AU2013202288B2 - High affinity HIV T cell receptors - Google Patents

High affinity HIV T cell receptors Download PDF

Info

Publication number
AU2013202288B2
AU2013202288B2 AU2013202288A AU2013202288A AU2013202288B2 AU 2013202288 B2 AU2013202288 B2 AU 2013202288B2 AU 2013202288 A AU2013202288 A AU 2013202288A AU 2013202288 A AU2013202288 A AU 2013202288A AU 2013202288 B2 AU2013202288 B2 AU 2013202288B2
Authority
AU
Australia
Prior art keywords
tcr
tcrs
chain
cells
hla
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.)
Ceased
Application number
AU2013202288A
Other versions
AU2013202288A1 (en
Inventor
Steven Mark Dunn
Bent Karsten Jakobsen
Yi Li
Peter Eamon Molloy
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.)
Immunocore Ltd
Adaptimmune Ltd
Original Assignee
Adaptimmune Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2012211503A external-priority patent/AU2012211503B2/en
Application filed by Adaptimmune Ltd filed Critical Adaptimmune Ltd
Priority to AU2013202288A priority Critical patent/AU2013202288B2/en
Publication of AU2013202288A1 publication Critical patent/AU2013202288A1/en
Assigned to IMMUNOCORE LIMITED, ADAPTIMMUNE LIMITED reassignment IMMUNOCORE LIMITED Request for Assignment Assignors: IMMUNOCORE LIMITED
Application granted granted Critical
Publication of AU2013202288B2 publication Critical patent/AU2013202288B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

C:WRPortbrDCC\AM 4527388.l.DOC-9/0/2012 The present invention provides TCRs having an affinity (KD) of less than or equal to IpM, and/or an off-rate (koff) of 1x10 3 S~1 or slower, for the SLYNTVATL-HLA- A* 0201 complex provided that when the said TCR is presented by cell and comprises SEQ ID NOs: 1 and 2, the cell is not a native T cell. Such TCRs are useful, either alone or associated with a therapeutic agent, for targeting HIV infected cells presenting that complex.

