CA2582963A1 - T-cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents - Google Patents

T-cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents Download PDF

Info

Publication number
CA2582963A1
CA2582963A1 CA002582963A CA2582963A CA2582963A1 CA 2582963 A1 CA2582963 A1 CA 2582963A1 CA 002582963 A CA002582963 A CA 002582963A CA 2582963 A CA2582963 A CA 2582963A CA 2582963 A1 CA2582963 A1 CA 2582963A1
Authority
CA
Canada
Prior art keywords
antibody
tcr
therapeutic agent
gly
chain
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.)
Abandoned
Application number
CA002582963A
Other languages
French (fr)
Inventor
Bent Karsten Jakobsen
Torben Bent Andersen
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.)
Medigene Ltd
Original Assignee
Individual
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 GBGB0421836.8A external-priority patent/GB0421836D0/en
Priority claimed from GB0427584A external-priority patent/GB0427584D0/en
Application filed by Individual filed Critical Individual
Publication of CA2582963A1 publication Critical patent/CA2582963A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Abstract

The present invention provides a dimeric TCR (dTCR) or single-chain TCR
(scTCR) associated with selected therapeutic agents, wherein said TCR
comprises a first segment constituted by an amino acid sequence corresponding to a TCR .alpha. chain variable domain sequence fused to the N terminus of an amino acid sequence corresponding to a TCR .alpha. chain constant domain extracellular sequence, a second segment constituted by an amino acid sequence corresponding to a TCR ~ chain variable domain fused to the N terminus of an amino acid sequence corresponding to TCR ~ chain constant domain extracellular sequence, a disulfide bond between the first and second chains, said disulfide bond being one which has no equivalent in native .alpha..beta. T cell receptors, and in the case of said scTCRs further comprising a linker sequence linking the C terminus of the first segment to the N terminus of the second segment, or vice versa, the length of the linker sequence and the position of the disulfide bond being such that the variable domain sequences of the first and second segments are mutually orientated substantially as in native .alpha..beta. T cell receptors.

Description

'I' cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents The present invention relates to T cell receptors (TCRs) containing a non-native disulphide interchain bond associated with therapeutic agents.

Back2round to the Invention The novel TCR therapeutic combinations disclosed herein will be of use in the treatment of autoimmune disease, organ rejection, Graft Versus Host Disease (GVHD) and cancer. The TCR portion of the TCR therapeutic agent combinations disclosed herein are targeting moieties.

Brief Description of the Invention This invention makes available for the first time a dimeric TCR (dTCR) or single-chain TCR (scTCR) associated with a therapeutic agent, wherein said agent is selected from IL-1, IL-la, IL-3, IL-4, IL-5, IL-6, IL-7, IL-l0, IL-11, IL-12, IL-13, IL-15, IL-21, IL-23, TGF-(3, IFN-y, Lymphotoxin, TNFa, Anti-CD2 antibody, Anti-CD3 antibody, Anti-CD4 antibody, Anti-CD8 antibody, Anti-CD44 antibody, Anti-CD45RA antibody, Anti-CD45RB antibody, Anti-CD45RO antibody, Anti-Thy 1.2 antibody, Antilymphocyte globulin, Anti-a(3TCR antibody, Anti-y8TCR antibody, Anti-CD49a antibody, Anti-CD49b antibody, Anti-CD49c antibody, Anti-CD49d antibody, Anti-CD49e antibody, Anti-CD49f antibody, Anti-TCR V(38 antibody, Anti-CD16 antibody, Anti-CD28 antibody, CTLA-4-Ig, Anti-B7.2 antibody, Anti-CD40L antibody, Anti-ICAM-1 antibody, ICAM-1, Anti-Mac antibody, Anti-LFA-1 antibody, Anti-IFN-y antibody IFN-y, IFN-yR/IgGl fusions, Anti-IL-2R
antibodies, IL-2R antibody, IL-2 Diptheria-toxin protein, Anti-IL-12 antibody, IL-12 Antagonist (p40), Anti-IL-1 antibody, IL-1 Antagonist, Glutamic acid decarboxylase (GAD), Anti-GAD antibody, Viral proteins and peptides, Bacterial proteins or peptides, A-Galactosyl-ceramide, Calcitonin, Nicotinamide, Anti-oxidants (Vitamin E, Probucol analog, Probucol + deflazacoert or Aminoguanidine), Anti-Inflammatory agents (Pentoxifylline or Rolipram), Immunomodulators (Linomide, Ling-zhi-8, D-Glucan, Multi-functional protein 14, Ciamexon, Cholera toxin B, Vanadate or Vitamin D3 analogue, small molecule CD80 inhibitors, Androgens, IGF-1, Immunomanipulation (Natural antibodies), Lupus idiotype, Lipopolysaccaride), Sulfatide, Bee venom, Kampo formulation, Silica, Ganglioside, Antiasialo GM-1 antibody, Hyaluronidase, Concanavalin A, Anti-Class I MHC antibody, or Anti-Class II MHC antibody, Cyclosporin, FK-506, Azathioprine, Rapainycin or Deoxyspergualin, PE38 Pseudomonas exotoxin or a functional variant or fragment of any of the foregoing, and wherein said TCR comprises a first segment constituted by an amino acid sequence corresponding to a TCR a chain variable domain sequence fused to the N
terminus of an amino acid sequence corresponding to a TCR a chain constant domain extracellular sequence, a second segment constituted by an amino acid sequence corresponding to a TCR (3 chain variable domain fused to the N terminus of an arnino acid sequence corresponding to TCR P chain constant domain extracellular sequence, a disulfide bond between the first and second chains, said disulfide bond being one which has no equivalent in native a(3T cell receptors, and in the case of said scTCRs further comprising a linker sequence linking the C terrninus of the first segment to the N terminus of the second segment, or vice versa, the length of the linker sequence and the position of the disulfide bond being such that the variable domain sequences of the first and second segments are mutually orientated substantially as in native a(3 T cell receptors.

Detailed Description of the Invention The present invention provides a dimeric TCR (dTCR) or single-chain TCR
(scTCR) associated with a tlierapeutic agent, wherein said agent selected from one of IL-l, IL-la, IL-3, IL-4, IL-5, IL-6, IL-7, IL-10, IL-11, IL-12, IL-13, IL-15, IL-21, IL-23, TGF-(3, IFN-y, Lymphotoxin, TNFa, Anti-CD2 antibody, Anti-CD3 antibody, Anti-CD4 antibody, Anti-CD8 antibody, Anti-CD44 antibody, Anti-CD45RA antibody, Anti-CD45RB antibody, Anti-CD45RO antibody, Anti-Thy 1.2 antibody, Antilymphocyte globulin, Anti-a(3TCR antibody, Anti-ySTCR antibody, Anti-CD49a antibody, Anti-CD49b antibody, Anti-CD49c antibody, Anti-CD49d antibody, Anti-CD49e antibody, Anti-CD49f antibody, Anti-TCR V(38 antibody, Anti-CD16 antibody, Anti-CD28 antibody, CTLA-4-Ig, Anti-B7.2 antibody, Anti-CD40L antibody, Anti-ICAM-1 antibody, ICAM-1, Anti-Mac antibody, Anti-LFA-1 antibody, Anti-IFN-,y antibody IFN-y, IFN-yR/IgGl fusions, Anti-IL-2R antibodies, IL-2R antibody, IL-2 Diptheria-toxin protein, Anti-IL-12 antibody, IL-12 Antagonist (p40), Anti-IL-1 antibody, IL-1 Antagonist, Glutamic acid decarboxylase (GAD), Anti-GAD antibody, Viral proteins and peptides, Bacterial proteins or peptides, A-Galactosyl-ceramide, Calcitonin, Nicotinamide, Anti-oxidants (Vitamin E, Probucol analog, Probucol +
deflazacoert or Aminoguanidine), Anti-In.flammatory agents (Pentoxifylline or Rolipram), Immunomodulators (Linomide, Ling-zhi-8, D-Glucan, Multi-functional protein 14, Ciamexon, Cholera toxin B, Vanadate or Vitamin D3 analogue, small molecule inhibitors, Androgens, IGF-1, Immunomanipulation (Natural antibodies), Lupus idiotype, Lipopolysaccaride), Sulfatide, Bee venom, Kampo formulation, Silica, Ganglioside, Antiasialo GM-I antibody, Hyaluronidase, Concanavalin A, Anti-Class I MHC antibody, or Anti-Class II MHC antibody, Cyclosporin, FK-506, Azathioprine, Rapamycin or Deoxyspergualin, PE38 Pseudomonas exotoxin, or a functional variant or fragment of any of the foregoing, and wherein said TCR
comprises a first segment constituted by an amino acid sequence corresponding to a TCR a chain variable domain sequence fused to the N terminus of an amino acid sequence corresponding to a TCR a chain constant domain extracellular sequence, a second segment constituted by an amino acid sequence corresponding to a TCR (3 chain variable domain fused to the N terminus of an amino acid sequence corresponding to TCR (3 chain constant domain extracellular sequence, a disulfide bond between the first and second chains, said disulfide bond being one which has no equivalent in native a(3 T cell receptors, and in the case of said scTCRs further comprising a linker sequence linking the C terminus of the first segment to the N
terminus of the second segment, or vice versa, the length of the linker sequence and the position of the disulfide bond being such that the variable domain sequences of the first and second segments are mutually orientated substantially as in native a(3 T cell receptors.

As used herein the term "a dimeric TCR (dTCR) or single-chain TCR (seTCR) associated with an therapeutic agent" is understood to refer to a TCR
covalently or otherwise linked to an therapeutic agent. The therapeutic agent may either be directly linked to the TCR, or indirectly via a linker moiety.
As used herein the term "functional variant" is understood to refer to analogues of the disclosed therapeutic agents which have the same therapeutic effect. For example, as is known to those skilled in the art, it may be possible to produce therapeutics that incorporate minor changes in the chemical structure or amino acid sequence thereof compared to those disclosed without altering the therapeutic effect of the agents. Such trivial variants are included in the scope of this invention.

Functional antibody fYagnaents and variants Antibody fragments and variants/analogues which are suitable for use in the compositions and methods described herein include, but are not limited to, the following.

Antibody Fragments As is known to those skilled in the art, it is possible to produce fragments of a given antibody wllich retain substantially the same binding characteristics as those of the parent antibody. The following provides details of such fragments:

Minibodies - These constructs consist of antibodies with a truncated Fc portion. As such they retain the complete binding domains of the antibody from which are derived.

