domain and a beta chain variable domain linked by a flexible short peptide sequence (linker).
In another preferred embodiment, the C-or N-terminus of the α chain and/or β chain of the TCR is conjugated to a conjugate, preferably a detectable label, a therapeutic agent, a PK modifying moiety or a combination of any of these.
In another preferred embodiment, the therapeutic agent that binds to the TCR is an anti-CD 3 antibody linked to the C-or N-terminus of the α or β chain of the TCR.
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 TCR molecule is a TCR as claimed in any one of the preceding claims.
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 a multivalent TCR complex according to the second aspect of the invention, or a complement thereof.
In a fourth aspect of the invention, there is provided a vector comprising the nucleic acid molecule of the third aspect of the invention.
In a fifth aspect of the invention, there is provided a host cell comprising a vector or chromosome of the fourth aspect of the invention and, integrated therein, an exogenous nucleic acid molecule of the third aspect of the invention.
In a sixth aspect of the invention there is provided an isolated cell expressing a TCR according to the first aspect of the invention, preferably the isolated cell is a T cell, an NK cell and an NKT cell, most preferably the isolated cell is a T 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, or a TCR complex according to the second aspect of the invention, or a cell according to the sixth aspect of the invention.
In an eighth 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 TCR according to the first aspect of the invention, or a TCR complex according to the second 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 an HPV 16E 7 positive tumour, more preferably the tumour is cervical cancer.
In a ninth aspect of the invention there is provided the use of a TCR of the first aspect of the invention, or a TCR complex of the second aspect of the invention, or a cell of the sixth aspect of the invention, in the manufacture of a medicament for the treatment of a tumour, preferably the disease is a HPV 16E 7 positive tumour, more preferably the tumour is cervical cancer.
In a tenth aspect of the invention, there is provided a method of preparing a T cell receptor according to the first aspect of the invention, comprising the steps of:
(i) culturing a host cell according to the fifth aspect of the invention, thereby expressing a T-cell receptor according to the first aspect of the invention;
(ii) isolating or purifying said T cell receptor.
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 be limited to space.
Drawings
FIGS. 1a and 1b show the amino acid sequences of the wild-type TCR alpha and beta chain variable domains, respectively, capable of specifically binding to the YMLDLQPET-HLA A0201 complex.
FIGS. 2a and 2b show the amino acid sequence of the alpha chain variable domain and the amino acid sequence of the beta chain variable domain, respectively, of a single chain template TCR constructed in accordance with the invention.
FIGS. 3a and 3b are the DNA sequences of the alpha chain variable domain and beta chain variable domain, respectively, of a single-chain template TCR constructed in accordance with the invention.
FIGS. 4a and 4b show the amino acid sequence and DNA sequence of the linker peptide (linker) of the single-chain template TCR constructed according to the present invention.
FIGS. 5a and 5b show the amino acid sequence and DNA sequence, respectively, of a single-stranded template TCR constructed in accordance with the invention.
FIGS. 6a and 6b are amino acid sequences of soluble reference TCR α and β chains, respectively, of the invention.
Fig. 7(1) - (9) show the α chain variable domain amino acid sequences of the heterodimeric TCRs with high affinity for YMLDLQPET-HLA a0201 complex, respectively, with mutated residues underlined.
Fig. 8(1) - (12) show the β chain variable domain amino acid sequences of the heterodimeric TCRs with high affinity for YMLDLQPET-HLA a0201 complex, respectively, with mutated residues underlined.
FIGS. 9a and 9b show the extracellular amino acid sequences of the wild-type TCR α chain and β chain, respectively, capable of binding specifically to the YMLDLQPET-HLA A0201 complex.
FIGS. 10a and 10b show the amino acid sequences of the wild-type TCR α chain and β chain, respectively, capable of binding specifically to the YMLDLQPET-HLA A0201 complex.
FIG. 11 is a graph of the binding of soluble reference TCR, i.e., wild-type TCR, to the YMLDLQPET-HLA A0201 complex.
Fig. 12a and 12b are experimental results of the activation function of effector cells transfected with the high affinity TCR of the present invention against short peptide-loaded T2 cells.
FIGS. 13a and 13b are results of experiments on the activation function of effector cells transfected with the high affinity TCR of the invention against tumor cell lines.
Fig. 14 is an experimental result of the killing function of effector cells transfected with the high affinity TCR of the present invention against gradient short peptide-loaded T2 cells.
Fig. 15a, 15b and 15c are results of LDH experiments with the killing function of effector cells transfected with the high affinity TCRs of the invention against tumor cell lines.
Fig. 16a and 16b are results of the killing function IncuCyte experiment on effector cells transfected with the high affinity TCR of the invention against tumor cell lines.
Detailed Description
The present inventors, through extensive and intensive studies, have obtained a high-affinity T Cell Receptor (TCR) recognizing YMLDLQPET short peptide (derived from HPV 16E 7 protein), said YMLDLQPET short peptide being presented in the form of a peptide-HLA a0201 complex. The high affinity TCR has 3 CDR regions in its alpha chain variable domain:
CDR1α:DRVSQS
CDR2α:IYSNGD
CDR3 α: AVNPRYGNKLV; and/or in the 3 CDR regions of its beta chain variable domain:
CDR1β:KGHDR
CDR2β:SFDVKD
CDR3 β: ATSDRGQGAFGEQY; and, the affinity and/or binding half-life of the inventive TCR after mutation to the YMLDLQPET-HLA a0201 complex described above is at least 2-fold that of the wild-type TCR.
Before the present invention is described, it is to be understood that this invention is not limited to the particular methodology and experimental conditions described, as such methodologies and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now exemplified.
Term(s) for
T Cell Receptor (TCR)
The TCR may be described using the international immunogenetics information system (IMGT). Native α β heterodimeric TCRs have an α chain and a β chain. In a broad sense, each chain comprises a variable region, a linker region and a constant region, and the beta chain also typically contains a short diversity region between the variable region and the linker region, but the diversity region is often considered part of the linker region. The TCR connecting region is defined by the unique TRAJ and TRBJ of IMGT, and the TCR constant region is defined by the TRAC and TRBC of IMGT.
Each variable region comprises 3 CDRs (complementarity determining regions), CDR1, CDR2, and CDR3, chimeric in a framework sequence. In the IMGT nomenclature, the different numbers of TRAV and TRBV refer to different types of V α and V β, respectively. In the IMGT system, the α chain constant domain has the following symbols: TRAC 01, wherein "TR" denotes a T cell receptor gene; "A" represents an alpha chain gene; c represents a constant region; ". 01" indicates allele 1. The beta-strand constant domain has the following symbols: TRBC1 x 01 or TRBC2 x 01, wherein "TR" denotes a T cell receptor gene; "B" represents a beta chain gene; c represents a constant region; ". 01" indicates allele 1. The constant region of the alpha chain is uniquely defined, and in the form of the beta chain, there are two possible constant region genes, "C1" and "C2". The constant region gene sequences of the TCR alpha and beta chains can be obtained by those skilled in the art from published IMGT databases.
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 is composed of linked variable regions and linked regions. Thus, in the description and claims of this application, the "TCR α chain variable domain" refers to the linked TRAV and TRAJ regions, and likewise the "TCR β chain variable domain" refers to the linked TRBV and TRBD/TRBJ regions. The 3 CDRs of the TCR α chain variable domain are CDR1 α, CDR2 α and CDR3 α, respectively; the 3 CDRs of the TCR β chain variable domain are CDR1 β, CDR2 β and CDR3 β, respectively. The framework sequences of the TCR variable domains of the invention may be murine or human, preferably human. The constant domain of the TCR comprises an intracellular portion, a transmembrane region, and an extracellular portion.
In the present invention, the amino acid sequences of the α and β chain variable domains of the wild-type TCR capable of binding YMLDLQPET-HLA A0201 complex are SEQ ID NO 1 and SEQ ID NO 2, respectively, as shown in FIG. 1a and FIG. 1 b. The alpha chain amino acid sequence and the beta chain amino acid sequence of the soluble "reference TCR" are respectively SEQ ID NO. 11 and SEQ ID NO. 12, as shown in FIG. 6a and FIG. 6 b. The extracellular amino acid sequence of the alpha chain and the extracellular amino acid sequence of the beta chain of the wild-type TCR are respectively SEQ ID NO. 34 and SEQ ID NO. 35, as shown in FIG. 9a and FIG. 9 b. The TCR sequences used in the present invention are of human origin. The alpha chain amino acid sequence and the beta chain amino acid sequence of the wild-type TCR are respectively SEQ ID NO 36 and SEQ ID NO 37, as shown in FIGS. 10a and 10 b. 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, the amino acid sequences of TRAC 01 and TRBC1 × 01 or TRBC2 × 01 are position-numbered in the order from the N-terminus to the C-terminus, for example, in TRBC1 × 01 or TRBC2 × 01, 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 may be described as TRBC1 × 01 or TRBC2 × 01 exon 1 Pro60, and also as TRBC1 × 01 or TRBC2 × 01 exon 1, and further, in TRBC1 × 01 or TRBC2 × 01, the 61 st amino acid in the order from the N-terminus to the C-terminus is Q (glutamine), and thus, in the present invention, it may be described as TRBC1 × 01 or TRBC2, and further, as glbc 8201, and similarly, it may be described as TRBC1, glbc 6301, and further, as TRBC 8561 st amino acid. 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, position number 46 listed in IMGT, 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.
Tumor(s)
The term "tumor" is meant to include all types of cancer cell growth or carcinogenic processes, metastatic or malignantly transformed cells, tissues or organs, regardless of the type of pathology or the stage of infestation. Examples of tumors include, but are not limited to: solid tumors, soft tissue tumors, and metastatic lesions. Examples of solid tumors include: malignancies of different organ systems, such as sarcomas, squamous carcinomas of the lung and cancers. For example: infected prostate, lung, breast, lymph, gastrointestinal (e.g., colon), and genitourinary tract (e.g., kidney, epithelial cells), pharynx. Squamous carcinoma of the lung includes malignant tumors, such as, for example, most colon cancers, rectal cancers, renal cell carcinomas, liver cancers, non-small cell carcinomas of the lung, small bowel cancers, and esophageal cancers. Metastatic lesions of the above-mentioned cancers can likewise be treated and prevented using the methods and compositions of the present invention.
Detailed Description
It is well known that the α chain variable domain and β chain variable domain of a TCR each contain 3 CDRs, similar to the complementarity determining regions of an antibody. CDR3 interacts with antigen short peptides, CDR1 and CDR2 interact with HLA. Thus, the CDRs of the TCR molecule determine their interaction with the antigen short peptide-HLA complex. The amino acid sequences of the alpha chain variable domain and the beta chain variable domain of the wild-type TCR capable of binding the antigen short peptide YMLDLQPET and the HLA A0201 complex (i.e., YMLDLQPET-HLA A0201 complex) are SEQ ID NO 1 and SEQ ID NO 2, respectively, which was discovered by the inventors for the first time. It has the following CDR regions:
alpha chain variable domain CDR1 a: DRVSQS
CDR2α:IYSNGD
CDR3α:AVNPRYGNKLV
And the beta chain variable domain CDR1 beta: KGHDR
CDR2 β: SFDVKD and
CDR3β:ATSDRGQGAFGEQY。
the invention obtains the high affinity TCR with the affinity of YMLDLQPET-HLA A0201 complex being at least 2 times that of the wild type TCR and YMLDLQPET-HLA A0201 complex by carrying out mutation screening on the CDR region.
Further, the TCR of the invention is an α β heterodimeric TCR, the α chain variable domain of which comprises at least 85% of the amino acid sequence set forth in SEQ ID No. 1; preferably, at least 90%; more preferably, at least 92%; more preferably, at least 94% (e.g., can be at least 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence homology) of sequence homology; and/or the β chain variable domain of the TCR comprises at least 90%, preferably at least 92%, of the amino acid sequence set forth as SEQ ID No. 2; more preferably, at least 94% (e.g., may be at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence homology) of sequence homology.