Description

H MVntenvoevn\NRPortbl\DCCAhM 5022575 1. do-28/03/2013 1 High Affinity HIV T cell receptors The present invention relates to T-cell receptors (TCRs) having the property of binding to HIV Gag polypeptide-derived SLYNTVATL-HLA-A*0201. The TCRs comprise at least one TCR a chain variable domain and/or at least one TCR P chain variable domain and have a KD for the said SLYNTVATL-HLA-A*0201 complex of less than or equal to 1 sM and/or has an off-rate (kof) for the SLYNTVATL-HLA A*0201 complex of 1x10 3 S"or slower. This is a divisional of Australian patent application No. 2012211503, which in turn is a divisional of 2006228308, the entire content of which is incorporated herein by reference. Background to the Invention The Human Immuno-deficiency Virus (HIV) is the causative agent of Acquired Immuno-deficiency Disease Syndrome (AIDS). The virus is an enveloped retrovirus belonging to the lentivirus group. The SLYNTVATL (SEQ ID NO: 16) peptide is derived from the g17 gene product of the Gag gene, one of nine genes which make up the Human Immuno-deficiency Virus-1 (HIV-1) The peptide is loaded by HLA A* 0201 and presented on the surface of HIV infected cells. Therefore, the SLYNTVATL-HLA-A2*0201 complex provides an HIV marker that TCRs can target, for example for the purpose of delivering cytotoxic or immuno-stimulatory agents to the infected cells. However, for that purpose it would be desirable if the TCR had a high affinity and/or a slow off-rate for the peptide-HLA complex. Brief Description of the Invention This invention makes available for the first time TCRs having an affinity (KD) of less than or equal to 1 pM, and/or an off-rate (koff) of 1xiO SI or slower, for the SLYNTVATL-HLA-A*0201 complex PROVIDED THAT when the said TCR is presented by cell and comprises SEQ ID NOs: 1 and 2, the cell is not a native T cell. Such TCRs are useful, either alone or associated with a therapeutic agent, for targeting HIV infected cells presenting that complex 2 Detailed Description of the Invention The present invention provides a T-cell receptor (TCR) having the property of binding to SLYNTVATL-HLA-A*0201 and comprising at least one TCR a chain variable domain and/or at least one TCR p chain variable domain CHARACTERISED IN 5 THAT said TCR has a KD for the said SLYNTVATL-HLA-A*0201 complex of less than or equal to 1pM and/or has an off-rate (kff) for the SLYNTVATL-HLA-A*0201 complex of x10- 3 S'or slower, PROVIDED THAT when the said TCR is presented by cell and comprises SEQ ID NOs: 1 and 2, the cell is not a native T cell. The KD and/or (koff) measurement can be made by any of the known methods. A 10 preferred method is the Surface Plasmon Resonance (Biacore) method of Example 4. For comparison, the interaction of a disulfide-linked soluble variant of the parental HIV gag TCR (see SEQ ID NO: 9 for TCR a chain and SEQ ID NO: 10 for TCR s chain) and the SLYNTVATL-HLA-A*0201 complex has a KD of approximately 15 85nM and an off-rate (koff) of 2.21 x 10-2 S- as measured by the Biacore-base method of Example 4. The parental HIV Gag TCR specific for the SLYNTVATL-HLA-A*0201 complex has the following Valpha chain and Vbeta chain gene usage: 20 Alpha chain - TRAV12.2 Beta chain: - TRBV 5.6 The parental HIV Gag TCR can be used as a template from which other TCRs of the invention with high affinity and/or a slow off-rate for the interaction between said 25 TCRs and the SLYNTVATL-HLA-A*0201 complex can be produced. Thus the invention includes TCRs which are mutated relative to the parental HIV Gag TCR a chain variable domain (see Figure la and SEQ ID No: 1) and/or s chain variable domain (see Figure lb and SEQ ID NO: 2) in at least one complementarity determining region (CDR) and/or variable domain framework region thereof. It is also 30 contemplated that other hypervariable regions in the variable domains of the TCRs of 3 the invention, such as the hypervariable 4 (HV4) regions, may be mutated within a high affinity mutant TCR. Phage display provides one means by which libraries of TCR variants can be 5 generated. Methods suitable for the phage display and subsequent screening of libraries of TCR variants each containing a non-native disulfide interchain bond are detailed in (Li et al., (2005) Nature Biotech 23 (3): 349-354) and WO 2004/04404. Native TCRs exist in heterodimeric ap or y8 forms. However, recombinant TCRs 10 consisting of a single TCR a or TCR P chain have previously been shown to bind to peptide MHC molecules. In one embodiment the TCR of the invention comprise both an a chain variable domain and an TCR P chain variable domain. 15 As will be obvious to those skilled in the art the mutation(s) in the TCR a chain sequence and/or TCR P chain sequence may be one or more of substitution(s), deletion(s) or insertion(s). These mutations can be carried out using any appropriate method including, but not limited to, those based on polymerase chain reaction (PCR), 20 restriction enzyme-based cloning, or ligation independent cloning (LIC) procedures. These methods are detailed in many of the standard molecular biology texts. For further details regarding polymerase chain reaction (PCR) mutagenesis and restriction enzyme-based cloning see (Sambrook & Russell, (2001) Molecular Cloning - A Laboratory Manual ( 3 rd Ed.) CSHL Press) Further information on LIC procedures can 25 be found in (Rashtchian, (1995) Curr Opin Biotechnol 6 (1): 30-6) It should be noted that any as TCR that comprises similar Valpha and Vbeta gene usage and therefore amino acid sequence to that of the HIV Gag TCR could make a convenient template TCR. It would then be possible to introduce into the DNA 30 encoding one or both of the variable domains of the template as TCR the changes required to produce the mutated high affinity TCRs of the invention. As will be 4 obvious to those skilled in the art, the necessary mutations could be introduced by a number of methods, for example site-directed mutagenesis. The TCRs of the invention include those in which one or more of the TCR alpha chain 5 variable domain amino acids corresponding to those listed below are mutated relative to the amino acid occurring at these positions in the sequence provided for the parental HIV Gag TCR alpha chain variable domain in Figure la and SEQ ID No: 1. Unless stated to the contrary, the TCR amino acid sequences herein are generally 10 provided including an N-terminal methionine (Met or M) residue. As will be known to those skilled in the art this residue may be removed during the production of recombinant proteins. As will also be obvious to those skilled in the art, it may be possible to truncate the sequences provided at the C-terminus and/or N-terminus thereof, by 1, 2, 3, 4, 5 or more residues, without substantially affecting the pMHC 15 binding characteristics of the TCR, all such trivial variants are encompassed by the present invention. As used herein the term "variable region" is understood to encompass all amino acids of a given TCR which are not included within the constant domain as encoded by the 20 TRAC gene for TCR a chains and either the TRBC 1 or TRBC2 genes for TCR p chains. (T cell receptor Factsbook, (2001) LeFranc and LeFranc, Academic Press, ISBN 0-12-441352-8) As used herein the term "variable domain" is understood to encompass all amino acids 25 of a given TCR which are encoded by a TRAV gene for TCR a chains and a TRBV gene for TCR p chains. (T cell receptor Factsbook, (2001) LeFranc and LeFranc, Academic Press, ISBN 0-12-441352-8) As is known to those skilled in the art, part of the diversity of the TCR repertoire is 30 due to variations which occur in the amino acid encoded by the codon at the boundary between the variable region, as defined herein, and the constant domain. For example, 5 the codon that is present at this boundary in the parental HIV Gag TCR sequence results in the presence of the Histidine (H) residue at the C-terminal of the variable region sequences herein. This Histidine replaces the N-terminal Asparagine (N) residue encoded by the TRAC gene shown in Figure 8a. 5 Embodiments of the invention include mutated TCRs which comprise mutation of one or more of alpha chain variable region amino acids corresponding to: 95T, 96N, 97S, 98G, and 1OA, for example the amino acids: 10 95S or G 96A 97H 98D 100S 15 The numbering used above is the same as that shown in Figure 1 a and SEQ ID No: 1 Embodiments of the invention also include TCRs which comprise mutation of one or more of the TCR beta chain variable region amino acids corresponding to those listed 20 below, are relative to the amino acid occurring at these positions in the sequence provided for the native HIV Gag TCR alpha chain variable region of the native HIV Gag TCR beta chain in Figure lb and SEQ ID No: 2. The amino acids referred to which may be mutated are: 51Y, 52E, 53E and 54E, for example: 25 51VorA 52R or L 53G 54V The numbering used above is the same as that shown in Figure Ib and SEQ ID No: 2 30 6 Further preferred embodiments of the invention are provided by TCRs comprising one of the mutated alpha chain variable region amino acid sequences shown in Fig 6 (SEQ ID Nos: 11 to 13). Phenotypically silent variants of such TCRs also form part of this invention. 5 Additional preferred embodiments of the invention are provided by TCRs comprising one of the mutated beta chain variable region amino acid sequences shown in Figure 7. (SEQ ID Nos: 14 and 15). Phenotypically silent variants of such TCRs also form part of this invention. 10 Native TCRs exist in heterodimeric as or 75 forms. However, recombinant TCRs consisting of aa or PP homodimers have previously been shown to bind to peptide MHC molecules. Therefore, one embodiment of the invention is provided by TCR aa or TCR PP homodimers. 15 Further preferred embodiments are provided by TCRs of the invention comprising the alpha chain variable region amino acid sequence and the beta chain variable region amino acid sequence combinations listed below, phenotypically silent variants of such TCRs also form part of this invention: 20 Alpha chain variable region sequence, Beta chain variable region sequence, SEQ ID NO: SEQ ID NO: 12 114 1 15 11 2 12 2 13 2 12 15 13 15 7 12 14 13 14 In another preferred embodiment TCRs of the invention comprising the variable regioi combinations detailed above further comprise the alpha chain constant domain amino acid sequence shown in Figure 8a (SEQ ID NO: 19) and one of the beta chain amino 5 acid constant domain sequences shown in Figures 8b and 8c (SEQ ID NOs: 20 and 21) or phenotypically silent variants thereof. As used herein the term "phenotypically silent variants" is understood to refer to those TCRs which have a KD for the said SLYNTVATL-HLA-A*0201 complex of less-than 10 or equal to IpM and/or have an off-rate (kff) of 1x10- 3 S-1 or slower. For example, as is known to those skilled in the art, it may be possible to produce TCRs that incorporate minor changes in the constant domain and/or variable regions thereof compared to those detailed above without altering the affinity and/or off-rate for the interaction with the SLYNTVATL-HLA-A*0201 complex. Such trivial variants are 15 included in the scope of this invention. Those TCRs in which one or more conservative substitutions have been made also form part of this invention. In one broad aspect, the TCRs of the invention are in the form of either single chain TCRs (scTCRs) or dimeric TCRs (dTCRs) as described in WO 04/033685 and WO 20 03/020763. A suitable scTCR form comprises a first segment constituted by an amino acid sequence corresponding to a TCR a chain variable region, a second segment constituted by an amino acid sequence corresponding to a TCR P chain variable region 25 sequence fused to the N terminus of an amino acid sequence corresponding to a TCR p chain constant domain extracellular sequence, and a linker sequence linking the C terminus of the first segment to the N terminus of the second segment.