Fab fragments - These comprise a single immunoglobulin light chain covalently-linked to part of an immunoglobulin heavy chain. As such, Fab fragments coinprise a single antigen combining site. Fab fragments are defined by the portion of an IgG that can be liberated by treatment with papain. Such fragments are commonly produced via recombinant DNA techniques. (Reeves et al., (2000) Lecture Notes on Inznaunology (4th Edition) Published by Blackwell Science) F(ab')2 fragments - These comprise both antigen combining sites and the hinge region from a single antibody. F(ab')2 fragments are defined by the portion of an IgG
that can be liberated by treatment with pepsin. Such fragments are commonly produced via recombinant DNA techniques. (Reeves et al., (2000) Lecture Notes on Inamunology (4tth Edition) Published by Blackwell Science) Fv fragments - These comprise an immunoglobulin variable heavy domain linked to an immunoglobulin variable light domain. A number of Fv designs have been produced. These include dsFvs, in which the association between the two domains is enhanced by an introduced disulfide bond. Alternatively, scFVs can be formed using a peptide linker to bind the two domains together as a single polypeptide. Fvs constructs containing a variable domain of a heavy or light immunoglobulin chain associated to the variable and constant domain of the corresponding immunoglobulin heavy or light chain have also been produced. FV have also been multimerised to form diabodies and triabodies (Maynard et al., (2000) Annu Rev Bionzed Eng 2 339-376) NanobodiesTM - These constructs, marketed by Ablynx (Belgium), comprise synthetic single immunoglobulin variable heavy domain derived from a camelid (e.g. camel or llama) antibody.

Domain Antibodies - These constructs, marketed by Domantis (Belgium), comprise an affinity matured single immunoglobulin variable heavy domain or immunoglobulin variable light domain.
Antibody variants and analogues The defining functional characteristic of antibodies in the context of the present invention is their ability to bind specifically to a target ligand. As is known to those skilled in the art it is possible to engineer such binding characteristics into a range of other proteins. Examples of antibody variants and analogues suitable for use in the compositions and methods of the present invention include, but are not limited to, the following.
Protein scaffold-based binding polypeptides - This family of binding constructs comprise mutated analogues of proteins which contain native binding loops.
Examples include Affibodies, marketed by Affibody (Sweden), which are based on a three-helix motif derived from one of the IgG binding domains of Staphylococcus aureus Protein A. Another example is provided by Evibodies, marketed by EvoGenix (Australia) which are based on the extracellular domains of CTLA-4into which domains similar to antibody binding loops are grafted. A final example, Cytokine Traps marketed by Regeneron Pharmaceuticals (US), graft cytokine receptor domains into antibody scaffolds. (Nygren et al., (2000) Current Opinion in Structural biology 7 463-469) provides a review of the uses of scaffolds for engineering novel binding sites in proteins. This review mentions the following proteins as sources of scaffolds:
CP1 zinc finger, Tendamistat, Z domain (a protein A analogue), PST1, Coiled coils, LACI-D 1 and cytochrome b562. Other protein scaffold studies have reported the use of Fibronectin, Green fluorescent protein (GFP) and ankyrin repeats.

As is known to those skilled in the art antibodies or fragments, variants or analogues thereof can be produced which bind to various parts of a given protein ligand.
For example, anti-CD3 antibodies can be raised to any of the polypeptide chains from which this complex is formed (i.e.y, 8, s, ~, and 11 CD3 chains) Antibodies which bind to the s CD3 chain are the preferred anti-CD3 antibodies for use in the compositions and methods of the present invention.

Another aspect of the invention provides a dTCR or scTCR associated with a therapeutic agent, wherein the therapeutic agent is selected from IL-1, IL-la, IL-3, IL-5, IL-6, IL-7, IL-11, IL-12, TGF-0, Lymphotoxin, TNFa, Anti-CD2 antibody, Anti-CD4 antibody, Anti-CD8 antibody, Anti-CD44 antibody, Anti-CD45RA
antibody, Anti-CD45RB antibody, Anti-CD45RO antibody, Anti-Thy 1.2 antibody, Antilylnphocyte globulin, Anti-a(3TCR antibody, Anti-y6TCR antibody, Anti-CD49a antibody, Anti-CD49b antibody, Anti-CD49c antibody, Anti-CD49d antibody, Anti-CD49e antibody,Anti-CD49f antibody, Anti-TCR V(38 antibody, Anti-CD 16 antibody, Anti-CD28 antibody, CTLA-4-Ig, Anti-B7.2 antibody, Anti-CD40L
antibody, Anti-ICAM-1 antibody, ICAM-1, Anti-Mac antibody, Anti-LFA-1 antibody, Anti-IFN-y antibody IFN-y, IFN-yR/IgGl fusions, Anti-IL-2R
antibodies, IL-2R antibody, IL-2 Diptheria-toxin protein, Anti-IL-12 antibody, IL-12 Antagonist (p40), Anti-IL-1 antibody, IL-1 Antagonist, Glutamic acid decarboxylase (GAD), Anti-GAD antibody, Viral proteins and peptides, Bacterial proteins or peptides, A-Galactosyl-ceramide, Calcitonin, Nicotinamide, Anti-oxidants (Vitamin E, Probucol analog, Probucol + deflazacoert or Aminoguanidine), Anti-Inflammatory agents (Pentoxifylline or Rolipram), Immunomodulators (Linomide, Ling-zhi-8, D-Glucan, Multi-functional protein 14, Ciamexon, Cholera toxin B, Vanadate or Vitamin D3 analogue, small molecule CD80 inhibitors, Androgens, IGF-1, Immunomanipulation (Natural antibodies), Lupus idiotype, Lipopolysaccaride), Sulfatide, Bee venom, Kampo formulation, Silica, Ganglioside, Antiasialo GM-1 antibody, Hyaluronidase, Concanavalin A, Anti-Class I MHC antibody, or Anti-Class II MHC antibody, Cyclosporin, FK-506, Azathioprine, Rapamycin or Deoxyspergualin, or a functional variant or fragment of any of the foregoing.

"Anti-T cell" antibodies One preferred group of the immunomodulatory agents of the invention are antibodies or functional fragrnents or variants/analogues thereof which bind epitopes presented only by T cells or Natural Killer (NK) cells. The following are the antibodies which will specifically target these cells:

Anti-CD3 antibody, Anti-CD4 antibody, Anti-CD8 antibody, Anti-a(3TCR antibody, Anti-CD49a antibody, Anti-CD49b antibody, Anti-CD49c antibody, Anti-CD49d antibody, Anti-CD49e antibody, Anti-CD49f antibody, Anti-ySTCR antibody, Anti-TCR V08 antibody and Anti-CD28 antibody.

As will be known to those skilled in the art particular subsets of T cells and/or NK
cells are targeted by the majority of the above antibodies. Only anti-CD3 antibodies will target all NK cells and T cells.

Such antibodies, linked to a soluble TCR to form a bifunctional composition of the invention, will cause T cells and/or NK cells to be localised to the cells expressing the cognate peptide-MHC ligand for the soluble TCR. Without wishing to be limited by theory, the binding of these antibodies to the T cells or NK cells may cause these cells to be activated.
Another aspect of the invention provides a dTCR or scTCR associated with a therapeutic agent selected from IL-10, IL-4 or IL-13 or a functional variant or fragrnent of any of the foregoing.
One aspect of the invention is provided wherein the dTCR or scTCR is tissue-specific.
In a one embodiment of the present aspect the dTCR or scTCR is specific for a tissue which is a target for auto-reactive T cells in autoimmune disease, organ rejection or Graft Versus Host Disease (GVHD). In a specific embodiment of the present aspect the dTCR or scTCR is islet cell-specific. The T cell clones NY8.3 (Santamaria et al., J. bn7nunology (1995) 154 2494-2503) and (Nagata et al., (1995) Jlninzunologv 2042-2050) and G9C8 (Wong et al., JExp Med 1996) 183 67-76) are examples of murine T cell clones that are islet cell-specific. The NY8.3 T cell clone is specific for a glucose-6-phosphatase catalytic subunit-related protein (IGRP)-derived peptide presented by the murine H2-Kd MHC and the G9C8 T cell clone is specific for an insulin-derived peptide presented by the murine H2-Ka MHC.

A further aspect of the invention provides a dTCR or scTCR associated with a therapeutic agent, wherein the therapeutic agent is selected from IL-15, IL-21, IL-23, PE3 8 Pseudomonas exotoxin, IFN-y or Anti-CD3 antibody or a functional variant or fragrnent of any of the foregoing.

In one aspect of the invention the TCR associated with a therapeutic agent is a dTCR.
In an alternative aspect of the invention the TCR associated with a therapeutic agent is a scTCR.

There are two classes of linker that are preferred for the association of TCRs and therapeutic agents of the present invention. A TCR of the invention in which the TCR
is linked by a polyalkylene glycol chain to the therapeutic agent provides one embodiment of the present aspect. Peptidic linkers are the other class of TCR
linkers.
These two classes of linker are discussed in detail below in relation to their use in the formation of TCR multimers. Example 6 herein provides two examples of peptidic linkers which may be used to form the association between the TCR and therapeutic agent. As is known to those skilled in the art a variety of peptide linkers may be suitable to link the TCR (3 chains to the required therapeutic agents. The following are additional examples linker sequences which may be used for this purpose ggcggtccg - which encodes a Gly-Gly-Pro linker.
ccc - which encodes a Pro-Gly linker including a Xmal restriction enzyme site As mentioned above, the TCR portions of the TCR therapeutic agent combinations disclosed herein are targeting moieties. The TCRs of the invention target TCR
ligands such as peptide-MHC or CD1-antigen complexes. As such, it would be desirable if these TCR had a higher affinity and/or a slower off-rate for the TCR ligands than native TCRs specific for that ligand. The inventors co-ending application WO
2004/044004 details methods of producing TCR having a higher affinity and/or a slower off-rate for the TCR ligand than native TCRs specific for that ligand.
Preferably, the affinity (KD) of the TCR for the TCR ligand is higher than 1 M, and/or the off-rate (kOFF) is slower than 1 x 10-3 S-1. More preferably, the affinity (KD) of the TCR for the TCR ligand is higher than 10nM, and/or the off-rate (koff) is slower than 1 x 10"4 S-1. Most preferably, the affmity (KD) of the TCR for the TCR
ligand is higher than 1nM, and/or the off-rate (koff) is slower than 1 x 10"5 S-1.
The affinity (KD) and/or off-rate (koff) measurement can be made by any of the known methods. A preferred method is the Surface Plasmon Resonance (Biacore) method of Example 3.

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
03/020763.