Further, the TCR of the invention is a single chain TCR, the α chain variable domain of which comprises at least 85%, preferably at least 90% of the amino acid sequence shown in SEQ ID No. 3; more preferably, at least 92%; most preferably, at least 94% (e.g., can be at least 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence homology) of sequence homology; and/or the β chain variable domain of the TCR comprises at least 85%, preferably at least 90% of the amino acid sequence set forth as SEQ ID No. 4; more preferably, at least 92%; most preferably, at least 94%; (e.g., can be at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence homology) of sequence homology.
The 3 CDRs of the variable domain of the wild-type TCR alpha chain of SEQ ID NO. 1, i.e., CDR1, CDR2 and CDR3, are located at positions 27-32, 50-55 and 90-100 of SEQ ID NO. 1, respectively. Accordingly, the amino acid residue numbering is as shown in SEQ ID NO 1, wherein 29V is the 3 rd position V of CDR1 alpha, 30S is the 4 th position S of CDR1 alpha, 31Q is the 5 th position Q of CDR1 alpha, 32S is the 6 th position S of CDR1 alpha, 51Y is the 2 nd position Y of CDR2 alpha, 52S is the 3 rd position S of CDR2 alpha, and 53N is the 4 th position N of CDR2 alpha.
The invention provides a TCR having the property of binding YMLDLQPET-HLA a0201 complex and comprising an alpha chain variable domain and a beta chain variable domain, wherein the TCR is mutated in the alpha chain variable domain as shown in SEQ ID No. 1, the mutated amino acid residue positions comprising one or more of 29V, 30S, 31Q, 32S, 51Y, 52S and 53N, wherein the numbering of the amino acid residues is as shown in SEQ ID No. 1.
Preferably, the TCR α chain variable domain after mutation comprises one or more amino acid residues selected from the group consisting of: 29L or 29M or 29H; 30A or 30T or 30G; 31N or 31V or 31Y; 32A or 32T or 32V; 51F; 52N; 53P, wherein the numbering of the amino acid residues adopts the numbering shown in SEQ ID NO. 1.
More specifically, the specific forms of the mutations described in the variable domains of the alpha chain include one or several of V29L/M/H, S30A/T/G, Q31N/V/Y, S32A/T/V, Y51F, S52N and N53P.
The 3 CDRs of the variable domain of wild-type TCR beta chain of the invention SEQ ID NO. 2, i.e., CDR1, CDR2 and CDR3, are located at positions 27-31, 49-54 and 92-105 of SEQ ID NO. 2, respectively. Accordingly, the amino acid residue numbering is as shown in SEQ ID NO. 2, with 97G being the 6 th G and 98Q of CDR3 β being the 7 th Q of CDR3 β, 102G being the 11 th G and 104Q of CDR3 β being the 13 th Q of CDR3 β, and 105Y being the 14 th Y of CDR3 β.
The invention provides a TCR having the property of binding YMLDLQPET-HLA a0201 complex and comprising a β chain variable domain and a β chain variable domain, wherein the TCR is mutated in the β chain variable domain as shown in SEQ ID No. 2, the mutated amino acid residue positions comprising one or more of 97G, 98Q, 102G, 104Q, 105Y, wherein the numbering of the amino acid residues is as shown in SEQ ID No. 2.
Preferably, the TCR β chain variable domain after mutation comprises one or more amino acid residues selected from the group consisting of: 97Q; 98Y or 98W or 98F; 102A; 104Y; 105F or 105H, wherein the numbering of the amino acid residues is as shown in SEQ ID NO 2.
More specifically, the specific forms of the mutations described in the variable domains of the beta chain include one or several of G97Q, Q98Y/W/F, G102A, Q104Y, Y105F/H.
It should be understood that the amino acid names herein are given by the international common single-letter symbols, and the three-letter symbols corresponding to the amino acid names are: 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);
in the present invention, Pro60 or 60P both represent proline at position 60. In addition, the expression of a specific form of the mutation described in the present invention, for example, "G97Q" means that G at position 97 is substituted with Q, and so on.
The reference TCR was obtained by mutating Thr48 of exon 1 of the α chain constant region TRAC 01 of the wild-type TCR to cysteine and Ser57 of exon 1 of the β chain constant region TRBC1 x 01 or TRBC2 x 01 to cysteine according to site-directed mutagenesis methods well known to those skilled in the art, the amino acid sequences of which are SEQ ID NO:11 and SEQ ID NO:12, respectively, as shown in fig. 6a and 6b, and the mutated cysteine residues are shown in bold letters. The cysteine substitutions described above enable the formation of artificial interchain disulfide bonds between the constant regions of the α and β chains of the reference TCR to form a more stable soluble TCR, thereby enabling a more convenient assessment of the binding affinity and/or binding half-life between the TCR and the YMLDLQPET-HLA a0201 complex. It will be appreciated that the CDR regions of the TCR variable region determine their affinity for the pMHC complex and therefore cysteine substitutions in the TCR constant region as described above do not have an effect on the binding affinity and/or binding half-life of the TCR. Therefore, in the present invention, the binding affinity measured between the reference TCR and the YMLDLQPET-HLA A0201 complex is considered to be the binding affinity between the wild-type TCR and the YMLDLQPET-HLA A0201 complex. Similarly, if the binding affinity between the inventive TCR and the YMLDLQPET-HLA A0201 complex is determined to be at least 10 times greater than the binding affinity between the reference TCR and the YMLDLQPET-HLA A0201 complex, i.e. equivalent to the binding affinity between the inventive TCR and the YMLDLQPET-HLA A0201 complex being at least 10 times greater than the binding affinity between the wild-type TCR and the YMLDLQPET-HLA A0201 complex.
Binding affinity (equilibrium constant K to dissociation) can be determined by any suitable methodDInversely proportional) and binding half-life (denoted T)1/2) For example, surface plasmon resonance. It will be appreciated that doubling the affinity of the TCR will result in KDAnd (4) halving. T is1/2Calculated as In2 divided by dissociation rate (K)off). Thus, T1/2Doubling can result in KoffAnd (4) halving. Preferably, the binding affinity or binding half-life of a given TCR is measured several times, e.g. 3 times or more, using the same assay protocol, and the results are averaged. In a preferred embodiment, the affinity of the soluble TCR is measured using the surface plasmon resonance (BIAcore) method in the examples herein, provided that: the temperature is 25 ℃, and the PH value is 7.1-7.5. The method detects the dissociation equilibrium constant K of the reference TCR to YMLDLQPET-HLA A0201 complexD9.37E-05M, 93.7. mu.M, the dissociation equilibrium constant K of the wild-type TCR for the YMLDLQPET-HLA A0201 complex is considered in the present inventionDAlso 93.7. mu.M. Doubling of the affinity due to TCR will result in KDHalving, so if the dissociation equilibrium constant K of the high affinity TCR for the YMLDLQPET-HLA A0201 complex is detectedD9.37E-06M, i.e., 9.37. mu.M, indicates that the affinity of the high affinity TCR for the YMLDLQPET-HLA A0201 complex is wild typeTCR 10 times affinity for YMLDLQPET-HLA A0201 complex. K is well known to those skilled in the artDConversion between units of value, i.e. 1M 106μ M, 1000nM for 1 μ M and 1000pM for 1 nM. In the present invention, the affinity of the TCR to the YMLDLQPET-HLA a0201 complex is at least 2-fold that of the wild-type TCR.
The mutation may be performed by any suitable method, including but not limited to those according to the Polymerase Chain Reaction (PCR), cloning according to restriction enzymes, or Ligation Independent Cloning (LIC) methods. These methods are detailed in a number of standard molecular biology texts. For more details on Polymerase Chain Reaction (PCR) mutagenesis and Cloning by restriction enzymes, see Sambrook and Russell, (2001) Molecular Cloning-A Laboratory Manual (third edition) CSHL Press. More information on the LIC method can be found (Rashtchian, (1995) Curr Opin Biotechnol 6(1): 30-6).
The method of generating the TCRs of the invention may be, but is not limited to, screening a diverse library of phage particles displaying such TCRs for a TCR with high affinity for the YMLDLQPET-HLA-A0201 complex, as described in the literature (Li, et al (2005) Nature Biotech 23(3): 349-.
It will be appreciated that genes expressing the α and β chain variable domain amino acids of a wild type TCR, or genes expressing slightly modified α and β chain variable domain amino acids of a wild type TCR, may be used to make a template TCR. The alterations required to produce the high affinity TCRs of the invention are then introduced into the DNA encoding the variable domain of the template TCR.
The high affinity TCR of the invention comprises an alpha chain variable domain amino acid sequence of one of SEQ ID NO 1, 13-21; and/or the amino acid sequence of the variable domain of the beta chain of the TCR is one of SEQ ID NO 2, 22-33. In the present invention, the amino acid sequences of the α chain variable domain and the β chain variable domain that form the hetero-dimeric TCR molecule are preferably selected from table 1 below:
TABLE 1
Figure BDA0002544355500000131
Figure BDA0002544355500000141
For the purposes of the present invention, the inventive TCRs are moieties having at least one TCR α and/or TCR β chain variable domain. They typically comprise both a TCR α chain variable domain and a TCR β chain variable domain. They may be α β heterodimers or in single-stranded form or in any other form that is stable. In adoptive immunotherapy, the full-length chain (comprising the cytoplasmic and transmembrane domains) of the α β heterodimeric TCR can be transfected. The TCRs of the invention are useful as targeting agents for delivering therapeutic agents to antigen presenting cells or in combination with other molecules to produce bifunctional polypeptides for targeting effector cells, where the TCRs are preferably in soluble form.
For stability, it is disclosed in the prior art that introduction of artificial interchain disulfide bonds between the α and β chain constant domains of the TCR enables soluble and stable TCR molecules to be obtained, as described in patent document PCT/CN 2015/093806. Thus, the inventive TCR may be one in which an artificial interchain 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 substitution of Thr48 for exon 1 of TRAC × 01 and a substitution of Ser57 for exon 1 of TRBC1 × 01 or TRBC2 × 01 form disulfide bonds. 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 15, or up to 10, or up to 8 or fewer amino acids, so as not to include cysteine residues for the purpose of deleting the native interchain disulfide bond, or by mutating the cysteine residue forming the native interchain disulfide bond to another amino acid.
As noted above, the TCRs of the invention may comprise artificial interchain 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 interchain disulfide bonds present in the TCR.
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 α chain variable region and the β chain constant region of the high affinity TCR of the invention may also comprise an artificial interchain disulfide bond. 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.
For stability, on the other hand, the inventive TCR also includes TCRs having mutations in their hydrophobic core region, preferably mutations that improve the stability of the inventive 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.
More specifically, the TCR with the mutated hydrophobic core region of the present invention may be a high stability single chain TCR comprising a flexible peptide chain connecting the variable domains of the α chain and β chain of the TCR. The CDR regions of the TCR variable region determine the affinity of the TCR variable region with the short peptide-HLA complex, and the mutation of the hydrophobic core can stabilize the TCR without affecting the affinity of the TCR variable region with the short peptide-HLA complex. 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. The template chain for screening high affinity TCR constructed in example 1 of the present invention is the above-described high stability single chain TCR comprising the hydrophobic core mutation. Using a TCR with higher stability, the affinity between the TCR and the YMLDLQPET-HLA-A0201 complex can be more conveniently assessed.
The CDR regions of the alpha chain variable domain and the beta chain variable domain of the single chain template TCR are completely identical to the CDR regions of the wild type TCR. That is, the 3 CDRs of the α chain variable domain are CDR1 α: DRVSQS; CDR2 α: IYSNGD; CDR3 α: AVNPRYGNKLV and the 3 CDRs of the β chain variable domain are CDR1 β: KGHDR; CDR2 β: SFDVKD; CDR3 β: ATSDRGQGAFGEQY are provided. The amino acid sequence (SEQ ID NO:9) and the nucleotide sequence (SEQ ID NO:10) of the single-chain template TCR are shown in FIGS. 5a and 5b, respectively. Thus, a single-chain TCR composed of an alpha chain variable domain and a beta chain variable domain having high affinity for the YMLDLQPET-HLA A0201 complex was selected.
