[go: up one dir, main page]

WO2024219973A1 - Néo-antigènes teipp dérivés de rcn1 et leurs utilisations - Google Patents

Néo-antigènes teipp dérivés de rcn1 et leurs utilisations Download PDF

Info

Publication number
WO2024219973A1
WO2024219973A1 PCT/NL2024/050206 NL2024050206W WO2024219973A1 WO 2024219973 A1 WO2024219973 A1 WO 2024219973A1 NL 2024050206 W NL2024050206 W NL 2024050206W WO 2024219973 A1 WO2024219973 A1 WO 2024219973A1
Authority
WO
WIPO (PCT)
Prior art keywords
hla
complex
seq
acid sequence
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/NL2024/050206
Other languages
English (en)
Inventor
Sjoerd Henricus Van Der Burg
Thorbald Van Hall
Koen Abraham MARIJT
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.)
Leids Universitair Medisch Centrum LUMC
Original Assignee
Leids Universitair Medisch Centrum LUMC
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 Leids Universitair Medisch Centrum LUMC filed Critical Leids Universitair Medisch Centrum LUMC
Priority to AU2024257340A priority Critical patent/AU2024257340A1/en
Publication of WO2024219973A1 publication Critical patent/WO2024219973A1/fr
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4728Calcium binding proteins, e.g. calmodulin
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • RCN1-derived TEIPP neoantigens and uses thereof Novel nucleic acid sequences, vectors, modified cells, binding agents, peptides and pharmaceutical compositions are provided that are useful as a medicament, for example in the prevention or treatment of cancer or viral infections associated with impaired HLA class I antigen presentation.
  • Corresponding methods and uses are also provided.
  • Background Many T-cell based immunotherapies used for treatment of cancer in humans are based on recognition of tumor antigens presented in HLA class I (HLA-I) molecules by tumor cells (Robbins et al., 2013; Schumacher et al., 2015). Point mutated peptides constitute daunting tumor antigens due to their non-self nature for which a non-curtailed T cell repertoire is available.
  • T cells An absolute requirement for such T cells to exert their action against cancer is the display of HLA-I at the surface of tumor cells.
  • HLA-I down modulation on cancer cells is observed in many immune-escaped cancers, often caused by epigenetic silencing of antigen processing components, like the peptide transporter TAP (Ritter et al., 2017; Setiadi et al., 2007; Garrido et al., 2016).
  • TAP tumor antigen processing components
  • a novel category of tumor antigens referred to as TEIPP (T cell epitopes associated with impaired peptide processing), are presented at the surface of tumor cells carrying defects in antigen processing (Van Hall et al., 2006; Seidel et al., 2012).
  • TEIPPs are derived from ubiquitously expressed non-mutated ‘self’ proteins, however, their processed peptides fail to be loaded up to T-cell detectable levels into HLA-I in healthy cells. Their surface presentation is highly promoted by defects in the antigen processing machinery, especially in the absence of the peptide transporter TAP. TAP-impairment is frequently found in cancer cells and thus TEIPP peptides constitute tumor-specific antigens.
  • TEIPP peptides may also be considered as target antigens for treating or preventing a viral infection associated with lowered TAP expression and/or impaired antigen presentation in HLA class I molecules.
  • US2009/0220534 and Weinzierl describe screening methods for identifying T cell epitopes that are presented on the surface of cells via a TAP independent mechanism.
  • Human TEIPP neoantigens as potential targets for T-cell based immunotherapies have been described previously in WO2019231326A1 and WO2021107775A1.
  • TEIPP neoantigens for preventing and/or treating cancer are needed.
  • the inventors have previously developed a hybrid forward-reverse immunologic screen to identify novel TAP-independent non-mutated neoantigens that are selectively presented by immune- escaped cancers (Marijt et al., 2018). Their approach encompassed an in silico prediction of TEIPP neoantigen-candidates from the whole human proteome, matching of candidates to the cancer-specific peptidome, and an ex vivo screen to confirm the presence of a TEIPP T cell repertoire in healthy donors.
  • TEIPP neoantigen candidates were selected for their capacity to activate CD8+ T cells present in peripheral blood mononuclear cells (PBMC) of healthy human donors.
  • PBMC peripheral blood mononuclear cells
  • HLA- A*02:01-specific peptide was identified that showed a particularly strong immunogencity and a general availability of T cells to this peptide in the human T-cell repertoire.
  • This peptide was identified to be derived from Reticulocalbin 1 (RCN1) and as having the sequence VLAPRVLRA (SEQ ID NO: 2).
  • RCN1 Reticulocalbin 1
  • VLAPRVLRA SEQ ID NO: 2
  • the CD8+ T cells did not recognize the peptides presented herein in cells with normal TAP expression, but did so when confronted with cells having impaired TAP expression.
  • the peptides presented herein are therefore ideal candidates for inducing a T-cell based immune response in vivo against cells associated with impaired antigen presentation in HLA class I presentation molecules and/or impaired TAP function.
  • the data presented herein therefore provides evidence that these peptides could be used as a novel therapy to induce a tumor-specific T cell based immune response in vivo (by activating the na ⁇ ve cognate T cells present within the natural T cell repertoire of the patient).
  • binding agents such as antibodies, TCRs or CARs (or modified cells expressing the same) that specifically bind to these peptides may advantageously be used as a novel immunotherapy for the prevention or treatment of cancer or viral infections associated with impaired antigen presentation in HLA class I molecules and/or impaired TAP function.
  • the peptides described herein are not derived from the mutanome of cancers, but are of ‘self’ origin and therefore constitute universal neoantigens that may be presented on the cell surface of any cell with impaired antigen presentation in HLA class I molecules.
  • SLP synthetic long peptide
  • VLAPRVLRA peptide results in a higher binding affinity of the peptide to HLA-A*02:01 and more efficient cross-presentation by monocyte- derived dendritic cells.
  • this specific amino acid change allows the peptide to be used as a more effective vaccine.
  • the data presented herein shows that small alterations to signal peptide-epitopes retain immunogenicity of TEIPP antigens and render them suitable candidates for the SLP vaccine format. Such vaccines may represent a salvage therapy for immune-escaped cancer by activating RCN1-specific T cells.
  • the invention therefore provides functional variants of VLAPRVLRA (SEQ ID NO: 2) for use as a medicament.
  • a functional variant refers to a variant of SEQ ID NO:2 that still elicits the desired immune response (i.e. a T cell response that is able to recognize and bind to the natural TEIPP of SEQ ID NO:2).
  • the second amino acid, L, in this peptide functions as the main anchor of the peptide in the HLA groove and therefore replacement of this amino acid with alternative amino acids is possible without adversely affecting TCR binding to the peptide:HLA complex.
  • this amino acid may be varied when providing a peptide for a peptide vaccine, as the variant peptide should still elicit the desired immune response (that will be able to recognise and bind to the natural TEIPP, VLAPRVLRA).
  • This also applies to the C-terminal amino acid (A), which, as shown herein, may also be varied without adversely affecting TCR specificity.
  • VLAPRVLRA VLAPRVLRA
  • SEQ ID NO: 2 VX 1 APRVLRX 2
  • X 1 and X 2 are any amino acid (i.e A or R or N or D or C or Q or E or G or H or I or L or K or M or F or P or O or S or U or T or W or Y or V).
  • Reference to “SEQ ID NO: 1” herein therefore encompasses the amino acid sequences of SEQ ID NO: 2 - 21.
  • a peptide is therefore provided comprising the amino acid sequence of SEQ ID NO: 1.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 1.
  • nucleic acid sequences encoding the peptides of the present invention.
  • a nucleic acid sequence is therefore provided encoding the amino acid sequence of SEQ ID NO: 1.
  • a vaccine is also provided comprising: (a) a peptide comprising or consisting of the amino acid sequence of SEQ ID NO: 1, or a nucleic acid encoding the peptide; and (b) a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • the vaccine encodes or comprises an immunogenic peptide.
  • an immunogenic peptide is a peptide that is capable of eliciting an immune response, wherein the immune response is directed against the peptide or an epitope presented by the peptide, optionally with the help of a suitable vaccine adjuvant.
  • a vaccine as described herein is an example of a pharmaceutical composition described herein.
  • the term “pharmaceutical composition” herein therefore encompasses the vaccines described herein.
  • the peptide comprises the amino acid sequence VLAPRVLRA (SEQ ID NO: 2).
  • the peptide may consist of the amino acid sequence VLAPRVLRA (SEQ ID NO: 2).
  • the peptide may comprise or consist of the amino acid sequence of any one of SEQ ID NO: 22, 23 or 24.
  • the peptide comprises the amino acid sequence VLAPRVLRV (SEQ ID NO: 3).
  • the peptide may consist of the amino acid sequence VLAPRVLRV (SEQ ID NO: 3).
  • the peptide may comprise or consist of the amino acid sequence of any one of SEQ ID NO: 25, 26, or 27.
  • the peptide comprises the amino acid sequence VLAPRVLRI (SEQ ID NO: 4).
  • the peptide may consist of the amino acid sequence VLAPRVLRI (SEQ ID NO: 4).
  • the peptide comprises the amino acid sequence VLAPRVLRL (SEQ ID NO: 5).
  • the peptide may consist of the amino acid sequence VLAPRVLRL (SEQ ID NO: 5). In one embodiment, the peptide comprises the amino acid sequence of any one of SEQ ID NOs: 6 - 21. Suitably, the peptide may consist of the amino acid sequence of any one of SEQ ID NOs: 6 - 21. In one embodiment, the peptide comprises the amino acid sequence of SEQ ID NO: 22. Suitably, the peptide may consist of the amino acid sequence of SEQ ID NO: 22. In one embodiment, the peptide comprises the amino acid sequence of SEQ ID NO: 23. Suitably, the peptide may consist of the amino acid sequence of SEQ ID NO: 23.
  • the peptide comprises the amino acid sequence of SEQ ID NO: 24.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 24.
  • the peptide comprises the amino acid sequence of SEQ ID NO: 25.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 25.
  • the peptide comprises the amino acid sequence of SEQ ID NO: 26.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 26.
  • the peptide comprises the amino acid sequence of SEQ ID NO: 27.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 27.
  • the peptide may have no more than 35 amino acids.
  • the peptide may consist of from 10 to 35 amino acids.
  • the peptide may consist of from 20 to 35 amino acids.
  • the peptide may be conjugated to an immune stimulatory compound.
  • the immune stimulatory compound may comprise a TLR, NLR, RLR, CLR, or ALR ligand.
  • a nucleic acid encoding the peptide of the invention is also provided.
  • the nucleic acid may be mRNA or DNA.
  • the nucleic acid may be an isolated nucleic acid.
  • the invention provides a complex comprising: a) a peptide comprising the amino acid sequence of SEQ ID NO: 1, and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1.
  • the complex may be an isolated complex.
  • the binding agent may be an HLA-A*02 molecule.
  • the complex may be a complex of HLA-A*02 with any one of SEQ ID NOs: 2 - 21.
  • the complex may be an HLA- A*02:VLAPRVLRA complex, an HLA-A*02:VLAPRVLRV complex, an HLA-A*02:VLAPRVLRI complex, or an HLA-A*02:VLAPRVLRL complex.
  • the binding agent may be an HLA-A*02:01 molecule.
  • the complex may be a complex of HLA-A*02:01 with any one of SEQ ID NOs: 2 - 21.
  • the complex may be an HLA-A*02:01:VLAPRVLRA complex, an HLA-A*02:01:VLAPRVLRV complex, an HLA- A*02:01:VLAPRVLRI complex, an HLA-A*02:01:VLAPRVLRL complex.
  • the complex may be: a.
  • VLAPRVLRA:HLA-A*02 complex a VLAPRVLRV:HLA-A*02 complex, a VLAPRVLRI:HLA- A*02 complex, a VLAPRVLRL:HLA-A*02 complex, a VLAPRVLRR:HLA-A*02 complex, a VLAPRVLRN:HLA-A*02 complex, a VLAPRVLRD:HLA-A*02 complex, a VLAPRVLRC:HLA-A*02 complex, a VLAPRVLRQ:HLA-A*02 complex, a VLAPRVLRE:HLA-A*02 complex, a VLAPRVLRG:HLA-A*02 complex, a VLAPRVLRH:HLA-A*02 complex, a VLAPRVLRK:HLA-A*02 complex, a VLAPRVLRM:HLA-A*02 complex, a VLAPRVLRF:HLA-A*02 complex,
  • VLAPRVLRA:HLA-A*02:01 complex a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, a VLAPRVLRL:HLA-A*02:01 complex, a VLAPRVLRR:HLA- A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, a VLAPRVLRD:HLA-A*02:01 complex, a VLAPRVLRC:HLA-A*02:01 complex, a VLAPRVLRQ:HLA-A*02:01 complex, a VLAPRVLRE:HLA-A*02:01 complex, a VLAPRVLRG:HLA-A*02:01 complex, a VLAPRVLRH:HLA-A*02:01 complex, a VLAPRVLRK:HLA-A*02:01 complex, a VLAPRVLRM
  • the invention provides a cell loaded with a peptide according to the invention or a cell loaded with or expressing the complex according to the invention.
  • the cell may be an antigen presenting cell.
  • the antigen presenting cell may be selected from a macrophage, dendritic cell, a monocyte, a B-cell or a synthetic form of antigen presenting cell.
  • the cell may be loaded with a peptide according to the invention comprising an amino acid sequence selected from the group consisting of:VLAPRVLRA (SEQ ID NO: 2), VLAPRVLRV (SEQ ID NO: 3), VLAPRVLRI (SEQ ID NO: 4) and VLAPRVLRL (SEQ ID NO: 5).
  • the cell may be loaded with a peptide comprising an amino acid sequence selected from any one of SEQ ID NO: 22 to 27.
  • the cell may be loaded with or expressing a complex according to the invention selected from the group consisting of: a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex and a VLAPRVLRL:HLA-A*02:01.
  • the invention provides an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain, the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence; wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 1 (e.g. when the peptide is complexed with HLA).
  • the CDR3 sequences together may specifically bind to a peptide comprising the amino acid sequence of any one of SEQ ID NOs: 2 – 21 (e.g. when the peptide is complexed with HLA).
  • the CDR3 sequences together may specifically bind to a peptide comprising the amino acid sequence VLAPRVLRA (SEQ ID NO: 2), VLAPRVLRV (SEQ ID NO: 3), VLAPRVLRI (SEQ ID NO: 4) or VLAPRVLRL (SEQ ID NO: 5) (e.g. when the peptide is complexed with HLA).
  • the CDR3 sequences together may specifically bind to the peptide when it is complexed with HLA-A*02:01.
  • the nucleic acid sequence may encode a T cell receptor.
  • the invention provides an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain, the composition comprising: (i) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:42, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:45, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 2 (e.g. when the peptide is complexed with HLA)); or (ii) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:52, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 55, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 2 (e.g. when the peptide is complexed with HLA)); or (iii) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:62, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:65, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 2 (e.g. when the peptide is complexed with HLA)); or (v) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:82, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 85, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 2 (e.g. when the peptide is complexed with HLA)); or (vi) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:92, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 95, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 2 (e.g. when the peptide is complexed with HLA)); or (vii) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:102, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 105, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 2 (e.g. when the peptide is complexed with HLA)).
  • the peptide may comprise an amino acid sequence selected from the group consisting of: SEQ ID NOs: 2 - 21.
  • the peptide may comprise an amino acid sequence selected from the group consisting of: SEQ ID NOs: 2 - 5.
  • the peptide may comprise an amino acid sequence selected from the group consisting of: SEQ ID NOs: 2 - 4.
  • the peptide may comprise an amino acid sequence selected from the group consisting of: SEQ ID NOs: 3 - 5.
  • the isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain may comprise: (i) a V ⁇ domain CDR3 that comprises or consists of the amino acid sequence of SEQ ID NO: 42, and the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO:45; or (ii) a V ⁇ domain CDR3 that comprises or consists of the amino acid sequence of SEQ ID NO: 52, and the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO:55; or (iii) a V ⁇ domain CDR3 that comprises or consists of the amino acid sequence of SEQ ID NO: 62, and the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO:65; or (iv) a V ⁇ domain CDR3 that comprises or consists of the amino amino acid sequence of SEQ ID
  • the isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain may comprise: (i) a V ⁇ domain that comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 46; and the V ⁇ domain comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 48; or (ii) a V ⁇ domain that comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 56; and the V ⁇ domain comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 58; or (iii) a V ⁇ domain that comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 66; and the V ⁇ domain comprises:
  • the encoded RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is capable of specifically binding to a peptide:HLA complex selected from the group consisting of: a VX1APRVLRX2:HLA-A*02 complex (wherein X1 and X2 are any amino acid), a VLAPRVLRA:HLA-A*02 complex, a VLAPRVLRV:HLA- A*02 complex, a VLAPRVLRI:HLA-A*02 complex, a VLAPRVLRL:HLA-A*02 complex, a VLAPRVLRR:HLA-A*02 complex, a VLAPRVLRN:HLA-A*02 complex, a VLAPRVLRD:HLA-A*02 complex, a VLAPRVLRC:HLA-A*02 complex, a VLAPRVLRQ:HLA-A*02 complex,
  • the encoded RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is capable of specifically binding to a peptide:HLA complex selected from the group consisting of: a VX 1 APRVLRX 2 :HLA-A*02:01 complex (wherein X1 and X2 are any amino acid), a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, a VLAPRVLRL:HLA- A*02:01 complex, a VLAPRVLRR:HLA-A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, a VLAPRVLRD:HLA-A*02:01 complex, a VLAPRVLRC:HLA-A*02:01 complex
  • the encoded RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is capable of specifically binding to a peptide:HLA complex selected from the group consisting of: a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, and a VLAPRVLRL:HLA-A*02:01 complex.
  • the encoded RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is capable of specifically binding to a peptide:HLA complex selected from the group consisting of: a VLAPRVLRA:HLA-A*02:01 complex and a VLAPRVLRV:HLA-A*02:01 complex.
  • the CDR3 sequences together may specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 1 (e.g. when the peptide is complexed with HLA).
  • the CDR3 sequences together may specifically bind to a peptide comprising the amino acid sequence of any one of SEQ ID NOs: 2 – 21 ((e.g. when the peptide is complexed with HLA).
  • the peptide may comprise the amino acid sequence VLAPRVLRA (SEQ ID NO: 2), VLAPRVLRV (SEQ ID NO: 3), VLAPRVLRI (SEQ ID NO: 4) or VLAPRVLRL (SEQ ID NO: 5).
  • the CDR3 sequences together may specifically bind to the peptide when it is complexed with HLA-A*02 or HLA-A*02:01.
  • the encoded binding protein comprises a TCR, an antigen binding fragment of a TCR, a chimeric antigen receptor (CAR), or an ImmTAC.
  • the invention provides an isolated nucleic acid encoding a chimeric antigen receptor protein comprising a target binding moiety that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1.
  • the peptide may comprise the amino acid sequence of any one of SEQ ID NOs: 2 - 21.
  • the peptide may comprise the amino acid sequence VLAPRVLRA (SEQ ID NO: 2), VLAPRVLRV (SEQ ID NO: 3), VLAPRVLRI (SEQ ID NO: 4) or VLAPRVLRL (SEQ ID NO: 5).
  • a modified cell transformed, transfected or transduced with a nucleic acid comprising a nucleic acid sequence encoding a peptide described herein, a nucleic acid composition described herein, a nucleic acid described herein, or a nucleic acid sequence described herein or a vector described herein is also provided.
  • the modified cell may be a human cell.
  • the modified cell may be selected from the group consisting of a CD8 T cell, a CD4 T cell, an NK cell, an NKT cell, a gamma-delta T cell, an innate lymphoid cell (ILC), a hematopoietic stem cell, a progenitor cell, a T cell line or a NK-92 cell line.
  • the invention provides a pharmaceutical composition comprising a) a peptide, b) a nucleic acid, a nucleic acid sequence, or a nucleic acid composition, c) a complex, or d) a cell according to the invention, and a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • any one of a), b) or c) may be present in the pharmaceutical composition by virtue of them being encoded or expressed (as appropriate) by a cell that is present within the pharmaceutical composition.
  • any one of b) or c) may be encoded by a cell that is combined with a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier to generate the pharmaceutical composition; or any of a) or b) may be expressed by a cell that is combined with a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier to generate the pharmaceutical composition. More details on this are provided below under “detailed description”.
  • the composition may be formulated as a vaccine.
  • the composition may be a peptide vaccine.
  • the vaccine, peptide vaccine, or composition may comprise an immune stimulatory compound.
  • a pharmaceutical composition described herein is provided for use as a medicament.
  • a vaccine or peptide vaccine described herein is provided for use as a medicament.
  • a pharmaceutical composition described herein (comprising a nucleic acid, a nucleic acid composition, a complex, or a cell according to the invention) and a vaccine or peptide vaccine described herein are provided for use as a medicament.
  • the pharmaceutical composition described herein and the vaccine or peptide vaccine described herein may be combined for use as a medicament.
  • the combination may comprise administering the vaccine or peptide vaccine prior to, during, and/or after administration of the pharmaceutical composition.
  • the pharmaceutical composition, vaccine, and/or peptide vaccine may be for use in the prevention or treatment of a pre-cancer, a cancer or a viral infection associated with impaired HLA class I antigen presentation in a human subject.
  • the pre-cancer or cancer may be a pre-cancer or cancer with impaired peptide processing machinery.
  • the pharmaceutical composition, vaccine, and/or peptide vaccine may be for use in treating or preventing a pre-cancer, a cancer or viral infection associated with impaired HLA class I antigen presentation in a human subject, wherein the subject has been identified as having a pre-cancer, a cancer or viral infection associated with impaired HLA class I antigen presentation by the presence of a VX 1 APRVLRX 2 :HLA-A*02 complex (wherein X 1 and X 2 are any amino acid), a VLAPRVLRA:HLA-A*02 complex, a VLAPRVLRV:HLA-A*02 complex, a VLAPRVLRI:HLA- A*02 complex, a VLAPRVLRL:HLA-A*02 complex, a VLAPRVLRR:HLA-A*02 complex, a VLAPRVLRN:HLA-A*02 complex, a VLAPRVLRD:HLA-A*02 complex, a VLAPRVLRC:
  • the pharmaceutical composition, vaccine, and/or peptide vaccine may be for use in treating or preventing a pre-cancer, a cancer or viral infection associated with impaired HLA class I antigen presentation in a human subject, wherein the subject has been identified as having a pre-cancer, a cancer or viral infection associated with impaired HLA class I antigen presentation by the presence of a VX 1 APRVLRX 2 :HLA-A*02:01 complex (wherein X 1 and X 2 are any amino acid), a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, a VLAPRVLRL:HLA-A*02:01 complex, a VLAPRVLRR:HLA- A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, a VLAPRVLRD:HLA-
  • the invention provides a method of treating a condition in a human subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition, vaccine, and/or peptide vaccine according to the invention.
  • the method of treating may be for the prevention or treatment of a pre-cancer, a cancer or a viral infection associated with impaired HLA class I antigen presentation.
  • a method of generating a T cell receptor comprising contacting a nucleic acid sequence or composition according to the invention with a cell under conditions in which the nucleic acid sequence is incorporated and expressed by the cell to generate the T cell receptor that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1 (or to the peptide when complexed with HLA).
  • the peptide may comprise the amino acid sequence of any one of SEQ ID NOs: 2 - 21.
  • the method is ex vivo.
  • the invention provides a use of a peptide comprising an amino acid sequence of SEQ ID NO: 1, or a complex according to the invention, for identifying a therapeutic binding protein.
  • the peptide may comprise or consist of an amino acid sequence of any one of SEQ ID NOs: 2 – 27.
  • the therapeutic binding protein may be capable of preventing or treating a pre-cancer, a cancer or a viral infection associated with impaired HLA class I antigen presentation.
  • the therapeutic binding protein may comprise: a TCR, an antigen binding fragment of a TCR, or a chimeric antigen receptor (CAR), or an ImmTAC; or the therapeutic binding protein may be an antibody.
  • CAR chimeric antigen receptor
  • FIG 1 shows specificity of two T cell cultures for the peptide VLAPRVLRA.
  • T cell cultures of donors 2028 and 4409 were stimulated with the peptide VLAPRVLRA (SEQ ID NO:2) and analyzed via flow cytometry for the percentage of CD8+ T cells and the percentage therein of CD8+ T cells that stained positive with the HLA-A*02:01 tetramer presenting peptide VLAPRVLRA.
  • Both VLAPRVLRA-specific T cell cultures comprise more than 85% CD8+ T cells that stained for more than 98% positive with VLAPRVLRA-loaded HLA-A*02:01 tetramers (indicated by HLA-A*02:01: VLAPRVLRA TM in Figure 1).
  • VLAPRVLRA-specific T cells were used in subsequent experiments described below.
  • peptide VLAPRVLRA-specific T cells were used in subsequent experiments described below.
  • peptide VLAPRVLRA-specific T cells preferential recognition of TAP impaired tumor cells by VLAPRVLRA-specific T cells.
  • VLAPRVLRA-specific T cells preferentially recognized 518A2 melanoma and 08.11 melanoma cells when TAP is knocked out, but not non-tumor cells such as fibroblasts, immortalized Human Embryonic Kidney 293 (HEK-293T) cells or proximal tubular cells derived from a normal human adult male kidney (HK2), unless TAP was knocked out (HK2 TAP KO).
  • HEK-293T immortalized Human Embryonic Kidney 293
  • HK2 TAP KO proximal tubular cells derived from a normal human adult male kidney
  • FIGS. 2A and 2B shows the following: the T cell cultures of donor 2028 and 4409, stimulated with peptide VLAPRVLRA (respectively indicated by 2028 p113 and 4409 p113), were stimulated with HLA-A*02:01 matched wild type (WT) melanoma cells (518A2), its variant in which TAP was knocked out (518A2 TAP KO), and with 25 ug/ml cognate peptide.
  • WT wild type
  • TAP KO wild type melanoma cells
  • culture 4409 was stimulated with HLA-A*02:01 matched HK2 cells, fibroblasts and HEK293T cells.
  • FIG. 2C shows the following: T cell culture 4409 stimulated with peptide VLAPRVLRA (indicated by 4409 p113) was stimulated with both wild type (WT) melanoma cells (518A2, 08.11) and their variants in which TAP was knocked out (518A2 TAP KO, 8.11 TAP KO) as well as with wild type HK2 cells and its variant with TAP knocked out (HK2 TAP KO).
  • FIG. 3 shows that no recognition of Epstein-Barr virus (EBV) transformed B-cells, monocytes or monocyte-derived dendritic cells (DCs) by VLAPRVLRA-specific T cells was observed.
  • EBV Epstein-Barr virus
  • FIG. 3A the p113 VLAPRVLRA-specific T cell culture of donor 4409 was stimulated with HLA- A*02:01 matched EBV-transformed B-cell line (EBVs) (indicated by “T cells + EBV”). Negative controls are indicated by “T cells” and “EBVs”.
  • T cells + EBVs + p113 As a control for T-cell reactivity peptide p113 VLAPRVLRA was added (indicated by “T cells + EBVs + p113”).
  • the p113 VLAPRVLRA-specific T cell culture of donor 4409 was stimulated with monocytes (indicated by “Mono’s 7110”).
  • Monocytes As a control for T-cell reactivity peptide p113 VLAPRVLRA was added (indicated by “Mono’s 7110 p113 loaded and washed”).
  • FIG 3C shows the following: the p113 VLAPRVLRA-specific T cell culture of donor 4409 was stimulated with monocyte-derived DCs of two healthy donors 0559 and 4553 (indicated by “T cells + moDCs 0559” and “T cells + moDCs 4553”. T cells with 25 ug/ml peptide served as control for reactivity (indicated by “T cells + p113”). T cells only served as a negative control (indicated by “T cells only”). The amount of GM-CSF produced is shown on the y-axis in Figure 3.
  • Figure 4 shows expression levels of TAP1 and of RCN1, the gene coding for peptide 113.
  • RCN1 and TAP1 were determined in monocytes of two different donors 2752 and 4409 (indicated by “2752 mono’s and 4409 mono’s”), monocyte-derived dendritic cells of two different donors 2752 and 4409 (indicated by “2752 moDCs” and “4409 moDCs”), 518A2 melanoma cells (indicated by “518A2 WT”) and its TAP knock-out derivative (inidcated by “518A2 TAP KO”) as well as HK2 cells (indicated by “HK2 WT”) and its TAP knock-out derivative (indicated by “HK2 TAP KO”).
  • the expression of the indicated genes was tested by qPCR and measurements were performed in triplicates.
  • Figure 5 shows expression levels of RCN1 and TAP1 in 518A2 wild type cells, 518A2 TAP KO cells and 518A2 TAP KO RCN1 KO cells.
  • the expression levels of RCN1 and TAP1 were determined in 518A2 wild type melanoma cells (518A2 WT), 518A2 cells in which TAP was knocked out (518A2 TAP KO) and 518A2 cells in which both TAP and RCN1 were knocked out (518A2 TAP/RCN1 KO clone 2A4).
  • the expression of the indicated genes was tested by qPCR and measurements were performed in triplicates. Ct-values were normalized to the expression of the housekeeping genes and CPSF6.
  • the peptide 113 (VLAPRVLRA)-specific T-cell cultures from two different donors were stimulated with 518A2 wild type melanoma cells (518A2 WT), 518A2 cells in which TAP was knocked out (518A2 TAP KO) and 518A2 cells in which both TAP and RCN1 were knocked out (518A2 TAP/RCN1 KO clone 2A4).
  • 518A2 WT 518A2 wild type melanoma cells
  • 518A2 cells in which TAP was knocked out 518A2 TAP KO
  • 518A2 cells in which both TAP and RCN1 were knocked out 518A2 TAP/RCN1 KO clone 2A4
  • the amount of interferon-gamma (IFNy) or GM-CSF produced in pg/ml is shown on the y-axis.
  • the dashed line indicates the maximal value of the GM-CSF dose
  • the peptide 113 (VLAPRVLRA)-specific T-cell culture from donor 4409 was stimulated with monocyte- derived dendritic cells (MoDCs) from three different donors (indicated by MoDCs 0559, MoDC 4553 and MoDC 7401), which were loaded with (10uM for 20-24h) the indicated synthetic long peptides (SLP) (“p113 long 1” (SEQ ID NO: 22), “p113 long 2” (SEQ ID NO: 23), “p113 long 3” (SEQ ID NO: 24)) or with no peptide as background control (indicated by “No peptide”).
  • SLP synthetic long peptides
  • T cells + p113 The T cell culture incubated with 25ug/ml p113-peptide VLAPRVLRA (indicated by “T cells + p113”) served as positive control for reactivity.
  • the amount of interferon-gamma (IFNy) or GM-CSF produced is shown on the y-axis in pg/ml. None of the SLP variants were cross-presented to T cells, whereas exogenous pulsing of the short VLAPRVLRA peptide did stimulate the T cells. These results suggested that cross-presentation of the VLAPRVLRA epitope from its longer peptide stretch is not efficient and had to be optimized for vaccine applications.
  • T cell culture incubated with 25ug/ml of the wild type p113-peptide VLAPRVLRA (indicated by “T cells + p113” or “p113”) or its V-variant VLAPRVLRV (p113V: SEQ ID NO: 3) (indicated by “T cells + p113V” or “p113V”) served as positive control for T-cell reactivity, while T cells incubated without any added peptide served as a negative control (indicated by “T cells”).
  • T cells incubated without any added peptide served as a negative control (indicated by “T cells”).
  • the amount of interferon-gamma (IFNy) or GM-CSF produced is shown on the y-axis in pg/ml.
  • FIG. 9 shows that T cells transduced in order to express different unique HLA-A*02:01-restricted RCN121–29 CD8 T cell receptors as identified by the inventors, can bind to murine TCRb antibody and MHC multimers of HLA-A*02:01-molecules presenting the peptide RCN121–29 VLAPRVLRA.
  • TCR-alpha and TCR-beta chains of each respective identified TCR were cloned into a retroviral expression vector, which was used to transduce T cells, resulting in the generation of T cells expressing the respective TCRs for peptide RCN121–29 VLAPRVLRA. These T cells were analyzed via flow cytometry.
  • the introduced genes contain the murine TCR-C ⁇ domain which enhances correct pairing of transgenic alpha and beta chains. Expression of this domain is confirmed in TCR-transduced T cells after incubation with murine TCRb antibody.
  • FIG. 10 shows the identification of HLA-presented peptides FLGPWPAAS, FLGPWPAAV, VLAPRVLRA, and SLGDWGAEA in different isolated human primary tumour samples.
  • Various human primary tumour samples were obtained after which their HLA ligandomes were determined via mass spectrometry by standard methods in the art.