A suitable scTCR form comprises a first segment constituted by an amino acid sequence corresponding to a TCR a chain variable domain, a second segment constituted by an amino acid sequence corresponding to a TCR P chain variable domain sequence fused to the N terminus of an amino acid sequence corresponding to a TCR (3 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.
Alternatively the first segment may be constituted by an amino acid sequence corresponding to a TCR (3 chain variable domain, the second segment may be constituted by an amino acid sequence corresponding to a TCR a chain variable domain sequence fused to the N terminus of an amino acid sequence corresponding to a TCR a chain constant domain extracellular sequence More specifically the first segment rnay be constituted by an amino acid sequence corresponding to a TCR a chain variable domain sequence fused to the N
terminus of 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 (3 chain variable domain 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 disulfide bond being one which has no equivalent in native a(3 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)5-P- (SEQ ID NO: 1) or -PGGG-(SGGGG)6-P- (SEQ ID NO: 2) wherein P
is proline, G is glycine and S is serine.

A suitable dTCR form of the TCRs of the present invention coinprises a first polypeptide wherein a sequence corresponding to a TCR a chain variable domain 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 var-iable domain sequence fused to the N
terminus a sequence corresponding to a TCR (3 chain constant domain extracellular sequence, the first and second polypeptides being linked by a disulfide bond which has no equivalent in native a(3 T cell receptors.
The first polypeptide may comprise a TCR a chain variable domain 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 domain sequence is fused to the N terminus a sequence corresponding to a TCR (3 chain constant domain extracellular sequence, the first and second polypeptides being linked by a disulfide bond between cysteine residues substituted for Thr 48 of exon 1 of TRAC*01 and Ser 57 of exon 1 of TRBCI*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) The dTCR or scTCR form of the TCRs of the invention may have amino acid sequences corresponding to human a(3 TCR extracellular constant and variable domain 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 residues whose (3 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 TIRAC*01 and Ser 57 of exon 1 of TRBC1*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 TRBC1'1401 or TRBC2*01 for the TCR
(3 chain:

TCR a chain TCR (3 chain Native (3 carbon separation (nm) Thr 45 Ser 77 0.533 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
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.
One embodiment of the invention provides a TCR associated with a therapeutic agent, wherein said therapeutic agent is a PE38 exotoxin.
PE38 exotoxin is a truncated form of a Pseudomonas exotoxin. The native polypeptide is a 66kDa protein consisting of domains IA, II, IB and III. The derivative consists of domain II, amino acids 380-399 of domain IB and domain III.
As will be obvious to those skilled in the art other triuicated forms of Pseudomonas exotoxin may be of use in the present invention. (For example PE40).The preferred variant of PE38 for use in the present invention contains mutations in the domain III
thereof such that the C-terminus amino acids are KDEL. These C-terminal mutations have previously been shown to increase the toxicity of the Pseudomona.s exotoxin.
(Kreitman et al (1995) JBiochem 307 29-37) In a preferred embodiment said TCR associated with a PE38 exotoxin comprises the amino acid sequences of (SEQ ID NO: 73) and (SEQ ID NO: 71). (Figures 29b and 28b respectively).

PEGylated TCR Monomers In one particular embodiment a TCR associated with a therapeutic agent 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 repeating units.
1Vlultivalent TCR Complexes One aspect of the invention provides a multivalent TCR complex comprising at least two TCRs associated with a therapeutic agent. In one embodiment of this aspect, at least two TCR molecules are linked via linker moieties to form multivalent complexes. Such multivalent TCR complexes may be linked by either a non-peptidic polymer chain or a peptidic linker sequence. 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 defmed 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.
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.

Examples of linker moieties which fulfil the above desirable criteria are known in the art, for example the art of linking antibody fragments.

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 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.

HOCH2CH2O (CH2CH2O)n CH2CH2OH
Wherein n is greater than two. However, others are based on other suitable, optionally substituted, polyalkylene glycols include polypropylene glycol, and copolymers of 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.. 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 4 244) The size of the hydrophilic polymer used my in particular be selected on the basis of the intended therapeutic use of the TCR complex. Thus for example, where the product is intended to leave the circulation and penetrate tissue, for example for use in the treatment of a tumour, it may be advantageous to use low molecular weight polymers in the order of 5 KDa. There are numerous review papers and books that detail the use of PEG and similar molecules in pharmaceutical formulations.
For example, see Harris & Zalipsky (1997) Chemistry and Biological Applications of Polyethylene Glycol ACS Books, Washington, D.C.

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.

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 below:

Reactive chemistry-Hydrophilic polymer-Reactive chemistry Reactive chemistry-Spacer-Hydrophilic polymer-Spacer-Reactive chemistry 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:

-(CH2)õ- wherein n= 2 to 5 -(CH2)3NHCO(CH2)2 A multivalent 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 associated with a therapeutic agent provides a further embodiment of the present aspect.

A multivalent TCR complex of the invention in which the polyalkylene glycol chain coinprises at least two polyethylene glycol repeating units provides a further embodiment of the present aspect.

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 chemistry is largely dependant on the desired coupling site. For example N-maleimide, Vinyl sulfone, Benzotriazole carbonate, Succinimidyl proprionate, Succinimidyl butanoate, Thio-ester, Acetaldehyde, Acrylate, Biotin and Primary amine coupling chemistries have been used attached to one or more of the termini of PEG molecules (Source: Nektar Molecular Engineering Catalogue 2003):
As stated above non-PEG based polymers also provide suitable linkers for multimerising the TCRs of the present invention. For example, moieties containing maleimide termini linlced 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 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.
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. 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. Biol. Chem. 276 (17) 14385-14392) Haemoglobin also has a tetramerisation domain that could potentially be used for this kind of application.
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).
Therapeutic Use The invention also provides a method for delivering a therapeutic agent to a target cell, which method comprises contacting potential target cells with a TCR or multivalent TCR complex in accordance with the invention under conditions to allow attachment of the TCR or multivalent TCR complex to the target cell, said TCR
or multivalent TCR complex being specific for a given peptide-MHC complex.

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, for example, against tumours or sites of autoiminune disease. A therapeutic agent could be delivered such that it would exercise its effect locally but not only on the cell to which it binds.
Thus, one particular strategy envisages iinmunostimulatory molecules linked to TCRs or multivalent TCR complexes according to the invention specific for tumour antigens. For cancer treatment, the localisation in the vicinity of tumours or metastasis would enhance the effect of toxins or immunostimulants. Alternatively, the soluble TCR or multivalent TCR complex of the present invention can be used to deliver immunoinhibitory agents to the location of cells presenting a particular antigen related to an autoiminune disease. For example, an Islet cell-specific TCR could be used to deliver an immunoinhibitory agent, such as IL- 10, IL-4 or IL- 13 or a functional variant or fragment of any of the foregoing to the Islet cells of a patient suffering from diabetes.

For vaccine delivery, the vaccine antigen could be localised in the vicinity of antigen presenting cells, thus enliancing the efficacy of the antigen.

It is envisaged that the administration of an interferon (IFN), such as IFN-y, to a patient prior to, and/or simultaneously with, the administration of the TCR
associated with a therapeutic agent may increase levels of peptide-MHC expression on the target cells. This may be of particular benefit in the treatment of cancer.

Further embodiments of the invention are provided by a pharmaceutical composition comprising a TCR associated with a therapeutic agent or a multivalent TCR
complex thereof together with a pharmaceutically acceptable carrier.

The invention also provides a method of treatment of cancer comprising administering to a subject suffering such cancer disease an effective amount of a TCR
associated with a therapeutic agent or a multivalent TCR complex thereof. In a related embodiment the invention provides for the use of a TCR associated with a therapeutic agent or a multivalent TCR complex thereof, in the preparation of a composition for the treatment of cancer. IL-15, IL-21 or Anti-CD3 antibody or a functional variant or fragment of the foregoing, are particularly preferred therapeutic agents for use in the treatment of cancer.
The invention also provides a method of treatment of autoimmune disease, organ rejection or GVHD comprising administering to a subject suffering such an autoimmune disease, organ rejection or GVHD an effective amount of a TCR
associated with a therapeutic agent or a multivalent TCR complex thereof. In a related embodiment the invention provides for the use of a TCR associated with a therapeutic agent or a multivalent TCR complex thereof, in the preparation of a composition for the treatment of autoimmune disease, organ rejection or GVHD. Preferred therapeutic agents for use in the treatment of autoiminune disease, organ rejection or GVHD are IL- 10, IL-4 and IL- 13 or a functional variant or fragment of any of the foregoing. In another related embodiment the dTCR or scTCR of the invention is tissue-specific. In further related embodiment the dTCR or scTCR is specific for a tissue which is a target for auto-reactive T
cells in autoimmune disease, organ rejection or Graft Versus Host Disease (GVHD). In a specific embodiment the invention provides a method of treating diabetes, wherein the dTCR or scTCR is islet cell-specific.

Cancers which may benefit the methods of the present invention include:
leukaemia, head, neck, lung, breast, colon, cervical, liver, pancreatic, ovarian and testicular) Auto-immune diseases which may benefit the methods of the following invention include:
Acute disseminated encephalomyelitis Adrenal insufficiency Allergic angiitis and granulomatosis Amylodosis Ankylosing spondylitis Asthma Autoimmune Addison's disease Autoimmune alopecia Autoimmune chronic active hepatitis Autoimmune haemolytic anaemia Autoimmune Neutrogena Autoimmune thrombocytopenic purpura Behget's disease Cerebellar degeneration Chronic active hepatitis Chronic inflammatory demyelinating polyradiculoneuropathy Chronic neuropathy with monoclonal gaminopathy Classic polyarteritis nodosa Congenital adrenal hyperplasia Cryopathies Dermatitis herpetiformis Diabetes Eaton-Lambert myasthenic syndrome Encephalomyelitis Epidermolysis bullosa acquisita Erytherna nodosa Gluten-sensitive enteropathy Goodpasture's syndrome Guillain-Barre syndrome Hashirnoto's thyroiditis Hyperthyroidism Idiopathic hemachromatosis Idiopathic membranous glomerulonephritis Isolated vasculitis of the central nervous system Kawasaki's disease Miniinal change renal disease Miscellaneous vasculitides Mixed connective tissue disease Multifocal motor neuropathy with conduction block Multiple sclerosis Myasthenia gravis Opsoclonus-myoclonus syndrome Pemphigoid Pemphigus pernicious anaemia Polymyositis/dermatomyositis Post-infective arthritides Primary biliary sclerosis Psoriasis Reactive arthritides Reiter's disease Retinopathy Rheumatoid arthritis Sclerosing cholangitis Sjogren's syndrome Stiff-man syndrome Subacute thyroiditis Systemic lupus erytllematosis Systeinic necrotizing vasculitides Systemic sclerosis (scleroderma) Takayasu's arteritis Temporal arteritis Thromboangiitis obliterans Type I and type II autoil=une polyglandular syndrome Ulcerative colitis Uveitis Wegener's granulomatosis Therapeutic compositions in accordance with the invention will usually be supplied as part of a sterile, phaimaceutical composition which will normally include a pharmaceutically acceptable carrier. This pharmaceutical composition may be in any suitable form, (depending upon the desired metliod 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) include instructions for use. It may include a plurality of said unit dosage forms.
The pharmaceutical composition may be adapted for administration by any appropriate route, for example pareiiteral, 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.