The α β heterodimer having high affinity for the YMLDLQPET-HLA-A0201 complex of the present invention was obtained by transferring the CDR regions of the α and β chain variable domains of the selected high affinity single-chain TCR to the corresponding positions of the α chain variable domain (SEQ ID NO:1) and β chain variable domain (SEQ ID NO:2) of the wild-type TCR.
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, wherein the TCR is used to detect the presence of cells presenting the YMLDLQPET-HLA-a0201 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.
Antibodies or fragments thereof that bind to the TCRs of the invention include anti-T cell or NK-cell determining antibodies, such as anti-CD 3 or anti-CD 28 or anti-CD 16 antibodies, whose binding to the TCR directs effector cells to better target cells. A preferred embodiment is a TCR of the invention conjugated to an anti-CD 3 antibody or a functional fragment or variant of said anti-CD 3 antibody. Specifically, the fusion molecule of the TCR and the anti-CD 3 single-chain antibody comprises a TCR alpha chain variable domain amino acid sequence which is one of SEQ ID NO 1 and SEQ ID NO 13-21; and/or the amino acid sequence of the variable domain of the beta chain of the TCR is one of SEQ ID NO 2, 22-33.
The invention also relates to nucleic acid molecules encoding the inventive TCRs. The nucleic acid molecules of the invention may be in the form of DNA or in the form of RNA. The DNA may be the coding strand or the non-coding strand. For example, the nucleic acid sequence encoding the TCR of the invention may be identical to or a degenerate variant of the nucleic acid sequence shown in the figures of the invention. By way of illustration of the meaning of "degenerate variant", as used herein, is meant a nucleic acid sequence which encodes a protein sequence having SEQ ID NO. 3, but differs from the sequence of SEQ ID NO. 5.
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 invention also relates to vectors comprising the nucleic acid molecules of the invention, as well as to host cells genetically engineered with the vectors or coding sequences of the invention.
The invention also includes isolated cells, particularly T cells, expressing a TCR of the invention. There are many methods suitable for T cell transfection using DNA or RNA encoding the high affinity TCRs of the invention (e.g., Robbins et al, (2008) J.Immunol. 180: 6116-. T cells expressing the high affinity 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 Cancer 8(4): 299-.
The invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a TCR of the invention, or a TCR complex of the invention, or a cell presenting a TCR of the invention.
The invention also provides a method of treating a disease comprising administering to a subject in need thereof an amount of a TCR of the invention, or a TCR complex of the invention, or a cell presenting a TCR of the invention, or a pharmaceutical composition of the invention.
In the art, substitutions with amino acids of similar or similar properties will not generally alter the function of the protein. Addition of one or several amino acids at the C-terminus and/or N-terminus will not generally alter the structure and function of the protein. Thus, the inventive TCR also includes TCRs in which up to 5, preferably up to 3, more preferably up to 2, most preferably up to 1 amino acid (particularly amino acids outside the CDR regions) of the inventive TCR is replaced by amino acids having similar or analogous properties, and still retain its functionality.
The invention also includes TCRs that are slightly modified from the TCRs of the invention. Modified (generally without altering primary structure) forms include: chemically derivatized forms of the inventive TCR, such as acetylation or carboxylation. Modifications also include glycosylation, such as those that result from glycosylation modifications made during synthesis and processing or during further processing steps of the inventive TCR. Such modification may be accomplished by exposing the TCR to an enzyme that effects glycosylation, such as mammalian glycosylase or deglycosylase. Modified forms also include sequences having phosphorylated amino acid residues (e.g., phosphotyrosine, phosphoserine, phosphothreonine). Also included are TCRs that have been modified to increase their resistance to proteolysis or to optimize solubility.
The TCR of the invention, the TCR complex or the TCR-transfected T cell of the invention may be provided in a pharmaceutical composition together with a pharmaceutically acceptable carrier. The TCRs, multivalent TCR complexes or cells of the invention are typically provided as part of a sterile pharmaceutical composition, which typically includes a pharmaceutically acceptable carrier. The pharmaceutical composition may be in any suitable form (depending on the desired method of administration to the patient). It may be provided in unit dosage form, typically in a sealed container, and may be provided as part of a kit. Such kits (but not necessarily) include instructions for use. It may comprise a plurality of said unit dosage forms.
In addition, the TCRs of the invention may be used alone, or in combination or coupling with other therapeutic agents (e.g., formulated in the same pharmaceutical composition).
The pharmaceutical composition may further comprise a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" refers to a carrier for administration of a therapeutic agent. The term refers to such pharmaceutical carriers: they do not themselves induce the production of antibodies harmful to the individual receiving the composition and are not unduly toxic after administration. Such vectors are well known to those of ordinary skill in the art. A thorough discussion of pharmaceutically acceptable excipients can be found in Remington's Pharmaceutical Sciences (Mack pub. co., n.j.1991). Such vectors include (but are not limited to): saline, buffer, glucose, water, glycerol, ethanol, adjuvants, and combinations thereof.
Pharmaceutically acceptable carriers in therapeutic compositions can comprise liquids such as water, saline, glycerol and ethanol. In addition, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances and the like, may also be present in these carriers.
Generally, the therapeutic compositions can be prepared as injectables, e.g., as liquid solutions or suspensions; solid forms suitable for constitution with a solution or suspension, or liquid carrier, before injection, may also be prepared.
Once formulated, the compositions of the present invention may be administered by conventional routes including, but not limited to: ophthalmic, intramuscular, intravenous, subcutaneous, intradermal, or topical administration, preferably parenteral including subcutaneous, intramuscular, or intravenous. The subject to be prevented or treated may be an animal; especially a human.
When the pharmaceutical composition of the present invention is used for practical treatment, various dosage forms of the pharmaceutical composition may be used depending on the use case. Preferably, injections, oral agents and the like are exemplified.
These pharmaceutical compositions may be formulated by mixing, dilution or dissolution according to a conventional method, and occasionally, suitable pharmaceutical additives such as excipients, disintegrants, binders, lubricants, diluents, buffers, isotonic agents (isotonicities), preservatives, wetting agents, emulsifiers, dispersants, stabilizers and solubilizing agents are added, and the formulation process may be carried out in a conventional manner according to a dosage form.
The pharmaceutical compositions of the present invention may also be administered in the form of sustained release formulations. For example, the inventive TCR may be incorporated into a pellet or microcapsule carried by a slow-release polymer, which pellet or microcapsule is then surgically implanted into the tissue to be treated. As examples of the sustained-release polymer, ethylene-vinylacetate copolymer, polyhydroxymethacrylate, polyacrylamide, polyvinylpyrrolidone, methylcellulose, lactic acid polymer, lactic acid-glycolic acid copolymer, and the like can be exemplified, and biodegradable polymers such as lactic acid polymer and lactic acid-glycolic acid copolymer can be preferably exemplified.
When the pharmaceutical composition of the present invention is used for practical treatment, the TCR or TCR complex of the present invention or the cells presenting the TCR of the present invention as an active ingredient can be determined reasonably according to the body weight, age, sex, degree of symptoms of each patient to be treated, and finally the reasonable amount is decided by a physician.
The main advantages of the invention are:
(1) the high affinity TCRs of the invention have at least 2-fold greater affinity and/or binding half-life for the YMLDLQPET-HLA-a0201 complex than wild-type TCRs.
(2) The high affinity TCR of the invention was able to specifically bind to the YMLDLQPET-HLA a0201, while cells transfected with the high affinity TCR of the invention were able to be specifically activated.
(3) The effector cells transfected with the high affinity TCRs of the invention have a strong specific killing effect.
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.
Materials and methods
The experimental materials used in the examples of the present invention are commercially available as such, unless otherwise specified, wherein e.coli DH5 α is available from Tiangen, e.coli BL21(DE3) is available from Tiangen, e.coli Tuner (DE3) is available from Novagen, and plasmid pET28a is available from Novagen.
Example 1 Generation of Stable Single chain TCR template chains with hydrophobic core mutations
The invention utilizes a site-directed mutagenesis method, and constructs a stable single-chain TCR molecule formed by connecting TCR alpha and beta chain variable domains by a flexible short peptide (linker) according to the patent document WO2014/206304, wherein the amino acid and DNA sequences of the stable single-chain TCR molecule are SEQ ID NO:9 and SEQ ID NO:10 respectively, as shown in FIG. 5a and FIG. 5 b. And the single-chain TCR molecule is used as a template to screen the high-affinity TCR molecule. The amino acid sequences of the alpha chain variable domain (SEQ ID NO:3) and the beta chain variable domain (SEQ ID NO:4) of the template strand are shown in FIGS. 2a and 2 b; the corresponding DNA sequences are SEQ ID NO 5 and SEQ ID NO 6, respectively, as shown in FIGS. 3a and 3 b; the amino acid sequence and DNA sequence of the flexible short peptide (linker) are shown in SEQ ID NO 7 and 8, respectively, as shown in FIGS. 4a and 4 b.
The target gene carrying the template strand was digested simultaneously with Nco I and Not I, and ligated to pET28a vector digested simultaneously with Nco I and Not I. The ligation product was transformed into e.coli DH5 α, spread on LB plates containing kanamycin, cultured at 37 ℃ for overnight inversion, positive clones were selected for PCR screening, positive recombinants were sequenced, and after the sequence was determined to be correct, recombinant plasmids were extracted and transformed into e.coli BL21(DE3) for expression.
Example 2 expression, renaturation and purification of the Stable Single-chain TCR constructed in example 1
The BL21(DE3) colonies containing the recombinant plasmid pET28 a-template strand prepared in example 1 were all inoculated in LB medium containing kanamycin and cultured at 37 ℃ to OD6000.6-0.8, IPTG was added to a final concentration of 0.5mM and incubation continued at 37 ℃ for 4 h. The cell pellet was harvested by centrifugation at 5000rpm for 15min, lysed by Bugbuster Master Mix (Merck), inclusion bodies were recovered by centrifugation at 6000rpm for 15min, washed with Bugbuster (Merck) to remove cell debris and membrane fraction, and centrifuged at 6000rpm for 15min to collect the inclusion bodies. The inclusion bodies were dissolved in buffer (20mM Tris-HCl pH 8.0,8M urea), the insoluble material was removed by high speed centrifugation, the supernatant was quantified by BCA method and split charged, and stored at-80 ℃ for further use.
To 5mg of solubilized single-chain TCR inclusion body protein, 2.5mL of buffer (6M Gua-HCl, 50mM Tris-HCl pH 8.1, 100mM NaCl, 10mM EDTA) was added, DTT was added to a final concentration of 10mM, and treatment was carried out at 37 ℃ for 30 min. The treated single-chain TCR was added dropwise to 125mL of renaturation buffer (100mM Tris-HCl pH 8.1,0.4M L-arginine, 5M urea, 2mM EDTA,6.5mM beta-mercaptoethylamine, 1.87mM Cystamine) with a syringe, stirred at 4 ℃ for 10min, and then the renaturation solution was filled into a cellulose membrane dialysis bag with a cut-off of 4kDa, and the bag was placed in 1L of precooled water and stirred slowly at 4 ℃ overnight. After 17 hours, the dialysate was changed to 1L of pre-chilled buffer (20mM Tris-HCl pH 8.0), dialysis was continued at 4 ℃ for 8h, and then dialysis was continued overnight with the same fresh buffer. After 17 hours, the sample was filtered through a 0.45 μ M filter, vacuum degassed and then passed through an anion exchange column (HiTrap Q HP, GE Healthcare), the protein was purified using a 0-1M NaCl linear gradient eluent formulated in 20mM Tris-HCl pH 8.0, the collected fractions were subjected to SDS-PAGE analysis, the fractions containing single-chain TCR were concentrated and then further purified using a gel filtration column (Superdex 7510/300, GE Healthcare), and the target fraction was also subjected to SDS-PAGE analysis.