  • the primary tumour samples comprise four melanoma samples (Mel0401, Mel1207, Mel1213, and Mel1502), three ovarian cancer samples (OVAL1, OVAL10, and OVAL23), and two cervical cancer samples (COV362-4 and COV413b).
  • the table in figure 10 shows the presence (+) or absence (no) of four possible HLA- presented peptides (FLGPWPAAS, FLGPWPAAV, VLAPRVLRA, and SLGDWGAEA) in the samples after analysis of their HLA ligandomes. It can be devisved that VLAPRVLRA is present in a large proportion of the samples and therefore may be an attractive target for therapeutic interventions.
  • FIG 11 shows that T cells transduced in order to express different unique HLA-A*02:01- restricted RCN121–29 CD8 T cell receptors as identified by the inventors herein can be activated by TAP-deficient tumor cells.
  • Figure 11A TCR generated by TCR-gene transfer and enriched after tetramer-guided MACS sort were cultured overnight with (from left to right in the figure) HLA- A*0201-matched wild type (WT), TAP-deficient (TAP KO) and TAP and RCN1-deficient (TAP/Ag KO) 518A2 tumor cells, cultured by themselves (T cells only), or cultured with 25 ug/ml cognate peptide (T cells + peptide).
  • WT wild type
  • TAP KO TAP-deficient
  • TAP/Ag KO TAP and RCN1-deficient
  • TCR1 The amount of GM-CSF, IFNy and CCL4 produced is shown for TCR1, 2, 4, 5, 6, 9 and 13.
  • Figure 11B TCR generated by TCR-gene transfer and enriched after tetramer-guided MACS sort were cultured overnight with (from left to right in the figure) HLA- A*0201-matched wild type (WT), or TAP-deficient (TAP KO) 08.11 tumor cells.
  • WT HLA- A*0201-matched wild type
  • the amount of CCL4 produced is shown for TCR 4 and 9.
  • the experiments of figure 11 were performed using the materials and methods as set out in the section Examples below.
  • the TCRs used in figure 11 are the same as in the Examples and their sequences can be found in tables 7-13.
  • a peptide comprising the amino acid sequence of SEQ ID NO: 1 is provided herein.
  • a peptide comprising the amino acid sequence VLAPRVLRA is provided herein.
  • a peptide comprising the amino acid sequence VLAPRVLRV is provided herein.
  • a peptide comprising the amino acid sequence VLAPRVLRI is provided herein.
  • a peptide comprising the amino acid sequence VLAPRVLRL is provided herein.
  • a peptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 2 - 21 is provided herein, preferably wherein the amino acid sequence is selected from the group consisting of SEQ ID NO: 2 - 5, more preferably wherein the amino acid sequence is selected from the group consisting of SEQ ID NO: 2 - 4, or from the group consisting of SEQ ID NO: 3 - 5.
  • a peptide comprising the amino acid sequence of SEQ ID NO:2 includes the peptides of SEQ ID NO: 22 to 24. Reference to a peptide comprising SEQ ID NO:2 therefore expressly encompasses the peptides of SEQ ID NO:22 to 24.
  • a peptide comprising the amino acid sequence of SEQ ID NO:3 includes the peptides of SEQ ID NO: 25 to 27. Reference to a peptide comprising SEQ ID NO:3 therefore expressly encompasses the peptides of SEQ ID NO:25 to 27. Preferably, the peptide is an isolated peptide. Also provided herein are nucleic acid molecules encoding the peptides of the present invention. A nucleic acid is provided encoding the amino acid sequence of SEQ ID NO: 1. A nucleic acid is provided encoding any one of the amino acid sequences of SEQ ID NO: 1 - 27.
  • any suitable nucleic acid sequence encoding any one of the amino acid sequences of SEQ ID NO: 1 - 27 may be used accordingly. These can suitably be selected by the skilled person.
  • an “isolated peptide” refers to a peptide that is not in its natural environment. The peptide may therefore be of synthetic origin (or alternatively, of natural original, but isolated from its natural environment). In the context of this disclosure, the natural environment of these peptides is within the human body. Accordingly, when the peptides are present e.g. in a pharmaceutical composition (comprising adjuvants etc) they are considered to be in isolated form, as they are not in their natural environment.
  • compositions may be part of a pharmaceutical composition, wherein the composition comprises: (a) the peptide; and (b) a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • compositions may be referred to as vaccines (i.e. peptide vaccines) herein.
  • nucleic acids described herein may be part of a pharmaceutical composition, wherein the compostion comprises: (a) a nucleic acid encoding a peptide of the invention; and (b) a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • vaccines i.e. nucleic acid vaccines
  • an example of such a vaccine comprising a nucleic acid sequence encoding the peptide, optionally comprising the peptide, is an mRNA vaccine.
  • the vaccine according to the invention is an mRNA vaccine.
  • the peptide according to the invention may consist of the amino acid sequence of any one of SEQ ID NOs: 1 - 27 only. Alternatively, the peptide may include additional amino acids, and thus comprise the amino acid sequence of any one of SEQ ID NOs: 1 - 27.
  • peptides comprising the amino acid sequence VLAPRVLRV (SEQ ID NO:3) (such as any one of the peptides of SEQ ID NO: 25 to 27) may have a higher binding affinity to HLA- A*02 than an equivalent peptide in which the VLAPRVLRV (SEQ ID NO:3) sequence is replaced with VLAPRVLRA (SEQ ID NO:2).
  • peptides comprising VLAPRVLRV may have a higher binding affinity to HLA-A*02:01 than an equivalent peptide in which the VLAPRVLRV (SEQ ID NO:3) sequence is replaced with VLAPRVLRA (SEQ ID NO:2).
  • VLAPRVLRV SEQ ID NO:3
  • VLAPRVLRA VLAPRVLRA
  • VLAPRVLRV amino acid sequence VLAPRVLRV
  • monocyte-derived dendritic cells may be more effective (improved/higher) than the cross- presentation of an equivalent peptide in which the VLAPRVLRV (SEQ ID NO:3) sequence is replaced with VLAPRVLRA (SEQ ID NO:2).
  • a peptide comprising the amino acid sequence VLAPRVLRV (SEQ ID NO:3) (such as any one of the peptides of SEQ ID NO: 25 to 27) may be more efficiently presented by monocyte-derived dendritic cells than an equivalent peptide in which the VLAPRVLRV (SEQ ID NO:3) sequence is replaced with VLAPRVLRA (SEQ ID NO:2).
  • cross-presentation of a 10-35 amino acid long peptide comprising the equivalent peptide epitope can be tested by the use of monocyte-derived dendritic cells (DC) and a CD8+ T cell clone recognizing the peptide VLAPRVLRA in the context of HLA class I.
  • Monocyte-derived DC are obtained by incubating peripheral blood mononuclear cells with anti-CD14 magnetic beads for 20 min at 4 °C followed by the isolation of CD14 positive monocytes were isolated using magnetic separation columns.
  • the CD14+ monocytes are cultured in RPMI medium supplemented with 10% FCS, GM-CSF (800 units/ml), and IL-4 (500 units/ml) for 6 days to generate immature monocyte-derived dendritic cells.
  • the immature monocyte- derived DCs are incubated with the synthetic long peptide at different doses (e.g.20 ⁇ g/ml, 10 ⁇ g/m, 5 ⁇ g/ml) for 24h, and matured with LPS (20 ng/ml) stimulation on day 7.
  • the cross- presentation of the peptide by these monocyte-derived DC is monitored by the reactivity of the CD8+ T cell clone co-cultured with the peptide-pulsed monocyte-derived DCs at different ratio’s (e.g.10 T cells to 1 DC; 5 T cells to 1 DC; 1 T cell to 1 DC).
  • the reactivity of a CD8+ T cell clone can be tested in different ways, including measurement of cytokine production (e.g. GM-CSF or Interferon-gamma) in the supernatant of the co-culture and can be compared to control co- cultures in which the monocyte-derived DC are pulsed with an irrelevant HLA class I binding peptide or with no peptide.
  • the monocyte-derived DC can be pulsed with the natural short epitope VLAPRVLRA.
  • the peptides described herein may have an equivalent or a higher binding affinity to HLA-A*02 (e.g. HLA-A*02:01) and/or be presented by monocyte-derived dendritic cells at a level that is equivalent or more efficient (improved/higher) than an equivalent peptide comprising VLAPRVLRA.
  • HLA-A*02 e.g. HLA-A*02:01
  • equivalent peptides are peptides with an identical amino acid sequence except for the recited difference.
  • the peptide may be no more than 35 amino acids in length; e.g. no more than 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10 or 9 amino acids.
  • the peptide may be no more than 30 amino acids in length.
  • the peptide may be no more than 29 amino acids in length.
  • the peptide may be no more than 28 amino acids in length.
  • the peptide may be no more than 27 amino acids in length.
  • the peptide may be no more than 26 amino acids in length.
  • the peptide may be no more than 25 amino acids in length.
  • the peptide may be no more than 24 amino acids in length. In another example, the peptide may be no more than 23 amino acids in length. Different lengths of peptide have been shown to be particularly effective as peptide vaccines. For example, Ossendorp (Ossendorp et al., (1998)) describe that 9-19 amino acid long peptides are able to induce a CD4+ helper T cell response. Accordingly, the peptide of the invention may be from 9 to 19 amino acids long. Peptides that are longer than the conventional 9mer sequence presented by HLA may be more efficient in inducing an immune response.
  • the peptides described herein may be from 10 to 19 amino acids long.
  • Bijker (Bijker et al., (2007)) demonstrate that a 9-mer HPV CTL epitope can induce a CD8+ response to RAHYNIVTF but that a 35-mer peptide comprising this epitope is more efficient. This is further supported by Beyranvand-Nejad (Beyranvand-Nejad et al., (2016)), showing that a 35 mer HPV peptide works well to induce RAHYNIVTF -specific CD8+ T cells.
  • the peptides described herein may be from 9 to 35 amino acids long.
  • the peptides have a total of 9 to 35 amino acids, which includes the sequence of any one of SEQ ID NO: 1 - 21.
  • the peptides have the sequence of any one of SEQ ID NO: 1 – 21 and 0 to 26 additional amino acids.
  • the 0 to 26 additional amino acids of the peptide may be located N- terminal or C-terminal to the sequence of any one of SEQ ID NO: 1 - 21.
  • the additional amino acids may flank the sequence of any one of SEQ ID NO: 1 - 21 (i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence of any one of SEQ ID NO: 1 - 21). Additional amino acids located N-terminal, C- terminal or flanking any one of SEQ ID NO: 1 - 21 are referred to collectively as “additional amino acids” herein.
  • the peptides may have a total of 10 to 35 amino acids, which includes the sequence of any one of SEQ ID NOs 1 - 21.
  • the peptides may have the sequence of any one of SEQ ID NOs 1 - 21 and 1 to 26 additional amino acids.
  • the 1 to 26 additional amino acids of the peptide may be located N-terminal or C- terminal to the sequence of any one of SEQ ID NOs 1 - 21.
  • the additional amino acids may flank the sequence of any one of SEQ ID NOs 1 - 21 (i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence of any one of SEQ ID NOs 1 - 21).
  • the peptides described herein may be from 15 to 30 amino acids long, where there are additional 6 to 21 additional amino acids.
  • the additional amino acids may be located N- terminal or C-terminal to sequence of any one of SEQ ID NO: 1 - 21.
  • the additional amino acids may flank the sequence of any one of SEQ ID NO: 1 - 21 (i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence of any one of SEQ ID NO: 1 - 21).
  • the peptides described herein may be from 18 to 27 amino acids long, where there are additional 9 to 18 additional amino acids.
  • the additional amino acids may be located N- terminal or C-terminal to sequence of any one of SEQ ID NO: 1 - 21.
  • the additional amino acids may flank the sequence of any one of SEQ ID NO: 1 - 21 (i.e.
  • the peptides described herein may be from 21 to 24 amino acids long, where there are additional 12 to 15 additional amino acids.
  • the additional amino acids may be located N-terminal or C-terminal to sequence of any one of SEQ ID NO: 1 - 21.
  • the additional amino acids may flank the sequence of any one of SEQ ID NO: 1 - 21 (i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence of any one of SEQ ID NO: 1 - 21).
  • Examples of peptides that comprise the amino acid sequence of SEQ ID NO:2 with additional amino acids are shown in SEQ ID NO: 22 to 24.
  • Examples of peptides that comprise the amino acid sequence of SEQ ID NO:3 with additional amino acids are shown in SEQ ID NO: 25 to 27.
  • the peptides described herein may be 27 amino acids long.
  • the peptides described herein may be 24 amino acids long.
  • the peptides described herein may be 21 amino acids long.
  • the peptides described herein may be 18 amino acids long.
  • the peptides described herein are 24 amino acids long.
  • the peptides have a total of e.g.27, 24, 21 or 18 amino acids, which includes the sequence of any one of SEQ ID NOs 1 - 21.
  • the peptides have the sequence of any one of SEQ ID NOs 1 - 21 and a suitable number of additional amino acids (i.e. to generate a peptide that is 27, 24, 21 or 18 amino acids long).
  • the N-terminus of a peptide (also known as the amino-terminus, NH2-terminus, N-terminal end or amine-terminus) is the start of a peptide terminated by an amino acid with a free amine group (-NH2).
  • peptide sequences are written N-terminus to C-terminus (from left to right).
  • the C-terminus also known as the carboxyl-terminus, carboxy-terminus, C-terminal tail, C- terminal end, or COOH-terminus
  • the C-terminus is the end of an amino acid chain (protein or polypeptide), terminated by a free carboxyl group (-COOH).
  • the terms “N-terminal” and “C-terminal” are used to describe the relative position of e.g. a sequence within a peptide. Accordingly, a sequence that is “N-terminal” is positioned closer (in relative terms) to the N-terminus than to the C-terminus of the peptide.
  • a domain that is “C-terminal” is positioned (in relative terms) closer to the C-terminus than to the N- terminus of the peptide.
  • the term “positioned” refers to the location of the sequence within the linear amino acid sequence of the peptide.
  • Peptides comprising an N-terminal amino acid sequence (A) and a C-terminal amino acid sequence (B) are conventionally written as A-B i.e. N-terminal to C-terminal (left to right).
  • the peptides described herein include additional amino acids located N-terminal, C- terminal or flanking the sequence of any one of SEQ ID NOs 1 - 21, any appropriate additional amino acid sequences may be included.
  • the additional amino acids may be amino acid sequences that are naturally located N-terminal, C-terminal or flanking the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may be all located N- terminal to the sequence of any one of SEQ ID NOs 1 - 21 and may be the natural sequence that is found N-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may be all be located C-terminal to the sequence of any one of SEQ ID NOs 1 - 21 and may be the natural sequence that is found C-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may flank the sequence of any one of SEQ ID NOs 1 - 21 (i.e.
  • the additional amino acids may be all located N-terminal to a VLAPRVLRV sequence, optionally wherein the additional amino acid sequences are amino acid sequences that are naturally located N-terminal of the VLAPRVLRA sequence in RCN1.
  • the peptide sequence of interest e.g. VLAPRVLRV
  • VLAPRVLRV is located at the C-terminus of the peptide according to the invention.
  • Peptides that comprise the amino acid sequence of any one of SEQ ID NOs 1 - 21 and consist of from 10 to 35 amino acids may include any appropriate additional amino acid sequences.
  • the additional amino acids may be amino acid sequences that are naturally located N- terminal, C-terminal or flanking the VLAPRVLRA sequence in RCN1.
  • the additional 1 to 26 amino acids may all be located N-terminal to any one of SEQ ID NOs 1 - 21, preferably the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence, and may be the natural sequence that is found N-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional 1 to 26 amino acids may be all be located C-terminal to any one of SEQ ID NOs 1 - 21, preferably the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence, and may be the natural sequence that is found C-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may flank any one of SEQ ID NOs 1 - 21, preferably the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence (i.e.
  • peptides that comprise the amino acid sequence of SEQ ID NO: 1 e.g. the amino acid sequence of VLAPRVLRA (SEQ ID NO:2), VLAPRVLRV (SEQ ID NO:3), VLAPRVLRI (SEQ ID NO:4) or VLAPRVLRL (SEQ ID NO:5)
  • SEQ ID NO: 1 e.g. the amino acid sequence of VLAPRVLRA (SEQ ID NO:2), VLAPRVLRV (SEQ ID NO:3), VLAPRVLRI (SEQ ID NO:4) or VLAPRVLRL (SEQ ID NO:5)
  • the additional amino acids may be amino acid sequences that are naturally located N-terminal, C-terminal or flanking the VLAPRVLRA sequence in RCN1.
  • the additional 6 to 21 amino acids may all be located N-terminal to the sequence of SEQ ID NO: 1 (e.g. the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence) and may be the natural sequence that is found N-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional 6 to 21 amino acids may be all be located C-terminal to the sequence of SEQ ID NO: 1 (e.g.
  • the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence may be the natural sequence that is found C-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may flank the sequence of SEQ ID NO: 1 (e.g. the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence) (i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence) and may be the natural sequence that flank the VLAPRVLRA sequence in RCN1.
  • peptides that comprise the amino acid sequence of SEQ ID NO: 1 may include any appropriate additional amino acid sequences.
  • the additional amino acids may be amino acid sequences that are naturally located N-terminal, C-terminal or flanking the VLAPRVLRA sequence in RCN1.
  • the additional 9 to 18 amino acids may all be located N-terminal to the sequence of SEQ ID NO: 1 (e.g.
  • the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence may be the natural sequence that is found N-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional 9 to 18 amino acids may be all be located C-terminal to the sequence of SEQ ID NO: 1 (e.g. the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence) and may be the natural sequence that is found C-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may flank the sequence of SEQ ID NO: 1 (e.g.
  • VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence
  • peptides that comprise the amino acid sequence of SEQ ID NO: 1 e.g. the amino acid sequence of VLAPRVLRA (SEQ ID NO:2), VLAPRVLRV (SEQ ID NO:3), VLAPRVLRI (SEQ ID NO:4) or VLAPRVLRL (SEQ ID NO:5)
  • SEQ ID NO: 1 e.g. the amino acid sequence of VLAPRVLRA (SEQ ID NO:2), VLAPRVLRV (SEQ ID NO:3), VLAPRVLRI (SEQ ID NO:4) or VLAPRVLRL (SEQ ID NO:5)
  • SEQ ID NO: 5 e.g. the amino acid sequence of VLAPRVLRA (SEQ ID NO:2), VLAPRVLRV (SEQ ID
  • the additional amino acids may be amino acid sequences that are naturally located N-terminal, C-terminal or flanking the VLAPRVLRA sequence in RCN1.
  • the additional 12 to 15 amino acids may all be located N-terminal to the sequence of SEQ ID NO: 1 (e.g. the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence) and may be the natural sequence that is found N-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional 12 to 15 amino acids may be all be located C-terminal to the sequence of SEQ ID NO: 1 (e.g.
  • the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence may be the natural sequence that is found C-terminal to the VLAPRVLRA sequence in RCN1.
  • the additional amino acids may flank the sequence of SEQ ID NO: 1 (e.g. the VLAPRVLRA, VLAPRVLRV, VLAPRVLRI or VLAPRVLRL sequence) (i.e. such that there are additional amino acid(s) N-terminal and C-terminal of the sequence) and may be the natural sequence that flank the VLAPRVLRA sequence in RCN1.
  • Suitable natural sequences from RCN1 are provided in the Examples section below.
  • the peptide may comprise N-terminal additional amino acids, for example it may comprise the sequence: GRGRRLGLALGLLLALVLAPRVLRA (SEQ ID NO:22). This is an example of a 25mer with N-terminal additional amino acids, although other lengths may also be acceptable e.g.18 mer, 21 mer, 24 mer, 27 mer etc.
  • the peptide may comprise additional amino acid(s) N-terminal and C-terminal of the peptide, for example it may comprise the sequence: ALGLLLALVLAPRVLRAKPTVRKER (SEQ ID NO:23).
  • peptide may comprise C-terminal additional amino acids, for example it may comprise the sequence VLAPRVLRAKPTVRKERVVRPDSEL (SEQ ID NO:24).
  • SEQ ID NO:24 the sequence VLAPRVLRAKPTVRKERVVRPDSEL
  • the peptide may comprise N-terminal additional amino acids, for example it may comprise the sequence: GRGRRLGLALGLLLALVLAPRVLRV (SEQ ID NO:25). This is an example of a 25mer with N-terminal additional amino acids, although other lengths may also be acceptable e.g.18 mer, 21 mer, 24 mer, 27 mer etc.
  • the peptide may comprise additional amino acid(s) N-terminal and C-terminal of the peptide, for example it may comprise the sequence: ALGLLLALVLAPRVLRVKPTVRKER (SEQ ID NO:26).
  • the peptide may comprise C-terminal additional amino acids, for example it may comprise the sequence VLAPRVLRVKPTVRKERVVRPDSEL (SEQ ID NO:27).
  • the peptide may comprise the amino acid sequence of SEQ ID NO: 22.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 22. In an example, the peptide may comprise the amino acid sequence of SEQ ID NO: 23. In another example, the peptide may consist of the amino acid sequence of SEQ ID NO: 23. In another example, the peptide may comprise the amino acid sequence of SEQ ID NO: 24. In yet another example, the peptide may consist of the amino acid sequence of SEQ ID NO: 24. In a further example, the peptide may comprise the amino acid sequence of SEQ ID NO: 25. In another example, the peptide may consist of the amino acid sequence of SEQ ID NO: 25. In a further example, the peptide may comprise the amino acid sequence of SEQ ID NO: 26.
  • the peptide may consist of the amino acid sequence of SEQ ID NO: 26. In a further example, the peptide may comprise the amino acid sequence of SEQ ID NO: 27. In another example, the peptide may consist of the amino acid sequence of SEQ ID NO: 27.
  • Alternative appropriate natural sequences from RCN1 may also be identified by a person of skilled in the art.
  • the additional amino acids may be amino acid sequences that are not naturally located N-terminal, C-terminal or flanking the VLAPRV
  • flanking sequences Both natural and non-natural flanking sequences have been shown to be useful in peptide vaccines and therefore either may be used in the peptides described herein.
  • the SIINFEKL epitope of the OVA antigen has been successfully used as a peptide vaccine when flanked by its natural sequence (Bijker et al., (2007), a non-natural C-terminal flanking sequence (Varypataki et al., (2015) or without an N-terminal flanking sequence but with a glycine linker attached to a helper epitope at the C-terminal position, so completely outside the context of its own natural flanking sequences (Masuko et al., (2015)).
  • N-terminal and/or C-terminal non-natural additional amino acid sequences may be acceptable in a peptide vaccine format.
  • the peptide may be a “natural peptide” i.e. a peptide composed of natural amino acids. Such peptides are composed of conventional amino acids defined by the genetic code, linked to each other by a normal peptide bond. Natural peptides may, for example, be produced by a cell (via protein expression, e.g.
  • the peptide may be a “synthetic peptide”.
  • a synthetic peptide may comprise a mix of natural amino acids and amino acids other than conventional amino acids defined by the genetic code (“synthetic amino acids”). Alternatively, it may be composed of synthetic amino acids only. Examples of synthetic amino acids are well known in the literature. Natural peptides and synthetic peptides may be modified. In other words, the peptide may comprise amino acids modified by natural processes, such as post-translational maturation processes or by chemical processes, which are well known to a person skilled in the art. Such modifications are fully detailed in the literature.
  • Non-limiting examples of peptide modifications include acetylation, acylation, ADP- ribosylation, amidation, covalent fixation of a nucleotide or of a nucleotide derivative, covalent fixation of a lipid or of a lipidic derivative, the covalent fixation of a phosphatidylinositol, covalent or non-covalent cross-linking, cyclization, disulfide bond formation, demethylation, glycosylation including pegylation, hydroxylation, iodization, methylation, myristoylation, oxidation, proteolytic processes, phosphorylation, prenylation, racemization, seneloylation, sulfatation, amino acid addition such as arginylation or ubiquitination.
  • peptide may include for example lipopeptides, lipoproteins, glycopeptides, glycoproteins and the like.
  • the peptide can be branched following ubiquitination or be cyclic with or without branching. This type of modification can be the result of natural or synthetic post-translational processes that are well known to a person skilled in the art.
  • the peptides described herein may be conjugated directly, or via a linker, to a therapeutic moiety, a polymer, a polypeptide, a ligand and/or any other moiety e.g. a detectable moiety.
  • peptide conjugates Such peptides are referred to herein as “peptide conjugates”.
  • the peptides described herein may be conjugated to an immune stimulatory compound.
  • the peptides described herein may be conjugated to a non-specific immune stimulatory compound.
  • the peptide conjugate may comprise a peptide covalently attached to an immune stimulatory compound.
  • the peptide conjugate may comprise a peptide covalently attached to a non-specific immune stimulatory compound.
  • the peptides described herein may alternatively be comprised in a composition or vaccine comprising an immune stimulatory compound.
  • composition or vaccine comprising any one of the peptides according to the invention (or a nucliec acid molecule encoding such a peptide) and an immune stimulatory compound.
  • composition or vaccine comprising any one of the peptides according to the invention or a nucleic acid sequence encoding the same, and an immune stimulatory compound.
  • Immune stimulatory compounds are compounds that stimulate the immune system by inducing activation or increasing activity of any of its components. Immune stimulatory compounds comprise specific immune stimulatory compounds and non-specific immune stimulatory compounds. Specific immune stimulatory compounds provide antigenic specificity in immune response, for example any antigen.
  • Non-specific immune stimulatory compounds act irrespective of antigenic specificity to augment immune response of other antigen or stimulate components of the immune system without antigenic specificity, for example adjuvants.
  • immune stimulatory compounds may activate receptors of the innate immune system.
  • immune stimulatory compounds may activate pattern recognition receptors.
  • the immune stimulatory compound may comprise a damage-associated molecular pattern (DAMP).
  • the immune stimulatory compound may comprise a pathogen-associated molecular pattern (PAMP).
  • the immune stimulatory compound may comprise a ligand for a nucleotide- binding oligomerization domain-like receptor (NLR), such as NOD1 or NOD2.
  • NLR nucleotide- binding oligomerization domain-like receptor
  • the peptide conjugate may comprise a peptide covalently attached to a NLR ligand.
  • the immune stimulatory compound may comprise a ligand for a RIG-I-like receptor (RLR), such as RIG-I, MDA5, or LGP2.
  • RLR RIG-I-like receptor
  • the peptides described herein may be conjugated to a RLR ligand.
  • the peptide conjugate may comprise a peptide covalently attached to a RLR ligand.
  • the immune stimulatory compound may comprise a ligand for a C-type lectin receptor (CLR), such as Dectin-1 or Dectin-2.
  • CLR C-type lectin receptor
  • the peptide conjugate may comprise a peptide covalently attached to a CLR ligand.
  • the immune stimulatory compound may comprise a ligand for an absent in melanoma-2-like receptor (ALR), such as any ALR.
  • ALR melanoma-2-like receptor
  • the peptides described herein may be conjugated to an ALR ligand.
  • the peptide conjugate may comprise a peptide covalently attached to an ALR ligand.
  • the immune stimulatory compound may comprise a Toll-Like Receptor (TLR) ligand.
  • TLR Toll-Like Receptor
  • the immune stimulatory compound may be a TLR ligand.
  • the peptides described herein may be conjugated to a TLR ligand.
  • the peptide conjugate may comprise a peptide covalently attached to a TLR ligand.
  • TLR ligands may also be referred to as TLR agonists.
  • a “TLR agonist” is an agonist of a TLR, i.e. it binds to a TLR and activates the TLR, in particular to produce a biological response.
  • Peptide conjugates comprising TLR agonists covalently bound to peptides, in particular TLR agonists that are covalently bound to synthetic peptides, are well known in the art.
  • Zom (Zom et al., (2016)) described a conjugate of the TLR2-ligand Pam3CSK4 to synthetic long peptides (SLPs). Furthermore, Zom (Zom et al., (2016)) described the conjugation of human papillomavirus type 16 (HPV16)-encoded synthetic long peptides to a Pam3CSK4-based TLR2 agonist.
  • Toll-like receptors (TLRs) are transmembrane proteins that are characterized by extracellular, transmembrane, and cytosolic domains. The extracellular domains containing leucine-rich repeats (LRRs) with horseshoe-like shapes are involved in recognition of common molecular patterns derived from diverse microbes.
  • TLRs1-10 Compounds capable of activating TLR receptors and modifications and derivatives thereof are well documented in the art.
  • TLR1 may be activated by bacterial lipoproteins and acetylated forms thereof
  • TLR2 may in addition be activated by Gram positive bacterial glycolipids, LPS, LPA, LTA, fimbriae, outer membrane proteins, heat shock proteins from bacteria or from the host, and Mycobacterial lipoarabinomannans.
  • TLR3 may be activated by dsRNA, in particular of viral origin, or by the chemical compound poly(LC).
  • TLR4 may be activated by Gram negative LPS, LTA, Heat shock proteins from the host or from bacterial origin, viral coat or envelope proteins, taxol or derivatives thereof, hyaluronan containing oligosaccharides and fibronectins.
  • TLR5 may be activated with bacterial flagellae or flagellin.
  • TLR6 may be activated by mycobacterial lipoproteins and group B streptococcus heat labile soluble factor (GBS-F) or staphylococcus modulins.
  • GSS-F group B streptococcus heat labile soluble factor
  • TLR7 may be activated by imidazoquinolines.
  • TLR9 may be activated by unmethylated CpG DNA or chromatin—lgG complexes.
  • TLRs are expressed either on the cell surface (TLR1, 2, 4, 5, 6, and 10) or on membranes of intracellular organelles, such as endosomes (TLR3, 4, 7, 8, and 9).
  • the natural ligands for the endosomal receptors are nucleic acid-based molecules (except for TLR4).
  • the cell surface- expressed TLR1, 2, 4, 5, 6, and 10 recognize molecular patterns of extracellular microbes (Monie et al., (2009)).
  • TLRs are expressed on several cell types but virtually all TLRs are expressed on DCs allowing these specialized cells to sense all possible pathogens and danger signals.
  • TLR2, 4, and 5 are constitutively expressed at the surface of DCs.
  • TLR2 can detect a wide variety of ligands derived from bacteria, viruses, parasites, and fungi.
  • the ligand specificity is often determined by the interaction of TLR2 with other TLRs, such as TLR1, 6, or 10, or non-TLR molecules, such as dectin-1, CD14, or CD36.
  • TLR1 TLR1, 6, or 10
  • non-TLR molecules such as dectin-1, CD14, or CD36.
  • TLR2 can identify triacyl lipoproteins or lipopeptides from (myco)bacterial origin, such as Pam3CSK4 and peptidoglycan (PGA) (Gay et al., (2007); Spohn et al., (2004)).
  • PGA peptidoglycan
  • TLR2 and 6 Heterodimerization of TLR2 and 6 enables the detection of diacyl lipopeptides and zymosan.
  • Lipopolysaccharide (LPS) and its derivatives are ligands for TLR4 and flagellin for TLR5 (Bryant et al., (2010)).
  • TLR2 interacts with a broad and structurally diverse range of ligands, including molecules expressed by microbes and fungi. Multiple TLR2 agonists have been identified, including natural and synthetic lipopeptides (e.g. Mycoplasma fermentas macrophage-activating lipopeptide (MALP-2)), peptidoglycans (PG such as those from S.
  • MALP-2 Mycoplasma fermentas macrophage-activating lipopeptide
  • PG peptidoglycans
  • lipopolysaccharides from various bacterial strains (LPS), polysaccharides (e.g. zymosan), glycosylphosphatidyl- inositol-anchored structures from gram positive bacteria (e.g. lipoteichoic acid (LTA) and lipo- arabinomannan from mycobacteria and lipomannas from M. tuberculosis).
  • LTA lipoteichoic acid
  • lipo- arabinomannan from mycobacteria and lipomannas from M. tuberculosis
  • Certain viral determinants may also trigger via TLR2 (Barbalat et al., (2009)).
  • Bacterial lipopeptides are structural components of cell walls. They consist of an acylated s-glycerylcysteine moiety to which a peptide can be conjugated via the cysteine residue.
  • TLR2 agonists which are bacterial lipopeptides, include MALP-2 and it's synthetic analogue di-palmitoyl-S-glyceryl cysteine (Pam2Cys) or tri-palmitoyl-S-glyceryl cysteine (Pam3Cys).
  • a diversity of ligands interact with TLR4, including Monophosphoryl Lipid A from Salmonella minnesota R595 (MPLA), lipopolysaccharides (LPS), mannans (Candida albicans), glycoinositolphospholipids (Trypanosoma), viral envelope proteins (RSV and MMTV) and endogenous antigens including fibrinogen and heat-shock proteins.
  • TLR4 Such agonists of TLR4 are for example described in Akira (Akira et al., 2006) or in Kumar (Kumar et al., 2009).