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 ultiunately determine appropriate dosages to be used.

Additiorzal Aspects A scTCR or dTCR associated with a therapeutic agent (which TCR preferably is constituted by constant and variable sequences corresponding to human sequences) may be provided in substantially pure form, or as a purified or isolated preparation. For example, it may be provided in a form which is substantially free of otlier proteins.
Preferred features of eac11 aspect of the invention are as for each of the other aspects inutatis mutandis. The prior art documents mentioned herein are incorporated to the fullest extent permitted by law.

Examples The invention is further described in the following examples, which do not limit the scope of the invention in any way.

Reference is made in the following to the accompanying drawings in which:

Figures 1a and lb show respectively the nucleic acid sequences of the a and P
chains of a soluble A6 TCR, mutated so as to introduce a cysteine codon. The shading indicates the introduced cysteine codon;
Figure 2a shows the A6 TCR a chain extracellular amino acid sequence, including the T48 -> C mutation (underlined) used to produce the novel disulphide inter-chain bond, and Figure 2b shows the A6 TCR (3 chain extracellular amino acid sequence, including the S57 -> C mutation (underlined) used to produce the novel disulphide inter-chain bond;

Figure 3a shows the A6 TCR a chain sequence including novel cysteine residue mutated to incorporate a BamHl restriction site. Shading indicates the mutations introduced to form the BamHl restriction site.

Figures 3b and 3c show the DNA sequence of a and (3 chain of the JM22 TCR
mutated to include additional cysteine residues to form a non-native disulphide bond;
Figures 4a and 4b show respectively the J1VI22 TCR a and (3 chain extracellular amino acid sequences produced from the DNA sequences of Figures 3b and 3c;

Figures 5a and 5b show respectively the DNA sequences of the a and (3 chains of a soluble AH-1.23 TCR, mutated so as to introduce a novel cysteine codon (indicated by shading).

Figures 6a and 6b show respectively the AH-1.23 TCR a and (3 chain extracellular amino acid sequences produced from the DNA sequences of Figures 5a and 5b;

Figure 7a - DNA sequence of mature human IL-10.

Figure 7b - Amino acid sequence of mature human IL-10.

Figure 8a - DNA sequence of AH1.23 TCR 0 chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-via a Pro-Gly linker. The introduced cysteine is indicated by shading. The DNA
sequence encoding the Pro-Gly linker is underlined.
Figure 8b - Amino acid sequence of AH1.23 TCR (3 chain corntaining a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-10 via a Pro-Gly linker. The introduced cysteine is indicated by shading.

The Pro-Gly linlcer is underlined.

Figure 9a - DNA sequence of AH1.23 TCR (3 chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-via a Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading.
The DNA sequence encoding the Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 9b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature 1luman IL-10 via a Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading. The Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 10a - DNA sequence of AH 1.23 TCR P chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-via a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker.
The introduced cysteine is indicated by shading. The DNA sequence encoding the Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure lOb - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-10 via a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading. The Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 11 a - DNA sequence of mature human IL-4.
Figure 1 1b - Amino acid sequence of mature human IL-4.
Figure 12a - DNA sequence of AH1.23 TCR (3 chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-4 via a Pro-Gly linker. The introduced cysteine is indicated by shading. The DNA
sequence encoding the Pro-Gly linker is underlined.

Figure 12b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-4 via a Pro-Gly linker. The introduced cysteine is indicated by shading. The Pro-Gly linker is underlined.

Figure 13 a - DNA sequence of AH 1.23 TCR (3 chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-4 via a Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading.
The DNA sequence encoding the Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 13b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-4 via a Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading. The Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 14a - DNA sequence of AHl.23 TCR 0 chain containing a non-native cysteine involved in the fonnation of a novel interchain bond linked to mature human IL-4 via a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker. 'The introduced cysteine is indicated by shading. The DNA sequence encoding the Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 14b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-4 via a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading. The Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker is underlined.
Figure 15a - DNA sequence of mature human IL-13.

Figure 15b - Amino acid sequence of mature human IL-13.

Figure 16a - DNA sequence of AH1.23 TCR P chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-via a Pro-Gly linker. The introduced cysteine is indicated by shading. The DNA
sequence encoding the Pro-Gly linker is underlined.

Figure 16b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-13 via a Pro-Gly linker. The introduced cysteine is indicated by shading.
The Pro-Gly linker is underlined.

Figure 17a - DNA sequence of AH1.23 TCR 0 chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-via a Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading.
The DNA sequence encoding the Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 17b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-13 via a Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading. The Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 18 a - DNA sequence of AH1.23 TCR (3 chain containing a non-native cysteine involved in the formation of a novel interchain bond linked to mature human IL-via a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker.
The introduced cysteine is indicated by shading. The DNA sequence encoding the Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 18b - Amino acid sequence of AH1.23 TCR (3 chain containing a non-native cysteine codon involved in the formation of a novel interchain bond linked to mature human IL-13 via a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker. The introduced cysteine is indicated by shading. The Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro linker is underlined.

Figure 19 details the DNA sequence of the pEX821 plasmid.

Figure 20 provides a plasmid map of the pEX821 vector, the DNA sequence of which is provided by Figure 19.

Figure 21 details the DNA sequence of the pEX954 plasmid.

Figure 22 provides a plasmid map of the pEX954 plasmid, the DNA sequence of which is provided by Figure 21.

Figure 23a details the DNA sequence encoding the higli affinity c61 NY-ESO
MTCR
beta chain and Figure 23b details the AA sequence encoded by the DNA sequence of Figure 23a.

Figure 24a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to IL-18.
Figure 24b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 25 a details the DNA sequence encoding IL-18 pro-protein linked at the C-terminus thereof via a peptide linker to the high affinity c61 NY-ESO MTCR
beta chain. The pro-IL- 18 DNA has been altered to encode a Factor X cleavage site.
Figure 25b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 26a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to IL-10.
Figure 26b details the AA sequence of this fusion protein, the peptide linker is underlined.
Figure 27a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to IL-13.
Figure 27b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 28a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to the "KDEL"
variant of the PE38 exotoxin. Figure 28b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 29a details the DNA sequence encoding the high affinity c58 NY-ESO MTCR
alpha chain and Figure 29b details the AA sequence encoded by the DNA sequence of Figure 29a.

Exatnple 1 - Design ofprimers and mutagenesis ofA6 Tax TCR a and,Q chains For mutating A6 Tax threonine 48 of exon 1 in TRAC*Olto cysteine, the following primers were designed (mutation shown in lower case):

5'-C ACA GAC AAA tgT GTG CTA GAC AT (SEQ ID NO: 3) 5'-AT GTC TAG CAC Aca TTT GTC TGT G (SEQ ID NO: 4) For inutating A6 Tax serine 57 of exon 1 in both TRBC1*01 and TRBC2*01 to cysteine, the following priiners were designed (mutation shown in lower case):
5'-C AGT GGG GTC tGC ACA GAC CC (SEQ ID NO: 5) 5'-GG GTC TGT GCa GAC CCC ACT G (SEQ ID NO: 6) PCR nzutagenesis:
Expression plasmids containing the genes for the A6 Tax TCR a or (3 chain were mutated using the a-chain primers or the (3-chain primers respectively, as follows.
100 ng of plasmid was mixed with 5 l 10 mM dNTP, 25 l lOxPfu-buffer (Stratagene), 10 units Pfu polymerase (Stratagene) and the final volume was adjusted to 240 l with H20. 48 l of this mix was supplemented with primers diluted to give a final concentration of 0.2 M in 50 l final reaction volume. After an initial denaturation step of 30 seconds at 95 C, the reaction mixture was subjected to rounds of denaturation (95 C, 30 sec.), annealing (55 C, 60 sec.), and elongation (73 C, 8 min.) in a Hybaid PCR express PCR machine. The product was then digested for 5 hours at 37 C with 10 units of Dpnl restriction enzyme (New England Biolabs). 10 l of the digested reaction was transformed into competent XL1-Blue bacteria and grown for 18 hours at 37 C. A single colony was picked and grown over night in 5 ml TYP + ampicillin (16 g/l Bacto-Tryptone, 16 g/l Yeast Extract, 5 g/l NaCI, 2.5 g/l K2HPO4, 100 mg/l Ampicillin). Plasmid DNA was purified on a Qiagen mini-prep column according to the manufacturer's instructions and the sequence was verified by automated sequencing. The respective mutated nucleic acid and amino acid sequences are shown in Figures la and 2a for the a chain and Figures lb and 2b for the (3 chain.

Exainple 2 - Expression, refolding and purification of soluble TCR

The expression plasmids containing the mutated a-chain and (3-chain respectively were transformed separately into E.coli strain BL21pLysS, and single ampicillin-resistant colonies were grown at 37 C in TYP (ampicillin 100 g/ml) medium to OD600 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) NaAzide, 10mM DTT, pH 8Ø
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 membrane components. Each time the inclusion body pellet was homogenised in a Triton buffer (50mM Tris-HCI, 0.5% Triton-X100, 200mM NaCI, 10mM NaEDTA, 0.1% (w/v) NaAzide, 2mM DTT, pH 8.0) before being pelleted by centrifugation for 15 minutes at 13000rpm in a Beckman J2-21. Detergent and salt was then removed by a similar wash in the following buffer: 50mM Tris-HCI, 1mM NaEDTA, 0.1% (w/v) NaAzide, 2mM DTT, pH 8Ø Finally, the inclusion bodies were divided into 30 mg aliquots and frozen at -70 C. Inclusion body protein yield was quantitated by solubilising with 6M guanidine-HCl and measurement with a Bradford dye-binding assay (PerBio).
Denaturation of soluble TCRs; 30mg of the solubilised TCR 0-chain inclusion body and 60mg of the solubilised TCR a-chain inclusion body was thawed from frozen stocks. The inclusion bodies were diluted to a final concentration of 5mg/ml in 6M
guanidine solution, and DTT (2M stock) was added to a final concentration of 10mM.
The mixture was incubated at 37 C for 30 min.
Ref lding of soluble TCRs: 1 L refolding buffer was stirred vigorously at 5 C
3 C.
The redox couple (2-mercaptoethylamine and cystamine (to final concentrations of 6.6inM and 3.7mM, respectively) were added approximately 5 minutes before addition of the denatured TCR chains. The protein was then allowed to refold for approximately 5 hours 15 minutes with stirring at 5 C :L 3 C.
Dialysis of refolded soluble TCRs: The refolded TCR was dialysed in Spectrapor membrane (Spectrum; 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.
Example 3 - BIAcore surface plasmon resonance characterisation of sTCR binding to 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-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.
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 binding of TCR is obtained even at low concentrations (at least 40 g/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 and also suggests that biotinylated pMHC complexes are biologically as active as non-biotinylated complexes.