The eluted fractions for BIAcore analysis were further tested for purity using gel filtration. The conditions are as follows: the chromatography column Agilent Bio SEC-3(300A,
Figure BDA0002544355500000201
) The mobile phase is 150mM phosphate buffer solution, the flow rate is 0.5mL/min, the column temperature is 25 ℃, and the ultraviolet detection wavelength is 214 nm.
Example 3 binding characterisation
BIAcore analysis
The BIAcore T200 real-time assay system was used to detect the binding activity of the TCR molecules to the YMLDLQPET-HLA-A0201 complex. 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 conditions are as follows: the temperature is 25 ℃, and the PH value is 7.1-7.5.
The low concentration of streptavidin was flowed over the antibody coated chip surface, then YMLDLQPET-HLA-A0201 complex was flowed over the detection channel, the other channel served as the 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 affinity was determined by single cycle kinetic assay, in which the TCR was diluted with HEPES-EP buffer (10mM HEPES,150mM NaCl, 3mM EDTA, 0.005% P20, pH 7.4) to several different concentrations, passed over the chip surface sequentially at a flow rate of 30. mu.L/min, with a binding time of 120s for each injection, and dissociated for 600s after the end of the last injection. At the end of each assay run, the chip was regenerated with 10mM Gly-HCl pH 1.75. Kinetic parameters were calculated using BIAcore Evaluation software.
The YMLDLQPET-HLA-A0201 complex is prepared as follows:
a. and (3) purification: 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 using 5ml BugBuster Master Mix Extraction Reagents (Merck) to violently shake and resuspend the thalli, rotatably incubating at room temperature for 20min, centrifuging at 4 ℃ for 15min at 6000g, discarding supernatant, and collecting inclusion bodies.
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 YMLDLQPET (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. YMLDLQPET peptide was added to 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), then 20mg/L light chain and 90mg/L heavy chain were added in sequence (final concentration, heavy chain was added in three portions, 8 h/time), and renaturation was performed at 4 ℃ for at least 3 days until completion, and SDS-PAGE checked for success or failure of renaturation.
c. And (3) 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: the purified pMHC molecules were concentrated using Millipore ultrafiltration tubes while the buffer was replaced with 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 if biotinylation was complete.
e. Purification of biotinylated complex: 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.)TM16/60S200 HR 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).
Example 4 Generation of high affinity TCR
Phage display technology is one means of generating libraries of TCR high affinity variants to screen for high affinity variants. The TCR phage display and screening methods described by Li et al ((2005) Nature Biotech 23(3):349-354) were applied to the single-chain TCR templates in example 1. Libraries of high affinity TCRs were created by mutating the CDR regions of the template chains and panning was performed. The phage library after several rounds of panning has specific binding with corresponding antigen, and single clone is picked and analyzed.
Mutations in the CDR regions of the selected high affinity single chain TCR were introduced into the corresponding sites in the variable domain of the α β heterodimeric TCR and their affinity for the YMLDLQPET-HLA-A0201 complex was determined by BIAcore. The introduction of the high affinity mutation points in the CDR regions described above is performed by site-directed mutagenesis methods well known to those skilled in the art. The amino acid sequences of the alpha chain and beta chain variable domains of the wild-type TCR are shown in FIG. 1a (SEQ ID NO:1) and FIG. 1b (SEQ ID NO:2), respectively.
It should be noted that to obtain a more stable soluble TCR, in order to more conveniently assess the binding affinity and/or binding half-life between the TCR and the YMLDLQPET-HLA A0201 complex, the α β heterodimeric TCR may be a TCR in which a cysteine residue is introduced in a constant region of the α and β chains, respectively, to form an artificial interchain disulfide bond, in this example the amino acid sequences of the TCR α and β chains after introduction of the cysteine residue are shown in FIGS. 6a (SEQ ID NO:11) and 6b (SEQ ID NO:12), respectively, and the introduced cysteine residue is shown in bold letters.
Extracellular sequence genes of TCR α and β chains to be expressed were synthesized and inserted into expression vectors pET28a + (Novagene) by standard methods described in Molecular Cloning a Laboratory Manual (third edition, Sambrook and Russell), with upstream and downstream Cloning sites being NcoI and NotI, respectively. Mutations in CDR regions are introduced by overlap pcr (overlap pcr), which is well known to those skilled in the art. The insert was confirmed by sequencing without error.
Example 5 expression, renaturation and purification of high affinity TCR
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) at 0.6 final concentration with 0.5mM IPTG and washed repeatedly with BugBuster solution several times, and finally dissolved in 6M guanidine hydrochloride, 10mM Dithiothreitol (DTT),10mM ethylenediaminetetraacetic acid (EDTA),20 mM 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.
Example 6 BIAcore analysis results
The affinity of the α β heterodimeric TCR with the YMLDLQPET-HLA-A0201 complex incorporating the high affinity CDR regions was determined using the method described in example 3.
The invention obtains the amino acid sequences of the alpha chain and beta chain variable domains of a novel TCR, which are respectively shown in figures 7(1) - (9) and figures 8(1) - (12). As shown. Since the CDR regions of the TCR molecules determine their affinity for the corresponding pMHC complex, one skilled in the art would be able to expect that α β heterodimeric TCRs incorporating high affinity mutations also have high affinity for the YMLDLQPET-HLA-a0201 complex. The expression vector was constructed using the method described in example 4, the above α β heterodimeric TCR introduced with high affinity mutation was expressed, renatured and purified using the method described in example 5, and then its affinity to YMLDLQPET-HLA-a0201 complex was determined using BIAcore T200, as shown in table 2 below.
TABLE 2
Figure BDA0002544355500000231
Figure BDA0002544355500000241
As can be seen from Table 2 above, the affinity of the heterodimeric TCR was at least 2-fold greater than the affinity of the wild-type TCR for the YMLDLQPET-HLA-A0201 complex.
Example 7 expression, renaturation and purification of fusions of anti-CD 3 antibodies with high affinity α β heterodimeric TCRs
Fusion molecules were prepared by fusing an anti-CD 3 single chain antibody (scFv) to an α β heterodimeric TCR. The scFv of anti-CD 3 is fused to the β chain of the TCR, which β chain may comprise the β chain variable domain of any of the above-described high affinity α β heterodimeric TCRs, and the TCR α chain of the fused molecule may comprise the α chain variable domain of any of the above-described high affinity α β heterodimeric TCRs.
Construction of fusion molecule expression vectors
1. Construction of alpha chain expression vector: the target gene carrying the alpha chain of the α β heterodimeric TCR was double-digested with Nco i and Not i, and ligated to pET28a vector double-digested with Nco i and Not i. The ligation product was transformed into e.coli DH5 α, spread on LB plates containing kanamycin, cultured at 37 ℃ for inversion overnight, positive clones were picked for PCR screening, positive recombinants were sequenced, and after the sequence was determined to be correct, recombinant plasmids were extracted and transformed into e.coli Tuner (DE3) for expression.
2. Construction of anti-CD 3(scFv) -beta chain expression vector: by the overlap PCR method, primers were designed to link the anti-CD 3 scFv and the high-affinity heterodimeric TCR beta chain gene with the short linker peptide GGGGGGS (SEQ ID NO:31) in between, and the anti-CD 3 scFv was fused to the high-affinity heterodimeric TCR beta chain to carry the restriction endonuclease sites Nco I (CCATGG (SEQ ID NO:32)) and Not I (GCGGCCGC (SEQ ID NO:33)) in the gene fragment. The PCR amplification product was digested simultaneously with Nco I and Not I, and ligated with pET28a vector digested simultaneously with Nco I and Not I. The ligation product was transformed into e.coli DH5 α competent cells, coated with LB plates containing kanamycin, inverted cultured overnight at 37 ℃, positive clones were selected for PCR screening, positive recombinants were sequenced, and after the sequence was determined to be correct, recombinant plasmids were extracted and transformed into e.coli Tuner (DE3) competent cells for expression.
Expression, renaturation and purification of fusion proteins
The expression plasmids were transformed into E.coli Tuner (DE3) competent cells, respectively, and LB plates (kanamycin 50. mu.g/mL) were plated and incubated at 37 ℃ overnight. The next day, the selected clones were inoculated into 10mL LB liquid medium (kanamycin 50. mu.g/mL) for 2-3h of culture, inoculated into 1L of LB medium at a volume ratio of 1:100, cultured until OD600 became 0.5-0.8, and added with 1mM IPTG to induce the expression of the target protein. After 4 hours of induction, cells were harvested by centrifugation at 6000rpm for 10 min. The cells were washed once with PBS buffer and aliquoted, and 200mL of the cells from the bacterial culture were lysed with 5mL of BugBuster Master Mix (Merck) and the inclusion bodies were collected by centrifugation at 6000g for 15 min. 4 detergent washes were then performed to remove cell debris and membrane components. The inclusion bodies are then washed with a buffer such as PBS to remove the detergent and salts. Finally, inclusion bodies were dissolved in a buffer solution containing 6M guanidine hydrochloride, 10mM Dithiothreitol (DTT),10mM ethylenediaminetetraacetic acid (EDTA),20 mM Tris, pH 8.1, and the inclusion body concentration was measured, and they were stored frozen at-80 ℃ after being dispensed.
The TCR α chain and the anti-CD 3(scFv) - β chain after solubilization were separated by a 2: 5 in 5M Urea (urea), 0.4M L-arginine (L-arginine),20mM Tris pH 8.1,3.7mM cystamine,6.6mM β -mer capoethylamine (4 ℃ C.), final concentrations of α chain and anti-CD 3(scFv) - β chain were 0.1mg/mL, 0.25mg/mL, respectively.
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 (10mM 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 eluted peaks contain TCR alpha chain and anti-CD 3(scFv) -beta chain dimers of which the renaturation was successful TCR alpha chain was confirmed by SDS-PAGE gel. The TCR fusion molecules were then further purified by size exclusion chromatography (S-10016/60, GE healthcare) and re-purified on an anion exchange column (HiTrap Q HP 5ml, GE healthcare). The purity of the purified TCR fusion molecule was greater than 90% as determined by SDS-PAGE and the concentration was determined by BCA.
Example 8 activation function experiment of Effector cells transfected with high affinity TCR of the invention against short peptide-loaded T2 cells
IFN- γ is a potent immunomodulatory factor produced by activated T lymphocytes, and therefore this example examines the IFN- γ numbers by ELISPOT assays well known to those skilled in the art to verify the activation function and antigen specificity of cells transfected with the high affinity TCR of the invention. The high affinity TCRs of the invention were transfected into CD3+ T cells isolated from the blood of healthy volunteers as effector cells and CD3+ T cells transfected with other TCRs (a6) from the same volunteer as controls. The target cells used were T2 cells loaded with HPV 16E 7 antigen short peptide YMLDLQPET, T2 cells loaded with other short peptides and unloaded T2 cells.
The following experiments were carried out in two batches (I), (II) one after the other:
(I) the high affinity TCR can be learned from Table 2, respectively
Figure BDA0002544355500000251
First, an ELISPOT plate was prepared. ELISPOT plate ethanol activation coating, 4 degrees C overnight. Day 1 of the experiment, coating was removed, washed and blocked, incubated at room temperature for two hours, blocking solution removed, and the components of the experiment were added to ELISPOT plates: target cells are 1 x 1041 x 10 effector cells per well3One well (calculated as the positive rate of antibody) and two duplicate wells were set. Adding corresponding short peptide to make the final concentration of short peptide in ELISPOT pore plate be 1X 10-6And M. Incubation overnight (37 ℃, 5% CO)2). On day 2 of the experiment, the plates were washed and subjected to secondary detection and color development, dried, and spots formed on the membrane were counted using an immuno-spot plate READER (ELISPOT READER system; AID20 Co.).