  • LPS which is found in the outer membrane of gram negative bacteria, is the most widely studied of the TLR4 ligands. Suitable LPS-derived TLR4 agonist peptides are described for example in WO 2013/120073 (A1).
  • TLR5 is triggered by a region of the flagellin molecule expressed by nearly all motile bacteria.
  • flagellin, or peptides or proteins derived from flagellin and/or variants or fragments of flagellin are also suitable as TLR peptide agonists comprised by the peptide conjugate of the present invention.
  • Non-limiting examples of TLR peptide agonists thus include the TLR2 lipopeptide agonists MALP- 2, Pam2Cys and Pam3Cys or modifications thereof, different forms of the TLR4 agonist LPS, e.g. N. meningitidis wild-type L3-LPS and mutant penta-acylated LpxL1-LPS, and the TLR5 agonist flagellin.
  • TLR2 peptide agonist is annexin II or an immunomodulatory fragment thereof, which is described in detail in WO 2012/048190 A1 and U.S. patent application Ser. No.13/033,1546.
  • high-mobility group box 1 protein HMGB1
  • peptide fragments thereof are assumed to be TLR4 agonists.
  • TLR4 agonists are for example disclosed in US 2011/0236406 A1.
  • the peptide conjugate according to the present invention may comprise at least one TLR agonist, preferably the peptide conjugate may comprise more than one TLR agonist, in particular 2, 3, 4, 5, 6, 7, 8, 9, 10 or more TLR agonists.
  • the at least one TLR, NLR, RLR, CLR, or ALR agonist comprised within the peptide conjugate according to the present invention may be the same or different.
  • the various TLR, NLR, RLR, CLR, and/or ALR agonists comprised within the peptide conjugate of the present invention are different from each other. It is understood that a number of different TLR agonists activating the same or different TLR receptors may be advantageously comprised within a single peptide conjugate according to the present invention.
  • the immune stimulatory compound may be an adjuvant.
  • the adjuvant may be combined with any one of the TLR, NLR, RLR, CLR, or ALR agonists as disclosed herein.
  • the adjuvant may be selected from the group of mineral salts, emulsions, and microparticles.
  • the mineral salt may be an aluminium salt.
  • the emulsion may be a water-in-oil emulsion or an oil-in-water emulsion.
  • suitable emulsions are Complete Freund’s adjuvant, Incomplete Freund’s adjuvant, MF59, AS03, and ISA51.
  • Montanide ISA51 (Seppic, France) is a water-in-oil emulsion composed of a mineral oil and a surfactant from the mannide monooleate family, and is a preferred adjuvant for the vaccines herein.
  • microparticles are virus-like particles and virosomes. Virus-like particles are non-infectious nanoparticles without genetic information having an outer region comprised of immunogenic epitopes.
  • the pharmaceutical composition described herein may be administered to a human subject (e.g. as a peptide vaccine) in order to treat or prevent a cancer or viral infection associated with impaired HLA class I antigen presentation.
  • the pharmaceutical composition described herein e.g. vaccine
  • the pharmaceutical composition e.g. vaccine
  • the pharmaceutical composition may be administered as a peptide vaccine for treating or preventing a cancer or viral infection associated with impaired HLA class I antigen presentation.
  • the pharmaceutical composition may be administered to induce or enhance activation of T cells specific for cancerous or virally infected cells.
  • Nucleic acid sequences and vectors encoding the peptides described herein may be administered as a nucleic acid vaccine for treating or preventing a cancer or viral infection associated with impaired HLA class I antigen presentation.
  • the isolated nucleic acid sequences and vectors may be administered to induce or enhance activation of T cells specific for cancerous or virally infected cells.
  • Peptide vaccines and nucleic acid vaccines are examples of vaccines.
  • Cross-presentation of long peptides (e.g. SLPs as described herein) by dendritic cells involves endocytosis, cytosolic cleavage of the SLP into short peptides by the proteasome, transport over the ER membrane by TAP and loading onto MHC-I molecules (Rosalia et al.2013).
  • the peptides described herein (and corresponding nucleic acid sequences or vectors encoding the same) may be particularly useful as an immunotherapy for human subjects that are positive for HLA-A*02.
  • HLA-A*02 is a globally common human leukocyte antigen serotype within the HLA-A serotype group.
  • HLA-A*02 group Several subtypes exist within the HLA-A*02 group, including HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, HLA-A*02:05, HLA-A*02:06, HLA-A*02:09, HLA-A*02:11, HLA- A*02:12, HLA-A*02:16, HLA-A*02:19, HLA-A*02:50.
  • HLA- A*02:01 The data presented herein focuses on HLA- A*02:01, however, as would be clear to a person of skill in the art, other subtypes within the HLA- A*02 group (including but not limited to those listed herein) may also bind to any one of the amino acid sequences of SEQ ID NOs: 1 - 21 (see for example Ressing et al., 1999, particularly Tables 3 and 2). All HLA-A*02 subtypes are therefore encompassed herein, although HLA-A*02:01 is preferred (see Table 1 below).
  • HLA-A*02 subtypes HLA-A*02:01, HLA-A*02:02, HLA- A*02:03, HLA-A*02:04, and HLA-A*02:09 are also preferred for applications of the invention herein.
  • These HLA-A2 variants were shown to display comparable binding characteristics in accordance with the A2 supertype (Ressing et al., 1999 and M. F. Del Guercio et al., J. Immunol. 1995.154: 685-693).
  • Predicted binding affinity of VLAPRVLRA, VLAPRVLRV, VLAPRVLRL, VLAPRVLRI, and VLAPRVLRM is determined using the NetMHCcons1.1 algorithm (https://services.healthtech.dtu.dk/service.php?NetMHCcons-1.1).
  • the prediction value under affinity measures the predicted binding affinity.
  • the rank (in %) is determined by comparing the predicted binding affinity to a set of 200.000 random natural peptides. Strong and weak binding peptides are indicated. Recognition indicates the concentration of the peptide for half maximal interferon-gamma production of VLAPRVLRA- specific T cells stimulated with the different variant peptides.
  • nucleic acid sequences Isolated nucleic acid sequences that encode peptides of the invention are described herein, as well as nucleic acid sequences encoding binding agents are described herein.
  • nucleic acid sequence polynucleotide”, “nucleic acid” and “nucleic acid molecule” are used interchangeably to refer to an oligonucleotide sequence or polynucleotide sequence.
  • the term nucleic acid sequence may therefore be replaced by the term nucleic acid herein.
  • the nucleotide sequence may be of genomic, synthetic or recombinant origin, and may be double-stranded or single-stranded (representing the sense or antisense strand).
  • nucleotide sequence includes genomic DNA, cDNA, synthetic DNA, and RNA (e.g. mRNA) and analogs of the DNA or RNA generated, e.g., by the use of nucleotide analogs.
  • the nucleotide sequence lacks introns. In other words, it is an intronless nucleic acid sequence.
  • the nucleotide sequence may be a DNA sequence that does not comprise intron sequences.
  • an isolated nucleic acid sequence/composition is not a native nucleotide sequence/composition, wherein "native nucleotide sequence/composition” means an entire nucleotide sequence that is in its native environment and when operatively linked to an entire promoter with which it is naturally associated, which promoter is also in its native environment.
  • Such a nucleic acid could be part of a vector and/or such nucleic acid or polypeptide could be part of a composition (e.g., a cell lysate), and still be isolated in that such vector or composition is not part of the natural environment for the nucleic acid or polypeptide.
  • the term "gene” means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region ("leader and trailer") as well as intervening sequences (introns) between individual coding segments (exons).
  • the nucleic acid sequences of the invention may be a non-naturally occurring nucleic acid sequence (e.g. it may be that the entire sequence does not occur in its entirety in nature).
  • the nucleic acid sequence of the invention may be operably linked to a promoter, wherein the promoter is not naturally associated with equivalent human nucleic acid sequences in nature (e.g. human TCR sequences or fragments thereof); i.e. it is not the entire promoter that is naturally associated with the nucleic acid in its natural environment.
  • the invention provides a vector that comprises a nucleic acid sequence described herein (e.g. a nucleic acid sequence that encodes a peptide comprising an amino acid sequence of SEQ ID NO:1).
  • a vector system is also provided which includes a nucleic acid composition described herein.
  • the vector system may have one or more vectors.
  • the binding protein components that are encoded by the nucleic acid composition may be encoded by one or more nucleic acid sequences in the nucleic acid composition.
  • the nucleic acid sequence may be present within a single vector (and thus the vector system described herein may comprise of one vector only).
  • the binding protein components are encoded by two or more nucleic acid sequences (wherein the plurality of nucleic acid sequences, together, encode all of the components of the binding protein) these two or more nucleic acid sequences may be present within one vector (e.g. in different open reading frames of the vector), or may be distributed over two or more vectors.
  • the vector system will comprise a plurality of distinct vectors (i.e. vectors with different nucleotide sequences).
  • a vector system comprising a nucleic acid composition described herein.
  • the vector may be a plasmid, a cosmid, or a viral vector, such as a retroviral vector or a lentiviral vector.
  • Adenovirus, adeno- associated virus, vaccinia virus, canary poxvirus, herpes virus, minicircle vectors and naked (synthetic) DNA/RNA may also be used (for details on minicircle vectors, see for example non- viral Sleeping Beauty transposition from minicircle vectors as published by Monjezi et al., Leukemia 2016).
  • the vector is a plasmid, a viral vector, or a cosmid, optionally wherein the vector is selected from the group consisting of a retrovirus, lentivirus, adeno-associated virus, adenovirus, vaccinia virus, canary poxvirus, herpes virus, minicircle vector and synthetic DNA or RNA.
  • the term “vector” refers to a nucleic acid sequence capable of transporting another nucleic acid sequence to which it has been operably linked.
  • the vector can be capable of autonomous replication or it can integrate into a host DNA.
  • the vector may include restriction enzyme sites for insertion of recombinant DNA and may include one or more selectable markers or suicide genes.
  • the vector can be a nucleic acid sequence in the form of a plasmid, a bacteriophage or a cosmid.
  • the vector is suitable for expression in a cell (i.e. the vector is an “expression vector”).
  • the vector is suitable for expression in a human antigen presenting cell.
  • the vector is suitable for expression in a human T cell such as a CD8 + T cell or CD4 + T cell, or stem cell, iPS cell, or NK cell.
  • the vector is a viral vector, such as a retroviral vector, a lentiviral vector or an adeno-associated vector.
  • the vector is selected from the group consisting of an adenovirus, vaccinia virus, canary poxvirus, herpes virus, minicircle vector and synthetic DNA or synthetic RNA.
  • the (expression) vector is capable of propagation in a host cell and is stably transmitted to future generations. Suitable vectors and expression vectors are well known in the art.
  • the vector may comprise regulatory sequences.
  • the vector comprises the nucleic acid sequence of interest operably linked to a promoter.
  • the promoter may be one that is not naturally found in the host cell (e.g. it may be an exogenous promoter).
  • “Operably linked” refers to a single or a combination of the below-described control elements together with a coding sequence in a functional relationship with one another, for example, in a linked relationship so as to direct expression of the coding sequence.
  • a person of skill in the art will be well aware of the molecular techniques available for the preparation of (expression) vectors and how the (expression) vectors may be transduced or transfected into an appropriate host cell (thereby generating a modified cell as described herein).
  • the (expression) vector of the present invention can be introduced into cells by conventional techniques such as transformation, transfection or transduction.
  • Transformation refer generally to techniques for introducing foreign (exogenous) nucleic acid sequences into a host cell, and therefore encompass methods such as electroporation, microinjection, gene gun delivery, transduction with retroviral, lentiviral or adeno-associated vectors, lipofection, superfection etc.
  • the specific method used typically depends on both the type of vector and the cell.
  • the host cell is contacted with the vector (e.g. viral vector) in vitro, ex vivo, and in some embodiments, the host cell is contacted with the vector (e.g. viral vector) in vivo.
  • the term "host cell” includes any cell into which the nucleic acid sequences or vectors described herein may be introduced (e.g. transduced). Once a nucleic acid molecule or vector has been introduced into the cell, it may be referred to as a “modified cell” herein. Once the nucleic acid molecule or vector is introduced into the host cell, the resultant modified cell should be capable of expressing the encoded polypeptide (and e.g.
  • the nucleic acid composition or vector system may be introduced into the cell using any conventional method known in the art.
  • the nucleic acid composition or vector system may be introduced using CRISPR technology. Insertion of the nucleic acid sequences at the endogenous TCR locus by engineering with CRISPR/Cas9 and homologous directed repair (HDR) or non-homologous end joining (NHEJ) is therefore encompassed. Other conventional methods such as transfection, transduction or transformation of the cell may also be used.
  • modified cell refers to a genetically altered (e.g.
  • the modified cell includes at least one exogenous nucleic acid sequence (i.e. a nucleic acid sequence that is not naturally found in the host cell).
  • the term refers to the particular subject cell and also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a modified cell comprises a nucleic acid composition or a vector system provided herein.
  • a modified cell comprises a nucleic acid comprising a nucleic acid sequence encoding a peptide provided herein.
  • the host cell may be a bacterial cell, it is typically a eukaryotic cell, and particularly a human cell which can overexpress the antigen for uptake by antigen presenting cells (APCs), more particularly an antigen presenting cell, such as dendritic cells (DCs), B cells, monocytes, macrophages.
  • APCs antigen presenting cells
  • DCs dendritic cells
  • B cells monocytes
  • macrophages presenting cells
  • the host cell (and thus the modified cell) may be an autologous cell, which refers to a cell derived from the same individual to which it is later administered. In other words, the host cell (and thus the modified cell) may be a cell from a subject to be treated.
  • the host cell (and thus the modified cell) may be isolated from a blood sample e.g. by leukaphoresis.
  • the modified cell is typically a human cell.
  • the host cell (and thus the modified cell) may be any cell that is able to confer anti-tumour immunity after TCR gene transfer.
  • Non limiting examples of appropriate cells include autologous or allogeneic CD8 T cells, CD4 T cells, Natural Killer (NK) cells, NKT cells, gamma-delta T cells, inducible pluripotent stem cells (iPSCs), hematopoietic stem cells or other progenitor cells and any other autologous or allogeneic cell or cell line (NK-92 for example or T cell lines) that is able to confer anti-tumor immunity after TCR gene transfer.
  • NK Natural Killer
  • iPSCs inducible pluripotent stem cells
  • hematopoietic stem cells or other progenitor cells and any other autologous or allogeneic cell or cell line (NK-92 for example or T cell lines) that is able to confer anti-tumor immunity after TCR gene transfer.
  • the modified cell is selected from the group consisting of a CD8 T cell, a CD4 T cell, an NK cell, an NK-T cell, a gamma-delta T cell, an innate lymphoid cell (ILC), a hematopoietic stem cell, an inducible pluripotent stem cell, a progenitor cell, a T cell line and a NK-92 cell line.
  • the host cell (and thus the modified cell) that is to be administered to the subject can either be autologous or allogeneic.
  • the modified cell is capable of expressing the polypeptide encoded by the nucleic acid sequence or vector described herein such that the modified cell provides an immunotherapy that specifically targets cancerous cells or virally infected cells associated with impaired HLA class I antigen presentation and this can be used to treat or prevent cancer or viral infections associated with impaired HLA class I antigen presentation. More details on this use are given below.
  • Methods for preparing peptides As described above, the peptide according to the present invention may be a natural peptide or a synthetic peptide. In another aspect, the peptide of the present invention may be modified. Methods of preparing a peptide of the invention are also provided herein. In one aspect, the methods of preparing a peptide of the invention provided herein may be natural methods.
  • the method of preparing a peptide of the invention may be synthetic methods.
  • the method of preparing a peptide of the invention may comprise natural and synthetic methods.
  • the methods of preparing a peptide of the invention provided herein may be natural methods. Such methods comprise cultivating a modified cell that has been transformed, transfected or transduced with a nucleic acid (e.g. vector) encoding the peptide of interest in a culture medium and separating the peptide from the culture medium or from the modified cell lysate after cell lysis.
  • the modified cell is used to express the peptide of interest. Examples of such cells include, but are not limited to, bacterial cells, e.g. E.
  • the cells are mammalian, e.g., human, CHO, HEK293T, PER.C6, NS0, myeloma or hybridoma cells. Dendritic cells and dendritic cell lines are particularly preferred.
  • the nucleic acid encoding the peptide of interest is present within a vector, such as an expression vector.
  • an appropriate secretion signal can be integrated in the vector, so that the peptide encoded by the nucleic acid will be directed, for example towards the lumen of the endoplasmic reticulum, towards the periplasmic space, on the membrane or towards the extracellular environment.
  • the choice of appropriate secretion signal may facilitate subsequent protein purification. Selection of appropriate secretion signals are well within the capabilities of a person with average skill in the art. Typically, the choice of a culture medium depends in particular on the choice of the cell type and/or the cell line that is used to express the peptide of interest. A person of skill in the art is well aware of suitable culture media, which are appropriate for a selected cell type and/or cell line.
  • the cells are cultivated in the appropriate culture medium for a period that is sufficient to induce expression of the encoded peptide. Suitable time periods and conditions for culturing cells are well known in the art and depend on the specific cell type and/or cell line that is used.
  • the peptide may be purified using standard methods. For example, commercially available kits and/or reagents for protein extraction may be used, for example BugBusterTM from Novagen. Alternative standard methods such as affinity chromatography, ion-exchange chromatography, hydrophobic interaction chromatography, and immunoaffinity methods may also be used.
  • the peptides of the invention may be prepared by synthetic methods. Such methods are well described in the literature.
  • Non-limiting examples include liquid phase peptide synthesis methods or solid peptide synthesis methods, e.g. solid peptide synthesis methods according to Merrifield, t-Boc solid-phase peptide synthesis, Fmoc solid-phase peptide synthesis, BOP (Benzotriazole-1-yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate) based solid- phase peptide synthesis, etc.
  • Peptide-loaded cells A cell loaded with a peptide described herein is also provided. These cells may advantageously be used in the therapeutic methods described below.
  • a cell “loaded” with peptide refers to a cell wherein the peptide is in association with an MHC (major histocompatibility complex) on the surface of the cell.
  • MHC major histocompatibility complex
  • cells loaded with peptide do not express the peptide themselves, but present exogenous peptides in the context of MHC.
  • Cells may be pulsed with exogenous peptide in order to “load” them with peptide.
  • Cells loaded with peptides may therefore also be referred to as cells comprising the peptide of interest (e.g. exogenous peptide), wherein the peptide of interest is part of an MHC complex on the surface of the cell.
  • such cells comprise extracellular (or cell surface) MHC complexed with the peptide of interest.
  • antigen presentation is the expression of antigen molecules on the surface of a macrophage or other antigen-presenting cell in association with MHC class II molecules when the antigen is being presented to a CD4+ helper T cell or in association with MHC class I molecules when presentation is to CD8+ cytotoxic T cells.
  • the cell may be a modified cell transformed, transfected or transduced with a nucleic acid comprising a nucleic acid sequence encoding a peptide described herein.
  • Such a modified cell expresses the peptide and is therefore also capable of loading the same cell or other cells with the peptide.
  • Cells loaded with the peptide as defined herein may be cells from a subject to be treated. In particular, they may be cells that have been isolated from a subject to be treated. Alternatively, cell lines, e.g. antigen presenting cell lines, may also be used.
  • the cell loaded with the peptide as defined herein is an antigen-presenting cell (APC).
  • the antigen presenting cell is selected from the group consisting of a dendritic cell (DC), a macrophage, a monocyte, a B-cell and a synthetic form of antigen presenting cell.
  • Dendritic cells in particular dendritic cells (conventional and/or plasmacytoid) isolated from a subject to be treated, are most preferred.
  • Methods to isolate antigen-presenting cells, in particular dendritic cells, from a subject are known to the skilled person. They include harvesting monocytes or hematopoietic stem cells from bone marrow, cord blood, or peripheral blood. They also include the use of embryonic stem (ES) cells and induced pluripotent stem cells (iPS).
  • ES embryonic stem
  • iPS induced pluripotent stem cells
  • Antigen presenting cells in particular dendritic cells or their precursors, can be enriched by methods including elutriation and magnetic bead based separation, which may involve enrichment for CD14+ precursor cells.
  • dendritic cells Methods to load the complex as defined herein into the cells, preferably into the above-mentioned antigen presenting cells, more preferably into dendritic cells, and further to prepare such cells before administration to a subject are known to one skilled in the art.
  • preparation of dendritic cells can include their culture or differentiation using cytokines that may include for example GM-CSF and IL-4.
  • Dendritic cell lines may also be employed.
  • Loading of the peptide into the cells, preferably into APC, more preferably into the dendritic cells can involve co-incubation of the peptide with the cells in culture. Further culture of the cells, e.g.
  • compositions comprising a) a peptide, b) a nucleic acid and/or a vector, c) a complex or a binding agent or d) a cell described herein, and a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • any one of a), b) or c) may be present in the pharmaceutical composition by virtue of them being encoded or expressed (as appropriate) by a cell that is present within the pharmaceutical composition.
  • any one of b) or c) may be encoded by a cell that is combined with a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier to generate the pharmaceutical composition; or any of a) or b) may be expressed by a cell that is combined with a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier to generate the pharmaceutical composition. More details on this are provided below.
  • a particularly suitable composition may be selected based on the HLA serotype of the human subject, as described in detail elsewhere herein.
  • a nucleic acid, vector, complex, cell, binding agent, and/or peptide as described herein may therefore be provided as part of a pharmaceutical composition.
  • such compositions may be administered to a human subject in order to treat or prevent a cancer or viral infection associated with impaired HLA class I antigen presentation (e.g. by inducing or enhancing a specific immune response to such cancerous or virally infected cells).
  • pharmaceutical composition and “composition” are used interchangeably herein, unless the context specifically requires otherwise.
  • a pharmaceutical composition may comprise a nucleic acid sequence, vector, complex, binding agent, cell, and/or peptide described herein along with a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
  • the nucleic acid sequence, vector, complex, binding agent and/or peptide may be present in the pharmaceutical composition as part of a cell.
  • the nucleic acid sequence or vector may be incorporated into a cell; or the binding agent, complex or peptide may be expressed by a cell.
  • the cell may be any suitable cell, for example a bacterial cell, or a eukaryotic cell such as a mammalian cell e.g.
  • a pharmaceutical composition comprising a nucleic acid sequence, vector, complex, binding agent and/or peptide described herein therefore encompasses a pharmaceutical composition comprising a cell (e.g. a bacterial cell, DC, etc) that encodes the nucleic acid sequence or vector, or is capable of expressing the peptide, complex or binding agent.
  • a cell e.g. a bacterial cell, DC, etc
  • the cell e.g. bacterial cell, DC, etc
  • the cell may be a cell that has been modified to introduce into the cell the appropriate nucleic acid sequence/vector (e.g.
  • the modified cell may be combined with a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier to generate a pharmaceutical composition of the invention.
  • the cell may modified ex vivo.
  • it may be an autologous cell that has been derived from the subject that is to be treated with the pharmaceutical composition described herein (e.g. for treating or preventing a cancer or viral infection associated with impaired HLA class I antigen presentation).
  • the cells may be modified ex vivo to introduce e.g.
  • compositions may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, supplementary immune potentiating agents such as adjuvants and cytokines and optionally other therapeutic agents or compounds.
  • pharmaceutically acceptable refers to a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the selected nucleic acid sequence, vector, cell, binding agent or peptide without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • Excipients are natural or synthetic substances formulated alongside an active ingredient (e.g. a nucleic acid sequence, a nucleic acid composition, vector or vector system, modified cell, or isolated nucleic acid as provided herein), included for the purpose of bulking-up the formulation or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption or solubility.
  • Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerned such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation over the expected shelf life.
  • Pharmaceutically acceptable excipients are well known in the art. A suitable excipient is therefore easily identifiable by one of ordinary skill in the art.
  • suitable pharmaceutically acceptable excipients include saline, water, aqueous dextrose, glycerol, ethanol, and the like.
  • Adjuvants are pharmacological and/or immunological agents that modify the effect of other agents in a formulation.
  • Pharmaceutically acceptable adjuvants are well known in the art.
  • a suitable adjuvant is therefore easily identifiable by one of ordinary skill in the art.
  • an excipient does not include water.
  • a pharmaceutical composition may comprise an immune stimulatory compound as disclosed elsewhere herein.
  • Diluents are diluting agents.
  • Pharmaceutically acceptable diluents are well known in the art.
  • a suitable diluent is therefore easily identifiable by one of ordinary skill in the art.
  • Carriers are non-toxic to recipients at the dosages and concentrations employed and are compatible with other ingredients of the formulation.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • Pharmaceutically acceptable carriers are well known in the art. A suitable carrier is therefore easily identifiable by one of ordinary skill in the art.
  • compositions described herein may be administered to a subject as a monotherapy or as part of a combination therapy.
  • combinations of the vaccines described herein with an immune checkpoint inhibitor or other immunomodulatory compounds may also be particularly useful for targeting immune-escaped TAP-deficient cancers, as has been shown for the combination of cancer-virus vaccination with PD-1 blockade.
  • the pharmaceutical compositions provided herein may be used in combination with an immune checkpoint inhibitor that blocks PD-1, CTLA-4, PD-L1, TIM3, TIGIT, VISTA, NKG2A or LAG-3.
  • the pharmaceutical compositions provided herein may be used in combination with an immune check point inhibitor that is selected from an antibody that blocks CTLA-1 OR PD-1/PD-L1 OR NKG2A.
  • an immune check point inhibitor that is selected from an antibody that blocks CTLA-1 OR PD-1/PD-L1 OR NKG2A.
  • the immune checkpoint inhibitor may be an inhibitor of PD-1 and/or PD- L1 activity.
  • the immune checkpoint inhibitor may result in PD-1 or PD-L1 blockade.
  • the inhibitor of PD-1 and/or PD-L1 activity may be e.g. an antibody that blocks PD-L1 binding to PD1 (or vice versa).
  • Administration of the pharmaceutical composition and the immune checkpoint inhibitor may be in any order.
  • the pharmaceutical composition is administered at the same time or after the immune checkpoint inhibitor.
  • the pharmaceutical composition is administered at the same time or before the immune checkpoint inhibitor.
  • a pharmaceutical composition described herein (comprising a nucleic acid, a nucleic acid composition, a complex, or a cell according to the invention) and a vaccine or peptide vaccine described herein are provided for use as a medicament.
  • the pharmaceutical composition described herein and the vaccine or peptide vaccine described herein may be combined for use as a medicament.
  • the combination may comprise administering the vaccine or peptide vaccine prior to, during, and/or after administration of the pharmaceutical composition. Treatment of a subject Pharmaceutical compositions or vaccines described herein may advantageously be used as a medicament.
  • compositions or vaccines may be used to treat or prevent a pre-cancer, a cancer or a viral infection associated with impaired HLA class I antigen presentation in a human subject.
  • the human subject is positive for HLA-A*02, such as HLA-A*02:01.
  • pre-cancer or "pre-cancerous” refers to a condition or a growth that typically precedes or develops into a cancer.
  • a "pre-cancerous" growth or “pre-cancer” will have cells that are characterized by abnormal cell cycle regulation, proliferation, or differentiation, which can be determined by markers of cell cycle regulation, cellular proliferation, or differentiation.
  • induced or enhanced immune response e.g. a cell mediated response
  • subject e.g. a targeted immune response to cancerous or virally infected cells that present the HLA-A restricted peptide.
  • induced or enhanced immune response refers to an increase in the immune response (e.g. a cell mediated immune response such as a T cell mediated immune response) of the subject during or after treatment compared to their immune response prior to treatment.
  • An “induced or enhanced” immune response therefore encompasses any measurable increase in the immune response that is directly or indirectly targeted to the pre-cancer, cancer or viral infection being treated (or prevented).
  • compositions of the invention may be used to treat or prevent a cancer associated with impaired HLA class I antigen presentation.
  • a person of skill in the art will be fully aware of cancers that are associated with impaired HLA class I antigen presentation and thus may be treated in accordance with the invention.
  • the pharmaceutical composition or vaccine may be for use in stimulating a cell mediated immune response to a target cell population or tissue in a human subject.
  • the pharmaceutical composition or vaccine may also be for use in providing anti-tumor immunity to a human subject.
  • the cancer is cancer with impaired peptide processing machinery.
  • the cancer is melanoma.
  • the cancer is lung cancer.
  • compositions or vaccines of the invention may be used to treat or prevent a pre-cancer associated with impaired HLA class I antigen presentation.
  • a pre-cancer associated with impaired HLA class I antigen presentation A person of skill in the art will be fully aware of pre-cancers that are associated with impaired HLA class I antigen presentation and thus may be treated in accordance with the invention.
  • the pre-cancer is a pre-cancer with impaired peptide processing machinery.
  • Compositions or vaccines of the invention may also be used to treat or prevent a viral infection associated with impaired HLA class I antigen presentation.
  • a person of skill in the art will be fully aware of viral infections that are associated with impaired HLA class I antigen presentation and thus may be treated in accordance with the invention.
  • a pre-cancer, cancer or viral infection “associated with impaired HLA class I antigen presentation” refers to a pre-cancer, cancer or viral infection that results in a change in the HLA class I antigen presentation pathway in the pre-cancerous, cancerous or virally infected cell, which results in a reduction in HLA class I antigen presentation in these cells.
  • a reduction encompasses a decrease of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% etc in the presentation of non-TEIPP HLA class I-restricted antigens at the cell surface of these cells (at a given time) compared to control cells (e.g.
  • TAP downregulation has also been observed in lung cancer specimens (“Different Expression Levels of the TAP Peptide Transporter Lead to Recognition of Different Antigenic Peptides by Tumor-Specific CTL”, A. Durgeau, F. El Hage, I. Vergnon, P. Validire, V. de Monterieville, B. Besse, J. Soria, T. van Hall and F. Mami-Chouaib, J Immunol December 1, 2011, 187 (11) 5532-5539, “Loss of antigen-presenting molecules (MHC class I and TAP-1) in lung cancer”, Korkolopoulou P., L. Kaklamanis, F. Pezzella, A. L. Harris, K. C. Gatter.1996. Br. J.
  • a pre-cancer, cancer or viral infection associated with impaired HLA class I antigen presentation may therefore be a pre-cancer, cancer or viral infection wherein the tumor cells or infected cells have a mutated TAP1 or TAP2 gene.
  • the mutation reduces TAP1 or TAP2 expression (such that the pre-tumor cell, tumor cell or virally infected cell has low TAP1 or TAP2 expression). In another example, the mutation reduces TAP1 or TAP2 activity in the cell (such that the pre-tumor cell, tumor cell or virally infected cell has reduced/low TAP1 or TAP2 activity). In other example, the mutation reduces TAP1 or TAP2 protein levels in the cell (e.g. the pre-tumor cell, the tumor cell or virally infected cell has reduced/low TAP1 or TAP2 protein expression and/or reduced/low TAP1 or TAP2 protein stability).
  • TAP1 or TAP2 expression may also be reduced/low in a pre-cancerous cell, cancerous cell or virally infected cell due to epigenetic silencing.
  • Methods for detecting TAP1 or TAP2 epigenetic silencing are well known in the art.
  • TAP1 or TAP2 expression, activity, protein level and/or protein stability may also be reduced/low in a pre-cancerous cell, cancerous cell or virally infected cell for other reasons than mutation of the TAP1 or TAP2 genes (e.g. due to the pre-cancer/cancer/virus altering the molecular machinery and pathways of the cell).
  • the pre-cancer, cancer or viral infection may therefore be a pre-cancer, cancer or viral infection associated with reduced (or low) TAP1 or TAP2 protein expression, activity, level, or stability.
  • Methods for determining the presence of mutations in TAP1 or TAP2 are well known in the art.
  • methods for determining TAP1 or TAP2 expression levels, TAP1 or TAP2 activity levels, TAP1 or TAP2 protein levels, and TAP1 or TAP2 protein stability are well known in the art.
  • the expression level may be detected by measuring mRNA (e.g. using Northern blot analysis, RNA probes (e.g. using spatial transcriptomics (e.g. as offered by NanoString) or using in situ hybridization (e.g.
  • RNAscope® RNAscope®
  • the level of protein may be detected using TAP1 or TAP2 specific antibodies (e.g. with a detectable label) and methods such as enzyme linked immunosorbent assays (ELISAs), immunoprecipitation, immunofluorescence, enzyme immunoassay (EIA), radioimmunoassay (RIA), spatial proteomics (e.g. as offered by NanoString), and Western blot analysis may be used. Other standard methods for determining these parameters are well known in the art.
  • the pre-cancer, cancer or viral infection may be a pre-cancer, cancer or viral infection associated with reduced (or low) TAP1 or TAP2 protein expression, activity, level, or stability.
  • reduced (or low) TAP1 or TAP2 protein expression, activity, level, or stability refers to a decrease in the protein expression activity, level, or stability compared to a control or a reference level (e.g. at least a 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% decrease).
  • reference level refers to a cell sample having a normal level of TAP1 or TAP2 protein expression, activity, level, or stability, for example a sample from a healthy subject not having or suspected of having pre-cancer, cancer or a viral infection or alternatively a cell sample from the same subject being tested, where the control or reference level cell sample is not (and is not suspected of being) pre-cancerous, cancerous or virally infected.
  • the reference level may be a TAP1 or TAP2 protein expression, activity, level, or stability value from a reference database, which may be used to generate a pre- determined cut off value, i.e.
  • a diagnostic score that is statistically predictive of a symptom or disease or lack thereof or may be a pre-determined reference level based on a standard population sample, or alternatively, a pre-determined reference level based on a subject's base line level of expression, i.e. prior to developing or being suspected of having pre-cancer, cancer or a viral infection.
  • reduced or low protein expression may be determined using immunohistochemistry, using anti-TAP1 or anti-TAP2 antibodies, such as the anti-TAP1 Antibody, clone mAb 148.3 (MABF125 EMD Millipore).
  • evaluation of TAP1 or TAP2 normal level protein expression in a sample as compared to reduced or low level of expression is determined by the ‘De Ruiter’ evaluation method.
  • the sample is a pre-cancer, pre-tumour, cancer or tumour sample.
  • the presence of immune modulatory viral gene products for example CMV, HSV or BVS, reduces the expression and / or activity of TAP function and the presence of such gene products can be used as a marker of reduced or low TAP1 or TAP2 expression and / or activity.
  • HLA class I antigen presentation Other molecular pathways that may be altered in the pre-cancerous, cancerous or virally infected cell to impair HLA class I antigen presentation include for example a deficiency in tapasin (a chaperone protein involved in TAP-mediated peptide loading of MHC class I molecules) and inhibition of proteasome-mediated degradation of proteins into peptides for MHC class I presentation (see for example US2009/0220534 for more details).
  • tapasin a chaperone protein involved in TAP-mediated peptide loading of MHC class I molecules
  • proteasome-mediated degradation of proteins into peptides for MHC class I presentation see for example US2009/0220534 for more details.
  • the terms “treat”, “treating” and “treatment” are taken to include an intervention performed with the intention of preventing the development or altering the pathology of a condition, disorder or symptom (i.e. in this case a pre-cancer, cancer or viral infection associated with impaired HLA class I antigen presentation).
  • treatment refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted condition, disorder or symptom.
  • Treatment therefore encompasses a reduction, slowing or inhibition of the amount or concentration of pre-malignant, malignant or virally infected cells, for example as measured in a sample obtained from the subject, of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% when compared to the amount or concentration of malignant cells (or pre-malignant cells or virally infected cells) before treatment.
  • Methods of measuring the amount or concentration of malignant cells include, for example, qRT-PCR, and quantification of specific biomarkers in a sample obtained from the subject.
  • subject refers to an individual, e.g., a human, having or at risk of having a specified condition, disorder or symptom.
  • the subject may be a patient i.e. a subject in need of treatment in accordance with the invention.
  • the subject may have received treatment for the condition, disorder or symptom. Alternatively, the subject has not been treated prior to treatment in accordance with the present invention.
  • the subject is a human subject, preferably a HLA*02 positive human subject, more preferably a HLA*02:01 positive human subject.
  • the compositions or vaccines described herein can be administered to the subject by any conventional route, including injection or by gradual infusion over time.
  • the administration may, for example, be by infusion or by intramuscular, intravascular, intracavity, intracerebral, intralesional, rectal, subcutaneous, intradermal, epidural, intrathecal, percutaneous administration.
  • the compositions or vaccines described herein may be in any form suitable for the above modes of administration.
  • compositions comprising cells may be in any form suitable for infusion.
  • suitable forms for parenteral injection include a sterile solution, suspension or emulsion
  • suitable forms for topical administration include an ointment or cream
  • suitable forms for rectal administration include a suppository.
  • the route of administration may be by direct injection into the target area, or by regional delivery or by local delivery.
  • the identification of suitable dosages of the compositions of the invention is well within the routine capabilities of a person of skill in the art.
  • the compositions of the invention may be formulated for use as a vaccine (e.g.
  • compositions comprising a peptide, wherein the peptide comprises the amino acid sequence of SEQ ID NO: 1 (or the corresponding nucleic acid sequence or vector) may be formulated as a pharmaceutical composition that is suitable for use as a (peptide) vaccine).
  • compositions comprising cells may also be formulated as pharmaceutical compositions that are suitable for use as a vaccine.
  • Suitable cell, binding agent (e.g. antibody), peptide and nucleic acid vaccine formulations are well known in the art.
  • the compositions described herein may be formulated for use in T cell receptor (TCR) gene transfer, an approach that is rapid, reliable and capable of generating large quantities of T cells with specificity for RCN1 antigenic peptides (e.g.
  • the pharmaceutical composition or vaccine is preferably for, and therefore formulated to be suitable for, administration to a subject, preferably a human or animal subject.
  • the administration is parenteral, e.g. intravenous, subcutaneous, intramuscular, intradermal intracutaneous and/or intratumoral administration, i.e. by injection.
  • the pharmaceutical composition or vaccine comprises or consists of an amount of active ingredient (e.g.
  • a pharmaceutical dosage unit is defined herein as the amount of active ingredients (i.e. the total amount of peptide in a peptide-based vaccine for example) that is applied to a subject at a given time point.
  • a pharmaceutical dosage unit may be applied to a subject in a single volume, i.e. a single shot, or may be applied in 2, 3, 4, 5 or more separate volumes or shots that are applied preferably at different locations of the body, for instance in the right and the left limb. It is to be understood herein that the separate volumes of a pharmaceutical dosage may differ in composition, i.e. may comprise different kinds or composition of active ingredients and/or adjuvants.
  • a single injection volume or shot (i.e. volume applied on one location at a certain time point), comprising a total pharmaceutical dosage, or part thereof in case multiple shots applied at substantially the same time point, may between 100 and 2 mL, or between 100 and 1 mL.
  • the single injection volume may be 100 ⁇ l, 200 ⁇ l, 300 ⁇ l, 400 ⁇ l, 500 ⁇ l, 600 ⁇ l, 700 ⁇ l, 800 ⁇ l, 900 ⁇ l, 1 mL, 1.1 mL, 1.2 mL, 1.3 mL, 1.4 mL, 1.5 mL, 1.6 mL, 1.7 mL, 1.8 mL, 1.9 mL, 2 mL, 3 mL or any value in between.
  • the pharmaceutical dosage unit, or total amount of active ingredient applied to a subject at a given time point will depend on the type of vaccine (e.g. peptide, cell, nucleic acid etc).
  • the pharmaceutical dosage unit, or total amount of peptide applied to a subject at a given time point comprises an amount of peptide in the range from 0.1 ⁇ g to 20 mg, such as about 0.1 ⁇ g, 0.5 ⁇ g, 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 15 ⁇ g, 20 ⁇ g, 30 ⁇ g, 40 ⁇ g, 50 ⁇ g, 60 ⁇ g, 70 ⁇ g, 80 ⁇ g, 90 ⁇ g, 100 ⁇ g, 150 ⁇ g, 200 ⁇ g, 250 ⁇ g, 300 ⁇ g, 350 ⁇ g, 400 ⁇ g, 450 ⁇ g, 500 ⁇ g, 650 ⁇ g, 700 ⁇ g, 750 ⁇ g, 800 ⁇ g,
  • compositions or vaccines described herein are for administration in an effective amount.
  • An “effective amount” is an amount that alone, or together with further doses, produces the desired (therapeutic or non-therapeutic) response.
  • the effective amount to be used will depend, for example, upon the therapeutic (or non-therapeutic) objectives, the route of administration, and the condition of the patient/subject.
  • the suitable dosage of the composition of the invention for a given patient/subject will be determined by the attending physician (or person administering the composition), taking into consideration various factors known to modify the action of the composition of the invention for example severity and type of haematological malignancy, body weight, sex, diet, time and route of administration, other medications and other relevant clinical factors.
  • the dosages and schedules may be varied according to the particular condition, disorder or symptom the overall condition of the patient/subject.
  • Effective dosages may be determined by either in vitro or in vivo methods.
  • the pharmaceutical compositions of the present invention are advantageously presented in unit dosage form. Binding agents are described herein that specifically bind to a peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO: 1.
  • binding agent is useful in the prevention or treatment of a pre-cancer, cancer or viral infection associated with impaired HLA class I antigen presentation in a human subject.
  • binding agents are described herein that specifically bind to a peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • the binding agent may specifically bind to an epitope within the amino acid sequence provided by SEQ ID NO: 1.
  • the binding agent may specifically bind to an epitope within the amino acid sequence provided by any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • epitope refers to a site on a target molecule (in this case the recited peptide) to which a binding agent binds.
  • Epitopes are groupings of molecules such as amino acids or sugar side chains and usually have specific structural characteristics, as well as specific charge characteristics. A single peptide (antigen) may have more than one epitope.
  • Epitopes can be formed both from contiguous or adjacent noncontiguous residues (e.g., amino acid residues) of the target molecule. Epitopes formed from contiguous residues (e.g., amino acid residues) typically are also called linear epitopes. An epitope typically includes at least 5 and up to about 12 residues, mostly between 6 and 10 residues (e.g. amino acid residues). Epitopes may also be conformational (i.e. non-linear).
  • the binding agent specifically binds to an epitope generated by the peptide itself.
  • the binding agent e.g. antibody
  • the binding agent binds to an epitope generated by the combination of the peptide and the HLA molecule that presents it (i.e.
  • the binding agent of the invention may be any appropriate binding agent that specifically binds to a peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO: 1.
  • the binding agent of the invention may be any appropriate binding agent that specifically binds to a peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • An example of a suitable binding agent of the invention includes an HLA-A*02 molecule that specifically binds to the peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO:1.
  • Another example of a suitable binding agent of the invention includes an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule that specifically binds to the peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO:1.
  • a suitable binding agent of the invention includes an HLA-A*02 molecule that specifically binds to the peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • a further example of a suitable binding agent of the invention includes an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule that specifically binds to the peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • HLA-A*02, HLA-A*02:02, HLA-A*02:03, HLA- A*02:04, or HLA-A*02:09, or HLA-A*02:01 molecules may be useful, for example, as part of a multimeric structure for use in administration to a subject for stimulating T cells in the subject (for example in the form of a synthetic DC).
  • the binding agent that specifically binds to a peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO: 1 comprises an HLA-A*02 molecule.
  • the HLA-A*02 molecule specifically binds to a peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO: 1.
  • the binding agent that specifically binds to a peptide comprising (or consisting of) the amino acid sequence of SEQ ID NO: 1 comprises an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA- A*02:09, preferably an HLA-A*02:01, molecule.
  • the binding agent that specifically binds to a peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5, comprises an HLA-A*02 molecule.
  • the HLA-A*02 molecule specifically binds to a peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • the binding agent that specifically binds to a peptide comprising (or consisting of) the amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 - 5, more preferably any one of SEQ ID NOs: 2 - 4 or any one of SEQ ID NO: 3 - 5, comprises an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA- A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • binding agents may be useful as pharmaceutical compositions, as described elsewhere herein.
  • a complex comprising: a) a peptide comprising the amino acid sequence of SEQ ID NO: 1, and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1; optionally wherein the binding agent is an HLA-A*02:01, HLA- A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRA (SEQ ID NO: 2), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRA (SEQ ID NO: 2); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRV (SEQ ID NO: 3), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRV (SEQ ID NO: 3); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRI (SEQ ID NO: 4), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRI (SEQ ID NO: 4); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA- A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRL (SEQ ID NO: 5), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRL (SEQ ID NO: 5); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRR (SEQ ID NO: 6), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRR (SEQ ID NO: 6); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRN (SEQ ID NO: 7), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRN (SEQ ID NO: 7); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRD (SEQ ID NO: 8), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRD (SEQ ID NO: 8); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRC (SEQ ID NO: 9), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRC (SEQ ID NO: 9); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRE (SEQ ID NO: 10), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRE (SEQ ID NO: 10); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRQ (SEQ ID NO: 11), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRQ (SEQ ID NO: 11); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRG (SEQ ID NO: 12), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRG (SEQ ID NO: 12); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRH (SEQ ID NO: 13), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRH (SEQ ID NO: 13); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRK (SEQ ID NO: 14), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRK (SEQ ID NO: 14); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRM (SEQ ID NO: 15), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRM (SEQ ID NO: 15); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRF (SEQ ID NO: 16), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRF (SEQ ID NO: 16); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRP (SEQ ID NO: 17), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRP (SEQ ID NO: 17); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRS (SEQ ID NO: 18), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRS (SEQ ID NO: 18); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRT (SEQ ID NO: 19), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRT (SEQ ID NO: 19); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRW (SEQ ID NO: 20), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRW (SEQ ID NO: 20); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01,molecule.
  • a complex comprising: a) a peptide comprising the amino acid sequence VLAPRVLRY (SEQ ID NO: 21), and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence VLAPRVLRY (SEQ ID NO: 21); optionally wherein the binding agent is an HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecule.
  • the binding agent is complexed with or bound to the peptide.
  • HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably an HLA-A*02:01, molecules described herein may be useful, for example, as part of a multimeric structure for use in administration to a subject for stimulating T cells in the subject, for example in the form of an artificial antigen presenting cell (aAPC).
  • aAPCs are synthetic structures loaded with tumor antigens.
  • aAPCs are designed to mimic dendritic cells (DCs), hence also called synthetic DCs, with the goal of triggering an efficient and specific T cell response directed against a tumor in the context of cancer treatment.
  • DCs dendritic cells
  • the complex may be present in a cell, where a binding agent is loaded with the peptide according to the invention.
  • a cell expresses the complex according to the invention.
  • the cell may be an antigen presenting cell.
  • the antigen presenting cell may be selected from a macrophage, dendritic cell, a monocyte, a B-cell or a synthetic form of antigen presenting cell.
  • Binding agents as described above may be useful as pharmaceutical compositions, as described elsewhere herein.
  • the binding agent is an isolated binding agent.
  • an “isolated binding agent” refers to a binding agent that is not in its natural environment.
  • the binding agent may therefore be a recombinant binding agent, or the binding agent may be of synthetic origin (or alternatively, of natural original, but isolated from its natural environment).
  • the natural environment of binding agents such as HLA-A2*02 or HLA-A2*02:01 molecules is within the human body. Accordingly, when the binding agent (e.g. HLA-A2*02 or HLA-A2*02:01 molecules) are present e.g. in a pharmaceutical composition (comprising adjuvants etc) they are considered to be in isolated form, as they are not in their natural environment.
  • the terms “specific binding” and “binding specifically” are used interchangeably to indicate that other biomolecules do not significantly bind to the region (that is specifically binding to the peptide of interest (e.g. the recited peptide comprising the amino acid sequence of SEQ ID NO:1)).
  • the level of binding to a biomolecule other than the peptide of interest results in a negligible (e.g., not determinable) binding affinity by means of ELISA or an affinity determination.
  • binding affinity of the binding agent to the peptide of interest (e.g. the recited peptide comprising the amino acid sequence of SEQ ID NO:1) may be determined using a standard binding assay, such as surface plasmon resonance technique (BIAcore®, GE-Healthcare Uppsala, Sweden).
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • BIAcore Phacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.
  • nucleic acid compositions that encode binding protein components
  • An isolated nucleic acid composition that encodes a Reticulocalbin 1 (RCN1) antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is provided herein, the composition comprising: (a) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence; and (b) a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • a RCN1 antigen according to the invention comprises a peptide comprising an amino acid sequence selected from the group consisting of: SEQ ID NOs 1 - 21, preferably selected from the group consisting of: SEQ ID NOs 1 – 5 or SEQ ID NOs 2 - 5, more preferably selected from the group consisting of: SEQ ID NOs 2 - 4 or any one of SEQ ID NO: 3 - 5.
  • the CDR3 sequences according to the invention preferably together specifically bind to a peptide comprising the amino acid sequence of any one of SEQ ID NOs 1 - 21, more preferably the amino acid sequence of any one of SEQ ID NOs 1 - 5 or any one of SEQ ID NOs 2 - 5, even more preferably the amino acid sequence of any one of SEQ ID NOs 2 - 4 or any one of SEQ ID NO: 3 – 5 (e.g. when the peptide is complexed with HLA).
  • the CDR3 amino acid sequences described herein specifically bind to their target (in this case a peptide comprising the amino acid sequence of SEQ ID NO:1, for example a VLAPRVLRA peptide, a VLAPRVLRV peptide, a VLAPRVLRI peptide or a VLAPRVLRL peptide), when the target (i.e. the appropriate peptide) is presented in the context of HLA.
  • the binding proteins (and CDR3 sequences specifically described herein) are therefore capable of specifically binding to an appropriate peptide:HLA complex. These complexes are described in more detail elsewhere herein.
  • the invention provides an isolated nucleic acid composition that encodes a binding protein comprising T cell receptor (TCR) components that specifically bind a RCN1 antigen (e.g. to a peptide comprising the sequence of SEQ ID NO: 1, such as a VLAPRVLRA peptide, a VLAPRVLRV peptide, a VLAPRVLRI peptide or a VLAPRVLRL peptide).
  • TCR T cell receptor
  • a peptide comprising an amino acid sequence selected from the group consisting of: SEQ ID NOs 2 - 21, preferably selected from the group consisting of: SEQ ID NOs 2 - 5, more preferably selected from the group consisting of: SEQ ID NOs 2 - 4 or from the group consisting of SEQ ID NO: 3 - 5) and does not bind to a peptide that does not contain the amino acid sequence of SEQ ID NO: 1 (e.g.
  • the nucleic acid composition comprises (a) a nucleic acid sequence that encodes a TCR V ⁇ domain with the specified features described herein and (b) a nucleic acid sequence that encodes a TCR V ⁇ domain with the specified features described herein.
  • the encoded TCR components form a RCN1 antigen-specific binding protein.
  • the nucleic acid sequences of (a) and (b) above may be distinct nucleic acid sequences within the nucleic acid composition.
  • the TCR components of the binding protein may therefore be encoded by two (or more) nucleic acid sequences (with distinct nucleotide sequences) which, together, encode all of the TCR components of the binding protein.
  • some of the TCR components may be encoded by one nucleic acid sequence in the nucleic acid composition, and others may be encoded by another (distinct) nucleic acid sequence within the nucleic acid composition.
  • the nucleic acid sequences of (a) and (b) may be part of a single nucleic acid sequence.
  • the TCR components of the binding protein may therefore all be encoded by a single nucleic acid sequence (for example with a single open reading frame, or with multiple (e.g.2 or more, three or more etc.) open reading frames).
  • Nucleic acid sequences described herein may form part of a larger nucleic acid sequence that encodes a larger component part of a functioning binding protein.
  • a nucleic acid sequence that encodes a TCR V ⁇ domain with the specified features described herein may be part of a larger nucleic acid sequence that encodes a functional TCR ⁇ chain (including the constant domain).
  • nucleic acid sequence that encodes a TCR V ⁇ domain with the specified features described herein may be part of a larger nucleic acid sequence that encodes a functional TCR ⁇ chain (including the constant domain).
  • both nucleic acid sequences (a) and (b) above may be part of a larger nucleic acid sequence that encodes a combination of a functional TCR ⁇ chain (including the constant domain) and a functional TCR ⁇ chain (including the constant domain), optionally wherein the sequence encoding the functional TCR ⁇ chain is separated from the sequence encoding the functional TCR ⁇ chain by a linker sequence that enables coordinate expression of two proteins or polypeptides in the same nucleic acid sequence.
  • the nucleic acid sequences described herein may alternatively encode a small component of a T cell receptor e.g. a TCR V ⁇ domain, or a TCR V ⁇ domain, only.
  • the nucleic acid sequences may be considered as “building blocks” that provide essential components for peptide binding specificity.
  • the nucleic acid sequences described herein may be incorporated into a distinct nucleic acid sequence (e.g. a vector) that encodes the other elements of a functional binding protein such as a TCR, such that when the nucleic acid sequence described herein is incorporated, a new nucleic acid sequence is generated that encodes e.g.
  • a TCR ⁇ chain and/or a TCR ⁇ chain that specifically binds to a RCN1 antigen e.g. wherein the RCN1 antigen comprises an amino acid sequence selected from the group consisting of: SEQ ID NOs 1 - 21, preferably selected from the group consisting of: SEQ ID NOs 1 - 5, or SEQ ID NOs 2 - 5, more preferably selected from the group consisting of: SEQ ID NOs 2 - 4 or from the group consisting of SEQ ID NO: 3 - 5).
  • the nucleic acid sequences described herein therefore have utility as essential components that confer binding specificity for a RCN1 antigen, and thus can be used to generate a larger nucleic acid sequence encoding a binding protein with the required antigen binding activity and specificity.
  • the nucleic acid sequences described herein may be codon optimised for expression in a host cell, for example they may be codon optimised for expression in a human cell, such as a cell of the immune system, an inducible pluripotent stem cell (iPSC), a hematopoietic stem cell, a T cell, a primary T cell, a T cell line, a NK cell, an innate lymphoid cell (ILC), or a natural killer T cell (Scholten et al, Clin.
  • iPSC inducible pluripotent stem cell
  • hematopoietic stem cell hematopoietic stem cell
  • T cell a primary T cell
  • T cell line a T cell line
  • the T cell can be a CD4+ or a CD8+ T cell.
  • Codon optimisation is a well-known method in the art for maximizing expression of a nucleic acid sequence in a particular host cell.
  • one or more cysteine residues may also be introduced into the encoded TCR alpha and beta chain components (e.g. to reduce the risk of mispairing with endogenous TCR chains).
  • the nucleic acid sequences described herein are codon optimised for expression in a suitable host cell, and/or are modified to introduce codons encoding one or more cysteine amino acids (e.g.
  • the nucleic acid sequences described herein are codon optimised for expression in a suitable host cell, optionally wherein the host cell is a human cell.
  • a TCR constant domain is modified to enhance pairing of desired TCR chains.
  • enhanced pairing between a heterologous TCR ⁇ chain and a heterologous TCR ⁇ chain due to a modification may result in the preferential assembly of a TCR comprising two heterologous chains over an undesired mispairing of a heterologous TCR chain with an endogenous TCR chain (see, e.g., Govers et al, Trends Mol. Med.16(2):11 (2010)).
  • Exemplary modifications to enhance pairing of heterologous TCR chains include the introduction of complementary cysteine residues in each of the heterologous TCR ⁇ chain and ⁇ chain.
  • a binding protein that is encoded by the nucleic acid compositions described herein is specific for a RCN1 antigen and comprises RCN1 antigen specific-TCR components.
  • the encoded binding protein is not limited to being a TCR.
  • Other appropriate binding proteins that comprise the specified RCN1 antigen specific-TCR components are also encompassed.
  • the encoded binding protein may comprise a TCR, an antigen binding fragment of a TCR, a chimeric antigen receptor (CAR), or an ImmTAC.
  • TCRs, antigen binding fragments thereof, CARs and ImmTACs are well defined in the art.
  • an antigen binding fragment of a TCR is a single chain TCR (scTCR) or a chimeric dimer composed of the antigen binding fragments of the TCR ⁇ and TCR ⁇ chain linked to transmembrane and intracellular domains of a dimeric complex so that the complex is a chimeric dimer TCR (cdTCR).
  • An ImmTAC comprises a TCR connected to an anti-CD3 antibody. ImmTACs are therefore bispecific, combining RCN1- recognizing TCR components with immune activating complexes.
  • an antigen-binding fragment of a TCR comprises a single chain TCR (scTCR), which comprises both the TCR V ⁇ and TCR V ⁇ domains, but only a single TCR constant domain.
  • an antigen-binding fragment of a TCR comprises a chimeric TCR dimer in which the antigen binding fragment of the TCR is linked to an alternative transmembrane and intracellular signalling domain (where the alternative transmembrane and intracellular signalling domain are not naturally found in TCRs).
  • an antigen-binding fragment of a TCR or a chimeric antigen receptor is chimeric (e.g., comprises amino acid residues or motifs from more than one donor or species), humanized (e.g., comprises residues from a non- human organism that are altered or substituted so as to reduce the risk of immunogenicity in a human), or human.
  • Chimeric antigen receptor refers to a fusion protein that is engineered to contain two or more naturally-occurring amino acid sequences linked together in a way that does not occur naturally or does not occur naturally in a host cell, which fusion protein can function as a receptor when present on a surface of a cell.
  • CARs described herein include an extracellular portion comprising an antigen binding domain (i.e., obtained or derived from an immunoglobulin or immunoglobulin-like molecule, such as an scFv derived from an antibody or TCR specific for an antigen (e.g. a cancer antigen etc), or an antigen binding domain derived or obtained from a killer immunoreceptor from an NK cell) linked to a transmembrane domain and one or more intracellular signalling domains (optionally containing co-stimulatory domain(s)) (see, e.g., Sadelain et al, Cancer Discov., 3(4):388 (2013); see also Harris and Kranz, Trends Pharmacol.
  • an antigen binding domain i.e., obtained or derived from an immunoglobulin or immunoglobulin-like molecule, such as an scFv derived from an antibody or TCR specific for an antigen (e.g. a cancer antigen etc), or an antigen binding domain derived or
  • TCR T cell receptor
  • the invention is directed to nucleic acid compositions that encode binding proteins comprising TCR components that interact with a particular peptide in the context of the appropriate serotype of MHC, i.e. a RCN1 antigen in the context of HLA-A*02 or HLA-A*02:01, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09, preferably HLA- A*02:01 (in other words, the encoded binding protein is capable of specifically binding to a RCN1 antigen: specific HLA complex).
  • the invention is directed to nucleic acid compositions that encode binding proteins comprising TCR components that interact with a particular peptide in the context of the appropriate serotype of MHC, i.e. the peptide of SEQ ID NO: 1 in the context of HLA-A*02:01; VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01; VLAPRVLRV (SEQ ID NO: 3) in the context of HLA-A*02:01; VLAPRVLRI (SEQ ID NO: 4) in the context of HLA-A*02:01; VLAPRVLRL (SEQ ID NO: 5) in the context of HLA-A*02:01; VLAPRVLRR (SEQ ID NO: 6) in the context of HLA-A*02:01; VLAPRVLRN (SEQ ID NO: 7) in the context of HLA-A*02:01; VLAPRVLRD (SEQ ID NO: 8) in the context of H
  • the TCR components interact with a peptide comprising an amino acid sequence selected from the group consisting of: SEQ ID NOs: 1 - 21 in the context of HLA-A*02:01.
  • HLA-A*02:01 is a globally common human leukocyte antigen serotype within the HLA-A serotype group.
  • Peptides that are presented by HLA-A*02:01 to TCRs are described as being “HLA- A*02:01 restricted”. Other respective serotypes are described accordingly.
  • the inventors have identified several RCN1 derived peptides presented on cells in HLA-A*02:01. Specifically, the inventors identified the RCN1 derived peptide SEQ ID NO: 2 (VLAPRVLRA).
  • the RCN1 antigen specifically bound by a binding protein described herein may comprise an amino acid sequence selected from the group consisting of: SEQ ID NOs: 1 - 21, preferably selected from the group consisting of: SEQ ID NOs 1 - 5 or SEQ ID NOs 2 - 5.
  • the antigen may be an antigenic fragment (i.e.
  • the inventors identified that the RCN1 derived peptide VLAPRVLRA (SEQ ID NO: 2) is capable of being presented by HLA-A*02:01.
  • the second amino acid, L, in this peptide may be varied without adversely affecting TCR binding to the peptide:HLA complex.
  • VLAPRVLRA SEQ ID NO: 2
  • VX1APRVLRX2 SEQ ID NO: 1
  • the binding protein described herein is capable of specifically binding to a peptide:HLA complex selected from the group consisting of: a VX1APRVLRX2:HLA- A*02:01 complex (wherein X1 and X2 are any amino acid), a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, a VLAPRVLRL:HLA-A*02:01 complex, a VLAPRVLRR:HLA-A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, a VLAPRVLRD:HLA-A*02:01 complex, a VLAPRVLRC:HLA-A*02:01 complex, a VLAPRVLRQ:HLA-A*02:01 complex, a VLAPRVLRE:HLA-
  • the encoded binding protein is capable of specifically binding to a peptide:HLA complex selected from the group consisting of: a VX 1 APRVLRX 2 :HLA-A*02 complex (wherein X 1 and X 2 are any amino acid), a VLAPRVLRA:HLA-A*02 complex, a VLAPRVLRV:HLA- A*02 complex, a VLAPRVLRI:HLA-A*02 complex, a VLAPRVLRL:HLA-A*02 complex, a VLAPRVLRR:HLA-A*02 complex, a VLAPRVLRN:HLA-A*02 complex, a VLAPRVLRD:HLA-A*02 complex, a VLAPRVLRC:HLA
  • the RCN1 derived peptide of the peptide:HLA complex comprises an antigenic fragment of an amino acid sequence selected from the group consisting of: SEQ ID NOs: 1 - 21, preferably selected from the group consisting of: SEQ ID NOs 1 - 5 or SEQ ID NOs 2 - 5, more preferably selected from the group consisting of: SEQ ID NOs 2 - 4 or from the group consisting of SEQ ID NO: 3 - 5.
  • the RCN1 derived peptide of the peptide:HLA complex comprises or consists of an amino acid sequence selected from the group consisting of: SEQ ID NOs: 1 - 21, preferably selected from the group consisting of: SEQ ID NOs 1 - 5 or SEQ ID NOs 2 - 5, preferably selected from the group consisting of: SEQ ID NOs 2 - 4 or from the group consisting of SEQ ID NO: 3 - 5.
  • the encoded binding protein is capable of interacting with (e.g. specifically binding to) the natural RCN1 amino acid sequence of SEQ ID NO:2 (e.g. wherein it does not bind to a peptide that does not include the amino acid sequence of SEQ ID NO:1).
  • the TCR is composed of two different polypeptide chains. In humans, 95% of TCRs consist of an alpha ( ⁇ ) chain and a beta ( ⁇ ) chain (encoded by TRA and TRB respectively). When the TCR engages with a peptide in the context of HLA (e.g. in the context of HLA-A*02:01), the T cell is activated through signal transduction.
  • the alpha and beta chains of the TCR are highly variable in sequence. Each chain is composed of two extracellular domains, a variable domain (V) and a constant domain (C). The constant domain is proximal to the T cell membrane followed by a transmembrane region and a short cytoplasmic tail while the variable domain binds to the peptide/HLA complex.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain.
  • the nucleic acid composition described herein may comprise a TCR ⁇ chain constant domain and/or a TCR ⁇ chain constant domain.
  • the variable domain of each chain has three hypervariable regions (also called complementarity determining regions (CDRs)).
  • CDRs complementarity determining regions
  • the TCR alpha variable domain (referred to herein as a TCR V ⁇ domain, TCR V alpha domain, V ⁇ domain or V alpha domain, alpha variable domain etc) comprises a CDR1, a CDR2 and CDR3 region.
  • the TCR beta variable domain (referred to herein as a TCR V ⁇ domain, TCR V beta domain, V ⁇ domain or V beta domain, beta variable domain etc) also comprises a (different) CDR1, CDR2, and CDR3 region. In each of the alpha and beta variable domains it is CDR3 that is mainly responsible for recognizing the peptide being presented by the HLA molecules.
  • TCR ⁇ chain variable domain refers to the variable (V) domain (extracellular domain) of a TCR alpha chain, and thus includes three hypervariable regions (CDR1, CDR2 and the specified CDR3), as well as the intervening sequences, but does not include the constant (C) domain of the alpha chain, which does not form part of the variable domain.
  • TCR ⁇ chain variable domain refers to the variable (V) domain (extracellular domain) of a TCR beta chain, and thus includes three hypervariable regions (CDR1, CDR2 and the specified CDR3), as well as the intervening sequences, but does not include the constant (C) domain of the beta chain, which does not form part of the variable domain.
  • TCR Components The isolated nucleic acid composition described herein encodes a RCN1 antigen-specific binding protein. As discussed herein, the inventors have identified several TCRs that interact with the natural RCN1 amino acid sequence VLAPRVLRA (SEQ ID NO: 2) when presented by HLA-A*02 (e.g.
  • TCR clone TCR-1 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2) As provided elsewhere herein, the inventors identified TCR clone TCR-1 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01. The sequences provided herein that correspond to TCR clone TCR-1 are SEQ ID NOs 40 to 49.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain
  • the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:42, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:45, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA (SEQ ID NO: 2)), is shown in SEQ ID NO:42.
  • variants of the CDR3 amino acid sequences provided herein may also be functional (i.e. retain their ability to confer specific binding to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 42, i.e. they may have at least 80%, at least 81%, at least 90%, or 100% sequence identity to SEQ ID NO: 42.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO:42 by one or several (e.g. two etc) amino acids.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO:42).
  • functional variants of a CDR3 amino acid sequence provided herein retain their ability to confer specific binding to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when the CDR3 is part of TCR V ⁇ domain.
  • Functional variants may be naturally occurring, synthetic, or synthetically improved functional variants of a recited CDR3 amino acid sequence.
  • the term “variant” also encompasses homologues and fragments. Functional variants will typically contain only conservative substitutions of one, two or more amino acids, or substitution, deletion or insertion of non-critical amino acids in non-critical regions of the CDR3.
  • Non-functional variants are amino acid sequence variants of the CDR3 amino acid sequence that do not specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • Non-functional variants will typically contain a non- conservative substitution, a deletion, or insertion or premature truncation of the amino acid sequence of the CDR3 or a substitution, insertion or deletion in critical amino acids or critical regions. Methods for identifying functional and non-functional variants are well known to a person of ordinary skill in the art.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 42.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 40, or a functional variant thereof.
  • a functional variant of a CDR1 sequence refers to a variant that retains the ability to specifically bind to the peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2)).
  • Such functional variants may be naturally occurring, synthetic, or synthetically improved functional variants of the original CDR1 sequence.
  • the term “variant” also encompasses homologues and fragments.
  • Non-functional variants will typically contain only conservative substitutions of one or more amino acids, or substitution, deletion or insertion of non- critical amino acids in non-critical regions of the protein.
  • Non-functional variants are amino acid sequence variants of the original CDR1 sequence that do not specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • Non-functional variants will typically contain a non-conservative substitution, a deletion, or insertion or premature truncation of the amino acid sequence of the original CDR1, or a substitution, insertion or deletion in critical amino acids or critical regions.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 40, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 40.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 1 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO:40.
  • the TCR V ⁇ domain CDR1 has the amino acid sequence of SEQ ID NO:40
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO:41, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • Functional variants may be naturally occurring, synthetic, or synthetically improved functional variants of the original CDR2 sequence.
  • the term “variant” also encompasses homologues and fragments. Functional variants will typically contain only conservative substitutions of one or more amino acids, or substitution, deletion or insertion of non-critical amino acids in non-critical regions of the protein.
  • Non-functional variants are amino acid sequence variants of the original CDR2 sequence that do not specifically bind to HLA-A*02, preferably HLA-A*02:01.
  • Non-functional variants will typically contain a non-conservative substitution, a deletion, or insertion or premature truncation of the amino acid sequence of the original CDR2 sequence or a substitution, insertion or deletion in critical amino acids or critical regions.
  • Methods for identifying functional and non-functional variants are well known to a person of ordinary skill in the art.
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 41, i.e. it may have at least 80%, at least 85%, or 100% sequence identity to SEQ ID NO: 41.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO:41 by one or several amino acids.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO:41).
  • the variant may comprise an amino acid substitution such as a conservative amino acid substitution compared to the original CDR2 sequence.
  • a functional variant of a CDR2 retains the ability to specifically bind to HLA-A*02, preferably HLA-A*02:01.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 41.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:42, SEQ ID NO: 40 and SEQ ID NO: 41, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO:46, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • Such functional variants may be naturally occurring, synthetic, or synthetically improved functional variants of SEQ ID NO:46.
  • Functional variants will typically contain only conservative substitutions of one or more amino acids, or substitution, deletion or insertion of non-critical amino acids in non-critical regions of the protein.
  • Non-functional variants are amino acid sequence variants that do not specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • Non-functional variants will typically contain a non-conservative substitution, a deletion, or insertion or premature truncation of the amino acid sequence of the original TCR V ⁇ domain or a substitution, insertion or deletion in critical amino acids or critical regions.
  • Methods for identifying functional and non-functional variants are well known to a person of ordinary skill in the art.
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 46, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO:46 is also encompassed.
  • the amino acid substitution may be a conservative amino acid substitution.
  • the variability in sequence compared to SEQ ID NO:46 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 42, SEQ ID NO: 40 and/or SEQ ID NO: 41, and still have 25% (or less) sequence variability compared to SEQ ID NO:46).
  • the sequence of the CDRs of SEQ ID NO: 46 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • percent identity can be calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 46).
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g. at least 75%, at least 80%, at least 85%, at least 90%, at least 95% etc) sequence identity to the amino acid sequence of SEQ ID NO: 46, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 42.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 40 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 41.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 46, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions), wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 42.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 40 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 41.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO:47, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • a genetically degenerate sequence thereof i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code.
  • the phrase “genetically degenerate sequence thereof” is used interchangeably with “derivative thereof” herein.
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain.
  • a suitable constant domain for either a TCR ⁇ chain or a TCR ⁇ chain
  • the constant domain may be murine derived, human derived or humanised. Methods for identifying or generating appropriate constant domains are well known to a person of skill in the art and are well within their routine capabilities.
  • the constant domain may be encoded by or derived from a vector, such as a lentiviral, retroviral or plasmid vector but also adenovirus, adeno-associated virus, vaccinia virus, canary poxvirus or herpes virus vectors in which murine or human constant domains are pre-cloned.
  • a vector such as a lentiviral, retroviral or plasmid vector but also adenovirus, adeno-associated virus, vaccinia virus, canary poxvirus or herpes virus vectors in which murine or human constant domains are pre-cloned.
  • minicircles have also been described for TCR gene transfer (non-viral Sleeping Beauty transposition from minicircle vectors as published by R Monjezi, et al., 2017).
  • naked (synthetic) DNA/RNA can also be used to introduce the TCR.
  • nucleic acid composition comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:42, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 42.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 46.
  • the inventors identified TCR clone TCR-1 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-1 are SEQ ID NOs: 40 to 49.
  • variants of the amino acid sequence shown in SEQ ID NO:45 may also be functional (i.e. retain their ability to confer specific binding to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when the CDR3 is part of TCR V ⁇ domain). Such functional variants are therefore encompassed herein.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 45, i.e. they may have at least 80%, at least 83%, at least 91%, or 100% sequence identity to SEQ ID NO: 45.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 45 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 45.
  • the TCR V ⁇ domain CDR3 has the amino acid sequence of SEQ ID NO:45
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 43, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 43, i.e. it may have at least 80%, or 100% sequence identity to SEQ ID NO: 43.
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:43 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 43.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 44, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 44, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 44.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 44 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 44.
  • the TCR V ⁇ domain CDR2 has the amino acid sequence of SEQ ID NO:44
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:45, SEQ ID NO: 43 and SEQ ID NO: 44, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 48, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 48, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 48 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:48 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 45, SEQ ID NO: 43 and/or SEQ ID NO: 44, and still have 25% (or less) sequence variability compared to SEQ ID NO: 48).
  • the sequence of the CDRs of SEQ ID NO: 48 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • percent identity can be calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 48).
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g. at least 75%, at least 80%, at least 85%, at least 90%, at least 95% etc) sequence identity to the amino acid sequence of SEQ ID NO: 48, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 45.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:43 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 44.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO:49, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:45, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:45.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 48.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:42, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:45, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 42; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:45.
  • the peptide comprising the amino acid sequence of SEQ ID NO:1 may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 46; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 48.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 46 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 48.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 47; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 49.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:40 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:41.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:43 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 44.
  • this particular example encompasses components of TCR clone TCR- 1 exemplified herein. The different components of TCR clone TCR-1 and their respective SEQ ID Nos are summarised in Table 7 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker, e.g. a linker that enables expression of two proteins or polypeptides from the same vector.
  • a linker comprising a porcine teschovirus-12A (P2A) sequence may be used, such as 2A sequences from foot-and-mouth disease virus (F2A), equine rhinitis A virus (E2A) or Thosea asigna virus (T2A) as published by A.L. Szymczak et al., Nature Biotechnology 22, 589 - 594 (2004) or 2A-like sequences.2A and 2A-like sequences are linkers that are cleavable once the nucleic acid molecule has been transcribed and translated.
  • Another example of a linker is an internal ribosomal entry sites (IRES) which enables translation of two proteins or polypeptides from the same transcript.
  • IRS internal ribosomal entry sites
  • any other appropriate linker may also be used.
  • the nucleic acid sequence encoding the TCR V ⁇ domain and nucleic acid sequence encoding the TCR V ⁇ domain may be cloned into a vector with dual internal promoters (see e.g. S Jones et al., Human Gene Ther 2009).
  • the identification of appropriate linkers and vectors that enable expression of both the TCR V ⁇ domain and the TCR V ⁇ domain is well within the routine capabilities of a person of skill in the art.
  • Additional appropriate polypeptide domains may also be encoded by the nucleic acid sequences that encode the TCR V ⁇ domain and/or the TCR V ⁇ domain.
  • the nucleic acid sequence may comprise a membrane targeting sequence that provides for transport of the encoded polypeptide to the cell surface membrane of the modified cell.
  • Other appropriate additional domains are well known and are described, for example, in WO2016/071758.
  • the nucleic acid composition described herein may encode a soluble TCR.
  • the nucleic acid composition may encode the variable domain of the TCR alpha and beta chains respectively together with an immune-modulator molecule such as a CD3 agonist (e.g. an anti-CD3 scFv).
  • a CD3 agonist e.g. an anti-CD3 scFv.
  • the CD3 antigen is present on mature human T cells, thymocytes and a subset of natural killer cells. It is associated with the TCR and is involved in signal transduction of the TCR.
  • Antibodies specific for the human CD3 antigen are well known.
  • One such antibody is the murine monoclonal antibody OKT3, which is the first monoclonal antibody approved by the FDA.
  • Other antibodies specific for CD3 have also been reported (see e.g. WO2004/106380; U.S. Patent Application Publication No.2004/0202657; U.S. Pat. No.6,750,325).
  • Immune mobilising mTCR against Cancer (ImmTAC; Immunocore Limited, Milton Partk, Abington, Oxon, United Kingdom) are bifunctional proteins that combine affinity monoclonal T cell receptor (mTCR) targeting with a therapeutic mechanism of action (i.e., an anti-CD3 scFv).
  • a soluble TCR of the invention may be combined with a radioisotope or a toxic drug.
  • a radioisotope or a toxic drug are well known in the art and are readily identifiable by a person of ordinary skill in the art.
  • the nucleic acid composition may encode a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • the linker is non-cleavable.
  • the nucleic acid composition may encode a chimeric two chain TCR in which the TCR alpha chain variable domain and the TCR beta chain variable domain are each linked to a CD3 zeta signalling domain or other transmembrane and intracellular domains.
  • Methods for preparing such single chain TCRs and two chain TCRs are well known in the art; see for example RA Willemsen et al, Gene Therapy 2000.
  • TCR clone TCR-2 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2) As provided elsewhere herein, the inventors have also identified TCR clone TCR-2 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01. The sequences provided herein that correspond to TCR clone TCR-2 are SEQ ID NO:s 50 to 59.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain
  • the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:52, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 55, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA (SEQ ID NO: 2)), is shown in SEQ ID NO:52.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO:52, i.e. they may have at least 80%, at least 83%, at least 91%, or 100% sequence identity to SEQ ID NO:52.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO:52 by one or several (e.g. two etc) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 52.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 50, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 50, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 50.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 50 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO:50.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO:51, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO:51, i.e.
  • V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO:51 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 51.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO:56, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO:56, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO:56 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:56 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 52, SEQ ID NO:50 and/or SEQ ID NO:51, and still have 25% (or less) sequence variability compared to SEQ ID NO:56).
  • the sequence of the CDRs of SEQ ID NO: 56 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 52.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 50 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 51.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 56, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 52.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 50 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 51.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 57, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:52, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 52.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 56.
  • TCR clone TCR-2 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-2 are SEQ ID NOs: 50 to 59.
  • variants of the amino acid sequence shown in SEQ ID NO:55 may also be functional (i.e.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 55, i.e. they may have at least 80%, at least 84%, at least 92%, or 100% sequence identity to SEQ ID NO: 55.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 55 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 55.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 53, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 53, i.e. it may have at least 80%, or 100% sequence identity to SEQ ID NO: 53.
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:53 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 53.
  • the TCR V ⁇ domain CDR1 has the amino acid sequence of SEQ ID NO:53
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 54, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 54, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 54.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 54 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 54.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:55, SEQ ID NO: 53 and SEQ ID NO: 54, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 58, or a functional variant thereof (i.e.
  • variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 58, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 58 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:58 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 55, SEQ ID NO: 53 and/or SEQ ID NO: 54, and still have 25% (or less) sequence variability compared to SEQ ID NO: 58).
  • the sequence of the CDRs of SEQ ID NO: 58 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 55.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:53 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 54.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO:59, or a genetically degenerate sequence thereof (i.e.
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain.
  • suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:55, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:55.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 58.
  • the TCR V ⁇ domain sequences derived from TCR clone TCR-2 discussed above are particularly compatible with the TCR V ⁇ domain sequences derived from TCR clone TCR-2 discussed elsewhere herein.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:52, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:55, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 52; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:55.
  • the peptide comprising the amino acid sequence of SEQ ID NO:1 may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 56; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 58.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 56 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 58.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 57; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 59.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:50 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:51.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:53 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 54.
  • this particular example encompasses components of TCR clone TCR- 2 exemplified herein. The different components of TCR clone TCR-2 and their respective SEQ ID Nos are summarised in Table 8 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker. Suitable linkers are discussed generally elsewhere herein. Additional appropriate polypeptide domains that may also be encoded by the nucleic acid sequences that encode the TCR V ⁇ domain and/or the TCR V ⁇ domain are also discussed generally elsewhere herein.
  • the nucleic acid composition described herein may encode a soluble TCR or a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • TCR clone TCR-4 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2)
  • VLAPRVLRA SEQ ID NO: 2
  • the inventors have also identified TCR clone TCR-4 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is provided, the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:62, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:65, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA SEQ ID NO:2), is shown in SEQ ID NO: 62.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 62, i.e. they may have at least 80%, at least 84%, at least 92%, or 100% sequence identity to SEQ ID NO: 62.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g.
  • V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 62 by one or several (e.g. two etc) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 62.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 60, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 60, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 60.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 60).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 60 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 60.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO: 61, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 61, i.e. it may have at least 80%, at least 85%, or 100% sequence identity to SEQ ID NO: 61.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 61 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 61.
  • the TCR V ⁇ domain CDR2 has the amino acid sequence of SEQ ID NO: 61
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:62, SEQ ID NO: 60 and SEQ ID NO: 61, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO: 66, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 66, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 66 is also encompassed.
  • the variability in sequence compared to SEQ ID NO: 66 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 62, SEQ ID NO: 60 and/or SEQ ID NO: 61, and still have 25% (or less) sequence variability compared to SEQ ID NO:66).
  • the sequence of the CDRs of SEQ ID NO: 66 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 62.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 60 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 61.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 66, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 62.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 60 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 61.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 67, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:62, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 62.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 66.
  • TCR clone TCR-4 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-4 are SEQ ID NO:s 60 to 69.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 65, i.e.
  • V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 65 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 65.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 63, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 63, i.e. it may have at least 80%, or 100% sequence identity to SEQ ID NO: 63.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 63).
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:63 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 63.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 64, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 64, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 64.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 64 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 64.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:65, SEQ ID NO: 63 and SEQ ID NO: 64, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 68, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 68, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 68 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:68 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 65, SEQ ID NO: 63 and/or SEQ ID NO: 64, and still have 25% (or less) sequence variability compared to SEQ ID NO: 68).