Biotinylated class I HLA-A2 - peptide complexes 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. Bioehem. 266: 9-15). HLA-heavy chain was expressed with a C-terminal biotinylation tag which replaces the transmembrane and cytoplasrnic domains of the protein in an appropriate construct. Inclusion body expression levels of -75 mg/litre bacterial culture were obtained. The HLA
light-chain or (32-microglobulin was also expressed as inclusion bodies in E.coli from an appropriate construct, at a level of -500 mg/litre bacterial culture.

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 naM NaCI, 10 mM DTT, 10 mM EDTA, and was refolded at a concentration of 30 mg/litre heavy chain, 30 mg/litre (32in into 0.4 M L-Arginine-HCI, 100 mM Tris pH 8.1, 3.7 m1VI cystamine, mM cysteamine, 4 mg/ml peptide (e.g. tax 11-19), by addition of a single 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 of buffer were necessary to reduce the ionic strength of the solution sufficiently. The protein solution was then filtered through a 1.5 m cellulose acetate filter and loaded onto a POROS 50HQ anion exchange column (8 ml bed voluine). Protein was eluted with a linear 0-500 mM NaCl gradient. HLA-A2-peptide complex eluted at approximately 250 mM NaCI, and peak fractions were collected, a cocktail of protease inhibitors (Calbiochem) was added and the fractions were chilled on ice.
Biotinylation tagged HLA complexes were buffer exchanged into 10 mM Tris pH
8.1, 5 mM NaCI using a Pharmacia fast desalting column equilibrated in the same buffer.
Immediately upon elution, the protein-containing fractions were chilled on ice 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 MgC12, and 5 g/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.
Biotinylated HLA complexes 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 HLA coinplexes eluted as a single peak at approximately 15 ml. Fractions containing 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 HLA complexes were stored frozen at -20 C. Streptavidin was immobilised by standard amine coupling methods.

The interactions between A6 Tax sTCR containing a novel inter-chain bond and its ligand/ MHC complex or an irrelevant HLA-peptide combination, the production of which is described above, were analysed on a BlAcore 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-peptide complexes in separate flow cells via binding between the biotin cross linked onto (32m 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. Initially, the specificity of the interaction was verified by passing sTCR at a constant flow rate of 5 1 min-1 over two different surfaces; one coated with -5000 RU of specific peptide-HLA complex, the second coated with -5000 RU of non-specific peptide-HLA complex. Injections of soluble sTCR at constant flow rate and different concentrations over the peptide-HLA complex were used to define the background resonance. The values of these control measurements were subtracted from the values obtained with specific peptide-HLA complex and used to calculate binding affinities expressed as the dissociation constant, Kd (Price &
Dwek, Principles and Problems in Physical Chemistry for Biochemists (2 a Edition) 1979, Clarendon Press, Oxford).
The Kd value obtained (1.8 M) is close to that reported for the interaction between A6 Tax sTCR without the novel di-sulphide bond and pMHC (0.91 M - Ding et al, 1999, Inzmunity 11:45-56).

Exanaple 4- Production of soluble JM22 TCR c ntaining a novel disulphide bond.
The (3 chain of the soluble A6 TCR prepared in Example 1 contains in the native sequence a BglII restriction site (AAGCTT) suitable for use as a ligation site.

PCR mutagenesis was carried as detailed below to introduce a BamHl restriction site (GGATCC) into the a chain of soluble A6 TCR, 5' of the novel cysteine codon.
The sequence described in Figure 1 a was used as a template for this mutagenesis.
The following primers were used:

IBamxI I
5'-ATATCCAGAACCCgGAtCCTGCCGTGTA- 3'(SEQ ID NO: 7) 5' -TACACGGCAGGAaTCcGGGTTCTGGATAT-3' (SEQ IDNO: 8) 100 ng of plasmid was mixed with 5 l 10 mM dNTP, 25 l l OxPfu-buffer (Stratagene), 10 units Pfu polymerase (Stratagerxe) and the final volume was adjusted to 240 l with H2O. 48 l of this mix was supplemented with primers diluted to give a final concentration of 0.2 M in 50 l final reaction volume. After an initial denaturation step of 30 seconds at 95 C, the reaction mixture was subjected to rounds of denaturation (95 C, 30 sec.), annealing (55 C, 60 sec.), and elongation (73 C, 8 min.) in a Hybaid PCR express PCR machine. The product was then digested for 5 hours at 37 C with 10 units of Dpnl restriction enzyme (New England Biolabs). 10 l of the digested reaction was transformed into competent XL1 -Blue bacteria and grown for 18 hours at 37 C. A single colony was picked and grown over night in 5 ml TYP + ampicillin (16 g/1 Bacto-Tryptone, 16 g/l Yeast Extract, 5 g/l NaCI, 2.5 g/1 K2HPO4, 100 mg/1 Ainpicillin). Plasmid DNA was purified on a Qiagen mini-prep colunm according to the manufacturer's instructions and the sequence was verified by automated sequencing. The mutations introduced into the a chain were "silent", therefore the amino acid sequence of this chain remained unchanged from that detailed in Figure 2a. The DNA sequence for the mutated a chain is shown in Figure 3a.

In order to produce a soluble JM22 TCR incorporating a novel disulphide bond, TCR plasmids containing the a chain BamHl and (3 chain Bg11I restriction sites were used as templates. The following primers were used:

I Nde1 1 5'-GGAGATATACATATGCAACTACTAGAACAA-3'(SEQ IDN :9) 5'-TACACGGCAGGATCCGGGTTCTGGATATT- 3'(SEQ ID NO : 10) BamHII

~Nde1 ~

5 ' -GGAGATATACATATGGTGGATGGTGGAATC-3 ' (SEQ ID NO: 11) 5'-CCCAAGCTTAGTCTGCTCTACCCCAGGCCTCGGC-3'(SEQ ID NO: 12) 1 sglII1 JM22 TCR a and (3-chain constructs were obtained by PCR cloning as follows.
PCR reactions were performed using the primers as shown above, and templates containing the JM22 TCR chains. The PCR products were restriction digested with the relevant restriction enzymes, and cloned into pGMT7 to obtain expression plasmids. The sequence of the plasmid inserts were confirmed by automated DNA
sequencing. Figures 3b and 3c show the DNA sequence of the mutated a and (3 cliains of the JM22 TCR respectively, and Figures 4a and 4b show the resulting amino acid sequences.

The respective TCR chains were expressed, co-refolded and purified as described in Exainples 1 and 2.

A Biacore analysis of the binding of the JM22 TCR to pMHC was carried out as described in Example 3. The Kd of this disulphide-linked TCR for the HLA-flu complex was determined to be 7.9 0.51 M

Example S- Production of soluble AH-1.23 TCR containing a novel disulphide intey--chain bond cDNA encoding AH-1.23 TCR was isolated from T cells supplied by Hill Gaston (Medical School, Addenbrooke's Hospital, Cambridge) according to known techniques. cDNA encoding NY-ESO TCR was produced by treatment of the mRNA
with reverse transcriptase.

In order to produce a soluble AH-1.23 TCR incorporating a novel disulphide bond, TCR plasmids containing the a chain BamHI and (3 chain BglII restriction sites were used as a framework as described in Example 4. The following primers were used:

I Ndel 1 5'-GGGAAGCTTACATATGAAGGAGGTGGAGCAGAATTCTGG-3'(SEQID NO: 13) 5'-TACACGGCAGGATCCGGGTTCTGGATATT- 3'(SEQ ID NO: 14) BamHil Ndei 5'-TTGGAATTCACATATGGGCGTCATGCAGAA.CCCAAGACAC-3 (SEQ ID NO: 15) 5' -CCCAAGCTTAGTCTGCTCTACCCCAGGCCTCGGC-3' (SEQ ID NO: 16) lBglIII
AH-1.23 TCR a and (3-chain constructs were obtained by PCR cloning as follows.
PCR reactions were performed using the primers as shown above, and templates containing the AH-1.23 TCR chains. The PCR products were restriction digested with the relevant restriction enzymes, and cloned into pGMT7 to obtain expression plasmids. The sequence of the plasmid inserts were confirmed by automated DNA
sequencing. Figures 5a and 5b show the DNA sequence of the mutated a and (3 chains of the AH-1.23 TCR respectively, and Figures 6a and 6b show the resulting amino acid sequences.

The respective TCR chains were expressed, co-refolded and purified as described in Example 2.

Exanaple 6- Production of a soluble AH-1.213 TCR - IL-1 D fusion protein.

Synthetic genes including the mature human IL-10 DNA sequence detailed in Figure 7a and one of a number of DNA extensions at the 5' end of the IL- 10 DNA
sequence can then be produced. The 5' DNA extensions are linker sequences used to attach the IL-10 DNA to that encoding the AH1.23 TCR (3 chain.

Linker sequences:

ccc - which encodes a Pro-Gly linker including a Xmal restriction enzyme site ggatccggcggtccg - (SEQ ID NO: 17) which encodes a Gly-Ser-Gly-Gly-Pro (SEQ ID
NO: 18) linker including a BamHl restriction enzyme site.
ggatccggtgggggcggaagtggaggcagcggtg.g_atccggcggtccg - (SEQ ID NO:19) which encodes a Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro (SEQ ID NO: 20) linker including two BamHl restriction enzyme sites.

One of the above synthetic genes is then sub-cloned into the pGMT7 plasmid containing the AH1.23 TCR P chain, produced as described in Example 5 to form a DNA sequence encoding the TCR (3 chain-linker-IL-10 fusion protein.
The DNA and amino acid sequence of the AH1.23 TCR (3 chain - Pro-Gly - IL- 10 fusion is detailed in Figures 8a and 8b respectively.

The DNA and amino acid sequence of the AH1.23 TCR (3 chain - Gly-Ser-Gly-Gly-Pro (SEQ ID NO: 18) - IL-10 fusion is detailed in Figures 9a and 9b respectively.
The DNA and amino acid sequence of the AH1.23 TCR P chain - Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly -Pro- (SEQ ID NO: 20) IL- 10 fusion is detailed in Figures 10a and 10b respectively.

These AH1.23 TCR (3 chain - Linker- IL-10 fusion proteins are then refolded with the AH1.23 TCR a chain using the methods detailed in Example 2 to produce the coinplete soluble AH1.23 TCR-IL-10 fusion protein.