(II) the high affinity TCR is shown in Table 2 as
Figure BDA0002544355500000261
The components added to the ELISPOT plate were: target cells are 1 x 1042 x 10 effector cells per well3One cell/well (calculated as the antibody positivity) and two duplicate wells were set. The rest of the steps are the same as in this example (I).
As shown in fig. 12a and 12b, the effector cells transfected with the high affinity TCR of the present invention have significant activation effect on target cells loaded with HPV 16E 7 antigen short peptide YMLDLQPET, while effector cells transfected with other TCRs have no activation effect; meanwhile, the effector cells transfected with the high affinity TCR of the invention have substantially no activating effect on target cells loaded with other short peptides or unloaded.
Example 9 functional assay for activation of Effector cells transfected with high affinity TCRs of the invention against tumor cell lines
This example again demonstrates the activation function and specificity of effector cells transfected with the high affinity TCRs of the invention using tumor cell lines. Again, detection is by ELISPOT assays well known to those skilled in the art. The high affinity TCRs of the invention were transfected into CD3+ T cells isolated from the blood of healthy volunteers as effector cells and CD3+ T cells transfected with other TCRs (a6) or with wild-type TCRs (WT-TCRs) from the same volunteer as negative controls. The following experiments were carried out in two batches (I), (II) one after the other:
(I) the high affinity TCRs can be gathered from Table 2 as TCR1 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:22), TCR2 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:23) and TCR3 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:24), respectively.
The HPV 16E 7 positive tumor cell lines used in this batch were A375-E7(HPV 16E 7 overexpression) and HK-2, and the negative tumor cell lines were SK-MEL-28-E7(HPV 16E 7 overexpression), SK-MEL-28 and A375.
(II) the high affinity TCR is shown in Table 2 as
Figure BDA0002544355500000262
The HPV 16E 7 positive tumor cell lines used in this batch were A375-E7(HPV 16E 7 over-expressed) and the negative tumor cell lines were A375 and SK-MEL-28.
The two batches were all subjected to the following steps: first, an ELISPOT plate was prepared. The ELISPOT plates were ethanol activated coated overnight at 4 ℃. Day 1 of the experiment, the coating solution was removed, washed and sealed, incubated at room temperature for two hours, the sealing solution was removed, and the components of the experiment were added to ELISPOTPlate: the target cells are 2 x 1042 x 10 effector cells per well3One well (calculated as the positive rate of antibody) and two duplicate wells were set. Incubation overnight (37 ℃, 5% CO)2). On day 2 of the experiment, the plates were washed and subjected to secondary detection and color development, the plates were dried, and spots formed on the membrane were counted using an immuno-spot plate READER (ELISPOT READER system; AID20 Co.).
As shown in fig. 13a and fig. 13b, for HPV 16E 7 positive tumor cell line, the effector cells transfected with the high affinity TCR of the present invention have a more significant activation effect than the wild-type effector cells, while the effector cells transfected with other TCRs have no activation state; meanwhile, effector cells transfected with the high affinity TCR of the invention had essentially no activating effect on HPV 16E 7 negative tumor cell line.
Example 10 killing function experiment of effector cells transfected with high affinity TCR of the invention against gradient short peptide-loaded T2 cells
Lactate Dehydrogenase (LDH) is abundant in the cytoplasm, normally cannot pass through the cell membrane, and is released to the outside of the cell when the cell is damaged or dies, and the LDH activity in the cell culture solution is proportional to the number of cell death. This example demonstrates the killing function of cells transfected with a TCR of the invention by measuring LDH release by non-radioactive cytotoxicity experiments well known to those skilled in the art. This test is a colorimetric substitution test for the 51 Cr-release cytotoxicity test, and quantitatively determines LDH released after cell lysis. LDH released in the medium was detected using a 30min 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. The formula is calculated as% cytotoxicity 100% × (experiment-effector cell spontaneous-target cell spontaneous)/(target cell maximal-target cell spontaneous).
This example uses CD3+ T cells transfected with the high affinity TCRs of the invention into blood of healthy volunteers as effector cells and CD3+ T cells null-transfected (NC) or transfected with other TCRs (a6) from the same volunteer as negative controls. Wherein the high affinity TCR and its numbering is taken from Table 2 as TCR1 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:22) and TCR2 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:23), respectively. The target cells were T2 cells loaded with YMLDLQPET peptide, other short peptides or empty.
LDH plates were prepared first, 3X 10 target cells were first aligned4Single cell/well, effector cell 3X 104Adding each cell/well (calculated according to the positive rate of the antibody) into the corresponding well, then adding HPV 16E 7 antigen short peptide YMLDLQPET into the experimental group, and enabling the final concentration of the short peptide in an ELISPOT pore plate to be 1 x 10 in sequence-15M to 1X 10-8M, 8 gradients in total; adding other short peptides into the control group, and making the final concentration of the short peptides to be 1 × 10-9M to 1X 10-8M, 2 gradients are provided, and three multiple wells are provided. Meanwhile, an effector cell spontaneous hole, a target cell maximum hole, a volume correction control hole and a culture medium background control hole are arranged. Incubation overnight (37 ℃, 5% CO)2). On day 2 of the experiment, color development was detected, and after termination of the reaction, the absorbance was recorded at 490nm using a microplate reader (Bioteck).
The experimental result is shown in fig. 14, aiming at the target cells loaded with HPV 16E 7 antigen short peptide YMLDLQPET in a gradient manner, the effector cells transfected with the high-affinity TCR of the invention show strong killing effect, and react when the concentration of the specific short peptide is lower, while the effector cells transfected with other TCRs or empty transfection have no killing effect basically; meanwhile, effector cells transfected with the high affinity TCR of the invention have no killing effect on target cells loaded with other short peptides or unloaded.
Example 11 killing function experiment of Effector cells transfected with high affinity TCR of the invention against tumor cell lines
This example also demonstrates the killing function of cells transfected with a TCR of the invention by measuring LDH release by nonradioactive cytotoxicity assays well known to those skilled in the art. The LDH assay of this example transfected the high affinity TCR of the invention as effector cells with CD3+ T cells isolated from the blood of healthy volunteers and as negative control CD3+ T cells of the same volunteer transfected with other TCR (A6) or with wild type TCR (WT-TCR).
The following experiments were carried out in three batches (I), (II), (III) one after the other:
(I) the high affinity TCRs and their numbering are known from Table 2 as TCR1 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:22), TCR2 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:23) and TCR3 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:24), respectively. The HPV 16E 7 positive tumor cell line used for this batch was: CASKI and HK-2, negative tumor cell lines SK-MEL-28, MEL526 and HCC 827. The experimental procedure was as follows: LDH plates were first prepared and the individual components of the assay were added to the plates in the following order: target cell 2X 104 2X 10 cells/well, Effector cell4Individual cells/well (calculated as antibody positivity) were added to the corresponding wells and three replicate wells were set. Meanwhile, an effector cell spontaneous hole, a target cell maximum hole, a volume correction control hole and a culture medium background control hole are arranged. Incubation overnight (37 ℃, 5% CO)2). On day 2 of the experiment, color development was detected, and after termination of the reaction, the absorbance was recorded at 490nm using a microplate reader (Bioteck).
(II) the high affinity TCRs and their numbering are known from Table 2 as TCR5 (alpha chain variable domain SEQ ID NO: 14, beta chain variable domain SEQ ID NO:2) and TCR9 (alpha chain variable domain SEQ ID NO:15, beta chain variable domain SEQ ID NO:24), respectively. The HPV 16E 7 positive tumor cell line used in this batch was CASKI and the negative tumor cell line was HCCC 9810. The components added to the plate were as follows: target cell 3X 104Single cell/well, effector cell 3X 104Individual cells/well (calculated as antibody positivity), the remaining experimental procedure was the same as in example (i).
(III) the high affinity TCRs and their numbering are known from Table 2 as TCR17 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:26), TCR19 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:28) and TCR22 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:31), respectively. HPV 16E 7 positive tumor cell lines used in this batch were CASKI, and negative tumor cell lines were a375 and SiHa. The components added to the plate were as follows: target cell 3X 104Single cell/well, effector cell 3X 104Individual cells/well (calculated as antibody positivity), the rest of the experimental procedure was the same as in this example (I).
As shown in fig. 15a, fig. 15b and fig. 15c, the effector cells transfected with the high affinity TCR of the present invention still showed strong killing efficacy against HPV 16E 7 positive tumor cell line, and was significantly more potent than T cells transfected with wild-type TCR in killing function, while T cells transfected with other TCRs were essentially non-reactive; meanwhile, the T cells transfected with the high-affinity TCR of the invention basically have no killing effect on negative tumor cell lines, and further embody the good specific killing function of the cells transfected with the high-affinity TCR of the invention.
Example 12 killing function of Effector cells transfected with the high affinity TCR molecules of the invention against tumor cell lines (IncuCyte experiment)
This example further demonstrates that effector cells transfected with the high affinity TCRs of the invention have excellent specific killing of target cells and their sensitivity by IncuCyte assays well known to those skilled in the art. IncuCyte is a functional analysis system which can automatically analyze images at different time points and quantify real-time apoptosis number by real-time microscopic shooting in an incubator.
The TCRs of the invention were randomly selected to transfect CD3+ T cells isolated from the blood of healthy volunteers as effector cells, and the same volunteers were transfected with other TCR (a6) or null transfected (NC) CD3+ T cells as controls. The TCRs and their numbering are known from Table 2 as TCR1 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:22) and TCR2 (alpha chain variable domain SEQ ID NO:1, beta chain variable domain SEQ ID NO:23), respectively. In the target cell line, A375-E7(HPV 16E 7 is over-expressed) is a positive tumor cell line; a375 was a negative tumor cell line as control.
The first day of experiment, the target cells are digested and centrifuged; the target cells were resuspended in complete medium of RPMI1640+ 10% FBS without phenol red and plated evenly in 96-well plates: 2*104Per well; placing the mixture back into an incubator with 37 ℃ and 5% CO2 for overnight incubation; the following day the medium in the 96-well plate was discarded and replaced with a medium containing the dye caspase3/7reagent phenol red free RPMI1640+ 10% FBS medium, dye concentration of 2 drops/ml. Discarding old medium, replacing new phenol red-free RPMI1640+ 10% FBS medium, and adding effector cells 1 x 104(iii) co-incubation of wells (calculated as positive rate of transfection) with the experimental group plated with target cells; putting the plate into a real-time dynamic living cell imaging analyzer-Incucyte zooM special for Incucyte detection, and incubating for half an hour; starting to observe in real time and take a picture; and processing, analyzing data and exporting the detection result by adopting IncuCyte ZooM 2016A.
As shown in fig. 16a and fig. 16b, for HPV 16E 7 positive tumor cell line, cells transfected with the high affinity TCR of the present invention can show strong and effective killing effect in a short period, while effector cells transfected with other TCRs have no killing effect; at the same time, cells transfected with the high affinity TCR of the invention had essentially no killing of negative tumor cells.