  • the sequence of the CDRs of SEQ ID NO: 68 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g. at least 75%, at least 80%, at least 85%, at least 90%, at least 95% etc) sequence identity to the amino acid sequence of SEQ ID NO: 68, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 65.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:63 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 64.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO:69, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:65, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:65.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 68.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:62, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:65, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 62; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:65.
  • the a peptide comprising the amino acid sequence of SEQ ID NO:1 may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 66; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 68.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 66 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 68.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 67; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 69.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 60 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:61.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:63 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 64.
  • this particular example encompasses components of TCR clone TCR- 4 exemplified herein.
  • the different components of TCR clone TCR-4 and their respective SEQ ID Nos are summarised in Table 9 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker. Suitable linkers are discussed generally elsewhere herein.
  • nucleic acid composition described herein may encode a soluble TCR or a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • TCR clone TCR-5 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2) As provided elsewhere herein, the inventors have also identified TCR clone TCR-5 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01. The sequences provided herein that correspond to TCR clone TCR-5 are SEQ ID NO:s 70 to 79.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain
  • the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:72, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 75, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA (SEQ ID NO: 2)), is shown in SEQ ID NO: 72
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 72, i.e. they may have at least 80%, at least 84%, at least 92%, or 100% sequence identity to SEQ ID NO: 72.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g.
  • V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 72 by one or several (e.g. two etc) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 72.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 70, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 70, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 70.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 70).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 70 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 70.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO: 71, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 71, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 71.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 71 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 71.
  • the TCR V ⁇ domain CDR2 has the amino acid sequence of SEQ ID NO: 71
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:72, SEQ ID NO: 70 and SEQ ID NO: 71, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO: 76, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 76, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 76 is also encompassed.
  • the variability in sequence compared to SEQ ID NO: 76 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 72, SEQ ID NO: 70 and/or SEQ ID NO: 71, and still have 25% (or less) sequence variability compared to SEQ ID NO: 76).
  • the sequence of the CDRs of SEQ ID NO: 76 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 72.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 70 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 71.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 76, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 72.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 70 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 71.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 77, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:72, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 72.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 76.
  • TCR clone TCR-5 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-5 are SEQ ID NO:s 70 to 79.
  • An example of an appropriate TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) is shown in SEQ ID NO:75.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 75, i.e.
  • V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 75 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 75.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 73, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 73, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 73.
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:73 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 73.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 74, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 74, i.e. it may have at least 80%, at least 83, or 100% sequence identity to SEQ ID NO: 74.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 74).
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 74 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 74.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:75, SEQ ID NO: 73 and SEQ ID NO: 74, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 78, or a functional variant thereof (i.e.
  • variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 78, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 78 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:78 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 75, SEQ ID NO: 73 and/or SEQ ID NO: 74, and still have 25% (or less) sequence variability compared to SEQ ID NO: 78).
  • the sequence of the CDRs of SEQ ID NO: 78 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 75.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:73 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 74.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 79, or a genetically degenerate sequence thereof (i.e.
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain.
  • suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:75, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:75.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 78.
  • the TCR V ⁇ domain sequences derived from TCR clone TCR-5 discussed above are particularly compatible with the TCR V ⁇ domain sequences derived from TCR clone TCR-5 discussed elsewhere herein.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:72, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:75, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 72; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:75.
  • the peptide comprising the amino acid sequence of SEQ ID NO:1 may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 76; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 78.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 76 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 78.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 77; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 79.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:70 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:71.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:73 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 74.
  • this particular example encompasses components of TCR clone TCR- 5 exemplified herein. The different components of TCR clone TCR-5 and their respective SEQ ID Nos are summarised in Table 10 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker. Suitable linkers are discussed generally elsewhere herein. Additional appropriate polypeptide domains that may also be encoded by the nucleic acid sequences that encode the TCR V ⁇ domain and/or the TCR V ⁇ domain are also discussed generally elsewhere herein.
  • the nucleic acid composition described herein may encode a soluble TCR or a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • TCR clone TCR-9 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2)
  • the inventors have also identified TCR clone TCR-9 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain is provided, the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:82, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 85, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA (SEQ ID NO: 2)), is shown in SEQ ID NO: 82.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 82, i.e. they may have at least 80%, at least 82%,at least 88%, at least 94%, or 100% sequence identity to SEQ ID NO: 82.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 82.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 80, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2)).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 80, i.e. it may have at least 80%, at least 85%, or 100% sequence identity to SEQ ID NO: 80.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 80 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 80.
  • the TCR V ⁇ domain CDR1 has the amino acid sequence of SEQ ID NO: 80
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO: 81, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 81, i.e. it may have at least 80%, at least 87%, or 100% sequence identity to SEQ ID NO: 81.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 81 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 81.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:82, SEQ ID NO: 80 and SEQ ID NO: 81, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO: 86, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 86, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 86 is also encompassed.
  • the variability in sequence compared to SEQ ID NO: 86 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 82, SEQ ID NO: 80 and/or SEQ ID NO: 81, and still have 25% (or less) sequence variability compared to SEQ ID NO:86).
  • the sequence of the CDRs of SEQ ID NO: 86 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 82.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 80 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 81.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 86, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 82.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 80 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 81.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 87, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:82, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 82.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 86.
  • TCR clone TCR-9 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-9 are SEQ ID NO:s 80 to 89.
  • variants of the amino acid sequence shown in SEQ ID NO:85 may also be functional (i.e. retain their ability to confer specific binding to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when the CDR3 is part of TCR V ⁇ domain).
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 85, i.e. they may have at least 80%, at least 83%, at least 91%, or 100% sequence identity to SEQ ID NO: 85.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 85 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 85.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 83, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 83, i.e. it may have at least 80%, or 100% sequence identity to SEQ ID NO: 83.
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:83 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 83.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 84, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 84, i.e.
  • V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 84 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 84.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 88, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2)) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 88, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 88 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:88 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 85, SEQ ID NO: 83 and/or SEQ ID NO: 84, and still have 25% (or less) sequence variability compared to SEQ ID NO: 88).
  • the sequence of the CDRs of SEQ ID NO: 88 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 85.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:83 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 84.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 89, or a genetically degenerate sequence thereof (i.e.
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain.
  • suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:85, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:85.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 88.
  • the TCR V ⁇ domain sequences derived from TCR clone TCR-9 discussed above are particularly compatible with the TCR V ⁇ domain sequences derived from TCR clone TCR-9 discussed elsewhere herein.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:82, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:85, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:85.
  • the peptide comprising the amino acid sequence of SEQ ID NO:1 may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 86; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 88.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 86 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 88.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 87; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 89.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:80 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:81.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:83 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 84.
  • this particular example encompasses components of TCR clone TCR- 9 exemplified herein.
  • the different components of TCR clone TCR-9 and their respective SEQ ID Nos are summarised in Table 11 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker. Suitable linkers are discussed generally elsewhere herein.
  • nucleic acid composition described herein may encode a soluble TCR or a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • TCR clone TCR-12 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2) As provided elsewhere herein, the inventors have also identified TCR clone TCR-12 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01. The sequences provided herein that correspond to TCR clone TCR-12 are SEQ ID NOs 90 to 99.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain
  • the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:92, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 95, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA (SEQ ID NO: 2)), is shown in SEQ ID NO: 92.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 92, i.e. they may have at least 80%, at least 84%, at least 92%, or 100% sequence identity to SEQ ID NO: 92.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 92 by one or several (e.g. two etc) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 92.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 90, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 90, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 90.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 90).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 90 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 90.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO: 91, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 91, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 91.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 91).
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 91 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 91.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:92, SEQ ID NO: 90 and SEQ ID NO: 91, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO: 96, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 96, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 96 is also encompassed.
  • the variability in sequence compared to SEQ ID NO: 96 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 92, SEQ ID NO: 90 and/or SEQ ID NO: 91, and still have 25% (or less) sequence variability compared to SEQ ID NO:96).
  • the sequence of the CDRs of SEQ ID NO: 96 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g. at least 75%, at least 80%, at least 85%, at least 90%, at least 95% etc) sequence identity to the amino acid sequence of SEQ ID NO: 96, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 92.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 90 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 91.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 96, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 92.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 90 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 91.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 97, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:92, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 92.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 96.
  • TCR clone TCR-12 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-12 are SEQ ID NOs 90 to 99.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 95, i.e.
  • V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 95 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 95.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 93, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 93, i.e. it may have at least 80%, or 100% sequence identity to SEQ ID NO: 93.
  • percent identity is calculated as the percentage of identity to the entire length of the reference sequence (e.g. SEQ ID NO: 93).
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:93 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 93.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 94, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 94, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 94.
  • appropriate (functional) V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 94 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 94.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e. SEQ ID NO:95, SEQ ID NO: 93 and SEQ ID NO: 94, or functional variants thereof), with appropriate intervening sequences between the CDRs.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 98, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 98, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 98 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:98 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 95, SEQ ID NO: 93 and/or SEQ ID NO: 94, and still have 25% (or less) sequence variability compared to SEQ ID NO: 98).
  • the sequence of the CDRs of SEQ ID NO: 98 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g. at least 75%, at least 80%, at least 85%, at least 90%, at least 95% etc) sequence identity to the amino acid sequence of SEQ ID NO: 98, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 95.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:93 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 94.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 99, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:95, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:95.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 98.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:92, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:95, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 92; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:95.
  • the peptide comprising the amino acid sequence of SEQ ID NO:1 antigen may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 96; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 98.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 96 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 98.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 97; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 99.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:90 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:91.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:93 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 94.
  • this particular example encompasses components of TCR clone TCR- 12 exemplified herein.
  • the different components of TCR clone TCR-12 and their respective SEQ ID Nos are summarised in Table 12 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker. Suitable linkers are discussed generally elsewhere herein.
  • nucleic acid composition described herein may encode a soluble TCR or a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • TCR clone TCR-13 TCR components that interact with VLAPRVLRA (SEQ ID NO: 2) As provided elsewhere herein, the inventors have also identified TCR clone TCR-13 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01. The sequences provided herein that correspond to TCR clone TCR-13 are SEQ ID NOs 100 to 109.
  • an isolated nucleic acid composition that encodes a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain
  • the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:102, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO: 105, or a functional fragment thereof, wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g.
  • TCR V ⁇ domain CDR3 amino acid sequence that confers specific binding to a RCN1 antigen, in particular a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. to VLAPRVLRA (SEQ ID NO: 2)), is shown in SEQ ID NO: 102.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 102, i.e. they may have at least 80%, at least 84%, at least 92%, or 100% sequence identity to SEQ ID NO: 102.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 102 by one or several (e.g. two etc) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 102.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 100, or a functional variant thereof (i.e.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 100, i.e. it may have at least 80%, at least 83%, or 100% sequence identity to SEQ ID NO: 100.
  • appropriate functional V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO: 100 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 100.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 comprising an amino acid sequence of SEQ ID NO: 101, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 101, i.e.
  • V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 101 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 101.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence of SEQ ID NO: 106, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2)) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 106, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 106 is also encompassed.
  • the variability in sequence compared to SEQ ID NO: 106 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 102, SEQ ID NO: 100 and/or SEQ ID NO: 101, and still have 25% (or less) sequence variability compared to SEQ ID NO:106).
  • the sequence of the CDRs of SEQ ID NO: 106 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 102.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 100 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 101.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having the amino acid sequence of SEQ ID NO: 106, with 0 to 10 (or 0 to 5) amino acid substitutions, insertions or deletions, wherein the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 102.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO: 100 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 101.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 107, or a genetically degenerate sequence thereof (i.e. other nucleic acid sequences that encode the same protein as a result of the degeneracy of the genetic code).
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain. Examples of suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:102, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO: 102.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 106.
  • TCR clone TCR-13 which interacts with VLAPRVLRA (SEQ ID NO: 2) in the context of HLA-A*02:01.
  • the sequences provided herein that correspond to TCR clone TCR-13 are SEQ ID NOs 100 to 109.
  • variants of the amino acid sequence shown in SEQ ID NO:105 may also be functional (i.e. retain their ability to confer specific binding to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2) when the CDR3 is part of TCR V ⁇ domain).
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 105, i.e. they may have at least 80%, at least 86%, at least 93%, or 100% sequence identity to SEQ ID NO: 105.
  • appropriate (functional) V ⁇ domain CDR3 amino acid sequences may vary from the sequence shown in SEQ ID NO: 105 by one or several (e.g. two) amino acids.
  • the CDR3 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 105.
  • the CDR3 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may comprise, in addition to the specified CDR3, a CDR1 comprising an amino acid sequence of SEQ ID NO: 103, or a functional variant thereof (i.e.
  • V ⁇ domain CDR1 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 103, i.e. it may have at least 80%, or 100% sequence identity to SEQ ID NO: 103.
  • appropriate (functional) V ⁇ domain CDR1 amino acid sequences may vary from the sequence shown in SEQ ID NO:103 by one or several amino acids.
  • the CDR1 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 103.
  • the CDR1 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may also comprise, in addition to the specified CDR3 (and optionally the specified CDR1 above), a CDR2 having an amino acid sequence of SEQ ID NO: 104, or a functional variant thereof (i.e. wherein the variant retains the ability to specifically bind to HLA-A*02, most preferably HLA-A*02:01).
  • appropriate functional V ⁇ domain CDR2 amino acid sequences may have at least 80% sequence identity to SEQ ID NO: 104, i.e.
  • V ⁇ domain CDR2 amino acid sequences may vary from the sequence shown in SEQ ID NO: 104 by one or several amino acids.
  • the CDR2 of the V ⁇ domain comprises or consists of the amino acid sequence of SEQ ID NO: 104.
  • the CDR2 may be encoded by any appropriate nucleic acid sequence.
  • the encoded TCR V ⁇ domain may therefore comprise the CDRs mentioned in detail above (by SEQ ID specifically i.e.
  • the encoded TCR V ⁇ domain may have an amino acid sequence of SEQ ID NO: 108, or a functional variant thereof (i.e. wherein the variant TCR V ⁇ domain retains the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2)) when part of a binding protein described herein).
  • the encoded TCR V ⁇ domain may have an amino acid sequence having at least 75%, at least 80%, at least 85% or at least 90% (or at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 108, whilst retaining the ability to specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO:1 (e.g. the peptide shown in SEQ ID NO:2).
  • a functional TCR V ⁇ domain with one or several amino acid substitutions compared to the sequence of SEQ ID NO: 108 is also encompassed.
  • the variability in sequence compared to SEQ ID NO:108 may all be in regions of the TCR V ⁇ domain that do not form CDRs (i.e. the variant may have the CDRs of SEQ ID NO: 105, SEQ ID NO: 103 and/or SEQ ID NO: 104, and still have 25% (or less) sequence variability compared to SEQ ID NO: 108).
  • the sequence of the CDRs of SEQ ID NO: 108 may be retained whilst the rest of the sequence is varied, as appropriate within the “at least 75% identity” parameters specified above.
  • the encoded TCR V ⁇ domain may comprise an amino acid sequence having at least 75% (e.g.
  • the TCR V ⁇ domain comprises a CDR3 having an amino acid sequence of SEQ ID NO: 105.
  • the TCR V ⁇ domain CDR1 may have an amino acid sequence of SEQ ID NO:103 and the TCR V ⁇ domain CDR2 may have an amino acid sequence of SEQ ID NO: 104.
  • the TCR V ⁇ domain may be encoded by the nucleic acid sequence of SEQ ID NO: 109, or a genetically degenerate sequence thereof (i.e.
  • the nucleic acid sequence encoding the TCR V ⁇ domain may also encode a TCR ⁇ chain constant domain.
  • suitable constant domains are generally discussed above.
  • the nucleic acid composition provided herein comprises a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:105, or a functional fragment thereof.
  • the CDR3 of the V ⁇ domain of a nucleic acid composition provided herein comprises or consists of the amino acid sequence of SEQ ID NO:105.
  • the V ⁇ domain of a nucleic acid composition provided herein comprises an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 108.
  • the TCR V ⁇ domain sequences derived from TCR clone TCR-13 discussed above are particularly compatible with the TCR V ⁇ domain sequences derived from TCR clone TCR-13 discussed elsewhere herein.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:102, or a functional fragment thereof; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence having at least 80% sequence identity to SEQ ID NO:105, or a functional fragment thereof.
  • a nucleic acid composition described herein encodes a binding protein specific for a peptide comprising the amino acid sequence of SEQ ID NO:1 having a TCR V ⁇ domain with a CDR3 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 102; and a TCR V ⁇ domain with a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO:105.
  • the peptide comprising the amino acid sequence of SEQ ID NO:1 may comprise or consist of the sequence shown in SEQ ID NO: 2.
  • the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain and the TCR V ⁇ domain may be part of a TCR ⁇ chain having a constant domain.
  • the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 106; and the V ⁇ domain may comprise an amino acid sequence having at least 80% sequence identity to, comprising, or consisting of, SEQ ID NO: 108.
  • the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 106 and the V ⁇ domain comprises the amino acid sequence of SEQ ID NO: 108.
  • the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 107; and the V ⁇ domain may be encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 109.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:100 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:101.
  • the TCR V ⁇ domain may include a CDR1 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO:103 and a CDR2 amino acid sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 104.
  • this particular example encompasses components of TCR clone TCR- 13 exemplified herein.
  • the different components of TCR clone TCR-13 and their respective SEQ ID NOs are summarised in Table 13 below.
  • the nucleic acid composition described herein encodes both a TCR V ⁇ domain and a TCR V ⁇ domain, which form the binding protein that is capable of specifically binding to a RCN1 antigen.
  • the TCR V ⁇ domain and the TCR V ⁇ domain may be joined together via a linker. Suitable linkers are discussed generally elsewhere herein.
  • nucleic acid composition described herein may encode a soluble TCR or a chimeric single chain TCR wherein the TCR alpha chain variable domain is linked to the TCR beta chain variable domain and a constant domain which is e.g. fused to the CD3 zeta signalling domain.
  • a method of generating a binding protein that is capable of specifically binding to a peptide containing a RCN1 antigen comprising the amino acid sequence of SEQ ID NO: 1 and does not bind to a peptide that does not contain the RCN1 antigen comprising the amino acid sequence of SEQ ID NO: 1 comprising contacting a nucleic acid composition (or vector system) described herein with a cell under conditions in which the nucleic acid composition is incorporated and expressed by the cell.
  • the RCN1 antigen comprises or consists of a sequence comprising an amino acid sequence selected from the group consisting of: SEQ ID NO: 1 - 27, or a functional fragment or variant thereof.
  • the method may be carried out on the (host) cell ex vivo or in vitro. Alternatively, the method may be performed in vivo, wherein the nucleic acid composition (or vector system) is administered to the subject and is contacted with the cell in vivo, under conditions in which the nucleic acid sequence is incorporated and expressed by the cell to generate the binding protein. In one example, the method is not a method of treatment of the human or animal body.
  • the binding protein comprise a TCR, an antigen binding fragment of a TCR, a ImmTAC or a chimeric antigen receptor (CAR). Further details are provided elsewhere herein.
  • the binding proteins described herein may be used therapeutically, as described elsewhere herein.
  • the binding proteins may be used in a diagnostic setting, e.g. to detect the presence of RCN1 presented in the context of an appropriate HLA at the cell surface of diseased/malignant tissues.
  • a peptide according to the invention for identifying a therapeutic binding protein
  • Use of a peptide comprising an amino acid sequence of SEQ ID NO: 1, or a complex according to the invention, for identifying a therapeutic binding protein is also provided herein.
  • Use of a peptide comprising an amino acid sequence of any one of SEQ ID NOs: 2 - 21, preferably any one of SEQ ID NOs: 2 – 5, more preferably SEQ ID NO: 2, or a complex according to the invention, for identifying a therapeutic binding protein is also provided herein.
  • a peptide or complex according to the invention can be used to identify therapeutic binding proteins. For example, as disclosed in the Examples, T cell receptors can be identified that may enable therapeutic properties.
  • the complex according to the invention is a complex comprising: a) a peptide comprising the amino acid sequence of SEQ ID NO: 1, and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1; optionally wherein the binding agent is an HLA-A*02 molecule, further optionally wherein the binding agent is an HLA-A*02:01 molecule.
  • the complex according to the invention is a complex comprising: a) a peptide comprising the amino acid sequence of any one of SEQ ID NOs: 2 - 21, and b) a binding agent that specifically binds to a peptide comprising the amino acid sequence of any one of the respective SEQ ID NOs: 2 - 21; optionally wherein the binding agent is an HLA-A*02 molecule, further optionally wherein the binding agent is an HLA-A*02:01 molecule.
  • the complex is: a.
  • VLAPRVLRA:HLA-A*02 complex a VLAPRVLRV:HLA-A*02 complex, a VLAPRVLRI:HLA- A*02 complex, a VLAPRVLRL:HLA-A*02 complex, a VLAPRVLRR:HLA-A*02 complex, a VLAPRVLRN:HLA-A*02 complex, a VLAPRVLRD:HLA-A*02 complex, a VLAPRVLRC:HLA-A*02 complex, a VLAPRVLRQ:HLA-A*02 complex, a VLAPRVLRE:HLA-A*02 complex, a VLAPRVLRG:HLA-A*02 complex, a VLAPRVLRH:HLA-A*02 complex, a VLAPRVLRK:HLA-A*02 complex, a VLAPRVLRM:HLA-A*02 complex, a VLAPRVLRF:HLA-A*02 complex,
  • VLAPRVLRA:HLA-A*02:01 complex a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, a VLAPRVLRL:HLA-A*02:01 complex, a VLAPRVLRR:HLA- A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, a VLAPRVLRD:HLA-A*02:01 complex, a VLAPRVLRC:HLA-A*02:01 complex, a VLAPRVLRQ:HLA-A*02:01 complex, a VLAPRVLRE:HLA-A*02:01 complex, a VLAPRVLRG:HLA-A*02:01 complex, a VLAPRVLRH:HLA-A*02:01 complex, a VLAPRVLRK:HLA-A*02:01 complex, a VLAPRVLRM
  • the use of a peptide or a complex according to the invention, wherein the therapeutic binding protein is capable of preventing or treating a pre-cancer, a cancer or a viral infection associated with impaired HLA class I antigen presentation.
  • the use of a peptide or a complex according to the invention, wherein the therapeutic binding protein comprises: a TCR, an antigen binding fragment of a TCR, or a chimeric antigen receptor (CAR), or an ImmTAC; or the therapeutic binding protein is an antibody.
  • a peptide or a complex according to the invention, wherein: (a) the peptide comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 2 - 5; or (b) the complex is selected from the group consisting of: a VLAPRVLRA:HLA-A*02 complex, a VLAPRVLRV:HLA-A*02 complex, a VLAPRVLRI:HLA-A*02 complex; or a VLAPRVLRL:HLA- A*02 complex.
  • a vaccine comprising the peptide according to the invention; and a modified cell transformed, transfected or transduced with a nucleic acid described herein, a nucleic acid composition described herein, a nucleic acid sequence described herein, or a vector described herein; for use as a medicament is also provided herein.
  • Administering to a subject in need thereof a vaccine comprising the peptide according to the invention and a modified cell described herein expressing a binding moiety that binds the peptide according to the invention is expected to be more effective for treatment of the subject.
  • the vaccine and/or the modified cell may be in the form of a pharmaceutical composition.
  • the vaccine may be administered to a subject in need thereof prior to, during, and/or after administration of the modified cell.
  • Any of the vaccines and modified cells described herein may suitably be used in the combination.
  • the binding moiety may suitably be expressed by the nucleic acid, nucleic acid composition, nucleic acid sequence, or vector described herein.
  • the binding moiety may be a binding protein, comprising for example a TCR described herein, an antigen binding fragment of a TCR described herein, a CAR described herein, or an ImmTAC described herein.
  • the nucleic acid, nucleic acid composition, nucleic acid sequence, or vector may encode a RCN1 antigen- specific binding protein comprising a TCR described herein, an antigen binding fragment of a TCR described herein, a CAR described herein, or an ImmTAC described herein.
  • the nucleic acid composition may encode a RCN1 antigen-specific binding protein having a TCR ⁇ chain variable (V ⁇ ) domain and a TCR ⁇ chain variable (V ⁇ ) domain, the composition comprising: a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence; and a nucleic acid sequence that encodes a TCR V ⁇ domain comprising a CDR3 amino acid sequence; wherein the CDR3 sequences together specifically bind to a peptide comprising the amino acid sequence of SEQ ID NO: 1.
  • the vaccine and the modified cell may be for use in the prevention or treatment of a pre- cancer, a cancer or a viral infection associated with impaired HLA class I antigen presentation in a human subject.
  • the pre-cancer, cancer, or viral infection may be a pre-cancer, cancer, or viral infection with impaired peptide processing machinery.
  • the pre-cancer, cancer, or viral infection may be associated with reduced or low TAP1 or TAP2 protein expression, activity, level, or stability.
  • RCN1 antigen or “RCN1 peptide antigen” or “RCN1 containing peptide antigen” refers to a naturally or synthetically produced peptide portion of a RCN1 protein ranging in length from about 8 amino acids, about 9 amino acids, about 10 amino acids, preferably up to about 35 amino acids, which can form a complex with a MHC (e.g., HLA) molecule, and a binding protein of this disclosure specific for a RCN1 peptide:MHC (e.g., HLA) complex can specifically bind to such as complex.
  • MHC e.g., HLA
  • the RCN1 peptide antigen comprises or consists of an amino acid sequence selected from the group consisting of: SEQ ID NO: 1 - 27.
  • the RCN1 peptide antigen:HLA complex typically comprises a peptide:HLA complex selected from the group consisting of: a VX 1 APRVLRX 2 :HLA-A*02:01 complex (wherein X 1 and X 2 are any amino acid), a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA- A*02:01 complex, a VLAPRVLRL:HLA-A*02:01 complex, a VLAPRVLRR:HLA-A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, VLAPRVLRD:HLA-A*02:01 complex, VLAPRVLRD:HLA-A*02:01 complex
  • the HLA-A*02:01 molecule may instead also be HLA-A*02, HLA-A*02:02, HLA-A*02:03, HLA- A*02:04, or HLA-A*02:09.
  • binding agent that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1 refers to a binding agent, that specifically binds to a peptide comprising the amino acid sequence of SEQ ID NO: 1 (or to a peptide antigen:HLA complex where the peptide comprises the amino acide sequence of SEQ ID NO: 1, e.g., on a cell surface), and does not bind a peptide sequence that does not include the peptide comprising the amino acid sequence of SEQ ID NO: 1.
  • the peptide comprises or consists of an amino acid sequence selected from the group consisting of: SEQ ID NO: 1 - 27, and the peptide antigen:HLA complex comprises a peptide:HLA complex selected from the group consisting of: a VX1APRVLRX2:HLA-A*02:01 complex (wherein X1 and X2 are any amino acid), a VLAPRVLRA:HLA-A*02:01 complex, a VLAPRVLRV:HLA-A*02:01 complex, a VLAPRVLRI:HLA-A*02:01 complex, a VLAPRVLRL:HLA-A*02:01 complex, a VLAPRVLRR:HLA- A*02:01 complex, a VLAPRVLRN:HLA-A*02:01 complex, a VLAPRVLRD:HLA-A*02:01 complex, a VLAPRVLRC:HLA-A*02:01 complex (wherein X1 and
  • the HLA-A*02:01 molecule may instead also be HLA-A*02, HLA-A*02:02, HLA-A*02:03, HLA-A*02:04, or HLA-A*02:09.
  • RCN1 antigen-specific binding protein refers to a protein or polypeptide, such as a TCR or CAR, that specifically binds to a RCN1 peptide antigen comprising the amino acid sequence of SEQ ID NO: 1 (or to a peptide antigen:HLA complex, where the peptide comprises the amino acid sequence of SEQ ID NO: 1 e.g., on a cell surface), and does not bind a peptide sequence that does not include the RCN1 peptide antigen comprising the amino acid sequence of SEQ ID NO: 1.
  • a RCN1-specific binding protein specifically binds to a RCN1 peptide antigen (or a RCN1 peptide antigen:HLA complex) with a Kd of less than about 10 -8 M, less than about 10 -9 M, less than about 10 -10 M, less than about 10 -11 M, less than about 10 -12 M, or less than about 10 -13 M, or with an affinity that is about the same as, at least about the same as, or is greater than at or about the affinity exhibited by an exemplary RCN1-specific binding protein provided herein, such as any of the RCN1-specific TCRs provided herein, for example, as measured by the same assay.
  • a RCN1-specific binding protein comprises a RCN1-specific immunoglobulin superfamily binding protein or binding portion thereof.
  • the selective binding may be in the context of RCN1 antigen presentation by HLA-A*02:01.
  • a binding protein that “specifically binds to a RCN1 antigen” may only do so when it is being presented (i.e. it is bound by) by a specific HLA or is in an equivalent structural formation as when it is being presented by the specific HLA.
  • a binding protein that “specifically binds to a RCN1 antigen” may only do so when it is being presented (i.e.
  • HLA-A*02:01 or is in an equivalent structural formation as when it is being presented by HLA-A*02:01.
  • a binding protein that “specifically binds to a RCN1 antigen”, may only do so when it is being presented (i.e. it is bound by) by HLA-A*02:02 or is in an equivalent structural formation as when it is being presented by HLA-A*02:02.
  • a binding protein that “specifically binds to a RCN1 antigen” may only do so when it is being presented (i.e. it is bound by) by HLA- A*02:03 or is in an equivalent structural formation as when it is being presented by HLA-A*02:03.
  • a binding protein that “specifically binds to a RCN1 antigen”, may only do so when it is being presented (i.e. it is bound by) by HLA-A*02:04 or is in an equivalent structural formation as when it is being presented by HLA-A*02:04.
  • a binding protein that “specifically binds to a RCN1 antigen”, may only do so when it is being presented (i.e. it is bound by) by HLA-A*02:09 or is in an equivalent structural formation as when it is being presented by HLA-A*02:09.
  • a binding protein that “specifically binds to a RCN1 antigen” may only do so when it is being presented (i.e.
  • HLA-A*02 is bound by or is in an equivalent structural formation as when it is being presented by HLA-A*02.
  • a T cell receptor or as it refers to a recombinant T cell receptor, nucleic acid fragment, variant, or analog, or a modified cell, such as, for example, the RCN1 T cell receptors, and RCN1-expressing modified cells herein, is meant that the T cell receptor, or fragment thereof, recognizes, or binds selectively to a RCN1 antigen comprising the amino acid sequence of SEQ ID NO: 1 (e.g. wherein the RCN1 antigen comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1 - 27).
  • the T cell receptor binds to a RCN1 antigen comprising the amino acid sequence of SEQ ID NO: 1 (e.g. an amino acid sequence selected from the group consisting of: SEQ ID NO: 1 - 27) and does not bind in a significant amount to other polypeptides not comprising the amino acid sequence of SEQ ID NO: 1.
  • the T cell receptor may bind to a RCN1 antigen (e.g. an amino acid sequence selected from the group consisting of: SEQ ID NO: 1 - 27) with at least 10, 100, or 1000, fold more affinity than to a control antigenic polypeptide.
  • This binding may also be determined indirectly in the context of a modified T cell that expresses a nucleic acid or vector of the invention (i.e. a CAR T cell or T cell expressing a TCR specific for the sequence of SEQ ID NO: 1).
  • the modified cell is specifically reactive against a cell presenting a peptide comprising the amino acid sequence of SEQ ID NO: 1.
  • the modified RCN1-TCR expressing T cell may bind to a RCN1 expressing cell with at least 10, 100, or 1000, fold more reactivity when compared to its reactivity against a control cell line that is not expressing RCN1.
  • binding agent for example a binding protein (e.g., TCR receptor) or a binding domain (or fusion protein thereof), to a target molecule with an affinity or K a (i.e., an equilibrium association constant of a particular binding interaction with units of 1/M) equal to or greater than 10 5 M -1 (which equals the ratio of the on-rate [k on ] to the off-rate [k off ] for this association reaction), while not significantly associating or uniting with any other molecules or components in a sample.
  • a binding protein e.g., TCR receptor
  • K a i.e., an equilibrium association constant of a particular binding interaction with units of 1/M
  • Binding proteins or binding domains may be classified as “high affinity” binding proteins or binding domains (or fusion proteins thereof) or as “low affinity” binding proteins or binding domains (or fusion proteins thereof).
  • "High affinity” binding proteins or binding domains refer to those binding proteins or binding domains having a K a of at least 10 7 M -1 , at least 10 8 M -1 , at least 10 9 M -1 , at least 10 10 M -1 , at least 10 11 M -1 , at least 10 12 M -1 , or at least 10 13 M -1 .
  • Low affinity binding proteins or binding domains refer to those binding proteins or binding domains having a K a of up to 10 7 M -1, up to 10 6 M -1 , up to 10 5 M -1 .
  • affinity can be defined as an equilibrium dissociation constant (K d ) of a particular binding interaction with units of M (e.g., 10 -5 M to 10 -13 M).
  • a receptor or binding domain may have "enhanced affinity,” which refers to selected or engineered receptors or binding domains with stronger binding to a target antigen than a wild type (or parent) binding domain.
  • enhanced affinity may be due to a Ka (equilibrium association constant) for the target antigen that is higher than the wild type binding domain, due to a Kd (dissociation constant) for the target antigen that is less than that of the wild type binding domain, due to an off-rate (koff) for the target antigen that is less than that of the wild type binding domain, or a combination thereof.
  • Ka Equilibrium association constant
  • Kd dissociation constant
  • koff off-rate
  • enhanced affinity TCRs can be codon optimized to enhance expression in a particular host cell, such as a cell of the immune system, a inducible pluripotent stem cell (iPSC), a hematopoietic stem cell, a T cell, a primary T cell, a T cell line, a NK cell, or a natural killer T cell (Scholten et al, Clin. Immunol.119: 135, 2006).
  • the T cell can be a CD4+ or a CD8+ T cell, or gamma-delta T cell.
  • a protein and a gene encoding said protein may be referred to using the same term.
  • non-essential amino acid residue is a residue that can be altered from the wild-type sequence of (e.g., the sequence identified by SEQ ID NO 1, 2, or 3 herein) without abolishing or, more preferably, without substantially altering a biological activity, whereas an “essential” (or “critical”) amino acid residue results in such a change.
  • amino acid residues that are conserved are predicted to be particularly non-amenable to alteration, except that amino acid residues within the hydrophobic core of domains can generally be replaced by other residues having approximately equivalent hydrophobicity without significantly altering activity.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • non-polar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • a nonessential (or non-critical) amino acid residue in a protein is preferably replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly, and the resultant mutants can be screened for activity to identify mutants that retain activity.
  • Calculations of sequence homology or identity (the terms are used interchangeably herein) between sequences are performed as follows. To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 75%, 80%, 82%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% of the length of the reference sequence.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the percent identity between two amino acid sequences is determined using the Needleman et al. (1970) J. Mol. Biol.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • a particularly preferred set of parameters are a BLOSUM 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of Meyers et al. (1989) CABIOS 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • nucleic acid and protein sequences described herein can be used as a “query sequence” to perform a search against public databases to, for example, identify other family members or related sequences.
  • Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol.215:403-410).
  • gapped BLAST can be utilized as described in Altschul et al. (1997, Nucl. Acids Res. 25:3389-3402).
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the polypeptides and nucleic acid molecules described herein can have amino acid sequences or nucleic acid sequences sufficiently or substantially identical to the sequences identified by SEQ ID NOs 1 to 109.
  • amino acid sequences or nucleic acid sequences having one or several (e.g. two, three, four etc) amino acid or nucleic acid substitutions compared to the corresponding sequences identified by a SEQ ID NO may be sufficiently or substantially identical to the sequences identified by the SEQ ID NO (provided that they retain the requisite functionality).
  • the one or several (e.g. two, three, four etc) amino acid or nucleic acid substitutions may be conservative substitutions.
  • amino acid or nucleotide sequences that contain a common structural domain having at least about 60%, or 65% identity, likely 75% identity, more likely 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity are defined herein as sufficiently or substantially identical.
  • TCR sequences are defined according to IMGT. See the LeFranc references herein for further details i.e. [1] Lefranc M.-P. "Unique database numbering system for immunogenetic analysis" Immunology Today, 18: 509 (1997). [2] Lefranc M.-P.
  • ex vivo refers to “outside” the body.
  • in vitro can be used to encompass “ex vivo” components, compositions and methods.
  • 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 pertains. For example, Singleton and Sainsbury, Dictionary of Microbiology and Molecular Biology, 2d Ed., John Wiley and Sons, NY (1994); and Hale and Marham, The Harper Collins Dictionary of Biology, Harper Perennial, NY (1991) provide those of skill in the art with a general dictionary of many of the terms used in the invention.
  • Example 1 MATERIAL AND METHODS Cell culture Tumor cells were cultured in IMDM medium (Gibco) supplemented with 100ug/mL streptomycin, 100 U/mL penicillin, 2mM L-glutamine (Invitrogen) and 8% FCS (Gibco). Genetic disruption of the TAP1, or RCN1 gene in human tumor cell lines was performed with CRISPR/CAS9 and described before (Marijt et al.2018).
  • single guide RNAs targeting exon 1 of the human TAP1 gene (sgTAP1: 5′-GCT GCT ACT TCT CGC CGA CT-3′ (SEQ ID NO: 29)), or targeting exon 1 of the RCN1 gene (RCN1 21-29 : 5′- TAGCTGTCAGGTCACCATTG-3′ (SEQ ID NO: 30)) were designed and cloned into the lentiCRI SPR v2 vector.
  • Virus particles were generated by co- transfecting sgRNA/CAS9 containing plasmid together with PAX2/pMD2.G packaging vectors into HEK293T cells using Lipofectamine 2000 (Thermo Fisher).
  • TAP1 KO efficiency was analyzed by measuring surface HLA-ABC (w6/32; BioLegend) expression using flow cytometry.
  • Polyclonal TAP KO cell lines were generated by FACS sorting the HLA-I low cell population.
  • CRISPR/CAS9 TAP-WT control cells were generated by FACS sorting the HLA-I high cell population of the polyclonal bulk. Clones were made of the TAP/RCN1 KO tumor cells by limited dilution.
  • T cells were cultured in IMDM medium (Gibco) supplemented with 2mM L-glutamine, 10% human serum (Sanquin), and 100U/mL IL-2 (proleukine, Novartis). T cells were stimulated every 10-14 days using synthetic short peptide (Genscript) or 800 ng/ml PHA (Phytohaemagglutinin) (Murex Biotech), supplemented with 100 U/ml IL-2 and IL-7 (5ng/mL), and a feeder mix containing irradiated PBMCs (1 ⁇ 10 6 cells, 50 Gy), and EBV-JY cells (1 ⁇ 10 5 cells, 75 Gy). All cell types were maintained in humidified air incubator at 37 o C and 5% CO2.
  • PBMCs Approximately 500 ⁇ 10 6 PBMCs were incubated with PE-labeled pHLA- A*02:01 tetramers (Flex-TTM HLA-A*02:01 Monomer UVX van Biolegend) for 30 min at RT.
  • Anti- PE magnetic beads were used to pull out pHLA-A*02:01 tetramer–positive cells over a MACS LS column as instructed by the manufacturer (Miltenyi Biotech). T cells were cultured in complete IMDM containing 10% human serum (Sanquin blood facility) and 100 U/ml IL-2 (Proleukine; Novartis).
  • T cells were stimulated every 2 weeks with a mixture of T cells (10 6 ), 1 ⁇ g/ml synthetic peptide (VLAPRVLRA (SEQ ID NO: 2) (RCN1)), irradiated PBMCs (10 6 cells, 50 Gray), and EBV-JY (10 5 cells, 75 Gray) in complete T cell culture medium supplemented with 100 U/ml IL-2 in 24-well plates (Costar). Culture medium was replenished every 2–3 d with fresh complete T cell medium.
  • Cross-presentation of synthetic long peptides by MoDC HLA-A*02:01 positive PBMCs were isolated from buffy-coats from consented donors (Sanquin bloodbank, Amsterdam), using a gradient ficoll layer.
  • PBMCs were incubated with anti-CD14 magnetic beads for 20 min at 4 °C and the CD14 positive monocytes were isolated using magnetic separation columns (miltenyi).
  • CD14+ monocyte were cultured in RPMI medium supplemented with 10% FCS, GM-CSF (800 units/ml), and IL-4 (500 units/ml) for 6 days to generate immature monocyte-derived dendritic cells.
  • the immature moDCs were incubated with synthetic long peptide (10 ⁇ M, Genscript) for 2h, followed by overnight incubation with LPS (20 ng/ml).
  • TCR sequencing results were analyzed using the iPair analyzer platform.
  • Full-length cDNA transcripts for murinized TCRs for both TCR-alpha and TCR-beta chains were cloned into a retroviral pMP71 flex expression vector.
  • Retrovirus production Platinum-Amphotropic retrovirus production (Plat-A) retroviral packing cells (Cell Biolabs) were used for retrovirus production.
  • Plat-A cells were seeded in 6-well plates and incubated overnight until fully attached. Next, the cells were transfected with 2.5 ⁇ g pMP71_TCR vector using 5 ⁇ l lipofectamine 3000 reagent (Thermofisher). Next day, medium was refreshed, and retrovirus supernatant was harvested after 24h .
  • retrovirus supernatant was spun down to remove cells and debris and subsequently incubated overnight at 4 °C with lenti-X concentrator (Clontech; 3 volumes retrovirus with 1 volume Lenti-X).
  • lenti-X concentrator Clontech; 3 volumes retrovirus with 1 volume Lenti-X.
  • supernatant was spun down for 45 minutes at 1500xg at 4 °C, after which the pellet was gently dissolved in 50 ⁇ l PBS (for each 6 well) and concentrated virus was either stored at -80 °C in single-use aliquots or directly used for T cell transduction.
  • T cell transduction and culture of TEIPP-specific T cell lines CD8+ T cells were purified from PBMC using magnetic bead isolation (Miltenyi), and activated by stimulating them with aCD3/aCD28 expander beads (Thermofisher) at a 3:1 T cell:bead ratio in IMDM+ 8% human serum (Sanquin; hereafter referred to as T cell medium) in the presence of 100 U/ml IL-2, 10 ng/ml IL-7 and 5 ng/ml IL-15.
  • T cell medium human serum
  • CD8 T cells were plated in a retronectin (Takara) coated 24-well in 800 ⁇ l T cell medium together with 50 ⁇ l concentrated retrovirus supernatant and 100 U/ml IL-2. Subsequently, CD8 T cells and retrovirus containing supernatant was spun down for 90min at 430xg at 24 °C to increase the efficiency of transduction.24h after transduction, CD8+ T cells were harvested, spun down for 5 minutes at 1600 rpm to remove the virus, and cells were resuspended in fresh T cell medium containing 100 U/ml IL-2. After 96 hours, transduction efficiency was determined by combined tetramer and murine TCRb antibody staining.
  • TCR transduced T cells were replenished every 2–3 d with fresh complete T cell medium until sufficient number (>5x10e6) of T cells were obtained for further TCR enrichment by TCRb-mediated MACS.
  • CD8+ T cells were incubated for 30 minutes at RT with 1:200 TCRb-PE antibody, washed, and subsequently incubated with anti-PE microbeads for an additional 20 minutes at 4°C.
  • TCRb+ cells were isolated by separation over two consecutive MS columns, and subsequently expanded using irradiated PBMC (50 Gray) and EBV-JY cells (75 Gray) in complete T cell medium supplemented with 800 ng/ml PHA (Murex Biotech) and 100 U/ml IL-2. Culture medium was replenished every 2–3 d with fresh complete T cell medium.
  • T cell reactivity analysis TCR-transduced or bulk-cultured TEIPP T cells were tested for their ability to selectively recognize the indicated TEIPP epitope presented on TAP impaired tumors by co-culturing them for 18 hours with wild-type (wt), TAP-KO and/or TAP/RCN1-KO tumor cells at a 5-to-1 target-to-T cell ratio, after which supernatants were harvested and analyzed for cytokine production by GM-CSF, CCL4 and/or IFNg ELISA. Unstimulated and peptide-stimulated T cells were used as negative and positive controls.
  • TEIPP antigen-expressing healthy cells such as Epstein-Barr virus (EBV) transformed B-cells, monocytes or monocyte-derived dendritic cells were included as additional controls.
  • Test were performed in triplo in 96 well round bottom plates at 1:1 target to effector ratio.
  • Functional T cell affinity analysis Functional T cell affinity was determined by measuring GM-CSF and CCL4 production of activated T cells recognizing TEIPP-peptide pulsed HLA-A02:01+ JY target cells.
  • JY target cells were pulsed with increasing concentrations of peptide (10 pg/ml – 10 ug/ml) in serum-free medium for 2 hours at 37°C, after which T cells were directly added in T cell medium and incubated overnight at 37°C. Next day, supernatant was harvested and GM-CSF and CCL4 production was determined by ELISA. For each TCR, the IC50 value was determined by calculating the peptide concentration corresponding to 50% cytokine production of the maximum response using Graphpad Prism non-linear regression with dose-response – inhibition equations (four-parameter dose-response curve).
  • MoDCs were stained with anti-CD1a (clone HI149, BD), anti-CD14 (clone M5E2, BD), anti-CD80 (clone L307.4, BD), anti-CD83 (clone HB15e, BD), anti-CD86 (clone IT2.2, biolegend), and anti HLA-DR (clone G46-6, BD) antibodies for 30min at 4°C and washed three times with cold PBS/BSA. Samples were acquired using a BD LSRFortessaTM flow cytometry system and analyzed using FlowJo software (Tree Star).
  • 36 peptides (designated 101 to 136) were selected for their capacity to activate CD8+ T cells present in peripheral blood mononuclear cells (PBMC) of maximal 5 healthy donors, following the protocol described earlier (Marijt et al., 2018).
  • PBMC peripheral blood mononuclear cells
  • the PBMC of at least 3 out of 5 donors showed in vitro expansion of HLA-A*02:01-specific peptide multimer-positive CD8+ T cells.
  • 18 of the 21 cases could be tested for their capacity to preferentially recognize TAP-negative tumor cells over TAP-positive tumor cells.
  • Peptide 113-specific T cells preferentially recognized 518A2 melanoma and 08.11 melanoma cells when TAP is knocked out ( Figure 2), but not non-tumor cells such as fibroblasts, immortalized Human Embryonic Kidney 293 (HEK-293T) cells or proximal tubular cells derived from a normal human adult male kidney (HK2), unless TAP was knocked out (HK2-TAPKO; Figure 2).
  • HEK-293T immortalized Human Embryonic Kidney 293
  • HK2-TAPKO proximal tubular cells derived from a normal human adult male kidney
  • peptide 113 specific CD8+ T cells preferentially recognize tumor cells displaying lower levels of TAP, and as such peptide 113 can be classified as a genuine TEIPP.
  • Peptide 113-specific CD8+ T cell cultures are RCN1-specific.
  • An important feature for the clinical application of TEIPP-specific T cells is the absence of cross- reactivity to peptides-derived from other proteins present in cells. Therefore, the gene encoding peptide 113 (RCN1) was knocked out in 518A2 TAPKO melanoma cells ( Figure 5).
  • the synthetic long peptide (SLP) platform is highly immunogenic and successfully applied in therapeutic SLP vaccination for HPV-induced cancers (Kenter et al.2009; van Poelgeest et al. 2016). Therefore, the inventors first assessed the efficiency of cross-presentation of long versions of the RCN1-derived signal peptide VLAPRVLRA (SEQ ID NO: 2) in dendritic cells, using natural flanking amino acids extending the amino-terminus, the carboxy-terminus, or both ends (Table 3).
  • Monocyte-derived dendritic cells were incubated with these three SLPs (P113 long 1- 3; SEQ ID NOs: 22-24) or no peptide as control, then matured, and used as targets for the p113- specific CD8 T cell culture to assess correct processing and HLA-A2 presentation of the minimal TEIPP epitope. None of the SLP variants were cross-presented to T cells, whereas exogenous pulsing of the short VLAPRVLRA (SEQ ID NO: 2) peptide did stimulate the T cells (Figure 7). These results suggested that cross-presentation of the VLAPRVLRA (SEQ ID NO: 2) epitope from its longer peptide stretch is not efficient and had to be optimized for vaccine applications.
  • the c-terminal amino-acid substitution is underlined.
  • Cross-presentation of long peptides by dendritic cells involves endocytosis, cytosolic cleavage of the SLP into short peptides by the proteasome, transport over the ER membrane by TAP and loading onto MHC-I molecules (van Hall & van der Burg, 2012; Rosalia et al. 2013).
  • HLA- A*02:01 the C-terminal alanine is not the most optimal anchor residue as shown for the predicted binding affinity, ranking and binding level of this peptide (Table 4).
  • V C-terminal valine
  • L leucine
  • I isoleucine
  • M methionine
  • V-peptide variant elicited a strong T cell response at limiting peptide concentrations, albeit that this was almost similar the I-variant peptide despite its stronger binding affinity.
  • substitution of alanine (A) to valine (V) at the C-terminus of the RCN1 VLAPRVLRA peptide allowed for proper TCR interaction and resulted in a 23-fold better T cell activation.
  • cross- presentation of the V-variant was evaluated as SLP extended with natural flanking amino acids (Table 3).
  • the inventors reported the identification of the HLA-A*02:01-restricted RCN121–29 CD8 T cell epitope, present in the signal sequence of the RCN1 protein.
  • CD8 T cells reactive to this peptide preferentially recognize tumor cells displaying lower levels of TAP.
  • the inventors single cell sorted CD8 T cells specifically binding to MHC multimers of HLA-A*02:01-molecules presenting the peptide RCN121–29 VLAPRVLRA from 3 different RCN121–29 -specific bulk T-cell cultures displaying preferentially recognition of tumor cells with low levels of TAP. Then the TCR- alpha and TCR-beta chains were DNA sequenced in order to identify a series of unique T-cell receptors.
  • TCR-alpha and TCR-beta chains of each respective identified TCR were cloned into a retroviral expression vector, which was used to transduce T cells, resulting in the generation of T cells expressing the respective TCRs for peptide RCN121–29 VLAPRVLRA.
  • the introduced genes contain the murine TCR-C ⁇ domain which enhances correct pairing of transgenic alpha and beta chains. These T cells were analyzed via flow cytometry. Expression of this domain is confirmed in TCR-transduced T cells after incubation with murine TCRb antibody and the specificity of the T cells after binding MHC multimers of HLA-A*02:01-molecules presenting the peptide RCN1 21–29 VLAPRVLRA ( Figure 9).
  • SEQ ID NO: 1 - VX1APRVLRX2 wherein X1 and X2 are any amino acid (i.e A or R or N or D or C or Q or E or G or H or I or L or K or M or F or P or O or S or U or T or W or Y or V).
  • HLA class I peptide-binding assay based on competition for binding to class I molecules on intact human B cells. Identification of conserved HIV-1 polymerase peptides binding to HLA-A*0301. 17. van der Burg SH et al. J Immunol. 1996 May 1;156(9):3308-14, Immunogenicity of peptides bound to MHC class I molecules depends on the MHC-peptide complex stability. 18. Ossendorp, F., Mengedé, E., Camps, M., Filius, R. and Melief, C. (1998).
  • Varypataki E., van der Maaden, K., Bouwstra, J., Ossendorp, F. and Jiskoot, W. (2015).
  • Masuko K., Wakita, D., Togashi, Y., Kita, T., Kitamura, H. and Nishimura, T. (2015).
  • H/K-HELP Artificially synthesized helper/killer-hybrid epitope long peptide
  • Zom GG Willems MMJHP, Khan S, van der Sluis TC, Kleinovink JW, Camps MGM, van der Marel GA, Filippov DV, Melief CJM, Ossendorp F. Novel TLR2-binding adjuvant induces enhanced T cell responses and tumor eradication. J Immunother Cancer.2018 Dec 12;6(1):146. 24. Zom GG, Welters MJ, Loof NM, Goedemans R, Lougheed S, Valentijn RR, Zandvliet ML, Meeuwenoord NJ, Melief CJ, de Gruijl TD, Van der Marel GA, Filippov DV, Ossendorp F, Van der Burg SH.
  • TLR2 ligand-synthetic long peptide conjugates effectively stimulate tumor-draining lymph node T cells of cervical cancer patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne de nouvelles séquences d'acide nucléique, des vecteurs, des cellules modifiées, des agents de liaison, des peptides et des compositions pharmaceutiques qui sont utiles en tant que médicament, par exemple dans la prévention ou le traitement du cancer ou d'infections virales associées à une présentation d'antigène HLA de classe I altérée. L'invention concerne également des procédés et des utilisations correspondants.
PCT/NL2024/050206 2023-04-21 2024-04-22 Néo-antigènes teipp dérivés de rcn1 et leurs utilisations Pending WO2024219973A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2024257340A AU2024257340A1 (en) 2023-04-21 2024-04-22 Rcn1-derived teipp neoantigens and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NL2034657 2023-04-21
NL2034657A NL2034657B1 (en) 2023-04-21 2023-04-21 RCN1-derived TEIPP neoantigens and uses thereof

Publications (1)

Publication Number Publication Date
WO2024219973A1 true WO2024219973A1 (fr) 2024-10-24

Family

ID=86851941

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2024/050206 Pending WO2024219973A1 (fr) 2023-04-21 2024-04-22 Néo-antigènes teipp dérivés de rcn1 et leurs utilisations

Country Status (3)

Country Link
AU (1) AU2024257340A1 (fr)
NL (1) NL2034657B1 (fr)
WO (1) WO2024219973A1 (fr)

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
US6750325B1 (en) 1989-12-21 2004-06-15 Celltech R&D Limited CD3 specific recombinant antibody
US20040202657A1 (en) 1992-03-24 2004-10-14 Btg International Limited Humanized anti-CD3 specific antibodies
WO2004106380A2 (fr) 2003-05-31 2004-12-09 Micromet Ag Molecules humaines de liaison a cd3 anti-humain
WO2007105954A1 (fr) * 2006-03-16 2007-09-20 Leiden University Medical Center Procédés d'identification d'épitopes de lymphocytes t associés à une compromission de la transformation de peptides et applications des épitopes identifiés
US7446191B2 (en) 2001-04-11 2008-11-04 City Of Hope DNA construct encoding CE7-specific chimeric T cell receptor
US7514537B2 (en) 2001-04-30 2009-04-07 City Of Hope Chimeric immunoreceptor useful in treating human gliomas
US20100065818A1 (en) 2008-02-22 2010-03-18 Kim Jae-Hyun Layers and patterns of nanowire or carbon nanotube using chemical self assembly and fabricating method in liquid crystal display device thereby
US20110236406A1 (en) 2004-06-17 2011-09-29 Medimmune, Llc Immunogenic compositions comprising hmgb1 polypeptides
WO2012048190A1 (fr) 2010-10-08 2012-04-12 Regents Of The University Of Minnesota Composition d'annexine ii et procédés
WO2013120073A1 (fr) 2012-02-09 2013-08-15 Av Therapeutics, Inc. Agonistes peptidiques synthétiques du récepteur 4 de type toll (tlr-4)
WO2014031687A1 (fr) 2012-08-20 2014-02-27 Jensen, Michael Procédé et compositions pour l'immunothérapie cellulaire
US8822647B2 (en) 2008-08-26 2014-09-02 City Of Hope Method and compositions using a chimeric antigen receptor for enhanced anti-tumor effector functioning of T cells
WO2016071758A1 (fr) 2014-11-03 2016-05-12 Leiden University Medical Center Récepteurs de cellules t dirigées contre bob1 et leurs utilisations
WO2016156230A1 (fr) * 2015-03-31 2016-10-06 Immatics Biotechnologies Gmbh Nouveaux peptides et combinaisons de peptides et d'échafaudages destinés à être utilisés en immunothérapie contre le carcinome hépatocellulaire (chc) et d'autres cancers
WO2017182395A1 (fr) * 2016-04-21 2017-10-26 Immatics Biotechnologies Gmbh Immunothérapie contre le mélanome et d'autres cancers
WO2019231326A1 (fr) 2018-05-31 2019-12-05 ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n.LUMC) Néoantigènes teipp et leurs utilisations
WO2021107775A1 (fr) 2019-11-29 2021-06-03 ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC) Variant peptidique teipp et utilisations de celui-ci

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6750325B1 (en) 1989-12-21 2004-06-15 Celltech R&D Limited CD3 specific recombinant antibody
US20040202657A1 (en) 1992-03-24 2004-10-14 Btg International Limited Humanized anti-CD3 specific antibodies
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
US7446191B2 (en) 2001-04-11 2008-11-04 City Of Hope DNA construct encoding CE7-specific chimeric T cell receptor
US7514537B2 (en) 2001-04-30 2009-04-07 City Of Hope Chimeric immunoreceptor useful in treating human gliomas
WO2004106380A2 (fr) 2003-05-31 2004-12-09 Micromet Ag Molecules humaines de liaison a cd3 anti-humain
US20110236406A1 (en) 2004-06-17 2011-09-29 Medimmune, Llc Immunogenic compositions comprising hmgb1 polypeptides
WO2007105954A1 (fr) * 2006-03-16 2007-09-20 Leiden University Medical Center Procédés d'identification d'épitopes de lymphocytes t associés à une compromission de la transformation de peptides et applications des épitopes identifiés
US20090220534A1 (en) 2006-03-16 2009-09-03 Leiden University Medical Center Methods for identifying t-cell epitopes associated with impaired peptide processing and applications of the identified epitopes
US20100065818A1 (en) 2008-02-22 2010-03-18 Kim Jae-Hyun Layers and patterns of nanowire or carbon nanotube using chemical self assembly and fabricating method in liquid crystal display device thereby
US8822647B2 (en) 2008-08-26 2014-09-02 City Of Hope Method and compositions using a chimeric antigen receptor for enhanced anti-tumor effector functioning of T cells
WO2012048190A1 (fr) 2010-10-08 2012-04-12 Regents Of The University Of Minnesota Composition d'annexine ii et procédés
WO2013120073A1 (fr) 2012-02-09 2013-08-15 Av Therapeutics, Inc. Agonistes peptidiques synthétiques du récepteur 4 de type toll (tlr-4)
WO2014031687A1 (fr) 2012-08-20 2014-02-27 Jensen, Michael Procédé et compositions pour l'immunothérapie cellulaire
WO2016071758A1 (fr) 2014-11-03 2016-05-12 Leiden University Medical Center Récepteurs de cellules t dirigées contre bob1 et leurs utilisations
WO2016156230A1 (fr) * 2015-03-31 2016-10-06 Immatics Biotechnologies Gmbh Nouveaux peptides et combinaisons de peptides et d'échafaudages destinés à être utilisés en immunothérapie contre le carcinome hépatocellulaire (chc) et d'autres cancers
WO2017182395A1 (fr) * 2016-04-21 2017-10-26 Immatics Biotechnologies Gmbh Immunothérapie contre le mélanome et d'autres cancers
WO2019231326A1 (fr) 2018-05-31 2019-12-05 ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n.LUMC) Néoantigènes teipp et leurs utilisations
WO2021107775A1 (fr) 2019-11-29 2021-06-03 ACADEMISCH ZIEKENHUIS LEIDEN (h.o.d.n. LUMC) Variant peptidique teipp et utilisations de celui-ci

Non-Patent Citations (72)

* Cited by examiner, † Cited by third party
Title
A. DURGEAUF. EL HAGEI. VERGNONP. VALIDIREV. DE MONTPREVILLEB. BESSEJ. SORIAT. VAN HALLF. MAMI-CHOUAIB: "Different Expression Levels of the TAP Peptide Transporter Lead to Recognition of Different Antigenic Peptides by Tumor-Specific CTL", J IMMUNOL, vol. 187, no. 11, 1 December 2011 (2011-12-01), pages 5532 - 5539
A.L. SZYMCZAK ET AL., NATURE BIOTECHNOLOGY, vol. 22, 2004, pages 589 - 594
AKIRA SUEMATSU STAKEUCHI O: "Pathogen recognition and innate immunity", CELL, vol. 124, no. 4, 24 February 2006 (2006-02-24), pages 783 - 801, XP003018351, DOI: 10.1016/j.cell.2006.02.015
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCL. ACIDS RES., vol. 25, 1997, pages 3389 - 3402
ANDREAS O WEINZIERL ET AL: "Features of TAP-independent MHC class I ligands revealed by quantitative mass spectrometry", EUROPEAN JOURNAL OF IMMUNOLOGY, WILEY-VCH, HOBOKEN, USA, vol. 38, no. 6, 29 April 2008 (2008-04-29), pages 1503 - 1510, XP071223672, ISSN: 0014-2980, DOI: 10.1002/EJI.200838136 *
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1987, JOHN WILEY AND SONS, INC.
BARBALAT RLAU LLOCKSLEY R MBARTON G M: "Toll-like receptor 2 on inflammatory monocytes induces type I interferon in response to viral but not bacterial ligands", NAT IMMUNOL., vol. 10, no. 11, 2009, pages 1200 - 7, XP037924647, DOI: 10.1038/ni.1792
BEYRANVAND NEJAD, E.VAN DER SLUIS, T.VAN DUIKEREN, S.YAGITA, H.JANSSEN, G.VAN VEELEN, P.MELIEF, C.VAN DER BURG, S.ARENS, R.: "Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells", CANCER RESEARCH, vol. 76, no. 20, 2016, pages 6017 - 6029, XP055537015, DOI: 10.1158/0008-5472.CAN-16-0881
BIJKER, M. S. ET AL.: "CD8+ CTL priming by exact peptide epitopes in incomplete Freund's adjuvant induces a vanishing CTL response, whereas long peptides induce sustained CTL reactivity", J IMMUNOL, vol. 179, 2007, pages 5033 - 5040, XP055798792, DOI: 10.4049/jimmunol.179.8.5033
BOWERMAN ET AL., MOL. IMMUNOL, vol. 5, no. 15, 2009, pages 3000
BRENTJENS ET AL., CANCER RES., vol. 73, 2007, pages 5426
BRYANT, C. E.SPRING, D. R. ET AL.: "The molecular basis of the host response to lipopolysaccharide", NAT. REV. MICROBIOL., vol. 8, no. 1, 2010, pages 8 - 14
CHEN, X.LI, D.ZHONG, X.CHEN, B.DUAN, Z.WEN, J.: "Induction of multiple cytotoxic T lymphocyte responses in mice by a multiepitope DNA vaccine against dengue virus serotype 1", MICROBIOLOGY AND IMMUNOLOGY, vol. 60, no. 12, 2016, pages 835 - 845
COHEN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 69, 1972, pages 2110
DOORDUIJN, E.M. ET AL.: "T cells specific for a TAP-independent self-peptide remain naive in tumor-bearing mice and are fully exploitable for therapy", ONCOIMMUNOLOGY, vol. 7, 2018, pages e1382793, XP055546937, DOI: 10.1080/2162402X.2017.1382793
DOORDUIJN, E.M. ET AL.: "TAP-independent self-peptides enhance T cell recognition of immune-escaped tumors", J CLIN INVEST, vol. 126, 2016, pages 784 - 794
GARRIDO, F.APTSIAURI, N.DOORDUIJN, E.M.GARCIA LORA, A.M.VAN HALL: "T. The urgent need to recover MHC class I in cancers for effective immunotherapy", CURR OPIN IMMUNOL, vol. 39, 2016, pages 44 - 51, XP029463514, DOI: 10.1016/j.coi.2015.12.007
GAY, N.GANGLOFF, M.: "Structure and function of Toll receptors and their ligands", ANNU. REV. BIOCHEM., vol. 76, 2007, pages 141 - 165, XP009128143, DOI: 10.1146/annurev.biochem.76.060305.151318
GOVERS ET AL., TRENDS MOL. MED., vol. 16, no. 2, 2010, pages 11
HALEMARHAM: "The Harper Collins Dictionary of Biology", 1991, HARPER PERENNIAL
HARRISKRANZ, TRENDS PHARMACOL. SCI., vol. 37, no. 3, 2016, pages 220
JOHNNSON, B. ET AL., ANAL. BIOCHEM., vol. 198, 1991, pages 268 - 277
JOHNSSON, B. ET AL., J. MOL. RECOGNIT., vol. 8, 1995, pages 125 - 131
JONES BS ET AL.: "Improving the safety of cell therapy products by suicide gene transfer", FRONT PHARMACOL, vol. 5, 2014, pages 254
JONSSON, U. ET AL., ANN. BIOL. CLIN., vol. 51, 1993, pages 19 - 26
JONSSON, U. ET AL., BIOTECHNIQUES, vol. 11, 1991, pages 620 - 627
KENTER, G. G. ET AL.: "Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia", N ENGL J MED, vol. 361, 2009, pages 1838 - 1847, XP008157144, DOI: 10.1056/NEJMoa0810097
KORKOLOPOULOU P., L. KAKLAMANIS, F. PEZZELLA, A. L. HARRIS, K. C. GATTER: "Loss of antigen-presenting molecules (MHC class I and TAP-1) in lung cancer", BR. J. CANCER, vol. 73, 1996, pages 148 - 153, XP093140128
KUMAR HKAWAI TAKIRA S: "Toll-like receptors and innate immunity", BIOCHEM BIOPHYS RES COMMUN, vol. 388, no. 4, 30 October 2009 (2009-10-30), pages 621 - 5, XP026601489, DOI: 10.1016/j.bbrc.2009.08.062
LEFRANC M.-P. ET AL.: "IMGT unique numbering for immunoglobulin and T cell receptor constant domains and Ig superfamily C-like domains", DEV. COMP. IMMUNOL., vol. 29, 2005, pages 185 - 203, XP093138816, DOI: 10.1016/j.dci.2004.07.003
LEFRANC M.-P. ET AL.: "IMGT unique numbering for immunoglobulin and Tcell receptor variable domains and Ig superfamily V-like domains", DEV. COMP. IMMUNOL., vol. 27, 2003, pages 55 - 77
LEFRANC M.-P.: "The IMGT unique numbering for immunoglobulins, T cell Receptors and Ig-like domains", THE IMMUNOLOGIST, vol. 7, 1999, pages 132 - 136
LEFRANC M.-P.: "Unique database numbering system for immunogenetic analysis", IMMUNOLOGY TODAY, vol. 18, 1997, pages 509, XP004093509, DOI: 10.1016/S0167-5699(97)01163-8
LOU Y.T. Z. VITALISG. BASHAB. CAIS. S. CHENK. B. CHOIA. P. JEFFRIESW. M. ELLIOTTD. ATKINSB. SELIGER: "Restoration of the expression of transporters associated with antigen processing in lung carcinoma increases tumor-specific immune responses and survival", CANCER RES., vol. 65, 2005, pages 7926 - 7933, XP002714282, DOI: 10.1158/0008-5472.CAN-04-3977
LUCHANSKY ET AL., MOL. MICROBIOL., vol. 2, 1988, pages 637 - 646
LV COREN ET AL., BIOTECHNIQUES, 2015
M. F. DEL GUERCIO ET AL., J. IMMUNOL., vol. 154, 1995, pages 685 - 693
MARIJT, K. A. ET AL.: "Identification of non-mutated neoantigens presented by TAP-deficient tumors", J EXP MED, vol. 215, 2018, pages 2325 - 2337, XP009510574, DOI: 10.1084/jem.20180577
MASUKO, K.WAKITA, D.TOGASHI, Y.KITA, T.KITAMURA, H.NISHIMURA, T: "Artificially synthesized helper/killer-hybrid epitope long peptide (H/K-HELP): Preparation and immunological analysis of vaccine efficacy", IMMUNOLOGY LETTERS, vol. 163, no. 1, 2015, pages 102 - 112, XP055331195, DOI: 10.1016/j.imlet.2014.11.016
MEYER ET AL., CABIOS, vol. 4, 1989, pages 11 - 17
MONIE, T. P.BRYANT, C. E. ET AL.: "Activating immunity: Lessons from the TLRs and NLRs", TRENDS BIOCHEM. SCI., vol. 34, no. 11, 2009, pages 553 - 561, XP026736003, DOI: 10.1016/j.tibs.2009.06.011
MONJEZI R ET AL.: "Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors", LEUKEMIA, vol. 31, no. 1, 2016, pages 186 - 194, XP037653748, DOI: 10.1038/leu.2016.180
NEEDLEMAN ET AL., J. MOL. BIOL., vol. 48, 1970, pages 444 - 453
OSSENDORP, F.MENGEDE, E.CAMPS, M.FILIUS, R.MELIEF, C.: "Specific T Helper Cell Requirement for Optimal Induction of Cytotoxic T Lymphocytes against Major Histocompatibility Complex Class II Negative Tumors", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 187, no. 5, 1998, pages 693 - 702, XP002173925, DOI: 10.1084/jem.187.5.693
RA WILLEMSEN ET AL., GENE THERAPY, 2000
RAHIMIAN SFRANSEN MFKLEINOVINK JWCHRISTENSEN JRAMIDI MHENNINK WEOSSENDORP F: "Polymeric nanoparticles for co-delivery of synthetic long peptide antigen and poly IC as therapeutic cancer vaccine formulation", J CONTROL RELEASE, vol. 203, 10 April 2015 (2015-04-10), pages 16 - 22, XP029149030, DOI: 10.1016/j.jconrel.2015.02.006
RESSING ME ET AL.: "Differential binding of viral peptides to HLA-A2 alleles. Implications for human papillomavirus type 16 E7 peptide-based vaccination against cervical carcinoma", EUR J IMMUNOL., vol. 29, no. 4, April 1999 (1999-04-01), pages 1292 - 303
RITTER, C. ET AL.: "Epigenetic priming restores the HLA class-I antigen processing machinery expression in Merkel cell carcinoma", SCI REP, vol. 7, 2017, pages 2290
ROBBINS, P.F. ET AL.: "Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells.", NAT MED, vol. 19, 2013, pages 747 - 752, XP055161457, DOI: 10.1038/nm.3161
ROSALIA, R. A. ET AL.: "Dendritic cells process synthetic long peptides better than whole protein, improving antigen presentation and T-cell activation", EUR J IMMUNOL, vol. 43, 2013, pages 2554 - 2565, XP055469177, DOI: 10.1002/eji.201343324
ROTH ET AL., NATURE, vol. 559, 2018, pages 405
S JONES ET AL., HUMAN GENE THER, 2009
SADELAIN ET AL., CANCER DISCOV., vol. 3, no. 4, 2013, pages 388
SAMBROOK ET AL.: "A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY, article "Molecular Cloning"
SCHOLTEN ET AL., CLIN. IMMUNOL., vol. 119, 2006, pages 135
SCHUMACHER, T.N.SCHREIBER, R.D.: "Neoantigens in cancer immunotherapy.", SCIENCE, vol. 348, 2015, pages 69 - 74, XP055866872, DOI: 10.1126/science.aaa4971
SEIDEL, U.J.OLIVEIRA, C.C.LAMPEN, M.H.HALL, T.: "A novel category of antigens enabling CTL immunity to tumor escape variants: Cinderella antigens", CANCER IMMUNOL IMMUNOTHER, vol. 61, 2012, pages 119 - 125, XP019995543, DOI: 10.1007/s00262-011-1160-x
SETIADI, A.F. ET AL.: "Epigenetic control of the immune escape mechanisms in malignant carcinomas", MOL CELL BIOL, vol. 27, 2007, pages 7886 - 7894, XP008110261, DOI: 10.1128/MCB.01547-07
SINGLETONSAINSBURY: "Dictionary of Microbiology and Molecular Biology", 1994, JOHN WILEY AND SONS
SPOHN, R.BUWITT-BECKMANN, U. ET AL.: "Synthetic lipopeptide adjuvants and Toll-like receptor 2-Structure-activity relationships", VACCINE, vol. 22, no. 19, 2004, pages 2494 - 2499, XP004515469, DOI: 10.1016/j.vaccine.2003.11.074
STONE ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 63, no. 11, 2014, pages 1163
TOES, R. E.OFFRINGA, R.BLOM, R. J.MELIEF, C. J.KAST, W. M.: "Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction", PROC NATL ACAD SCI U S A, vol. 93, 1996, pages 7855 - 7860, XP001038340, DOI: 10.1073/pnas.93.15.7855
VAN DER BURG SH ET AL., HUM IMMUNOL., vol. 44, no. 4, December 1995 (1995-12-01), pages 189 - 98
VAN DER BURG SH ET AL.: "Immunogenicity of peptides bound to MHC class I molecules depends on the MHC-peptide complex stability", J IMMUNOL., vol. 156, no. 9, 1 May 1996 (1996-05-01), pages 3308 - 14
VAN HALL T, VAN DER BURG SH: "Mechanisms of peptide vaccination in mouse models: tolerance, immunity, and hyperreactivity", ADV IMMUNOL., vol. 114, 2012, pages 51 - 76
VAN HALL, T. ET AL.: "Selective cytotoxic T-lymphocyte targeting of tumor immune escape variants", NAT MED, vol. 12, 2006, pages 417 - 424
VAN POELGEEST MI ET AL.: "Vaccination against Oncoproteins of HPV16 for Noninvasive Vulvar/Vaginal Lesions: Lesion Clearance Is Related to the Strength of the T-Cell Response", CLIN CANCER RES., vol. 22, no. 10, 15 May 2016 (2016-05-15), pages 2342 - 50, XP055604485, DOI: 10.1158/1078-0432.CCR-15-2594
VARYPATAKI, E.VAN DER MAADEN, K.BOUWSTRA, J.OSSENDORP, F.JISKOOT, W.: "Cationic Liposomes Loaded with a Synthetic Long Peptide and Poly(l:C): a Defined Adjuvanted Vaccine for Induction of Antigen-Specific T Cell Cytotoxicity", THE AAPS JOURNAL, vol. 17, no. 1, 2015, pages 216 - 226, XP035420757, DOI: 10.1208/s12248-014-9686-4
WEINZIERL, A.O. ET AL.: "Features of TAP-independent MHC class I ligands revealed by quantitative mass spectrometry", EUR. J. IMMUNOL., vol. 38, 2008, pages 1503 - 1510
ZOM GGWELTERS MJLOOF NMGOEDEMANS RLOUGHEED SVALENTIJN RRZANDVLIET MLMEEUWENOORD NJMELIEF CJDE GRUIJL TD: "TLR2 ligand-synthetic long peptide conjugates effectively stimulate tumor-draining lymph node T cells of cervical cancer patients", ONCOTARGET, vol. 7, no. 41, 11 October 2016 (2016-10-11), pages 67087 - 67100
ZOM GGWILLEMS MMJHPKHAN SVAN DER SLUIS TCKLEINOVINK JWCAMPS MGMVAN DER MAREL GAFILIPPOV DVMELIEF CJMOSSENDORP F: "Novel TLR2-binding adjuvant induces enhanced T cell responses and tumor eradication", J IMMUNOTHER CANCER, vol. 6, no. 1, 12 December 2018 (2018-12-12), pages 146, XP021263809, DOI: 10.1186/s40425-018-0455-2

Also Published As

Publication number Publication date
AU2024257340A1 (en) 2025-11-13
NL2034657B1 (en) 2024-10-28

Similar Documents

Publication Publication Date Title
AU2022200745B2 (en) Novel peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
TWI730306B (zh) 用於抗cll及其他癌症之免疫治療的新穎胜肽及胜肽組合物
TWI670282B (zh) 治療幾種血液腫瘤特別是慢性淋巴白血病(cll)的新型免疫療法
KR20230004508A (ko) 코로나바이러스 백신 및 사용 방법
KR102171794B1 (ko) 헬퍼 t 세포의 활성화 방법
TW201841937A (zh) 用於白血病和其他癌症免疫治療的新穎肽和肽組合物
JP2003521245A (ja) ペプチドおよび核酸組成物を使用する、前立腺癌抗原に対する細胞性免疫応答の誘導
US20230035037A1 (en) Teipp peptide variant and uses thereof
NL2034657B1 (en) RCN1-derived TEIPP neoantigens and uses thereof
NL2034658B1 (en) TIMP3-derived TEIPP neoantigens and uses thereof
JP7785355B2 (ja) Teippペプチド変異体およびその使用

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24725025

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: AU2024257340

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2024257340

Country of ref document: AU

Date of ref document: 20240422

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2024725025

Country of ref document: EP