The methods detailed above describe the production of a soluble a(3TCR onto which an IL-10 monomer is attached to the C-terminus of the TCR P chain. IL-10 is often found in the form of a homodimer. Therefore, it may be advantageous to dimerise the IL-10 polypeptide attached to the soluble AH1.23 TCR. This can be achieved in a number of ways. For example, a single-chain version of the mature form human homodimer can be fused to the TCR 0 prior to refolding with the TCR a chain.
Alternatively, mature form human IL-10 can be added in solution to either the TCR (3 Chain-IL-10 fusion proteins formed as described above prior to refolding with the soluble TCR a chain, or to the refolded a(3TCR-IL-10 fusion proteins.
Alternatively, an additional IL-10 molecule can be added to the TCR a chain as a fusion protein using the methods described in this example for the production of the TCR P
chain -IL- 10 fusion protein. The two TCR chain-IL- 10 fusion proteins can then be re-folded together using the methods described in Example 2. Finally, complexes comprising two TCR, each containing a single IL-10 polypeptide linked to the TCR P chain, may be formed by homo-dimerisation of the IL-10 polypeptides. This would result in the formation of a complex of the following type:
a(3TCR-IL-10 homodimer-a(3TCR
Exafnple 7- Production of soluble AH-1.23 TCR - IL-4 and AH-1.23 TCR - IL-13 fusion proteins.

The methods detailed in Example 6 can also be used to produce fusion proteins containing the soluble AH-1.23 TCR linked to other polypeptides.

Synthetic genes including the mature human IL-4 DNA sequence detailed in Figure 11a and one of the 5' DNA extension sequences listed in Example 6 can be constructed and sub-cloned into the pGMT7 plasmid containing the AH1.23 TCR (3 chain, produced as described in Example 5 to form a DNA sequence encoding the TCR (3 chain-linker- IL-4 fusion proteins.

The DNA and amino acid sequence of the AH1.23 TCR (3 chain - Pro-Gly - IL-4 fusion is detailed in Figures 12a and 12b respectively.
The DNA and amino acid sequence of the AH1.23 TCR (3 chain - Gly-Ser-Gly-Gly-Pro- (SEQ ID NO: 18) - IL-4 fusion is detailed in Figures 13a and 13b respectively.
The DNA and amino acid sequence of the AH1.23 TCR (3 chain - Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro-(SEQ ID NO: 20)- IL-4 fusion is detailed in Figures 14a and 14b respectively.

Synthetic genes including the mature human IL-13 DNA sequence detailed in Figure 15a and one of the 5' DNA extension sequences listed in Example 6 can be constructed and sub-cloned into the pGMT7 plasmid containing the AH1.23 TCR (3 chain, produced as described in Example 5 to form a DNA sequence encoding the TCR (3 chain-linker- IL-4 fusion proteins.

The DNA and amino acid sequence of the AH1.23 TCR 0 chain - Pro-Gly - IL-13 fusion is detailed in Figures 16a and 16b respectively.

The DNA and amino acid sequence of the AH1.23 TCR 0 chain - Gly-Ser-Gly-Gly-Pro (SEQ ID NO: 18)- IL-13 fusion is detailed in Figures 17a and 17b respectively.
The DNA and amino acid sequence of the AH1.23 TCR 0 chain - Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Ser-Gly-Gly-Pro- (SEQ ID NO: 20)- IL-13 fusion is detailed in Figures 18a and 18b respectively.

These AH1.23 TCR (3 chain - Linker- interleukin fusion proteins are then refolded with the AH1.23 TCR a chain using the metllods detailed in Example 2 to produce the complete soluble AH1.23 TCR-interleukin fusion protein.

Example 10 - Tlzynaidine incorporation assay fof assessing the ability of AH1.23 TCR-IL-10 fusion proteins to cause Mast cell proliferation.

5 x106 cells of the D36 murine mast cell line which proliferates in response to huinan IL-10 in the presence of IL-4 are cultured in RPMI 1640 medium.

A range (0, 0.01, 0.1, 0.5 and 1 M of the AH1.23 TCR-IL-10 fusion protein prepared as described in Example 9 and IL-4 are added to the above culture. (Schlaak et al., (1994) J ITnnaunological Methods 168 49-54) 1.85 MBq / ml of H3 Thymidine is then added to 1 x 105 cells of the above culture in a 96 well plate. These cultures are then incubated for a further 8 hours at 37 C, 5% C02.
The cells are harvested using a cell-harvester, and the level of thymidine incorporation into the cells is measured using a Top Count (3 counter.

A reduction in thymidine incorporation into the D36 cells in the presence of the AH1.23 TCR-IL-10 fusion protein, compared to that seen in the absence of the fusion protein indicates that the IL-10 part of the fusion protein is active and causing D36 mast cell proliferation.

Exarnple 11 Prepar=ation of high affinity NY-ES MTCR - tlaerapeutic agent fusion proteins.
Synthetic genes comprising the DNA sequence encoding the soluble high affinity c6l NY-ESO TCR (3 chain detailed in Figure 23a linked via a DNA sequence encoding a peptide linker to DNA encoding a number of imunoinodulaotory agents were synthesised:
There are a nuinber of companies that provide a suitable DNA service, such as Geneart (Germany) Figure 24a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to IL-18.
Figure 24b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 25a details the DNA sequence encoding IL-18 pro-protein linked at the C-terminus thereof via a peptide linker to the high affinity c61 NY-ESO MTCR
beta chain. The pro-IL-18 DNA sequence has been altered to encode a Factor X
cleavage site which facilitates post translation removal of the amino acids within the pro-sequence. Figure 25b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 26a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to IL-10.
Figure 26b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 27a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus tliereof via a peptide linker to IL-13.
Figure 27b details the AA sequence of this fusion protein, the peptide linker is underlined.

Figure 28a details the DNA sequence encoding the high affinity c61 NY-ESO MTCR
beta chain linked at the C-terminus thereof via a peptide linker to the "KDEL"
variant of the PE38 exotoxin. Figure 28b details the AA sequence of this fusion protein the peptide linleer is underlined.
The DNA sequences above c61 NY-ESO TCR beta chain can be ligated into the pEX821 vector. (See Figures 19 and 20 for the DNA sequence and plasmid map of this vector respectively) Disulfide-linked a(3TCR-therapeutic agents are then produced following the methods substantially as described in Example 2. Briefly, DNA encoding the high affinity c61 NY-ESO alpha chain detailed in Figure 29a is synthesised and ligated into the pEX954 vector. (See Figures 21 and 22 for the DNA sequence and plasmid map of this vector respectively) The TCR beta chain fusion proteins described above are then refolded in the presence of the c61 NY-ESO TCR alpha chain.

Figure 29a details the DNA sequence encoding the high affinity c58 NY-ESO MTCR
alpha chain and Figure 29b details the AA sequence encoded by the DNA sequence of Figure 29a.
Exanzple 12 MTCR-PE-38 fusion protein cytotoxicity assay.

1x10"6 of the required target cells (e.g. SK-MEL tumour cells or J82 cells) were suspended in 10 ml RPMI media + 10% fetal calf serum (FCS). If required the target cells were then pulsed with 10 M of cognate peptide for 2 hours at 37 C. The samples were then washed three times in RPMI + 10% FCS, centrifuging at 1200 rpm for 5 min in between each wash. The washed cells were then re-counted and re-suspended in the appropriate volume of RPMI + 10% FCS media to provide a final cell density of 2 x 105 cells/ml.
The MTCR-PE38 fusion proteins prepared as described in Example 11 were diluted in RPMI media + 10% FCS to a final concentration of 2 x 10-6 M to provide a working standard. This working standard was then used to prepare a set of serial dilutions.

Preparation of experinaental and control samples in microtitre plate wells:
Experimental sample wells were filled with 50 l mTCR-PE38 in media and 50 l cells in medium. To produce a total volume of 100 l in 96 well flat bottom white opaque walled plates (Nunc 136101). The mTCR-PE38 serial dilutions prepared above were used to provide a range of mTCR-PE38 concentrations in these wells.

Control sample wells were prepared using either 100 l of cells (cell-only controls) or 100 l of mTCR-PE38 and media (effector-only controls).

The experimental and control samples were then incubated at 37 C, 5% CO2 for 48 or 96 hours. The nuinber of viable cells remaining in each well was then assessed using a CellTiter-Glo'E'Luminescent assay (Promega Cat No: G7572) following the manufacturers instructions.

Results Figures 30a and 30b demonstrate that the NY-ESO+ SK-MEL 37 and Mel 624 tumour cell lines can be killed by the 1G4 MTCR-PE38 fusion protein.

EC50 values for the effect of the 1G4 MTCR-PE38 fusion protein on the SK-MEL

and Mel 624 tumour cell lines after 48 hours incubation of 5.9 x 10-9 and 1 x respectively were calculated from the data presented in Figure 30a.

EC50 values for the effect of the 1G4 MTCR-PE38 fusion protein on the SK-MEL

and Mel 624 tumour cell lines after 96 hours incubation of 5.7 x 10-9 and 2.1 x 10-8 M
respectively were calculated from the data presented in Figure 30b.

The results provided by Figures 30a and 30b both demonstrate that pulsing the target cells with the cognate SLLMWITQC NY-ESO peptide leads to more efficient killing of these cells by the NY-ESO TCR-PE38 construct compared to that observed with unpulsed J82 target cells.

Claims (37)

1. A dimeric TCR (dTCR) or single-chain TCR (scTCR) associated with a therapeutic agent, wherein said agent is selected from IL-1, IL-1.alpha., IL-3, IL-4, IL-5, IL-6, IL-7, IL-10, IL-11, IL-12, IL-13, IL-15, IL-21, IL-23, TGF-.beta., IFN-.gamma., Lymphotoxin, TNF.alpha., Anti-CD2 antibody, Anti-CD3 antibody, Anti-CD4 antibody, Anti-CD8 antibody, Anti-CD44 antibody, Anti-CD45RA antibody, Anti-CD45RB
antibody, Anti-CD45RO antibody, Anti-Thy 1.2 antibody, Antilymphocyte globulin, Anti-.alpha..beta.TCR antibody, Anti-.gamma..delta.TCR antibody, Anti-CD49a antibody, Anti-CD49b antibody, Anti-CD49c antibody,Anti-CD49d antibody,Anti-CD49e antibody,Anti-CD49f antibody, Anti-TCR VP8 antibody, Anti-CD 16 antibody, Anti-CD28 antibody, CTLA-4-Ig, Anti-B7.2 antibody, Anti-CD40L antibody, Anti-ICAM-1 antibody, ICAM-1, Anti-Mac antibody, Anti-LFA-1 antibody, Anti-IFN-.gamma. antibody IFN-.gamma., IFN-.gamma.R/IgG1 fusions, Anti-IL-2R antibodies, IL-2R antibody, IL-2 Diptheria-toxin protein, Anti-IL-12 antibody, IL-12 Antagonist (p40), Anti-IL-1 antibody, IL-1 Antagonist, Glutamic acid decarboxylase (GAD), Anti-GAD antibody, Viral proteins and peptides, Bacterial proteins or peptides, A-Galactosyl-ceramide, Calcitonin, Nicotinamide, Anti-oxidants (Vitamin E, Probucol analog, Probucol +
deflazacoert or Aminoguanidine), Anti-Inflammatory agents (Pentoxifylline or Rolipram), Immunomodulators (Linomide, Ling-zhi-8, D-Glucan, Multi-functional protein 14, Ciamexon, Cholera toxin B, Vanadate or Vitamin D3 analogue, small molecule inhibitors, Androgens, IGF-1, Immunomanipulation (Natural antibodies), Lupus idiotype, Lipopolysaccaride), Sulfatide, Bee venom, Kampo formulation, Silica, Ganglioside, Antiasialo GM-1 antibody, Hyaluronidase, Concanavalin A, Anti-Class I MHC antibody, or Anti-Class II MHC antibody, Cyclosporin, FK-506, Azathioprine, Rapamycin or Deoxyspergualin, PE38 Pseudomonas exotoxin, and wherein said TCR comprises a first segment constituted by an amino acid sequence corresponding to a TCR
.alpha.
chain variable domain sequence fused to the N terminus of an amino acid sequence corresponding to a TCR .alpha. chain constant domain extracellular sequence, a second segment constituted by an amino acid sequence corresponding to a TCR
.beta.
chain variable domain fused to the N terminus of an amino acid sequence corresponding to TCR .beta. chain constant domain extracellular sequence, a disulfide bond between the first and second chains, said disulfide bond being one which has no equivalent in native .alpha..beta.T cell receptors, and in the case of said scTCRs further comprising a linker sequence linking the C
terminus of the first segment to the N terminus of the second segment, or vice versa, the length of the linker sequence and the position of the disulfide bond being such that the variable domain sequences of the first and second segments are mutually orientated substantially as in native .alpha..beta.T cell receptors.
2. A dTCR or scTCR associated with a therapeutic agent, as claimed in claim 1, wherein the therapeutic agent is selected from IL-1, IL-1.alpha., IL-3, IL-5, IL-6, IL-7, IL-11, IL-12, TGF-.beta., Lymphotoxin, TNF.alpha., Anti-CD2 antibody, Aiti-CD4 antibody, Anti-CD8 antibody, Anti-CD44 antibody, Anti-CD45RA antibody, Anti-CD45RB
antibody, Anti-CD45RO antibody, Anti-Thy 1.2 antibody, Antilymphocyte globulin, Anti-.alpha..beta.TCR antibody, Anti-.gamma..DELTA.TCR antibody, Anti-CD49a antibody, Anti-CD49b antibody, Anti-CD49c antibody,Anti-CD49d antibody,Anti-CD49e antibody,Anti-CD49f antibody, Anti-TCR V.beta.8 antibody, Anti-CD16 antibody, Anti-CD28 antibody, CTLA-4-Ig, Anti-B7.2 antibody, Anti-CD40L antibody, Anti-ICAM-1 antibody, ICAM-1, Anti-Mac antibody, Anti-LFA-1 antibody, Anti-IFN-.gamma. antibody IFN-.gamma., IFN-.gamma.R/IgG1 fusions, Anti-IL-2R antibodies, IL-2R antibody, IL-2 Diptheria-toxin protein, Anti-IL-12 antibody, IL-12 Antagonist (p40), Anti-IL-1 antibody, IL-1 Antagonist, Glutamic acid decarboxylase (GAD), Anti-GAD antibody, Viral proteins and peptides, Bacterial proteins or peptides, A-Galactosyl-ceramide, Calcitonin, Nicotinamide, Anti-oxidants (Vitamin E, Probucol analog, Probucol +
deflazacoert or Aminoguanidine), Anti-Inflammatory agents (Pentoxifylline or Rolipram), Immunomodulators (Linomide, Ling-zhi-8, D-Glucan, Multi-functional protein 14, Ciamexon, Cholera toxin B, Vanadate or Vitamin D3 analogue, small molecule inhibitors, Androgens, IGF-1, Immunomanipulation (Natural antibodies), Lupus idiotype, Lipopolysaccaride), Sulfatide, Bee venom, Kampo formulation, Silica, Ganglioside, Antiasialo GM-1 antibody, Hyaluronidase, Concanavalin A, Anti-Class I MHC antibody, or Anti-Class II MHC antibody, Cyclosporin, FK-506, Azathioprine, Rapamycin or Deoxyspergualin.
3. A dTCR or scTCR associated with a therapeutic agent, as claimed in claim 1, wherein the therapeutic agent is one of IL-10, IL-4 or IL-13.
4. A dTCR or scTCR associated with a therapeutic agent as claimed in any preceding claim, wherein the dTCR or scTCR is tissue-specific.
5. A dTCR or scTCR associated with a therapeutic agent, as claimed in claim 4, wherein the dTCR or scTCR is specific for a tissue which is a target for auto-reactive T cells in autoimmune disease, organ rejection or Graft Versus Host Disease (GVHD).
6. A dTCR or scTCR associated with a therapeutic agent, as claimed in claims 4 or 5, wherein the dTCR or scTCR is islet cell-specific.
7. A dTCR or scTCR associated with a therapeutic agent as claimed in claim 1, wherein the therapeutic agent consists of one of IL-15, IL-21, IL-23, PE38 Pseudomonas exotoxin, IFN-.gamma. or Anti-CD3 antibody.
8. A dTCR associated with a therapeutic agent as claimed in any preceding claim.
9. An scTCR associated with a therapeutic agent as claimed in any preceding claim.
10. An scTCR associated with a therapeutic agent, as claimed in claim 9 wherein the linker sequence links the C terminus of the first segment to the N
terminus of the second segment.
11. A scTCR associated with a therapeutic agent, as claimed in claim 9 wherein the linker sequence has the formula -PGGG-(SGGGG)n-P- wherein n is 5 or 6 and P
is proline, G is glycine and S is serine.
12. A dTCR associated with a therapeutic agent, as claimed in claim 8 which is a dTCR comprising a first polypeptide wherein a sequence corresponding to a TCR .alpha. chain variable domain sequence is fused to the N terminus of a sequence corresponding to a TCR .alpha.
chain constant domain extracellular sequence, and a second polypeptide wherein a sequence corresponding to a TCR .beta.chain variable domain sequence fused to the N terminus a sequence corresponding to a TCR
.beta. chain constant domain extracellular sequence, the first and second polypeptides being linked by a disulfide bond which has no equivalent in native .alpha..beta. T cell receptors.
13. A TCR associated with a therapeutic agent, as claimed in claim 8 which is a dTCR comprising a first polypeptide wherein a sequence corresponding to a TCR .alpha. chain variable domain sequence is fused to the N terminus of a sequence corresponding to .alpha. TCR .alpha.
chain constant domain extracellular sequence, and a second polypeptide wherein a sequence corresponding to a TCR .beta. chain variable domain sequence is fused to the N terminus a sequence corresponding to a TCR
.beta.
chain constant domain extracellular sequence, the first and second polypeptides being linked by a disulfide bond between cysteine residues 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.
14. A NCR associated with a therapeutic agent, as claimed in any preceding claim which darr or SST has amino acid sequences corresponding to human .alpha..beta. NCR
extracellular constant and variable domain sequences.
15. A NCR associated with a therapeutic agent, as claimed in claim 14 wherein a disulfide bond links amino acid residues of the said constant domain sequences, which disulfide bond has no equivalent in native torr.
16. A NCR associated with a therapeutic agent, as claimed in claim 15 wherein the said disulfide bond is between cysteine residues corresponding to amino acid residues whose .beta. carbon atoms are less than 0.6 nm apart in native torr.
17. A NCR associated with a therapeutic agent, as claimed in claim 15 wherein the said disulfide bond is between cysteine residues substituted for Thr 48 of exon 1 of trace*01 and Ser 57 of exon 1 of trub1*01 or trub2*01 or the non-human equivalent thereof.
18. A NCR associated with a therapeutic agent, as claimed in any preceding claim wherein the darr or SST includes a disulfide bond between residues corresponding to those linked by a disulfide bond in native torr.
19. A NCR associated with a therapeutic agent, as claimed in any preceding claim wherein the darr or SST does not contain a sequence corresponding to transmembrane or cytoplasmic sequences of native torr.
20. A NCR associated with a therapeutic agent, as claimed in any preceding claim wherein said therapeutic agent is a PE38 exotoxin.
21. A NCR associated with a PE38 exotoxin as claimed in claim 20 comprising the amino acid sequences of (SEQ ID NO: 73) and (SEQ ID NO: 71).
22. A NCR associated with a therapeutic agent, as claimed in any preceding claim wherein the NCR is associated with at least one polyalkylene glycol chain(s).
23. A TCR associated with a therapeutic agent, as claimed in claim 22, wherein the polyalkylene glycol chain(s) is/are covalently linked to the TCR.
24. A TCR associated with a therapeutic agent, as claimed in claim 22 or claim wherein the polyalkylene glycol chain(s) comprise(s) at least two polyethylene glycol repeating units.
25. A multivalent TCR complex comprising at least two TCRs associated with a therapeutic agent, as claimed in any preceding claim.
26. A multivalent TCR complex comprising at least two TCRs associated with a therapeutic agent, as claimed in any of claims 1 to 24 linked by a non-peptidic polymer chain or a peptidic linker sequence.
27. A multivalent TCR complex as claimed in claim 26 wherein the polymer chain or peptidic linker sequence extends between amino acid residues of each TCR
associated with a therapeutic agent or a functional variant or fragment thereof, which are not located in a variable region sequence of the TCR.
28. A multivalent TCR complex as claimed in either of claims 26 or 27 in which the TCRs associated with a therapeutic agent, are linked by a polyalkylene glycol chain or a peptidic linker derived from a human multimerisation domain.
29. A multivalent TCR complex as claimed in claim 28 wherein a divalent alkylene spacer radical is located between the polyalkylene glycol chain and its point of attachment to a TCR associated with a therapeutic agent, of the complex.
30. A multivalent TCR complex as claimed in claim 28 or claim 29 wherein the polyalkylene glycol chain comprises at least two polyethylene glycol repeating units.
31. A pharmaceutical composition comprising a TCR associated with a therapeutic agent, as claimed in any of claims 1 to 24 or a multivalent complex thereof as claimed in any of claims 25 to 30, together with a pharmaceutically acceptable carrier.
32. A method of treatment of cancer comprising administering to a subject suffering such cancer an effective amount of a TCR associated with a therapeutic agent, as claimed in any of claims 1 to 24 or a multivalent complex thereof as claimed in any of claims 25 to 30 wherein said therapeutic agent is selected from those defined in claim 2.
33. The use of a TCR associated with a therapeutic agent, as claimed in any of claims 1 to 24 or a multivalent complex thereof as claimed in any of claims 25 to 30, wherein said therapeutic agent is selected from those defined in claim 2 in the preparation of a composition for the treatment of cancer.
34. A method of treatment of cancer comprising administering to a subject suffering such cancer an effective amount of the fusion protein as claimed in claims 20 or 21.
35. The use of the fusion protein as claimed in claims 20 or 21 in the preparation of a composition for the treatment of cancer.
36. A method of treatment of autoimmune disease, organ rejection or GVHD
comprising administering to a subject suffering such autoimmune disease, organ rejection or GVHD an effective amount of a TCR associated with a therapeutic agent, as claimed in any of claims 1 to 24 or a multivalent complex thereof as claimed in any of claims 25 to 30, wherein said therapeutic agent is selected from those defined in claim 3.
37. The use of a TCR associated with a therapeutic agent, as claimed in as claimed in any of claims 1 to 24, wherein said therapeutic agent is selected from those defined in claim 3 in the preparation of a composition for the treatment of autoimmune disease, organ rejection or GVHD.
CA002582963A 2004-10-01 2005-09-29 T-cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents Abandoned CA2582963A1 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GBGB0421836.8A GB0421836D0 (en) 2004-10-01 2004-10-01 T cell receptors containing a non-native disulfide interchain bond linked to immunomodulatory agents
GB0421836.8 2004-10-01
US62106304P 2004-10-25 2004-10-25
US60/621,063 2004-10-25
GB0427584.8 2004-12-16
GB0427584A GB0427584D0 (en) 2004-12-16 2004-12-16 T cell receptors containing a novel disulfide interchain bond linked to immunomodulatory agents
PCT/GB2005/003752 WO2006037960A2 (en) 2004-10-01 2005-09-29 T-cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents

Publications (1)

Publication Number Publication Date
CA2582963A1 true CA2582963A1 (en) 2006-04-13

Family

ID=36000822

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002582963A Abandoned CA2582963A1 (en) 2004-10-01 2005-09-29 T-cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents

Country Status (6)

Country Link
EP (1) EP1809669A2 (en)
JP (1) JP2008514685A (en)
AU (1) AU2005291039A1 (en)
CA (1) CA2582963A1 (en)
MX (1) MX2007003910A (en)
WO (1) WO2006037960A2 (en)

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE602006005200D1 (en) 2005-01-05 2009-04-02 F Star Biotech Forsch & Entw Synthetic immunoglobulin domains with modified binding properties in regions of the molecule other than the complementarity determining regions
AT503889B1 (en) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F MULTIVALENT IMMUNE LOBULINE
AT503861B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING T-CELL RECEPTORS
CN101802006B (en) 2007-06-26 2013-08-14 F-星生物技术研究与开发有限公司 Display of binding agents
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
GB0908613D0 (en) * 2009-05-20 2009-06-24 Immunocore Ltd T Cell Reseptors
CN102161998B (en) 2011-01-14 2013-01-09 中国人民解放军军事医学科学院附属医院 Deoxyribonucleic acid (DNA) vaccine based on B7-1-PE40KDEL exotoxin fusion gene and application thereof
ES2635416T3 (en) 2011-06-09 2017-10-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Exotoxin A from pseudomonas with epitopes of T lymphocytes and / or less immunogenic B lymphocytes
EP3301110A1 (en) 2011-09-16 2018-04-04 The USA, as represented by The Secretary, Department of Health and Human Services Pseudomonas exotoxin a with less immunogenic b cell epitopes
JP2014530009A (en) * 2011-09-29 2014-11-17 エーピーオー‐ティー ビー.ヴイ. Multispecific binding molecules targeting abnormal cells
CN103130894B (en) * 2011-11-30 2017-04-12 中国医学科学院基础医学研究所 Recombinant single-chain antibody G5-4ScFv of anti-human gamma delta T cell receptor (TCR) monoclonal antibody and encoding gene and application thereof
JP2015504895A (en) 2012-01-13 2015-02-16 エーピーオー‐ティー ビー.ヴイ. Abnormal cell-restricted immunoglobulin with a toxic moiety
NZ715815A (en) * 2013-06-26 2017-02-24 Guangzhou Xiangxue Pharmaceutical Co Ltd High-stability t-cell receptor and preparation method and application thereof
WO2016070814A1 (en) * 2014-11-07 2016-05-12 广州市香雪制药股份有限公司 Soluble heterodimeric t cell receptor, and preparation method and use thereof
CN106279404A (en) * 2015-05-20 2017-01-04 广州市香雪制药股份有限公司 A kind of solvable and stable heterogeneous dimerization TCR
CA3022611A1 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
BR112018074748A2 (en) 2016-06-02 2019-03-06 Immunocore Limited bispecific therapeutic composition of t-cell redirection, and gp100 positive cancer treatment method in a patient
MA45455A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc PROCESS FOR IDENTIFYING PEPTIDIC EPITOPES, MOLECULES THAT BIND TO SUCH EPITOPES AND ASSOCIATED USES
MA45491A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc CMH-E RESTRICTED EPITOPES, BINDING MOLECULES AND RELATED METHODS AND USES
MA46354A (en) 2016-10-03 2019-08-07 Juno Therapeutics Inc MOLECULES BINDING SPECIFICALLY TO HPV
WO2018102795A2 (en) * 2016-12-02 2018-06-07 University Of Southern California Synthetic immune receptors and methods of use thereof
KR20240023449A (en) 2017-02-08 2024-02-21 드래곤플라이 쎄라퓨틱스, 인크. Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer
CA3054079A1 (en) 2017-02-20 2018-08-23 Dragonfly Therapeutics, Inc. Proteins binding her2, nkg2d and cd16
MA50613A (en) 2017-10-03 2020-08-12 Editas Medicine Inc HPV-SPECIFIC BINDING MOLECULES
SG11202003862PA (en) 2017-11-01 2020-05-28 Editas Medicine Inc Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
CN111556893A (en) 2017-11-06 2020-08-18 爱迪塔斯医药股份有限公司 Methods, compositions, and components for CRISPR-CAS9 editing of CBLB in immunotherapy T cells
EP3749346A4 (en) 2018-02-08 2021-12-15 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the nkg2d receptor
PE20201345A1 (en) 2018-04-05 2020-11-25 Juno Therapeutics Inc RECEIVERS OF T-CELLS, AND DESIGNED CELLS THAT EXPRESS THEM
KR20210020873A (en) 2018-04-05 2021-02-24 주노 쎄러퓨티크스 인코퍼레이티드 Τ cells expressing recombinant receptors, related polynucleotides and methods
US20210017249A1 (en) 2018-04-05 2021-01-21 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
EA202091977A1 (en) * 2018-05-28 2021-02-09 Драгонфлай Терапьютикс, Инк. MULTI-SPECIFIC BINDING PROTEINS THAT BIND CD33, NKG2D AND CD16 AND METHODS OF APPLICATION
SG11202101204TA (en) 2018-08-09 2021-03-30 Juno Therapeutics Inc Processes for generating engineered cells and compositions thereof
SG11202102108QA (en) 2018-09-11 2021-04-29 Juno Therapeutics Inc Methods for mass spectrometry analysis of engineered cell compositions
SG11202111360YA (en) 2019-05-01 2021-11-29 Juno Therapeutics Inc Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
EP4125943A1 (en) 2020-03-27 2023-02-08 Mendus B.V. In vivo use of modified cells of leukemic origin for enhancing the efficacy of adoptive cell therapy
CN115916963A (en) 2020-03-27 2023-04-04 门德斯有限公司 Ex vivo use of modified cells of leukemia origin for enhancing the efficacy of adoptive cell therapy
KR20230042283A (en) 2020-06-26 2023-03-28 주노 테라퓨틱스 게엠베하 Engineered T cells conditionally expressing recombinant receptors, related polynucleotides and methods
WO2022060904A1 (en) 2020-09-16 2022-03-24 Obsidian Therapeutics, Inc. Compositions and methods for expression of t-cell receptors with small molecule-regulated cd40l in t cells
JP2023547520A (en) 2020-11-05 2023-11-10 メンドゥス・ベスローテン・フェンノートシャップ Use of tumor-independent antigens in immunotherapy
WO2023081900A1 (en) 2021-11-08 2023-05-11 Juno Therapeutics, Inc. Engineered t cells expressing a recombinant t cell receptor (tcr) and related systems and methods
CN116496408A (en) * 2022-01-21 2023-07-28 广东菲鹏制药股份有限公司 Interleukin 21 and receptor complexes thereof
WO2023196884A1 (en) 2022-04-06 2023-10-12 Juno Therapeutics, Inc. Detection assay for human papillomavirus (hpv) type 16 (hpv-16)
US20240002800A1 (en) 2022-05-16 2024-01-04 Mendus B.V. Use of leukemia-derived cells for enhancing natural killer (nk) cell therapy

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ531208A (en) * 2001-08-31 2005-08-26 Avidex Ltd Multivalent soluble T cell receptor (TCR) complexes
JP4436319B2 (en) * 2002-10-09 2010-03-24 メディジーン リミテッド Single-chain recombinant T cell receptor
NZ570811A (en) * 2002-11-09 2009-11-27 Immunocore Ltd T cell receptor display
AU2003286263A1 (en) * 2002-12-03 2004-06-23 Avidex Ltd. Complexes of receptors
GB0304068D0 (en) * 2003-02-22 2003-03-26 Avidex Ltd Substances

Also Published As

Publication number Publication date
WO2006037960A2 (en) 2006-04-13
EP1809669A2 (en) 2007-07-25
WO2006037960A3 (en) 2006-08-03
MX2007003910A (en) 2007-06-07
JP2008514685A (en) 2008-05-08
AU2005291039A1 (en) 2006-04-13

Similar Documents

Publication Publication Date Title
CA2582963A1 (en) T-cell receptors containing a non-native disulfide interchain bond linked to therapeutic agents
US20210061878A1 (en) High affinity ny-eso 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
AU2006253941B2 (en) High affinity Melan-A T cell receptors
KR20200019211A (en) T cell receptor
US20080274133A1 (en) Soluble Bifunctional Proteins
EP1814992A1 (en) Gamma-delta t cell receptors
WO2019246593A2 (en) Compositions and methods to target cll-1 and cd123 for the treatment of acute myeloid leukemia and related disorders
EP1597273A2 (en) Soluble ctla4 polypeptides and methods for making the same

Legal Events

Date Code Title Description
FZDE Dead