All documents referred to herein are incorporated by reference into this application as if each had been 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> Guangdong Xiangxue accurate medical technology Limited
<120> a high affinity T cell receptor recognizing HPV antigen
<130> P2020-1132
<160> 37
<170> SIPOSequenceListing 1.0
<210> 1
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 2
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 3
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Glu Asn Val Ser Ile Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Val Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Phe Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Lys Leu Arg Val Lys Ser
100 105 110
<210> 4
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Leu Ser Val Lys Thr Gly
1 5 10 15
Lys Arg Val Thr Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Gln Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Arg Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Ile Glu Ser
65 70 75 80
Val Glu Pro Asn Asp Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 5
<211> 333
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 5
caaaaagaag ttgaacagaa tagtggcccg ctgagtgtgc cggaaggtga aaatgtgagt 60
attaattgta cctatagcga tcgcgttagt cagagctttt tctggtatcg tcagtatagc 120
ggtaaaagcc cggaactgat tatgagtatc tatagcaatg gcgataaaga agatggccgc 180
tttaccgcac agctgaataa ggcaagccag tatgtgagcc tgctgattcg cgatgtgcag 240
ccgagtgata gtgcaaccta tttttgtgca gtgaatccgc gttatggcaa taagctggtt 300
tttggtgccg gcaccaaact gcgcgttaaa agc 333
<210> 6
<211> 345
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 6
gacgcagatg ttacccagac cccgcgtaat ctgagcgtga aaaccggcaa acgcgtgacc 60
ctggaatgca gtcagaccaa aggccatgat cgcatgtatt ggtatcgtca agatccgggt 120
cagggcctgc gtctgatcta ttatagcttt gatgttaaag acatcaacaa gggcgaaatt 180
agtgatcgtt atagcgttag tcgtcaggcc caggccaaat tttcactgag tattgaaagc 240
gttgaaccga atgataccgc cctgtatttt tgcgcaacca gcgatcgcgg ccagggtgcc 300
tttggcgaac agtattttgg cccgggtacc cgcctgaccg ttacc 345
<210> 7
<211> 24
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Gly Gly Gly Ser Glu Gly Gly Gly Ser Glu Gly Gly Gly Ser Glu Gly
1 5 10 15
Gly Gly Ser Glu Gly Gly Thr Gly
20
<210> 8
<211> 72
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 8
ggtggcggta gcgaaggtgg cggtagtgaa ggtggcggca gtgaaggtgg tggcagcgaa 60
ggtggtaccg gt 72
<210> 9
<211> 250
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Glu Asn Val Ser Ile Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Val Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Phe Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Lys Leu Arg Val Lys Ser Gly
100 105 110
Gly Gly Ser Glu Gly Gly Gly Ser Glu Gly Gly Gly Ser Glu Gly Gly
115 120 125
Gly Ser Glu Gly Gly Thr Gly Asp Ala Asp Val Thr Gln Thr Pro Arg
130 135 140
Asn Leu Ser Val Lys Thr Gly Lys Arg Val Thr Leu Glu Cys Ser Gln
145 150 155 160
Thr Lys Gly His Asp Arg Met Tyr Trp Tyr Arg Gln Asp Pro Gly Gln
165 170 175
Gly Leu Arg Leu Ile Tyr Tyr Ser Phe Asp Val Lys Asp Ile Asn Lys
180 185 190
Gly Glu Ile Ser Asp Arg Tyr Ser Val Ser Arg Gln Ala Gln Ala Lys
195 200 205
Phe Ser Leu Ser Ile Glu Ser Val Glu Pro Asn Asp Thr Ala Leu Tyr
210 215 220
Phe Cys Ala Thr Ser Asp Arg Gly Gln Gly Ala Phe Gly Glu Gln Tyr
225 230 235 240
Phe Gly Pro Gly Thr Arg Leu Thr Val Thr
245 250
<210> 10
<211> 750
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 10
caaaaagaag ttgaacagaa tagtggcccg ctgagtgtgc cggaaggtga aaatgtgagt 60
attaattgta cctatagcga tcgcgttagt cagagctttt tctggtatcg tcagtatagc 120
ggtaaaagcc cggaactgat tatgagtatc tatagcaatg gcgataaaga agatggccgc 180
tttaccgcac agctgaataa ggcaagccag tatgtgagcc tgctgattcg cgatgtgcag 240
ccgagtgata gtgcaaccta tttttgtgca gtgaatccgc gttatggcaa taagctggtt 300
tttggtgccg gcaccaaact gcgcgttaaa agcggtggcg gtagcgaagg tggcggtagt 360
gaaggtggcg gcagtgaagg tggtggcagc gaaggtggta ccggtgacgc agatgttacc 420
cagaccccgc gtaatctgag cgtgaaaacc ggcaaacgcg tgaccctgga atgcagtcag 480
accaaaggcc atgatcgcat gtattggtat cgtcaagatc cgggtcaggg cctgcgtctg 540
atctattata gctttgatgt taaagacatc aacaagggcg aaattagtga tcgttatagc 600
gttagtcgtc aggcccaggc caaattttca ctgagtattg aaagcgttga accgaatgat 660
accgccctgt atttttgcgc aaccagcgat cgcggccagg gtgcctttgg cgaacagtat 720
tttggcccgg gtacccgcct gaccgttacc 750
<210> 11
<211> 206
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 11
Met Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu
1 5 10 15
Gly Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln
20 25 30
Ser Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile
35 40 45
Met Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala
50 55 60
Gln Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser
65 70 75 80
Gln Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr
85 90 95
Gly Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
Tyr Ile Gln Asn Pro Asp Pro Ala Val Tyr Gln Leu Arg Asp Ser Lys
115 120 125
Ser Ser Asp Lys Ser Val Cys Leu Phe Thr Asp Phe Asp Ser Gln Thr
130 135 140
Asn Val Ser Gln Ser Lys Asp Ser Asp Val Tyr Ile Thr Asp Lys Thr
145 150 155 160
Val Leu Asp Met Arg Ser Met Asp Phe Lys Ser Asn Ser Ala Val Ala
165 170 175
Trp Ser Asn Lys Ser Asp Phe Ala Cys Ala Asn Ala Phe Asn Asn Ser
180 185 190
Ile Ile Pro Glu Asp Thr Phe Phe Cys Ser Pro Glu Ser Ser
195 200 205
<210> 12
<211> 246
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 12
Met Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr
1 5 10 15
Gly Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg
20 25 30
Met Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr
35 40 45
Tyr Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly
50 55 60
Tyr Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu
65 70 75 80
Ser Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp
85 90 95
Arg Gly Gln Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg
100 105 110
Leu Thr Val Thr Glu Asp Leu Lys Asn Val Phe Pro Pro Glu Val Ala
115 120 125
Val Phe Glu Pro Ser Glu Cys Glu Ile Ser His Thr Gln Lys Ala Thr
130 135 140
Leu Val Cys Leu Ala Thr Gly Phe Tyr Pro Asp His Val Glu Leu Ser
145 150 155 160
Trp Trp Val Asn Gly Lys Glu Val His Ser Gly Val Ser Thr Asp Pro
165 170 175
Gln Pro Leu Lys Glu Gln Pro Ala Leu Asn Asp Ser Arg Tyr Ala Leu
180 185 190
Ser Ser Arg Leu Arg Val Ser Ala Thr Phe Trp Gln Asn Pro Arg Asn
195 200 205
His Phe Arg Cys Gln Val Gln Phe Tyr Gly Leu Ser Glu Asn Asp Glu
210 215 220
Trp Thr Gln Asp Arg Ala Lys Pro Val Thr Gln Ile Val Ser Ala Glu
225 230 235 240
Ala Trp Gly Arg Ala Asp
245
<210> 13
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 13
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Leu Ser Gln Thr
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 14
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 14
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Leu Ala Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 15
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 15
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Leu Thr Tyr Val
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 16
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 16
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Phe Asn Pro Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 17
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 17
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Met Ala Asn Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 18
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 18
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg His Ala Asn Thr
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 19
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 19
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Leu Ala Val Thr
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 20
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 20
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Leu Ala Asn Val
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 21
<211> 111
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 21
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Met Gly Asn Ala
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser
100 105 110
<210> 22
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 22
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gln Tyr Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 23
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 23
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gln Phe Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 24
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 24
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gln Gln Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 25
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 25
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gln Gln Gly Ala Phe Gly Glu Gln Phe Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 26
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 26
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gln Trp Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 27
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 27
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Tyr His Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 28
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 28
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Tyr Phe Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 29
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 29
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Ala Glu Tyr Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 30
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 30
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Ala Glu Tyr His Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 31
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 31
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Ala Glu Tyr Phe Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 32
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 32
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Tyr Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 33
<211> 115
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 33
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Gln Phe Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr
115
<210> 34
<211> 205
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 34
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser 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 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
195 200 205
<210> 35
<211> 245
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 35
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr Glu Asp Leu Lys Asn Val Phe Pro Pro Glu Val Ala Val
115 120 125
Phe Glu Pro Ser Glu Ala Glu Ile Ser His Thr Gln Lys Ala Thr Leu
130 135 140
Val Cys Leu Ala Thr Gly Phe Tyr Pro Asp His Val Glu Leu Ser Trp
145 150 155 160
Trp Val Asn Gly Lys Glu Val His Ser Gly Val Ser Thr Asp Pro Gln
165 170 175
Pro Leu Lys Glu Gln Pro Ala Leu Asn Asp Ser Arg Tyr Cys Leu Ser
180 185 190
Ser Arg Leu Arg Val Ser Ala Thr Phe Trp Gln Asn Pro Arg Asn His
195 200 205
Phe Arg Cys Gln Val Gln Phe Tyr Gly Leu Ser Glu Asn Asp Glu Trp
210 215 220
Thr Gln Asp Arg Ala Lys Pro Val Thr Gln Ile Val Ser Ala Glu Ala
225 230 235 240
Trp Gly Arg Ala Asp
245
<210> 36
<211> 252
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 36
Gln Lys Glu Val Glu Gln Asn Ser Gly Pro Leu Ser Val Pro Glu Gly
1 5 10 15
Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser Asp Arg Val Ser Gln Ser
20 25 30
Phe Phe Trp Tyr Arg Gln Tyr Ser Gly Lys Ser Pro Glu Leu Ile Met
35 40 45
Ser Ile Tyr Ser Asn Gly Asp Lys Glu Asp Gly Arg Phe Thr Ala Gln
50 55 60
Leu Asn Lys Ala Ser Gln Tyr Val Ser Leu Leu Ile Arg Asp Ser Gln
65 70 75 80
Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala Val Asn Pro Arg Tyr Gly
85 90 95
Asn Lys Leu Val Phe Gly Ala Gly Thr Ile Leu Arg Val Lys Ser 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 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> 37
<211> 294
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 37
Asp Ala Asp Val Thr Gln Thr Pro Arg Asn Arg Ile Thr Lys Thr Gly
1 5 10 15
Lys Arg Ile Met Leu Glu Cys Ser Gln Thr Lys Gly His Asp Arg Met
20 25 30
Tyr Trp Tyr Arg Gln Asp Pro Gly Leu Gly Leu Arg Leu Ile Tyr Tyr
35 40 45
Ser Phe Asp Val Lys Asp Ile Asn Lys Gly Glu Ile Ser Asp Gly Tyr
50 55 60
Ser Val Ser Arg Gln Ala Gln Ala Lys Phe Ser Leu Ser Leu Glu Ser
65 70 75 80
Ala Ile Pro Asn Gln Thr Ala Leu Tyr Phe Cys Ala Thr Ser Asp Arg
85 90 95
Gly Gln Gly Ala Phe Gly Glu Gln Tyr Phe Gly Pro Gly Thr Arg Leu
100 105 110
Thr Val Thr Glu Asp Leu Lys Asn Val Phe Pro Pro Glu Val Ala Val
115 120 125
Phe Glu Pro Ser Glu Ala Glu Ile Ser His Thr Gln Lys Ala Thr Leu
130 135 140
Val Cys Leu Ala Thr Gly Phe Tyr Pro Asp His Val Glu Leu Ser Trp
145 150 155 160
Trp Val Asn Gly Lys Glu Val His Ser Gly Val Ser Thr Asp Pro Gln
165 170 175
Pro Leu Lys Glu Gln Pro Ala Leu Asn Asp Ser Arg Tyr Cys Leu Ser
180 185 190
Ser Arg Leu Arg Val Ser Ala Thr Phe Trp Gln Asn Pro Arg Asn His
195 200 205
Phe Arg Cys Gln Val Gln Phe Tyr Gly Leu Ser Glu Asn Asp Glu Trp
210 215 220
Thr Gln Asp Arg Ala Lys Pro Val Thr Gln Ile Val Ser Ala Glu Ala
225 230 235 240
Trp Gly Arg Ala Asp Cys Gly Phe Thr Ser Glu Ser Tyr Gln Gln Gly
245 250 255
Val Leu Ser Ala Thr Ile Leu Tyr Glu Ile Leu Leu Gly Lys Ala Thr
260 265 270
Leu Tyr Ala Val Leu Val Ser Ala Leu Val Leu Met Ala Met Val Lys
275 280 285
Arg Lys Asp Ser Arg Gly
290