8 Alternatively the first segment may be constituted by an amino acid sequence corresponding to a TCR P chain variable region, the second segment may be constituted by an amino acid sequence corresponding to a TCR a chain variable region sequence fused to the N terminus of an amino acid sequence corresponding to a 5 TCR a chain constant domain extracellular sequence The above scTCRs may further comprise a disulfide bond between the first and second chains, said disulfide bond being one which has no equivalent in native apT cell receptors, and wherein the length of the linker sequence and the position of the 10 disulfide bond being such that the variable domain sequences of the first and second segments are mutually orientated substantially as in native us T cell receptors. More specifically the first segment may be constituted by an amino acid sequence corresponding to a TCR a chain variable region sequence fused to the N terminus of 15 an amino acid sequence corresponding to a TCR a chain constant domain extracellular sequence, the second segment may be constituted by an amino acid sequence corresponding to a TCR P chain variable region fused to the N terminus of an amino acid sequence corresponding to TCR P chain constant domain extracellular sequence, and a disulfide bond may be provided between the first and second chains, said 20 disulfide bond being one which has no equivalent in native us T cell receptors. In the above scTCR forms, the linker sequence may link the C terminus of the first segment to the N terminus of the second segment, and may have the formula -PGGG (SGGGG)n-P- wherein n is 5 or 6 and P is proline, G is glycine and S is serine. 25 -PGGG-SGGGGSGGGGSGGGGSGGGGSGGGG-P (SEQ ID NO: 17) -PGGG-SGGGGSGGGGSGGGGSGGGGSGGGGSGGGG-P (SEQ ID NO: 18) A suitable dTCR form of the TCRs of the present invention comprises 9 a first polypeptide wherein a sequence corresponding to a TCR a chain variable region sequence is fused to the N terminus of a sequence corresponding to a TCR a chain constant domain extracellular sequence, and a second polypeptide wherein a sequence corresponding to a TCR P chain variable region sequence fused to the N 5 terminus a sequence corresponding to a TCR P chain constant domain extracellular sequence, the first and second polypeptides being linked by a disulfide bond which has no equivalent in native afp T cell receptors. The first polypeptide may comprise a TCR a chain variable region sequence is fused 10 to the N terminus of a sequence corresponding to a TCR a chain constant domain extracellular sequence, and a second polypeptide wherein a sequence corresponding to a TCR P chain variable region sequence is fused to the N terminus a sequence corresponding to a TCR p chain constant domain extracellular sequence, the first and second polypeptides being linked by a disulfide bond between cysteine residues 15 substituted for Thr 48 of exon 1 of TRAC*01 and Ser 57 of exon 1 of TRBC1*01 or TRBC2*01 or the non-human equivalent thereof. ("TRAC" etc. nomenclature herein as per T cell receptor Factsbook, (2001) LeFranc and LeFranc, Academic Press, ISBN 0-12-441352-8) 20 The dTCR or scTCR form of the TCRs of the invention may have amino acid sequences corresponding to human as TCR extracellular constant domain and variable region sequences, and a disulfide bond may link amino acid residues of the said constant domain sequences, which disulfide bond has no equivalent in native TCRs. The disulfide bond is between cysteine residues corresponding to amino acid 25 residues whose p carbon atoms are less than 0.6 nm apart in native TCRs, for example between cysteine residues substituted for Thr 48 of exon 1 of TRAC*01 and Ser 57 of exon 1 of TRBC 1*01 or TRBC2*01 or the non-human equivalent thereof. Other sites where cysteines can be introduced to form the disulfide bond are the following residues in exon 1 of TRAC*01 for the TCR a chain and TRBCl*01 or TRBC2*01 30 for the TCR P chain: 10 Thr 45 Ser 77 0533 Tyr 10 Ser 17 0.359 Thr 45 Asp 59 0.560 Ser 15 Glu 15 0.59 In addition to the non-native disulfide bond referred to above, the dTCR or scTCR 5 form of the TCRs of the invention may include a disulfide bond between residues corresponding to those linked by a disulfide bond in native TCRs. The dTCR or scTCR form of the TCRs of the invention preferably does not contain a sequence corresponding to transmembrane or cytoplasmic sequences of native TCRs. TCRs of the invention bind strongly to the SLYNTVATL-HLA-A2*0201. These TCRs also bind to an altered, but still useful, extent to naturally occurring variants of the HIV Gag-derived SLYNTVATL when loaded by HLA-A*0201. Variants of the 15 SLYNTVATL which have been isolated from AIDs patients include the following (Sewell et al., (1997) Eur JImmunol. 27: 2323-2329): SLFNTVATL SLFNTVAVL 20 SLSNTVATL SSFNTVATL SLLNTVATL SLYNTIATL SLYNTIAVL 25 SLFNTIATL SLFNTIAVL
SLFNFVATL
The mutated amino acids are underlined. PEGylated TCR Monomers 5 In one particular embodiment a TCR of the invention is associated with at least one polyalkylene glycol chain(s). This association may be cause in a number of ways known to those skilled in the art. In a preferred embodiment the polyalkylene chain(s) is/are covalently linked to the TCR. In a further embodiment the polyethylene glycol chains of the present aspect of the invention comprise at least two polyethylene 10 repeating units. Multivalent TCR Complexes 15 One aspect of the invention provides a multivalent TCR complex comprising at least two TCRs of the invention. In one embodiment of this aspect, at least two TCR molecules are linked via linker moieties to form multivalent complexes. Preferably the complexes are water soluble, so the linker moiety should be selected accordingly. Furthermore, it is preferable that the linker moiety should be capable of attachment to 20 defined positions on the TCR molecules, so that the structural diversity of the complexes formed is minimised. One embodiment of the present aspect is provided by a TCR complex of the invention wherein the polymer chain or peptidic linker sequence extends between amino acid residues of each TCR which are not located in a variable region sequence of the TCR. 25 Since the complexes of the invention may be for use in medicine, the linker moieties should be chosen with due regard to their pharmaceutical suitability, for example their immunogenicity. 30 Examples of linker moieties which fulfil the above desirable criteria are known in the art, for example the art of linking antibody fragments.
12 There are two classes of linker that are preferred for use in the production of multivalent TCR molecules of the present invention. A TCR complex of the invention in which the TCRs are linked by a polyalkylene glycol chain provides one 5 embodiment of the present aspect. The first are hydrophilic polymers such as polyalkylene glycols. The most commonly used of this class are based on polyethylene glycol or PEG, the structure of which is shown below. 10
HOCH
2
CH
2 0 (CH 2
CH
2 0),-CH 2
CH
2 OH Wherein n is greater than two. However, others are based on other suitable, optionally substituted, polyalkylene glycols include polypropylene glycol, and copolymers of 15 ethylene glycol and propylene glycol. Such polymers may be used to treat or conjugate therapeutic agents, particularly polypeptide or protein therapeutics, to achieve beneficial changes to the PK profile of the therapeutic, for example reduced renal clearance, improved plasma half-life, 20 reduced immunogenicity, and improved solubility. Such improvements in the PK profile of the PEG-therapeutic conjugate are believe to result from the PEG molecule or molecules forming a 'shell' around the therapeutic which sterically hinders the reaction with the immune system and reduces proteolytic degradation. (Casey et al, (2000) Tumor Targetting 4 235-244) The size of the hydrophilic polymer used my in 25 particular be selected on the basis of the intended therapeutic use of the TCR complex. There are numerous review papers and books that detail the use of PEG and similar molecules in pharmaceutical formulations. For example, see Harris (1992) Polyethylene Glycol Chemistry - Biotechnical and Biomedical Applications, Plenum, New York, NY. or Harris & Zalipsky (1997) Chemistry and Biological Applications 30 of Polyethylene Glycol ACS Books, Washington, D.C.
13 The polymer used can have a linear or branched conformation. Branched PEG molecules, or derivatives thereof, can be induced by the addition of branching moieties including glycerol and glycerol oligomers, pentaerythritol, sorbitol and lysine. 5 Usually, the polymer will have a chemically reactive group or groups in its structure, for example at one or both termini, and/or on branches from the backbone, to enable the polymer to link to target sites in the TCR. This chemically reactive group or groups may be attached directly to the hydrophilic polymer, or there may be a spacer group/moiety between the hydrophilic polymer and the reactive chemistry as shown 10 below: Reactive chemistry-Hydrophilic polymer-Reactive chemistry Reactive chemistry-Spacer-Hydrophilic polymer-Spacer-Reactive chemistry 15 The spacer used in the formation of constructs of the type outlined above may be any organic moiety that is a non-reactive, chemically stable, chain, Such spacers include, by are not limited to the following:
-(CH
2 )- wherein n = 2 to 5 20 -(CH 2
)
3
NHCO(CH
2
)
2 A TCR complex of the invention in which a divalent alkylene spacer radical is located between the polyalkylene glycol chain and its point of attachment to a TCR of the complex provides a further embodiment of the present aspect. 25 A TCR complex of the invention in which the polyalkylene glycol chain comprises at least two polyethylene glycol repeating units provides a further embodiment of the present aspect. 30 There are a number of commercial suppliers of hydrophilic polymers linked, directly or via a spacer, to reactive chemistries that may be of use in the present invention.
14 These suppliers include Nektar Therapeutics (CA, USA), NOF Corporation (Japan), Sunbio (South Korea) and Enzon Pharmaceuticals (NJ, USA). Commercially available hydrophilic polymers linked, directly or via a spacer, to 5 reactive chemistries that may be of use in the present invention include, but are not limited to, the following: TCR Monomer attachment 5K linear (Maleimide) Nektar 2D2MOH01 20K linear (Maleimide) Nektar 2D2MOPO1 SUNBRIGHT 20K linear (Maleimide) NOF Corporation ME-200MA . . . SUNBRIGHT GL2- 20K branched (Maleimide) NOF Corporation 200MA . . .SUNBRIGHT ME 30K linear (Maleimide) NOF Corporation 300MA 40K branched PEG (Maleimide) Nektar 2D3XOTO1 SUNBRIGHT 5K-NP linear (for Lys attachment) NOF Corporation MENP-50H 10K-NP linear (for Lys attachment) NOF Corporation SUNBRIGHT ________________MENP- iOT SUNBRIGHT 20K-NP linear (for Lys attachment) NOF Corporation MENP-20T TCR dimer linkers 3.4K linear (Maleimide) Nektar 2D2DOF02 5K forked (Maleimide) Nektar 2D2DOHOF 10K linear (with orthopyridyl ds- Sunbio linkers in place of Maleimide) 20K forked (Maleimide) Nektar 2D2DOPOF 20K linear (Maleimide) NOF Corporation 40K forked (Maleimide) Nektar 2D3XOTOF Higher order TCR multimers 15K, 3 arms, Mal 3 (for trimer) Nektar OJOONO3 20K, 4 arms, Mal 4 (for tetramer) Nektar OJOOPO4 40 K, 8 arms, Mal 8 (for octamer) Nektar OJOOTO8 15 A wide variety of coupling chemistries can be used to couple polymer molecules to protein and peptide therapeutics. The choice of the most appropriate coupling 5 chemistry is largely dependant on the desired coupling site. For example, the following coupling chemistries have been used attached to one or more of the termini of PEG molecules (Source: Nektar Molecular Engineering Catalogue 2003): N-maleimide Vinyl sulfone 10 Benzotriazole carbonate Succinimidyl proprionate Succinimidyl butanoate Thio-ester Acetaldehydes 15 Acrylates Biotin Primary amines As stated above non-PEG based polymers also provide suitable linkers for 20 multimerising the TCRs of the present invention. For example, moieties containing maleimide termini linked by aliphatic chains such as BMH and BMOE (Pierce, products Nos. 22330 and 22323) can be used. Peptidic linkers are the other class of TCR linkers. These linkers are comprised of 25 chains of amino acids, and function to produce simple linkers or multimerisation domains onto which TCR molecules can be attached. The biotin / streptavidin system has previously been used to produce TCR tetramers (see WO/99/60119) for in-vitro binding studies. However, strepavidin is a microbially-derived polypeptide and as such not ideally suited to use in a therapeutic. 30 16 A TCR complex of the invention in which the TCRs are linked by a peptidic linker derived from a human multimerisation domain provides a further embodiment of the present aspect. 5 There are a number of human proteins that contain a multimerisation domain that could be used in the production of multivalent TCR complexes. For example the tetramerisation domain of p53 which has been utilised to produce tetramers of scFv antibody fragments which exhibited increased serum persistence and significantly reduced off-rate compared to the monomeric scFV fragment. (Willuda et al. (2001) J. 10 Biol. Chem. 276 (17) 14385-14392) Haemoglobin also has a tetramerisation domain that could potentially be used for this kind of application. A multivalent TCR complex of the invention comprising at least two TCRs provides a final embodiment of this aspect, wherein at least one of said TCRs is associated with a 15 therapeutic agent. In one aspect a TCR (or multivalent complex thereof) of the present invention may alternatively or additionally comprise a reactive cysteine at the C-terminal or N terminal of the alpha or beta chains thereof. 20 Diagnostic and therapeutic Use In one aspect the TCR of the invention may be associated with a therapeutic agent or detectable moiety. For example, said therapeutic agent or detectable moiety may be 25 covalently linked to the TCR. In one embodiment of the invention said therapeutic agent or detectable moiety is covalently linked to the C-terminus of one or both TCR chains. 30 In one aspect the scTCR or one or both of the dTCR chains of TCRs of the present invention may be labelled with an detectable moiety, for example a label that is 17 suitable for diagnostic purposes. Such labelled TCRs are useful in a method for detecting a SLYNTVATL-HLA-A*0201 complex which method comprises contacting the TCR ligand with a TCR (or a multimeric high affinity TCR complex) which is specific for the TCR ligand; and detecting binding to the TCR ligand. In tetrameric 5 TCR complexes formed for example, using biotinylated heterodimers, fluorescent streptavidin can be used to provide a detectable label. Such a fluorescently-labelled TCR tetramer is suitable for use in FACS analysis, for example to detect antigen presenting cells carrying the SLYNTVATL-HLA-A*0201 complex for which these high affinity TCRs are specific. 10 Another manner in which the soluble TCRs of the present invention may be detected is by the use of TCR-specific antibodies, in particular monoclonal antibodies. There are many commercially available anti-TCR antibodies, such as ccFl and pFl, which recognise the constant domains of the cc and P chains, respectively. 15 In a further aspect a TCR (or multivalent complex thereof) of the present invention may alternatively or additionally be associated with (e.g. covalently or otherwise linked to) a therapeutic agent which may be, for example, a toxic moiety for use in cell killing, or an immune effector molecule such as an interleukin or a cytokine. A 20 multivalent TCR complex of the invention may have enhanced binding capability for a TCR ligand compared to a non-multimeric wild-type or T cell receptor heterodimer of the invention. Thus, the multivalent TCR complexes according to the invention are particularly useful for tracking or targeting cells presenting SLYNTVATL-HLA A*0201 complexes in vitro or in vivo, and are also useful as intermediates for the 25 production of further multivalent TCR complexes having such uses. These TCRs or multivalent TCR complexes may therefore be provided in a pharmaceutically acceptable formulation for use in vivo. The invention also provides a method for delivering a therapeutic agent to a target cell, 30 which method comprises contacting potential target cells with a TCR or multivalent TCR complex in accordance with the invention under conditions to allow attachment 18 of the TCR or multivalent TCR complex to the target cell, said TCR or multivalent TCR complex being specific for the SLYNTVATL-HLA-A*0201 complex and having the therapeutic agent associated therewith. 5 In particular, the soluble TCR or multivalent TCR complex of the present invention can be used to deliver therapeutic agents to the location of cells presenting a particular antigen. This would be useful in many situations and, in particular, against HIV infected cells. A therapeutic agent could be delivered such that it would exercise its effect locally but not only on the cell it binds to. Thus, one particular strategy 10 envisages cytotoxic or immuno-stimulatory molecules linked to TCRs or multivalent TCR complexes according to the invention specific for the SLYNTVATL-HLA A*0201 complex. Many therapeutic agents could be employed for this use, for instance radioactive 15 compounds, enzymes (perforin for example) or chemotherapeutic agents (cis-platin for example). To ensure that toxic effects are exercised in the desired location the toxin could be inside a liposome linked to streptavidin so that the compound is released slowly. This will prevent damaging effects during the transport in the body and ensure that the toxin has maximum effect after binding of the TCR to the relevant antigen 20 presenting cells. Other suitable therapeutic agents include: e small molecule cytotoxic agents, i.e. compounds with the ability to kill mammalian cells having a molecular weight of less than 700 daltons. Such 25 compounds could also contain toxic metals capable of having a cytotoxic effect. Furthermore, it is to be understood that these small molecule cytotoxic agents also include pro-drugs, i.e. compounds that decay or are converted under physiological conditions to release cytotoxic agents. Examples of such agents include cis-platin, maytansine derivatives, rachelmycin, calichearnicin, 30 docetaxel, etoposide, gemcitabine, ifosfamide, irinotecan, melphalan, 19 mitoxantrone, sorfimer sodiumphotofrin II, temozolmide, topotecan, trimetreate glucuronate, auristatin E vincristine and doxorubicin; e peptide cytotoxins, i.e. proteins or fragments thereof with the ability to kill mammalian cells. 'Including but not limited to, ricin, diphtheria toxin, 5 pseudomonas bacterial exotoxin A, DNAase and RNAase; " radio-nuclides, i.e. unstable isotopes of elements which decay with the concurrent emission of one or more of a or P particles, or y rays. including but not limited to, iodine 131, rhenium 186, indium 111, yttrium 90, bismuth 210 and 213, actinium 225 and astatine 213; chelating agents may be used to 10 facilitate the association of these radio-nuclides to the high affinity TCRs, or multimers thereof; e prodrugs, including but not limited to, antibody directed enzyme pro-drugs; e immuno-stimulants, i.e. moieties which stimulate immune response. Including but not limited to, cytokines such as IL-2 and IFN, Superantigens and mutants 15 thereof, TCR-HLA fusions and chemokines such as IL-8, platelet factor 4, melanoma growth stimulatory protein, etc, antibodies or fragments thereof, complement activators, xenogeneic protein domains, allogeneic protein domains, viral/bacterial protein domains, viral/bacterial peptides and anti-T cell determinant antibodies (e.g. anti-CD3 or anti-CD28) or antibody analogues 20 such as NanobodiesTM and AffybodiesTM. Soluble TCRs or multivalent TCR complexes of the invention may be linked to an enzyme capable of converting a prodrug to a drug. This allows the prodrug to be converted to the drug only at the site where it is required (i.e. targeted by the sTCR). 25 It is expected that the high affinity SLYNTVATL (SEQ ID NO: 16)-HLA-A*0201 specific TCRs disclosed herein may be used in methods for the diagnosis and treatment of AIDS. For treatment, therapeutic agent localisation in the vicinity of HIV infected (CD4*) 30 cells would enhance the effect of toxins or immunostimulants. For vaccine delivery, 20 the vaccine antigen could be localised in the vicinity of antigen presenting cells, thus enhancing the efficacy of the antigen. The method can also be applied for imaging purposes. 5 One embodiment is provided by a membrane preparation comprising a TCR of the invention. Said membrane preparation may be prepared from cells or may comprise a synthetic membrane. Another embodiment is provided by a cell harbouring an expression vector comprising 10 nucleic acid encoding a TCR of the invention. For example, said cell may be a T cell. Further embodiments of the invention are provided by a pharmaceutical composition comprising: 15 a TCR or a multivalent TCR complex of the invention (optionally associated with a therapeutic agent), or a membrane preparation comprising a TCR of the invention , or a plurality of cells harbouring an expression vector comprising nucleic acid encoding a TCR of the invention, together with a pharmaceutically acceptable carrier; 20 The invention also provides a method of treatment of AIDS comprising administering to a subject suffering such AIDS an effective amount of a TCR or a multivalent TCR complex of the invention, or a membrane preparation comprising a TCR of the invention, or a plurality of cells harbouring an expression vector comprising nucleic acid encoding a TCR of the invention. In a related embodiment the invention provides 25 for the use of a TCR or a multivalent TCR complex of the invention, or a membrane preparation comprising a TCR of the invention, or a plurality of cells harbouring an expression vector comprising nucleic acid encoding a TCR of the invention, in the preparation of a composition for the treatment of AIDS. Further specific embodiments of these uses and methods of the invention are provided wherein the TCR, or 30 multivalent TCR complex of the invention, or a membrane preparation comprising a TCR of the invention is administered in a form which is associated with a therapeutic 21 agent. .In other preferred embodiments the cells harbouring an expression vector comprising nucleic acid encoding a TCR of the invention are CD8* T cells. Therapeutic or imaging TCRs in accordance with the invention will usually be supplied 5 as part of a sterile, pharmaceutical composition which will normally include a pharmaceutically acceptable carrier. This pharmaceutical composition may be in any suitable form, (depending upon the desired method of administering it to a patient). It may be provided in unit dosage form, will generally be provided in a sealed container and may be provided as part of a kit. Such a kit would normally (although not necessarily) 10 include instructions for use. It may include a plurality of said unit dosage forms. Without wishing to be limited by theory, it is expected that the TCRs of the invention will provide effective targeting agents capable of delivering therapeutic agents such as immunostimulants and/or cytotoxic agents to HIV infected (CD4*) cells. In particular, it 15 is expected that the administration of the TCRs of the present invention when associated with immunostimulants and/or cytotoxic agents in combination with conventional anti retrovirus drug therapies and/or IL-2 treatment will be able to target HIV infected cells. The following is a list of anti-retroviral drugs currently approved for use in the US: 20 Agenerase (amprenavir) - protease inhibitor Combivir -combination of Retrovir (300mg) and Epivir (1 50mg) Crixivan (indinavir) -pmtease inhibitor Epivir (3tc / lamivudine) - nucleoside analog reverse transcriptase inhibitor 25 Epzicom ( a combination of 2 nucleoside reverse transcriptase inhibitors (NRTIs in the same pill; 600mg of Ziagen (abacavir) and 300mg of Epivir (3TC). Emtriva [ emtricitabine (FTC)] Fortovase (saquinavir) - protease inhibitor Fuzeon (enfuvirtide) -Fusion inhibitor 30 Hivid (ddc / zalcitabine) - nucleoside analog reverse transcriptase inhibitor Invirase (saquinavir) - protease inhibitor 22 Kaletra (lopinavir) - protease inhibitor Lexiva (Fosamprenavir) - Protease Inhibitor approved 10/20/03 Norvir (ritonavir) - protease inhibitor Rescriptor (delavirdine) - non nucleoside analog reverse transcriptase inhibitor 5 Retrovir, AZT (zidovudine) - nucleoside analog reverse transcriptase inhibitor Reyataz (atazanavir; BMS-232632) - protease inhibitor Sustiva (efavirenz) - non nucleoside analog reverse transcriptase inhibitor Trizivir (3 non nucleosides in one tablet; abacavir + zidovudine + lamivudine Truvada (Emtricitabine + Tenofovir DF ) 10 Videx (ddl / didanosine) nucleoside analog reverse transcriptase inhibitor Videx EC; (ddl / didanosine) nucleoside analog reverse transcriptase inhibitor; Viracept (nelfinavir) - protease inhibitor Viramune (nevirapine) - non nucleoside analog Reverse transcriptase inhibitor Viread (tenofovir disoproxil fumarate) Nucleotide Reverse transcriptase inhibitor 15 (Adenosine Class) Zerit (d4t / stavudine) - nucleoside analog reverse transcriptase inhibitor Ziagen (abacavir) - nucleoside analog reverse transcriptase inhibitor The pharmaceutical composition may be adapted for administration by any appropriate 20 route, for example parenteral, transdermal or via inhalation, preferably a parenteral (including subcutaneous, intramuscular, or, most preferably intravenous) route. Such compositions may be prepared by any method known in the art of pharmacy, for example by mixing the active ingredient with the carrier(s) or excipient(s) under sterile conditions. 25 Dosages of the substances of the present invention can vary between wide limits, depending upon the disease or disorder to be treated, the age and condition of the individual to be treated, etc. and a physician will ultimately determine appropriate dosages to be used.
23 Additional Aspects A scTCR or dTCR (which preferably is constituted by constant and variable sequences corresponding to human sequences) of the present invention may be provided in 5 substantially pure form, or as a purified or isolated preparation. For example, it may be provided in a form which is substantially free of other proteins. The sequence(s) of the nucleic acid or nucleic acids encoding the TCRs of the invention may be altered so as to optimise the level of expression obtained in the host 10 cell. The host cell may be any appropriate prokaryotic or eukaryotic cell. For example, the host cell may be an E. coli cell or a human T cell. The alterations made to these genetic sequences are silent, that is they do not alter the amino acid sequence encoded. There are a number of companies which offer such expression optimisation services, including, GeneArt, Germany. 15 The invention also provides a method of producing a high affinity TCR having the property of binding to SLYNTVATL-HLA-A*0201.CHARACTERISED IN THAT the TCR (i) comprises at least one TCR a chain variable domain and/or at least one TCR 3 chain variable domain and (ii) has a KD for the said SLYNTVATL-HLA 20 A*0201 complex of less than or equal to 1 tM and/or an off-rate (koff) for the SLYNTVATL-HLA-A*0201 complex of 1x10 3 S- or slower, wherein the method comprises: (a) the production of a TCR comprising the a and 0 chain variable domains 25 of the parental HIV Gag TCR wherein one or both of the a and p chain variable domains comprise a mutation(s) in one or more of the amino acids identified in claims 7 and 8; (b) contacting said mutated TCR with SLYNTVATL-HLA-A*0201 under conditions suitable to allow the binding of the TCR to SLYNTVATL-HLA 30 A*0201; and measuring the KD and/or koff of the interaction.
24 Preferred features of each aspect of the invention aie as for each of the other aspects mutatis mutandis. The prior art documents mentioned herein are incorporated to the fullest extent permitted by law. 5 Examples The invention is further described in the following examples, which do not limit the scope of the invention in any way. 10 Reference is made in the following to the accompanying drawings in which: Figure 1 a and lb details the alpha chain variable domain amino acid and beta chain variable domain amino acid sequences of the parental HIV Gag TCR respectively. 15 Figures 2a and 2b show respectively the DNA sequence of soluble versions of the parental HIV Gag TCR a and P chains. Figures 3a and 3b show respectively the HIV Gag TCR a and P chain extracellular 20 amino acid sequences produced from the DNA sequences of Figures 2a and 2b. Figures 4a and 4b show respectively the DNA sequence of soluble versions of the HIV Gag TCR a and P chains mutated to enocode additional cysteine residues to form a non-native disulfide bond. The mutated codon is indicated by shading and The 25 introduced restriction enzyme recognition sites are underlined.. Figures 5a and 5b show respectively the HIV Gag TCR a and P chain extracellular amino acid sequences produced from the DNA sequences of Figures 4a and 4b. The introduced cysteine in each chain is indicated by shading. 30 25 Figure 6 details the alpha chain variable domain amino acid sequences of the high affinity HIV Gag TCR variants. Figure 7 details the beta chain variable domain amino acid sequences of the high 5 affinity HIV Gag TCR variants. Figure 8a details the amino acid sequence of a soluble portion of TRAC. Figure 8b details the amino acid sequence of a soluble portion of TRBC 1. 10 Figure 8c details the amino acid sequence of a soluble portion of TRBC2. Figure 9 details the DNA sequence of the pEX954 plasmid. 15 Figure 10 details the DNA sequence of the pEX821 plasmid. Figure 11 details the beta chain amino acid sequences of the parental soluble HIV Gag TCR variant fused via a peptide linker to wild-type human IL-2. The amino acids of the linker and IL-2 are indicated in italics. 20 Figure 12 provides the Biacore response curves generated for the interaction of the soluble disulfide-linked parental HIV Gag TCR and the SLYNTVATL-HLA-A*0201 complex. 25 Figure 13 provides a plasmid map of the pEX954 plasmid. Figure 14 provides a plasmid map of the pEX821 plasmid. Figure 15a provides the full-length DNA sequence of the parental HIV Gag TCR a 30 chain optimised for expression in human T cells.