Claims (10)

1. A T Cell Receptor (TCR) comprising a TCR alpha chain variable domain and a TCR beta chain variable domain, characterised in that it has the activity of binding YMLDLQPET-HLA a0201 complex and in that the amino acid sequence of the TCR alpha chain variable domain has at least 90% sequence homology with the amino acid sequence set out in SEQ ID No. 1 and the amino acid sequence of the TCR beta chain variable domain has at least 90% sequence homology with the amino acid sequence set out in SEQ ID No. 2.
2. A multivalent TCR complex comprising at least two TCR molecules, at least one of which is a TCR as claimed in claim 1.
3. A nucleic acid molecule comprising a nucleic acid sequence encoding a TCR as claimed in claim 1, or the complement thereof.
4. A vector comprising the nucleic acid molecule of claim 3.
5. A host cell comprising the vector of claim 4 or a nucleic acid molecule of claim 3 integrated into the chromosome.
6. An isolated cell expressing a TCR as claimed in claim 1, preferably wherein the isolated cell is a T cell.
7. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a TCR as claimed in claim 1, or a TCR complex as claimed in claim 2, or a cell as claimed in claim 6.
8. A method of treating a disease comprising administering to a subject in need thereof a TCR as claimed in claim 1 or a TCR complex as claimed in claim 2 or a cell as claimed in claim 6 or a pharmaceutical composition as claimed in claim 7, preferably wherein the disease is an HPV16 positive tumour, more preferably wherein the tumour is cervical cancer.
9. Use of a T cell receptor according to claim 1, a TCR complex according to claim 2 or a cell according to claim 6 for the preparation of a medicament for the treatment of a tumour, preferably wherein the tumour is a HPV16 positive tumour, more preferably wherein the tumour is cervical cancer.
10. A method of preparing a T cell receptor according to claim 1, comprising the steps of:
(i) culturing the host cell of claim 5 so as to express the T cell receptor of claim 1;
(ii) isolating or purifying said T cell receptor.
CN202010556192.XA 2020-06-17 2020-06-17 High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen Pending CN113801217A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202010556192.XA CN113801217A (en) 2020-06-17 2020-06-17 High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen
PCT/CN2021/100718 WO2021254458A1 (en) 2020-06-17 2021-06-17 High-affinity t-cell receptor for recognizing hpv antigen

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010556192.XA CN113801217A (en) 2020-06-17 2020-06-17 High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen

Publications (1)

Publication Number Publication Date
CN113801217A true CN113801217A (en) 2021-12-17

Family

ID=78892713

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010556192.XA Pending CN113801217A (en) 2020-06-17 2020-06-17 High-affinity T cell receptor for recognizing HPV (human papilloma Virus) antigen

Country Status (2)

Country Link
CN (1) CN113801217A (en)
WO (1) WO2021254458A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113321726A (en) * 2020-02-28 2021-08-31 香雪生命科学技术(广东)有限公司 T cell receptor for identifying HPV

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2645256C2 (en) * 2013-06-26 2018-02-19 Гуандун Сянсюэ Лайф Сайенсис, Лтд. High-stable t-cell receptor and method for its obtaining and application
WO2016182957A1 (en) * 2015-05-08 2016-11-17 Eureka Therapeutics, Inc. Constructs targeting hpv16-e7 peptide/mhc complexes and uses thereof
CN110139873A (en) * 2016-10-03 2019-08-16 朱诺治疗学股份有限公司 HPV specific binding molecules
CA3080546A1 (en) * 2017-10-03 2019-04-11 Juno Therapeutics, Inc. Hpv-specific binding molecules
CN110357952B (en) * 2019-07-17 2022-03-22 深圳市因诺转化医学研究院 TCR recognizing human papillomavirus HPV16-E7 antigen

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113321726A (en) * 2020-02-28 2021-08-31 香雪生命科学技术(广东)有限公司 T cell receptor for identifying HPV

Also Published As

Publication number Publication date
WO2021254458A1 (en) 2021-12-23

Similar Documents

Publication Publication Date Title
CN109879957B (en) High affinity T cell receptors for PRAME
CN112759641B (en) High-affinity TCR for recognizing Kras G12V
CN110198953B (en) High affinity TCRs for NY-ESO
US20230331805A1 (en) High-affinity tcr for recognizing afp antigen
EP4026844A1 (en) High-affinity t cell receptor that recognizes ssx2
CN112409474B (en) High-affinity TCR (T cell receptor) for identifying SSX2 antigen
CN110938136B (en) High-affinity T cell receptor for recognizing AFP antigen
CN111187345B (en) High-affinity TCR for identifying AFP
CN111662374B (en) High-affinity TCR for identifying AFP antigen
CN112390875B (en) High-affinity T cell receptor for identifying AFP
CN110156889B (en) High affinity HBs T cell receptor
CN114057864B (en) High affinity TCR for identifying AFP
WO2021254458A1 (en) High-affinity t-cell receptor for recognizing hpv antigen
WO2021204287A1 (en) High-affinity tcr for recognizing hpv16
CN112300268B (en) High affinity T cell receptors recognizing the NY-ESO-1 antigen
CN113667008A (en) High-affinity T cell receptor for recognizing AFP antigen
CN112940106A (en) High affinity AFPT cellular receptors
WO2022166905A1 (en) High-affinity tcr against hpv
WO2022206860A1 (en) T cell receptor for afp
WO2023221959A1 (en) High-affinity t cell receptors for recognizing mage and use thereof
CN115490773A (en) High-affinity T cell receptor aiming at AFP antigen
CN115677846A (en) High affinity T cell receptors for the antigen SSX2
CN115819555A (en) High-affinity TCR for identifying SSX2
CN116135879A (en) High affinity TCR against AFP
CN116836261A (en) High-affinity TCR (TCR) for recognizing MAGE-A4 antigen, and sequence and application thereof

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