26 Figure 15b provides the full-length DNA sequence of the parental HIV Gag TCR $ chain optimised for expression in human T cells. Figure 16a provides the full-length amino acid sequence of the parental HIV Gag TCR 5 a chain. Figure 16b provides the full-length amino acid sequence of the parental HIV Gag TCR P chain optimised for expression in human T cells. 10 Figures 17a provides FACS analysis data for untransduced control CD8* T cells. Figure 17b provides FACS analysis data demonstrating expression of the parental HIV Gag TCR on the surface of transduced CD8* T cells. 15 Figures 18a and 18b provide the amino acids sequences of the alpha and beta chains of a soluble disulfide-linked high affinity ci 1c6 HIV Gag TCR.respectively. Figure 19 demonstrates the ability of soluble disulfide-linked high affinity cl 1c6 HIV Gag TCRs to inhibit the activation of the SLYNTVATL-HLA-A*0201 reactive OX84 20 polyclonal T cell line in the presence of To cells infected with HIV as measured by IFN-y and TNF-a production. Figure 20 demonstrates the ability of soluble disulfide-linked high affinity c 11c6 HIV Gag TCRs to inhibit the activation of the SLYNTVATL-HLA-A*0201 reactive OX84 25 polyclonal T cell line in the presence of SLYNTVATL peptide-pulsed uninfected To cells as measured by IFN-y and TNF-a production. Figure 21 demonstrates the ability of soluble disulfide-linked high affinity cl 1c6 HIV Gag TCRs to stain SLYNTVATL peptide-pulsed T2 cells. 30 27 Example I - Production of soluble disulfide-linked TCRs comprising the parental HIV Gag TCR variable regions 5 Figures 4a and 4b provide the DNA sequences of soluble disulfide-linked alpha beta chains from a parental TCR which is specific for the SLYNTVATL-HLA-A*0201 complex. These DNA sequences can be synthesis de-novo by a number of contract research companies, for example GeneArt (Germany). Restriction enzyme recognition sites are also added to these DNA sequences in order to facilitate ligation of these 10 DNA sequences into the pGMT7-based expression plasmids, which contain the T7 promoter for high level expression in E.coli strain BL21-DE3(pLysS) (Pan et al., Biotechniques (2000) 29 (6): 1234-8) The TCR alpha chain sequences contain introduced ClaI and SalII restriction enzyme 15 recognition sites and this sequence was ligated into pEX954 (see Figures 9 and 13) cut with ClaI and XhoI. The TCR beta chain sequences contain introduced AseI and AgeI restriction enzyme recognition sites and were ligated into pEX821 (see Figures 10 and 14) cut with 20 NdeI/AgeL. Restriction enzyme recognition sites as introduced into DNA encoding the TCR chains ClaI- ATCGAT 25 SaII- GTCGAC AseI - ATTAAT AgeI - ACCGGT Ligation 30 The cut TCR alpha and beta chain DNA and cut vector were ligated using a rapid DNA ligation kit (Roche) following the manufacturers instructions.
28 Ligated plasmids were transformed into competent E.coli strain XL 1-blue cells and plated out on LB/agar plates containing 100mg/ml ampicillin. Following incubation overnight at 37"C, single colonies were picked and grown in 10 ml LB containing 5 100mg/ml ampicillin overnight at 37"C with shaking. Cloned plasmids were purified using a Miniprep kit (Qiagen) and the insert was sequenced using an automated DNA sequencer (Lark Technologies). Figures 5a and 5b show respectively the soluble disulfide linked parental HIV gag 10 TCR (x and P chain extracellular amino acid sequences produced from the DNA sequences of Figures 4a and 4b Example 2- Production of high affinity variants of the soluble disulfide linked HIV Gag TCR 15 The soluble disulfide-linked native HIV Gag TCR produced as described in Example 1 can be used a template from which to produce the TCRs of the invention which have an increased affinity for the SLYNTVATL (SEQ ID NO: 16) -HLA-A*0201 complex. 20 Phage display is one means by which libraries of HIV Gag TCR variants can be generated in order to identify high affinity mutants. For example, the TCR phage display and screening methods described in (Li et al., (2005) Nature Biotech 23 (3): 349-354) can be adapted and applied to HIV Gag TCRs. 25 The amino sequences of the mutated TCR alpha and beta chain variable domains which, when combined with an appropriate TCR chain, demonstrate high affinity for the SLYNTVATL-HLA-A*0201 complex, are listed in Figures 6 and 7 respectively. (SEQ ID Nos: 11-13 and 14-15 respectively) As is known to those skilled in the art the necessary codon changes required to produce these mutated chains can be introduced 30 into the DNA encoding these chains by site-directed mutagenesis. (QuickChange T Site-Directed Mutagenesis Kit from Stratagene) 29 Briefly, this is achieved by using primers that incorporate the desired codon change(s) and the plasmids containing the relevant TCR chain DNA as a template for the mutagenesis: 5 Mutagenesis was carried out using the following conditions : 50ng plasmid template, 1 [d of 10mM dNTP, 5 pl of 1 Ox Pfu DNA polymerase buffer as supplied by the manufacturer, 25 pmol of fwd primer, 25 pmol of rev primer, 1p l pfu DNA polymerase in total volume 50 pl. After an initial denaturation step of 2 mins at 95C, 10 the reaction was subjected to 25 cycles of denaturation (95C, 10 secs), annealing (55C 10 secs), and elongation (72C, 8 mins). The resulting product was digested with DpnI restriction enzyme to remove the template plasmid and transformed into E. coli strain XL1-blue. Mutagenesis was verified by sequencing. 15 Example 3 - Expression, refolding and purification of soluble TCR The expression plasmids containing the mutated a-chain and p-chain respectively as prepared in Examples 1 or 2 were transformed separately into E.coli strain 20 BL21pLysS, and single ampicillin-resistant colonies were grown at 37*C in TYP (ampicillin 100ptg/ml) medium to OD 600 of 0.4 before inducing protein expression with 0.5mM IPTG. Cells were harvested three hours post-induction by centrifugation for 30 minutes at 4000rpm in a Beckman J-6B. Cell pellets were re-suspended in a buffer containing 50mM Tris-HCI, 25% (w/v) sucrose, 1mM NaEDTA, 0.1% (w/v) 25 NaAzide, 10mM DTT, pH 8.0. After an overnight freeze-thaw step, re-suspended cells were sonicated in 1 minute bursts for a total of around 10 minutes in a Milsonix XL2020 sonicator using a standard 12mm diameter probe. Inclusion body pellets were recovered by centrifugation for 30 minutes at 13000rpm in a Beckman J2-21 centrifuge. Three detergent washes were then carried out to remove cell debris and 30 membrane components. Each time the inclusion body pellet was homogenised in a Triton buffer (50mM Tris-HCI, 0.5% Triton-XI 00, 200mM NaCI, 10mM NaEDTA, 30 0.1% (w/v) NaAzide, 2mM DTT, pH 8.0) before being pelleted by centrifugation for 15 minutes at 13000rpm in a Beckman J2-2 1. Detergent and salt was then removed by a similar wash in the following buffer: 50mM Tris-HC1, ImMNaEDTA, 0.1% (w/v) NaAzide, 2mM DTT, pH 8.0. Finally, the inclusion bodies were divided into 30 mg 5 aliquots and frozen at -70*C. Inclusion body protein yield was quantitated by solubilising with 6M guanidine-HCI and measurement with a Bradford dye-binding assay (PerBio). Approximately 30mg of TCR p chain and 60mg of TCR a chain solubilised inclusion 10 bodies were thawed from frozen stocks, samples were then mixed and the mixture diluted into 15ml of a guanidine solution (6 M Guanidine-hydrochloride, 10mM Sodium Acetate, 10mM EDTA), to ensure complete chain de-naturation. The guanidine solution containing fully reduced and denatured TCR chains was then injected into 1 litre of the following refolding buffer: 100mM Tris pH 8.5, 400mM L 15 Arginine, 2mM EDTA, 5mM reduced Glutathione, 0.5mM oxidised Glutathione, 5M urea, 0.2mM PMSF. The redox couple (2-mercaptoethylamine and cystamine (to final concentrations of 6.6mM and 3.7mM, respectively) were added approximately 5 minutes before addition of the denatured TCR chains. The solution was left for 5 hrs minutes. The refolded TCR was dialysed in Spectrapor 1 membrane (Spectrum; 20 Product No. 132670) against 10 L 10 mM Tris pH 8.1 at 5'C +3*C for 18-20 hours. After this time, the dialysis buffer was changed to fresh 10 mM Tris pH 8.1 (10 L) and dialysis was continued at 5*C +3*C for another 20-22 hours. sTCR was separated from degradation products and impurities by loading the dialysed 25 refold onto a POROS 50HQ anion exchange column and eluting bound protein with a gradient of 0-500mM NaCI over 50 column volumes using an Akta purifier (Pharmacia). Peak fractions were stored at 4'C and analysed by Coomassie-stained SDS-PAGE before being pooled and concentrated. Finally, the sTCR was purified and characterised using a Superdex 200HR gel filtration column pre-equilibrated in HBS 30 EP buffer (10 mM HEPES pH 7.4, 150 mM NaCl, 3.5 mM EDTA, 0.05% nonidet p40). The peak eluting at a relative molecular weight of approximately 50 kDa was 31 pooled and concentrated prior to characterisation by BIAcore surface plasmon resonance analysis. Example 4 - Biacore surface plasmon resonance characterisation ofsTCR binding to 5 specific pMHC A surface plasmon resonance biosensor (Biacore 3000TM) was used to analyse the binding of a sTCR to its peptide-MHC ligand. This was facilitated by producing single pMHC complexes (described below) which were immobilised to a streptavidin 10 coated binding surface in a semi-oriented fashion, allowing efficient testing of the binding of a soluble T-cell receptor to up to four different pMHC (immobilised on separate flow cells) simultaneously. Manual injection of HLA complex allows the precise level of immobilised class I molecules to be manipulated easily. 15 Biotinylated class I HLA-A*0201 molecules were refolded in vitro from bacterially expressed inclusion bodies containing the constituent subunit proteins and synthetic peptide, followed by purification and in vitro enzymatic biotinylation (O'Callaghan et al. (1999) Anal. Biochem. 266: 9-15). HLA-A*0201-heavy chain was expressed with a C-terminal biotinylation tag which replaces the transmembrane and cytoplasmic 20 domains of the protein in an appropriate construct. Inclusion body expression levels of -75 mg/litre bacterial culture were obtained. The MHC light-chain or p2 microglobulin was also expressed as inclusion bodies in E. coli from an appropriate construct, at a level of-500 mg/litre bacterial culture. 25 E. coli cells were lysed and inclusion bodies are purified to approximately 80% purity. Protein from inclusion bodies was denatured in 6 M guanidine-HCI, 50 mM Tris pH 8.1, 100 mM NaCl, 10 mM DTT, 10 mM EDTA, and was refolded at a concentration of 30 mg/litre heavy chain, 30 mg/litre p2m into 0.4 M L-Arginine-HCI, 100 mM Tris pH 8.1, 3.7 mM cystamine, 6.6 mM p-cysteamine, 4 mg/ml of the SLYNTVATL 30 peptide required to be loaded by the HLA-A*0201 molecule, by addition of a single 32 pulse of denatured protein into refold buffer at <5*C. Refolding was allowed to reach completion at 4*C for at least 1 hour. Buffer was exchanged by dialysis in 10 volumes of 10 mM Tris pH 8.1. Two changes 5 of buffer were necessary to reduce the ionic strength of the solution sufficiently. The protein solution was then filtered through a 1.5im cellulose acetate filter and loaded onto a POROS 50HQ anion exchange column (8 ml bed volume). Protein was eluted with a linear 0-500 mM NaCl gradient. HLA-A*0201-peptide complex eluted at approximately 250 mM NaCl, and peak fractions were collected, a cocktail of protease 10 inhibitors (Calbiochem) was added and the fractions were chilled on ice. Biotinylation tagged pMHC molecules were buffer exchanged into 10 mM Tris pH 8.1, 5 mM NaCl using a Pharmacia fast desalting column equilibrated in the same buffer. Immediately upon elution, the protein-containing fractions were chilled on ice 15 and protease inhibitor cocktail (Calbiochem) was added. Biotinylation reagents were then added: 1 mM biotin, 5 mM ATP (buffered to pH 8), 7.5 mM MgCl2, and 5 pg/ml BirA enzyme (purified according to O'Callaghan et al. (1999) Anal. Biochem. 266: 9 15). The mixture was then allowed to incubate at room temperature overnight. 20 The biotinylated pHLA-A*0201 molecules were purified using gel filtration chromatography. A Pharmacia Superdex 75 HR 10/30 column was pre-equilibrated with filtered PBS and 1 ml of the biotinylation reaction mixture was loaded and the column was developed with PBS at 0.5 ml/min. Biotinylated pHLA-A*0201 molecules eluted as a single peak at approximately 15 ml. Fractions containing 25 protein were pooled, chilled on ice, and protease inhibitor cocktail was added. Protein concentration was determined using a Coomassie-binding assay (PerBio) and aliquots of biotinylated pHLA-A*0201 molecules were stored frozen at -20*C. Streptavidin was immobilised by standard amine coupling methods. 30 Such immobilised complexes are capable of binding both T-cell receptors and the coreceptor CD8aa, both of which may be injected in the soluble phase. Specific 33 binding of TCR is obtained even at low concentrations (at least 40pg/ml), implying the TCR is relatively stable. The pMHC binding properties of sTCR are observed to be qualitatively and quantitatively similar if sTCR is used either in the soluble or immobilised phase. This is an important control for partial activity of soluble species 5 and also suggests that biotinylated pMHC complexes are biologically as active as non biotinylated complexes. The interactions between HIV Gag sTCR containing a novel inter-chain bond and its ligand/ MHC complex or an irrelevant HLA-peptide combination, the production of 10 which is described above, were analysed on a Biacore 3000TM surface plasmon resonance (SPR) biosensor. SPR measures changes in refractive index expressed in response units (RU) near a sensor surface within a small flow cell, a principle that can be used to detect receptor ligand interactions and to analyse their affinity and kinetic parameters. The probe flow cells were prepared by immobilising the individual HLA 15 peptide complexes in separate flow cells via binding between the biotin cross linked onto p2m and streptavidin which have been chemically cross linked to the activated surface of the flow cells. The assay was then performed by passing sTCR over the surfaces of the different flow cells at a constant flow rate, measuring the SPR response in doing so. 20 To measure Equilibrium binding constant Serial dilutions of the parental or mutated HIV Gag sTCR were prepared and injected at constant flow rate of 5 pl min-I over two different flow cells; one coated with 25 -1000 RU of specific SLYNTVATL-HLA-A*0201 complex, the second coated with -1000 RU of non-specific HLA-A2 -peptide complex. Response was normalised for each concentration using the measurement from the control cell. Normalised data response was plotted versus concentration of TCR sample and fitted to a hyperbola in order to calculate the equilibrium binding constant, KD. (Price & Dwek, Principles and 30 Problems in Physical Chemistry for Biochemists ( 2 nd Edition) 1979, Clarendon Press, Oxford).
34 To measure Kinetic Parameters 5 For high affinity TCRs KD was determined by experimentally measuring the dissociation rate constant, kd, and the association rate constant, ka. The equilibrium constant KD was calculated as kd/ka. TCR was injected over two different cells one coated with -300 RU of specific HLA 10 A2-nyeso peptide complex, the second coated with -300 RU of non-specific HLA-A2 -peptide complex. Flow rate was set at 50 pl/min. Typically 250 l of TCR at -3 pM concentration was injected. Buffer was then flowed over until the response had returned to baseline. Kinetic parameters were calculated using Biaevaluation software. The dissociation phase was also fitted to a single exponential decay equation enabling 15 calculation of half-life. Results The interaction between a soluble disulfide-linked native HIV Gag TCR (consisting of 20 the a and P TCR chains detailed in SEQ ID NOs 9 and 10 respectively) and the SLYNTVATL-HLA-A*0201 complex was analysed using the above methods and demonstrated a KD of 85 nM and an off-rate (koff) of 2.21 x 10-2 S-'. (See Figure 12 for Biacore response curves) 25 The TCRs specified in the following table have a KD of less than or equal to 1 tM and/or a kff of 1 x 10- S~1 or slower. Alpha chain variable domain sequence, Beta chain variable domain sequence, SEQ ID NO: SEQ ID NO: 1 2 114 35 1 15 11 2 12 2 13 2 12 15 13 15 12 14 13 14 Example 5 -Production of a soluble high affinity HIV Gag TCR --WT human IL-2 5 fusion protein The methods substantially as described in Examples 1 to 3 can be used to produce a soluble high affinity HIV Gag TCR-WT human IL-2 fusion protein. Briefly, the DNA encoding the desired linker and WT human IL-2 are added into the 3' end of the DNA 10 sequence of the soluble disulfide-linked parental HIV Gag TCR beta chain immediately prior to the TAA ("Stop") codon. Figure 11 provides the amino acid sequence of a fusion protein comprising a disulfide-linked parental HIV Gag TCR beta chain fused to WT human IL-2 via linker sequence.(SEQ ID NO: 24) The linker and IL-2 portion of this fusion protein are indicated in italics. The DNA encoding this 15 construct can then be ligated into pEX82 1. The soluble parental HIV Gag TCR- IL-2 fusion protein can then be expressed by combining this beta chain fusion protein with the soluble disulfide-linked parental HIV Gag alpha chain TCR chain detailed in Figure 5a (SEQ ID NO: 9) using the methods substantially as described in Example 3. 20 Example 6 - Recombinant expression of the parental HIV Gag TCR on the surface of T cells.
36 DNA constructs encoding the signal sequence, extracellular, transmembrane and intracellular domains of the parental HIV Gag TCR chains were synthesised (GeneArt, Germany). These TCR a chain and TCR p chain DNA sequences, provided in Figures 15a and 15b respectively, are altered from the parental HIV Gag TCR DNA sequences 5 so as to enhance expression levels of the encoded TCR chains in human T cells whilst maintaing the native amino acid sequence. Figures 16a and 16b provide the full-length amino acid sequences encoded by the DNA sequences of Figures 15a and 15b respectively. 10 TCR a chain and TCR p chain DNA sequences were then inserted together into a Lentiviral expression vector. This vector contains DNA encoding both the parental HIV Gag TCR a chain and P chain as a single open reading frame with the in-frame Foot and Mouth Disease Virus (FMDV) 2A cleaviage factor amino acid sequence (LLNFDLLKLAGDVESNPG (SEQ ID NO: 31)) separating the TCR chains. (de 15 Felipe et al., Genet Vaccines Ther (2004) 2 (1): 13) On mRNA translation the TCR a chain is produced with the 2A peptide sequence at its C-terminus and the TCR p chain is produced as a separate polypeptide. T cells were transduced with the above Lentiviral vector. Briefly, primary T cells were 20 stimulated for 24 hours using anti-CD3/anti-CD28 beads. A concentrated Lentivirus supernatant, expressing the TCR genes, was then incubated with the stimulated T cells to allow viral transduction. The anti-CD3/anti-CD28 beads were then removed and the transduced T cells were cultured until they attained a "resting volume" of 200-300 fL. 25 Presentation of parental HIV Gag TCRs on the surface of the transduced cells was confirmed by FACS analysis using HLA-A*0201-SLYNTVALT PE tetramer and anti-CD8 monoclonal antibody FITC co-staining. 30 37 Results Figure 17b provides the FACS analysis data which demonstrates the successful expression of the parental HIV Gag TCR on the surface of transduced CD8* T cells. 5 Figure 17a provides FACS analysis data generated using control untransduced T cells. Example 7 - Inhibition of CTL activation by soluble high affinity HIV Gag TCRs The following assays were carried out to demonstrate that the soluble high affinity 10 cl 1c6 HIV Gag TCR was capable of inhibiting activation of a SLYNTVATL-HLA A*0201 reactive polyclonal T cell line. Inhibition of activation ofthe OX84 SLYNTVA TL-HLA-A *0201 reactive polyclonal T cell line in the ofpresence of HIV infected cells 15 The soluble c 1c6 high affinity HIV Gag TCR utilised in this experiment contained the TCR alpha chain variable domain and TCR beta chain variable regions shown in Figure 6c (SEQ ID NO: 13) and Figure 7b (SEQ ID NO: 15) respectively. The full amino acid sequences of the TCR alpha and beta chains of this soluble TCR are 20 provided by Figure 18a (SEQ ID NO: 29) and Figure 18b (SEQ ID NO: 30) respectively. IFN-y and TNF-a production was used as the read-outs for CTL activation. 25 Reagents RI Assay media: 10% FCS (heat-inactivated, Gibco, cat# 10108-165), 88% RPMI 1640 (Gibco, cat# 42401-018), 1% glutamine (Gibco, cat# 25030-024) and 1% penicillin/streptomycin (Gibco, cat# 15070-063). 30 38 Peptide: (obtained from various sources) initially dissolved in DMSO (Sigma, cat# D2650) at 4mg/ml and frozen. The BDTM Cytometric Bead Array Kit, Human Thl/Th2 cytokine Kit II (BD 5 Biosciences, San Diego, US) contains all the reagents required for the assay. T cell activation Assay Chronically HIV infected To target cells (HXB2 and HIV3B HIV Lab strains) were 10 washed and re-suspended in R10 media. As a control uninfected To target cells were pulsed with 1 nM of SLYNTVATL peptide, for 30 minutes at 37 0 C, 5%C0 2 . Test Samples: 25,000 HIV infected To target cells in R10 media per well of a 96 well U-bottom 15 plate. 2 x 10- M high affinity c 11c6 HIV Gag TCR or parental HIV Gag TCR in R10 media per well. 20 5000 OX84 polyclonal effector T cell line in R10 media per well. Controls: As above substituting irrelevant soluble TCRs (HLA-A* 0201 -Tax specific and HLA A*0201-NY-ESO specific TCRs) or the high affinity HIV Gag TCRs. 25 The plate was then incubated for 4 hours at 37 0 C, 5% CO 2 . The culture supernatant was removed to measure the levels of IFN-y and TNF-a present using the following method. 30 IFN-7 and TNF-a assay 39 BDTM Cytometric Beads coated with (a) anti-IFNy capture antibodies and (b) anti TNFa capture antibodies were prepared according to the manufacturers instructions A number of assay tubes were then prepared containing the following additions: 5 50 pl of mixed anti-IFNy and anti-TNFa BDTM Cytometric Beads in BD Assay Diluent 50 pl of PE Dectection Reagent 10 Followed by either: 50 pl of the culture supernatant taken from the T cell activation assay wells. (Test Samples) Or 15 50 pl of mixed IFNy and TNFa standards prepared at a range of concentrations by serial dilution of stock standards. (Calibration Standards) The tube were then incubated in the dark for 3 hours prior to being washed with 1 ml 20 of BD Wash Buffer and centrifuged. Finally, the beads were re-suspended in 300 pl of the Wash Buffer and the level of IFNy and TNFa present was determined by Flow Cytometry according to manufacturer's instructions. Inhibition of the SLYNTVATL-HLA-A *0201 specific OX84 polyclonal T line inthe 25 presence of uninfected SLYNTVATL peptide pulsed To cells The same regents and methods as used for the above CTL activation assay were used except that: 30 2000 OX84 polyclonal effector T cells were used in each T cell activation assay.
40 Uninfected To lymphoblastoid cells, pulsed with 1010 -10~' M SLYNTVATL peptide were used as the target cells Results 5 The soluble high affinity c 11c6 HIV Gag TCR strongly inhibited activation of the SLYNTVATL-HLA-A*0201 reactive OX84 polyclonal T cell line in the presence of To cells infected by HIV as measured by IFN-y and TNF-a production. (See Figure 19) 10 The soluble high affinity c 1c6 HIV Gag TCR strongly inhibited activation of the SLYNTVATL-HLA-A*0201 reactive OX84 polyclonal T cell line in the presence of SLYNTVATL-pulsed uninfected To cells as measured by IFN-y and TNF-a production. (See Figure 20) 15 Example 8 - Quantification of cell surface SLYNTVATL-HLA- A*0201 antigens on peptide-pulsed T2 cells by fluorescence microscopy using high affinity cl1c6 HIV Gag TCR The number of SLYNTVATL-HLA-A*0201 antigens on peptide-pulsed T2 20 lymphoblastoid cell was determined (on the assumption that one fluorescence signal relates to a single labelled TCR bound to its cognate pMHC ligand on the surface of the target cell) by single molecule fluorescence microscopy using a soluble high affinity c 1c6 HIV Gag TCR. This was facilitated by using biotinylated TCR to target the antigen-expressing cancer cells and subsequent labelling of cell-bound TCR by 25 streptavidin-R phycoerythrin (PE) conjugates. Individual PE molecules were then imaged by 3-dimensional fluorescence microscopy. T2 lymphoblastoid cells were pulsed with the HIV Gag-derived SLYNTVATL peptide, or an irrelevant peptide (SLLMWITQC) at a range of concentrations (10~ 30 10' 0 M) for 90 minutes at 37'C. After pulsing the cells were washed twice with 500 1d of PBS. Cells were incubated in 200 tl of TCR solution (100 nM high-affinity c1 1c6 41 HIV Gag TCR), in PBS. 0.5% BSA albumin) for 30 min at room temperature. TCR solution was removed, and cells were washed three times with 500 pl of PBS. Cells were incubated in 200 pl of streptavidin-PE solution (5 pg ml- streptavidin-PE in PBS containing 0.5% BSA) at room temperature in the dark for 20 min. Streptavidin-PE 5 solution was removed and cells were washed three times with 500 pL of PBS. Wash media was removed, and cells kept in 400 pl of RI0, without Phenol Red before imaging by fluorescence microscopy. Fluorescence microscopy. Fluorescent microscopy was carried out using an Axiovert 10 200M (Zeiss) microscope with a 63x Oil objective (Zeiss). A Lambda LS light source containing a 300W Xenon Arc lamp (Sutter) was used for illumination, and light intensity was reduced to optimal levels by placing a 0.3 and a 0.6 neutral density filter into the light path. Excitation and emission spectra were separated using a TRITC/DiI filter set (Chroma). Cells were imaged in three dimensions by z-stack acquisition (21 15 planes, 1 pm apart). Image acquisition and analysis was performed using Metamorph software (Universal Imaging) as described (Irvine et al., Nature 419: p 84 5
-
9 , and Purbhoo et al., Nature Immunology 5: p524-30). Results 20 As shown by Figure 21 the above method was used successfully to image high affinity ci 1c6 HIV Gag TCR bound to SLYNTVATL -HLA-A*0201 antigens on the surface of peptide-pulsed T2 cells. These results show the threshold for counting epitopes on SLYNTVATL peptide-pulsed cells using the high affinity c6cl 1 HIV Gag TCR is 25 approxiamately 10- M peptide.
C:\NRPoztbRDCC\AM4527388 l.DOC-9/0/2012 42 Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

Claims (7)

1. Nucleic acid encoding a heterodimeric alpha-beta T-cell receptor (TCR) comprising the TCR alpha chain SEQ ID NO:27 and the TCR beta chain SEQ ID NO:28 except that YEE in positions 69-71 of SEQ ID NO:28 is mutated to ALG.
2. A T-cell transduced with a TCR expression vector which comprises nucleic acid as claimed in claim 1.
3. A T-cell displaying on its surface a TCR expressed by nucleic acid as claimed in claim 1.
4. A heterodimeric alpha-beta T-cell receptor (TCR) comprising the TCR alpha chain variable region SEQ ID NO: 1 and the TCR beta chain variable region SEQ ID NO: 15, optionally truncated at the C-terminus and/or N-terminus thereof, by 1, 2, 3, 4, or 5 residues.
5. A TCR as claimed in claim 4 which comprises the alpha chain constant domain SEQ ID NO: 19, and/or one of the beta chain amino acid constant domain SEQ ID NOs:20 and 21.
6. A TCR as claimed in claim 4 or claim 5 which lacks transmembrane and cytoplasmic regions.
7. The nucleic acid as claimed in claim 1, or the TCR as claimed in claim 4, substantially as herein described and with reference to any of the Examples and/or Figures.
AU2013202288A 2005-04-01 2013-04-02 High affinity HIV T cell receptors Ceased AU2013202288B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013202288A AU2013202288B2 (en) 2005-04-01 2013-04-02 High affinity HIV T cell receptors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0506760.8 2005-04-01
GB0516487.6 2005-08-10
AU2012211503A AU2012211503B2 (en) 2005-04-01 2012-08-10 High affinity HIV T cell receptors
AU2013202288A AU2013202288B2 (en) 2005-04-01 2013-04-02 High affinity HIV T cell receptors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2012211503A Division AU2012211503B2 (en) 2005-04-01 2012-08-10 High affinity HIV T cell receptors

Publications (2)

Publication Number Publication Date
AU2013202288A1 AU2013202288A1 (en) 2013-05-02
AU2013202288B2 true AU2013202288B2 (en) 2015-06-18

Family

ID=48347658

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013202288A Ceased AU2013202288B2 (en) 2005-04-01 2013-04-02 High affinity HIV T cell receptors

Country Status (1)

Country Link
AU (1) AU2013202288B2 (en)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Anikeeva, N. et al., "Soluble HIV-specific T cell receptor: expression, purification and analysis of the specificity", Journal of Immunological Methods, 2003, 277, 75-86 *

Also Published As

Publication number Publication date
AU2013202288A1 (en) 2013-05-02

Similar Documents

Publication Publication Date Title
AU2006228308B2 (en) High affinity HIV T cell receptors
US8217144B2 (en) High affinity Melan-A T cell receptors
EP1885754B1 (en) T cell receptors which specifically bind to vygfvracl-hla-a24
US8017730B2 (en) T cell receptors which specifically bind to VYGFVRACL-HLA-A24
CA2566363A1 (en) High affinity ny-eso t cell receptor
WO2006054096A2 (en) Soluble bifunctional proteins
US20100068186A1 (en) High affinity telomerase t cell receptors
CN101155829A (en) High affinity HIV T cell receptors
AU2012211503B2 (en) High affinity HIV T cell receptors
AU2013202288B2 (en) High affinity HIV T cell receptors

Legal Events

Date Code Title Description
PC1 Assignment before grant (sect. 113)

Owner name: IMMUNOCORE LIMITED; ADAPTIMMUNE LIMITED

Free format text: FORMER APPLICANT(S): IMMUNOCORE LIMITED

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired