Title of Invention

AGENT FOR THE PREPARATION OF A MEDICAMENT FOR TREATING OR PREVENTING COELIAC DISEASE

Abstract The invention herein disclosed is related to epitopes useful in methods of diagnosing, treating, and preventing coeliac disease. Therapeutic compositions which comprise at least one epitope are provided.
Full Text This application is a divisional out of the original application No. 32/DELNP/2005.
The invention relates to epitopes useful in the diagnosis and therapy of coeliac disease, including diagnostics, therapeutics, kits, and methods of using tile foregoing.
An immune reaction to gliadin (a component of gluten) in the diet causes coeiiac disease. It is known that immune responses in the intestinal tissue preferentially respond to gliadin which has been modified by an intestinal transglutaminase. Coeliac disease is diagnosed by detection of anti-endomysial antibodies, but this requires confirmation by me finding of a lymphocytic inflammation in intestinal biopsies. The taMng of such a biopsy is inconvenient for the patient
Investigators have previously assumed that only intestinal T cell responses provide an accurate indication of me immune response against Therefore they have concentrated on the investigation of T cell responses in intestinal tissue1. Gliadin epitopes which require Esghrtarninase modification (before they are recognised by the immune system) are known2.
The inventors have found the nruunnodorninant T cell A-gliadin epitope' recognised by the immune system in coeliac disease, and have shown fhat mis is recognised by T cells in me peripheral blood of individuals wifli coeliac disease {see WO 01/25793). Such T cells were found to be present at high enough frequencies to be detectable without restimula±ion{i.e. a 'fresh response' detection system could be used). The epitope was identified using a non-T cell cloning based method which provided a more accurate reflection of the epitopes being recognised. The epitope requires transglutaminase modification (causing substitution of a particular glutamine to glutamate) before immune system recognition.
Based on this work the inventors nave developed a. test which can be used to diagnose coeliac disease at an early stage. The test may be carried out on a sample from peripheral blood and therefore an intestinal biopsy is not required. The test is more sensitive than the antibody tests which are currently being used.
The invention thus provides a method of diagnosing coeliac disease, or susceptibility to coeliac disease, in an individual comprising:

(a) contacting a sample from the host with an agent selected from (f)' the epitope comprising sequence which is: SEQ ID NO:1 (PQPELPY)or SEQ ED NO:2 (QLQPFPQPELPYPQPQS), or an equivalent sequence from a naturally occurring homologue of the gliadin represented by SEQ ID N0:3, (ii) an epitope comprising sequence comprising: SEQ ID NO:1, or an equivalent sequence from a naturally occurring homologue of the gliadin represented by SEQ ID NO:3 (shown in Table 1), which epitope is an isolated oligopeptide derived from a gliadin protein, (in) an analogue of (i) or (ii) which is capable of being recognised by a T cell receptor that recognises (i) or (ii), which in the case of a pepti.de analogue is not more 50 ' amino acids in length, or (iv) a product comprising two or more agents as defined in (i), (ii) or (in), and (b) determining in vitro whether T cells in the sample recognise the agent, recognition by the T cells indicating that the individual has, or is susceptible to, coeliac disease.
Through comprehensive mapping of wheat gliadin T cell epitopes (see Example 13), the inventors have also found epitopes bioactive in coeliac disease in HLA-DQ2+ patients in other wheat gliadins, having similar core sequences (e.g., SEQ ID NOS: 18-22) and similar full length sequences (e.g., SEQ ID NOS31-36), as well as in rye secalins and barley hordeins (e.g., SEQ ID NOS'39-41); see also Tables 20 and 21. Additionally, several epitopes bioactive in coeliac disease in HLA-DQ8+ patients have been identified (e.g., SEQ ID NOS:42-44,46). This comprehensive mapping thus provides the dominant epitopes recognized by T cells • in coeliac patients. Thus, me above-described method and other methods of the invention described herein may be performed using any of these additional identified epitopes, and analogues and equivalents thereof) (i) and (Ii) herein include these additional epitopes. That is, the agents of the invention also include these novel epitopes.
The invention also provides use of the agent for the preparation of a diagnostic means for use in a method of diagnosing coeliac disease, or susceptibility to coeliac disease, in an individual, said method comprising determining whether T cells of the individual recognise the agent, recognition by the T cells indicating that the individual has, or is susceptible to, coeliac disease.

The finding of an immunodominant epitope which is modified by transglutaminase (as well as the additional other epitopes defined herein) also allows diagnosis of coeliac disease based-on determining whether other types of immune response to this epitope are present Thus the invention also provides a method of diagnosing coeliac disease, or susceptibility to coeliac disease, in an individual comprising determining the presence of an antibody that binds to the .epitope in a sample from the individual, the presence of the antibody indicating that the individual has, or is susceptible to, coeliac disease.
The invention additionally provides the agent, optionally in association with, a carrier, for use in a method of treating or preventing coeliac disease by tolerising T cells which recognise the agent Also provided is an antagonist of a T cell which has a T cell receptor that recognises (i) or (ii), optionally in association with a carrier, for use in a method of treating or preventing coeliac disease by antagonising such T • cells. Additionally provided is the agent or an analogue that binds an antibody (that binds the agent) for use in a method of treating or preventing coeliac disease in an individual by tolerising the individual to prevent me production of such an antibody.
The invention provides a method of determining whether a composition is capable of causing coeliac disease comprising determining whether a protein capable of being modified by a transglutaminase to an oligopepti.de sequence as defined above is present in me composition, the presence of the protein indicating that the composition is capable of causing coeliac disease.
The invention also provides a mutant gliadin protein whose wild-type sequence can be modified by a transglutaminase to a sequence mat comprises an epitope comprising sequence as defined above, but which mutant gliadin protein has been modified in such away that it does not contain sequence which can be modified by a transglutaminase to a sequence that comprises such an epitope comprising sequence; or a fragment of such a mutant gliadin protein which is at least 15 amino acids long and which comprises sequence which has been modified in said way.
The invention also provides a protein that comprises a sequence whiph is able to bind to a T cell receptor, "which T cell receptor recognises the agent, and which sequence is able to cause antagonism of a T cell that carries such a T cell receptor.

Additionally the invention provides a food that comprises the proteins defined above.
SUMMARY OF THE INVENTION
The present invention provides methods of preventing or treating coeliac disease comprising administering to an individual at least one agent selected from: a) a peptide comprising at least one epitope comprising a sequence selected from the group consisting of SEQ ID NOs: 18-22, 31-36, 39-44, and 46, and equivalents thereof; and b) an analogue of a) which, is capable of being recognised by a T cell receptor that recognises the peptide of a) and which is not more than 50 amino acids in length; and c) optionally, in addition to the agent selected from a) andb), a peptide comprising at least one epitope comprising a sequence selected from SEQ ID NO:1 and SEQ ID NO:2. In some embodiments, the agent is HLA-DQ2-restricted, HLA-DQ8-restricted or one agent is HLA-DQ2-restriclBd and a second agent is HLA-DQ8-restricted. In some embodiments, the agent comprises a wheat epitope, atye epitope, a barley epitope or any combination thereof either as a single agent or as multiple agents.
The present invention also provides methods of preventing or treating coeliac disease comprising administering to an individual a pharmaceutical composition comprising an agent above and pharmaceirtically acceptable carrier or diluent
The present invention also provides methods of preventing or treating coeliac disease comprising administering to an individual a pharmaceutical composition comprising an antagonist of a T cell which has a T cell receptor as defined above, and a phamiaceutically acceptable carrier or diluent
The present invention also provides methods of preventing or treating coeliac disease comprising administering to an individual a composition for tolerising an individual to a gliadin protein to suppress the production of a T cell or antibody response to an agent as denned above, which composition comprises an agent as defined above.
The present invention also provides methods of preventing or treating coeliac disease by 1) diagnosing coeliac disease in an individual by either: a) contacting a sample from the host with at least one agent selected from: i) a peptide comprising at

least one epjtope comprising a sequence selected from the group consisting of: SEQ ID NOS: 18-22, 31-36, 59-44, and 46, and equivalents thereof; and ii) an analogue of i) which is capable of being recognised by a T cell receptor that recognises i) and which is not more than 50 amino acids in length; and iii) optionally, in addition to the agent selected from i) and ii), a peptide comprising at least one epitope comprising a sequence selected from SEQ ID NOS:1 and 2; and determining 172 vitro whether T cells in the sample recognise the agent; recognition by the T cells indicating that the individual has, or is susceptible to, coeliac disease; or b) adrninistering an agent as defined above and determining in vivo whether T cells in the individual recognise the agent, recognition of the agent indicating that the individual has or is susceptible to coehac disease; and 2) administering to an individual diagnosed as having, or being susceptible to, coehac disease a therapeutic agent for preventing or treating coeliac disease.
The present invention also provides agents as defined above, optionally in association with a carrier, for use in a method of treating or preventing coeliac disease by tolerising T cells which recognise the agent.
The present invention also provides antagonists of a T cell which has a T cell -receptor as defined above, optionally in association with a carrier, for use in a method of treating-or preventing coehac disease by antagonising such T cells.
The present invention also providers proteins that comprises a sequence which is able to bind to a T cell receptor, which T cell receptor recognises an agent as defined above, and which sequence is able to cause antagonism of a T cell that carries suclia T cell receptor.
The present invention also provides pharmaceutical compositions comprising an agent or antagonist as defined and a pharmaceutically acceptable carrier or diluent
The present invention also provides compositions for tolerising an individual to a gliadin protein to suppress the production of a T cell or antibody response to an agent as defined above, which composition comprises an agent as defined above.
The present invention also provides compositions for antagonising a T cell response to an agent as defined above, which composition comprises an antagonist as defined above.

The present invention also provides mutant gliadin proteins whose wild-type sequence can be modified by a transglutaminase to a sequence which is an agent as defined in claim 1, which mutant gliadin protein comprises a mutation which prevents its modification by a transglutaminase to a sequence which is an agent as defined above; or a fragment of such a mutant gliadin protein which is at least 15 amino acids long and which comprises the mutation.
The present invention also provides polynucleotides that comprises a coding sequence that encodes a protein or fragment as defined above.
The present invention-alse provides cells comprising a polynucleotide as defined above or which has been transformed with such a polynucleotide.
The present invention also provides mammals that expresses a T cell1 receptor as defined above.
The present invention also provides methods of diagnosing coeliac disease, or susceptibility to coeliac disease, in an individual comprising: a) contacting a sample from the host with at least one agent selected from i) a peptide comprising at least one epftope "comprising a sequence selected from the group consisting of: SEQ ID NOS:18-22, 31-36, 39-44, and 46, and equivalents thereof; andii) an analogue off) which is capable of being recognised by a T cell receptor that recognises i) and which is not more than 50 amino acids in length; and iii) optionally, in addition to the agent selected from i) and ii), a peptide comprising at least one epitope comprising a sequence selected from SEQ ID NOS:1 and 2; andb) detennining in vitro whether T cells in the sample recognise the agent; recognition by the T cells indicating that the individual has, or is susceptible to, coeliac disease.
The present invention also provides methods of determining whether a composition is capable of causing coeliac disease comprising determining whether a protein capable of being modified by a transglutaminase to an oligopeptide sequence is present in the composition, the presence of the protein indicating that the composition is capable of causing coeliac disease.
The present invention also provides methods of identifying an antagonist of a T cell, which T ceH recognises an agent as defined above, comprising contacting a candidate substance with the T cell and detecting whether the substance causes a decrease in the ability of the T cell to undergo an antigen specific response, the

detecting of any such decrease in said ability indicating that the substance-is an antagonist.
The present invention also provides kits for carrying out any of the method described above comprising an. agent as defined above and a means to detect the recognition of the peptide by the T cell.
The present invention also provides methods of identifying a product which is therapeutic for coeliac disease comprising administering a candidate substance to a mammal as defined above which has, or which is susceptible to, coeliac disease and determining whether substance prevents or treats coeliac disease in the mammal, the prevention or treatment of coeliac disease indicating that the substance is a therapeutic product
The present invention also provides processes for the production of a protein encoded by a coding sequence as defined above which process comprises: a) cultivating a cell described above under conditions that allow the expression of the protein; and optionally b) recovering the expressed protein.
The present invention also provides methods of obtaining a transgenic plant cell comprising transforming a plant cell with, a vector as described above to give a transgenic plant cell.
The present invention also provides methods of obtaining a first-generation transgenic plant comprising regenerating a transgenic plant cell transformed with a vector as described above to give a transgenic plant
The present invention also provides methods of obtaining a transgenic plant seed comprising obtaining a transgenic seed from a transgenic plant obtainable as described above.
The present invention also provides methods of obtaining a transgenic progeny plant comprising obtaining a second-generation transgenic progeny plant from a first-generation transgenic plant obtainable by a method as described above, and optionally obtaining transgenic plants of one or more further generations from the second-generation progeny plant thus obtained.
The present invention also provides transgenic plant cells; plants, pknt seeds or progeny plants obtainable by any of the methods described above.

The present invention also provides transgenic plants or plant seeds comprising plant cells as described above.
The present invention also provides transgenic plant cell calluses comprising plant cells as described above obtainable from a transgenic plant cell, first-generation plant, plant seed or progeny as defined above.
The present invention also provides methods of obtaining a crop product comprising harvesting a crop product from a plant according to any method described above and optionally further processing the harvested product
The present invention-also provides food that comprises a protein as defined above.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention is illustrated by the accompanying drawings in which: Figure 1 snows freshly isolated PBMC (peripheral blood mononuclear ceH) IFNy ELISPOT responses (vertical axis shows spot forming cells per 106 PBMC) to transglutaminase (fTGHreated and untreated peptide pool 3 (each peptide 10 ug/ml) including five overlapping ISmers spanning A-gliadin 51-85 (see Table 1) and a-chymotrypsin-digested gliadin (40 ug/ml) in coeliac disease Subject 1, -initially in remission following a gluten free diet men challenged wifli 200g bread daily for three days from day 1 (a). PBMC IFNy ELISPOT responses by Subject 2 to tTG-treated A-gliadin peptide pools 1-10 spanning the complete A-gliadin protein daring ten day bread challenge (b). The horizontal axis shows days after commencing bread.
Figure 2 shows PBMC JFNy ELISPOT responses to tTG-treaied peptide pool 3 (spanning A-gliadin 51-85) in 7 individual coeliac disease subjects (vertical axis shows spot forming cells per 106 PBMC), initially in remission on gluten free diet, challenged with bread for three days (days 1 to 3). The horizontal axis shows days after commencing bread, (a). PBMC IFNy Elispot responses to tTG-treated overlapping ISmer peptides included in pool 3; bars represent the mean (± SEM) response to individual peptides (10 jig/ml) in 6 Coeliac disease subjects on day 6 or 7(b). (In individual subjects, ELISPOT responses to peptides were calculated as a % of response elicited by peptide 12 - as shown by the vertical axis.)

Figure 3 shows PBMC DFNy ELISPOT responses to tTG-treated truncations of A-gliadin 56-75 (0.1 pM). Bars represent the mean (± SEM) in 5 Coeliac disease subjects. (In individual subjects, responses were calculated as the % of the maximal response elicited by any of the peptides tested.)
Figure 4 shows how the minimal structure of the dominant A-gliadin epitope was mapped using tTG-treated 7-17mer A-gliadin peptides (0.1 pM) including the sequence, PQPOJLPY (SEQ ID NO:4) (A-gliadin 62-68) (a), and the same peptides without tTG treatment but with the substitution Q—»-E65 (b). Each line represents PBMC IFNy ELISPOT responses in each of three Coeliac disease subjects on day 6 or 7 after bread was ingested on days 1-3. (In individual subjects, ELISPOT responses were calculated as a % of the response elicited by the 17mer, A-gliadin 57-73.)
Figure 5 shows the amino acids that were deamidated by tTG. A-gliadin 56-75 LQLQPFPQPQLPYPQPQSFP (SEQ ID NO:5) (0.1 uM) was incubated with tTG (50 ug/ml) at 37°C for 2 hours. A single product was identified and purified by reverse phase HPLC. Amino acid analysis allowed % deamidation (Q—»E) of each Gin residue in A-gliadin 56-75 attributable to tTG to be calculated (vertical axis).
Figure 6 shows the effect of substituting Q—*E in A-gliadin 57-73 at other positions in addition to Q65 using the 17mers: QLQPFPQPELPYPQPEJS (SEQ ID N0:6) (E57,65), QLQPFPQPELPYPQPES (SEQ ID NO:7) (E65.72), ELQPFPQPELPYPQPES (SEQ IDNO:8) (E57, 65, 72), and QLQPFPQPELPYPQPQS (SEQ ID NO:2) (E65) in three Coeliac disease subjects on day 6 or 7 after bread was ingested on days 1-3. Vertical axis shows % of the E65 response.
Figure 7 shows that tTG treated A-gliadin 56-75 (0.1 uM) elicited IFN-g ELISPOT responses in (a) CD4 and CDS magnetic bead depleted PBMC. (Bars represent CD4 depleted PBMC responses as a % of CDS depleted PBMC responses; spot forming cells per million CDS depleted PBMC were: Subject 4: 29, and Subject 6:535). (b) PBMC EFNy ELISPOT responses (spot forming cells/million PBMC) after incubation with monoclonal antibodies to HLA-DR (L243), -DQ (L2) and -DP (B7.21) (10 ng/ml) lh prior to tTG-treated 56-75 (0.1 uM) in two coeliac disease subjects homozygous for HLA-DQ al*0501, bl*0201.

Figure 8 shows the effect of substituting Glu at position 65 for other amino acids in the irnmimodominant epitope. The vertical axis shows the % response in the 3 subjects in relation to the immunodominant epitope.
Figure 9 shows the' immunoreactivity of naturally occurring gliadinpeptides (measuring responses from 3 subjects) which contain the sequence PQLPY (SEQ ID NO: 12) with (shaded) and without (clear) transglutaminase treatment.
Figure 10 shows CDS, GEM, Pv, and CCE -specific immunomagnetic bead depletion of peripheral blood mononuclear cells from two coeliac subjects 6 days after commencing gluten challenge followed by interferon gamma ELISpot. A-gliadin 57-73 QE65 (25mcg/ml), tTG-rreated chymorrypsin-digested gliadin (100 meg/ml) or PPD (10 meg/ml) were used as antigen.
Figure 11 shows the optimal T cell epitope length. '
Figure 12 shows a comparison of A-gliadin 57-73 QE65 with other peptides in a dose response study.
Figure 13 shows a comparison of gliadin and A-gliadin 57-73 QE65 specific responses.
Figure 14 shows the bioactivity of gliadin polymorphisms in coeliac subjects.
Figures 15 and 16 show the defining of the core epitope sequence.
Figures 17 to 27 show the agonist activity of A-gliadin 57-73 QE65 variants.
Figure 28 shows responses in different patient groups.
Figure 29 shows bioactivity of prolamin homologues of A-gliadin 57-73.
Figure 30 shows, for healthy HLA-DQ2 subjects, the change in IFN-gamma EllSpot responses to tTG-deamidated gliadin peptide pools.
Figure 31 shows, for coeliac HLA-DQ2 snbjepts, the change in IFN-gamma ELJSpot responses to tTG-deamidated gliadin peptide pools.
Figure 32 shows individual peptide contributions to "summed" gliadin peptide response.
Figure 33 shows, for coeliac HLA-DQ2/8 subject COS, gluten challenge induced IFNy ELISpot responses to tTG-deamidated gliadin peptide pools.
Figure 34 shows, for coeliac HLA-DQ2/8 subject C07, gluten challenge induced IFNY ELISpot responses to tTG-deamidated gliadin peptide pools.

Figure 35 shows, for coeliac HLA-DQ8/7 subject C12, gluten challenge induced IFNy ELlSpot responses to tTG-deamidated gliadin peptide pools.
Figure 36 shows, for coeliac HLA-DQ6/8 subject Cl 1, gluten challenge induced IFNy ELISpot responses to tTG-deamidated gliadin peptide pools.
Detailed Description of the Invention
The term "coeliac disease" encompasses a spectrum of conditions caused by varying degrees of gluten sensitivity, including a severe form characterised by a flat small intestinal mucosa (hyperplastic villous atrophy) and other forms characterised by milder symptoms.
The individual mentioned above (in the context of diagnosis or therapy) is human. They may have coeliac disease (symptomatic or asymptomatic) or be suspected of having it They may be on. a gluten free diet They may be hi an acute phase response (for example they may have coeliac disease, but have only ingested gluten in the last 24 hours before which they had been on a gluten free diet for 14 to 28 days).
The individual may be susceptible to coeliac disease, such as a genetic susceptibility (determined for example by the individual having relatives with coeliac disease or possessing genes which cause predisposition to coeliac disease).
The agent
The agent is typically a peptide, for example of length 7 to 50 amino acids, such as 10 to 40, or 15 to 30 amino acids in length.
SEQ ID NO:1 is PQPELPY. SEQID NO2 is QLQPFPQPELPYPQPQS. SEQ ED N0:3 is shown in Table 1 and is me sequence of a whole A-gliadin. The glutamate at position 4 of SEQ ID NO: 1 (equivalent to position 9 of SEQ ID NO:2) is generated by transglutammase treatment of A-gliadrn.
The agent may be the peptide represented by SEQ ED NO: 1 or 2 or an epitope comprising sequence that comprises SEQ ID NO: 1 which is an isolated oligopeptide derived from a gliadin protein; or an equivalent of these sequences from a naturally occurring gliadin protein which is a homologue of SEQ ID NO:3. Thus the epitope may be a derivative of the protein represented by SEQ ID NO:3. Such a derivative is

typically a fragment of the gliadin, or a mutated derivative of the whole protein or fragment. Therefore the epitope of the invention does not include this naturally occurring whole gliadin protein, and does not include other whole naturally occurring gliadins.
The epitope may thus be a fragment of A-gliadin (e.g. SEQ ID NO:3), which comprises the'sequence of SEQ ID N0:ls obtainable by treating (fully or partially) with transglutaminase, i.e. with 1, 2,3 or more glutamines substituted to glutamates (including the substitution within SEQ EXNO.1).'
Such fragments may be or may include the sequences represented by positions 55 to 70, 58 to 73, 61 to 77 of SEQ ID NO:3 shown in Table 1. Typically such fragments will be recognised by T cells to at least the same extent that the peptides represented by SEQ ID NO:1 or 2 are recognised in any of the assays described herein using samples from coeliac disease patients.
Additionally, the agent may be the peptide represented by any of SEQ ID NOS:18-22, 31-36, 39-44, and46 oraprotein comprising a sequence corresponding to any of SEQ ID NOS:18-225 31-36,39-44, and 46 (such as fragments of a gliadin comprising any of SEQ ID NOS: 18-22, 31-36,39-44, and 46, for example after the gliadin has been treated with transglutaminase). Bioactive fragments of such sequences are also agents of the invention. Sequences equivalent to any of SEQ ID NOS:18-22, 31-36, 39-44, and 46 or analogues of these sequences are also agents of the invention.
In the case where the epitope comprises a sequence equivalent to the above epitopes (including fragments) from another gliadin protein (e.g. any of the gliadin proteins mentioned herein or any gliadins which cause coeliac disease), such equivalent sequences will correspond to a fragment of a gliadin protein typically treated (partially or fully) with transglutaminase. Such equivalent peptides can be determined by aligning the sequences of other gliadin proteins with the gliadin from which the original epitope derives, such as with SEQ ID NO:3 (for example using any of the programs mentioned-herein). Transglulaminase is commercially available (e.g. Sigma T-5398). Table 4 provides a few examples of suitable equivalent sequences.

The agent which is an analogue is capable of being recognised by a TCR which recognises (i) or (ii). Therefore generally when the analogue is added to T cells in the presence of (i) or (ii), typically also in the presence of an antigen presenting cell (APC) (such as any of the APCs mentioned herein), the analogue inhibits the recognition of (i) or (ii), i.e. the analogue is able to compete with (i) or (ii) in such a system.
The. analogue may be one which is capable of binding the TCR which recognises (i) or (ii). Such binding can be tested by standard techniques. Such TCRs can be isolated from X cells which have been shown to recognise (i) or (ii) (e.g. using the method of the invention). Demonstration of the binding of the analogue to the TCRs can then shown by determining whether the TCRs inhibit the binding of the analogue to a substance that binds the analogue, e.g. ari antibody to the analogue. Typically the analogue is bound to a class IIMHC molecule (e.g. HLA-DQ2) in such an inhibition of binding assay.
Typically the analogue inhibits the binding of (i) or (ii) to a TCR. In this case the amount of (i) or (ii) which can bind the TCR in the presence of the analogue is decreased. This is because the analogue is able to bind the TCR and therefore competes with (i) or (ii) for binding to the TCR.
T cells for use in the above binding experiments can be isolated from patients with coeliac disease, for example with the aid of the method of the invention.
Other binding characteristics of the analogue may also.be the same as (i) or (ii), and thus typically the analogue binds to the same MHC class n molecule to which the peptide binds (HLA-DQ2 or -DQ8). The analogue typically binds to antibodies specific for (i) or (ii), and thus inhibits binding of (i) or (ii) to such antibodies.
The analogue is typically a peptide. It may have homology with (i) or (ii), typically at least 70% homology, preferably at least 80, 90%, 95%, 97% or 99% homology with (i) or (ii), for example over a region of at least 15 more (such as the entire length of the analogue and/or (i) or (ii), or across the region which contacts the TCR or binds the MHC molecule) contiguous amino acids. Methods of measuring protein homology are well known in the art and it will be understood by those of skill

in the art that in the present context, homology is calculated on the basis of amino acid identity (sometimes referred to as "hard homology").
For example the UWGCG Package provides the BESTFIT program which can be used to calculate homology (for example used on its default settings) (Devereux et al (1984) Nucleic Acids Research 12, p387-395). The PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (typically on their default settings), for example as described in Altschul S. F. (1993) J Mol Evol 36:290-300; Altschul, S, F et al (1990) J Mol Biol 215:403-10.
Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information on the world wide web through the internet at, for example, "www.ncbLnhn.nih.gov/". This algorithm involves first identifying high scoring sequence pair (HSPs) by identifying short words of length W in the query sequence that either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighbourhood word score threshold (Altschul et al, supra). These initial neighbourhood word hits act as seeds for initiating searches to find HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; me cumulative score goes to zero or below, due to Hie accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment The BLAST program uses as defaults a word length (W) of 11, fee BLOSUM62 scoring matrix (see Henikoff and Henikoff (1992) Proc. Natl. Acad. ScL USA 89:10915-10919) alignments (B) of 50, expectation (E) of 10S M=5, N=4, and a comparison of both strands.
The BLAST algorithm performs a statistical analysis of the similarity between two sequences; see e.g., Karlin and Altschul (1993) Proc. Natl. Acad. ScL USA 90: 5873-5787. One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N))3 which provides an indication of the probability by which a match between two nucleotide or amino acid sequences

would occur by chance. For example, a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less man about 1, preferably less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
The homologous peptide analogues typically differ from (i) or (ii) by 1,2, 3, 4, 5, 6, 7, 8 of more mutations (which may be substitutions, deletions or insertions). These mutations may be measured across any of the regions mentioned above in relation to calculating homology. The substitutions are preferably 'conservative'. These are defined according to the following Table. Amino acids in the same block in the second column and preferably in the same line in the third column may be substituted for each other: (Figure Removed) Typically the ammo acids in the analogue at the equivalent positions to amino acids in (i) or (ii) mat contribute to binding the MHC molecule or are responsible for the recognition by the TCR, are the same or are conserved.
Typically the analogue peptide comprises one or more modifications, which may be natural post-translation modifications or artificial modifications. The modification may provide a chemical moiety (typically by substitution of a hydrogen, e.g. of a C-H bond), such as an amino, aceryl, hydroxy or halogen (e.g. fluorine) group or carbohydrate group. Typically the modification is present on the N or C terminus.
The analogue may comprise one or more non-natural ammo acids, for example amino acids with a side chain different from natural amino acids.

Generally, the non-natural amino acid will have an N terminus and/or a C terminus.. The non-natural amino acid may be an L- or a D- amino acid.
The analogue typically has a shape, size, flexibility or electronic configuration that'is substantially similar to (i)(or (ii). It is typically a derivative of (i) or (ii). hi one embodiment the analogue is a fusion protein comprising the sequence of SEQ ID NO:1 or 2, or any of the other peptides mentioned herein; and non-gliadin sequence.
In one embodiment the analogue is or mimics (i) or (ii) bound to a MHC class n molecule. 2,3, 4 or more of such complexes may be associated or bound to each other, for example using a biotin/streptavidin based system, in which typically 2, 3 or 4 biotin labelled MHC molecules bind to a streptavidin moiety. This analogue typically inhibits the binding of the (i) or (ii)/MHC Class IL complex to a TCR or antibody which is specific for me complex.
The analogue is typically an antibody or a fragment of an antibody, such as a Fab or (Fab)2 fragment The analogue may be immobilised on a solid support, particularly an analogue that mimics peptide bound to a MHC molecule.
The analogue is typically designed by computational means and then synthesised using methods known in the art Alternatively the analogue can be selected from a library of compounds. The library may be a combinatorial library or a display library, such as a phage display library. The library of compounds may be expressed in the display library in the form of being bound to a MHC class H molecule, such as HLA.-DQ2 or -DQ8. Analogues are generally selected from the library based on their ability to mimic the binding characteristics (i) or (ii). Thus they may be selected based on ability to bind a TCR or antibody which recognises (i) or (ii).
Typically analogues will be recognised by T cells to at least the same extent as any of the agents (i) or (ri), for example at least to the same extent as the equivalent epitope and preferably to the same extent as the peptide represented by SEQ ID N0:2, is recognised in any of the assays described herein, typically using T cells from coeliac disease patients. Analogues may be recognised to these extents in vivo and thus may be able to induce coeliac disease symptoms to at least the same

extent as any of the agents mentioned herein (e.g. in a human patient or animal model).
Analogues may be identified in a method comprising determining whether a candidate substance is recognised by a T cell receptor that recognises an epitope of the invention, recognition of the substance indicating that the substance is an analogue. Such TCRs may be any of the TCRs mentioned herein, and may be present on T cells. Any suitable assay mentioned herein can be used to identify the analogue. In one embodiment this method is carried out in vivo. As mentioned above preferred analogues are recognised to at least the same extent as the peptide SEQ ID N0:2, and so the method may be used to identify analogues which are recognised to this extent
In one embodiment the method comprises deteonining whether a candidate substance is able to inhibit the recognition of an epitope of the invention, inhibition of recognition indicating mat the substance is an analogue.
The agent may be a product comprising at least 2,5,10 or 20 agents as defined by (i), (ii) or (iii). Typically the composition comprises epitopes of the invention (or equivalent analogues) from different gliadins, such as any of me species or variety of or types of gliadin mentioned herein. Preferred compositions comprise at least one epitope of me invention, or equivalent analogue, from all of the gliadins present in any of the species or variety mentioned herein, or from 2,3,4 or more of the species mentioned herein (such as from the panel of species consisting of wheat, rye, barley, oats and triticale). Thus, the agent may be monovalent or multivalent
Diagnosis
As mentioned above the method of diagnosis of the invention may be based on the detection of T cells that bind the agent or on the detection of antibodies that recognise the agent
The T cells that recognise the agent in the method (which includes the use
mentioned above) are generally T cells that have beenpre-sensitised in vivo to gliadin. As mentioned above such antigen-experienced T cells have been found to be present in the peripheral blood.

In the method the T cells can be contacted with the agent in vitro or in vivo, and determining whether the T cells recognise the agent can be performed in vitro or in vivo. Thus the invention provides the agent for use in a method of diagnosis practiced on the human body. Different agents are provided for simultaneous, separate or sequential use in such a method.
The IK vitro method is typically carried out in aqueous solution into which the agent is added. The solution will also comprise the T cells (and in certain •embodiments the APCs discussed below). The term 'contacting' as used herein includes adding the particular substance to the solution.
Determination of whether the T cells recognise the agent is generally accomplished by detecting a change in the state of the T cells in the presence of the agent or detennining whether the T cells bind the agent. The change in state is generally caused by antigen specific functional activity of the T cell after the TCR binds the agent The change of state may be measured inside (e.g. change in intracellular expression of proteins) or outside (e.g. detection of secreted substances) the T cells.
The change in state of the T cell may be the start of or increase in secretion of a substance from the T cell, such as a cytokme, especially IFNy, IL-2 or TNF-a. Determination of IFNy secretion is particularly preferred. The substance can typically be detected by allowing it to bind to a specific binding agent and fhen measuring the presence of the specific binding agent/substance complex. The specific binding agent is typically an antibody, such as polyclonal or monoclonal antibodies. Antibodies to cytokbes are commercially available, or can be made. using standard techniques.
Typically me specific binding agent is immobilised on a solid support. After the substance is allowed to bind the solid support can optionally be washed to remove material which is not specifically bound to the agent The agent/substance complex may be detected by using a second binding agent that will bind the complex. Typically the second agent binds the substance at a site which is different from the site which binds the first agent The second agent is preferably an antibody and is labelled directly or indirectly by a detectable label

Thus the second agent may be detected by a third agent that is typically labelled directly or indirectly by a detectable label. For example the second agent may comprise a biotin moiety, allowing detection by a third agent which comprises a streptavidin moiety and typically alkaline phosphatase as a detectable label.
In one embodiment the detection system which is used is the ex-vivo ELISPOT assay described in WO 98/23960. In that assay IFN-secreted from the T cell is bound by a first ZFN-y specific antibody that is immobilised on a solid support. The bound IFN-y is then detected using a second IFNy specific antibody which is labelled with a detectable label. Such a labelled antibody can be obtained from MABTECK (Stockholm, Sweden). Other detectable kbels which can be used are discussed below.
The change in state of the T cell that can be measured may be the increase in the uptake of substances by the T cell, such as me uptake of thymidine. The change in state may be an increase in the size of the T cells, or proliferation of the T cells, or a change in cell surface markers on the T cell.
In one embodiment the change of state is detected by measuring the change in the intracellular expression of proteins, for example the increase in intracellular expression of any of me cytokines mentioned above. Such intracellular changes may be detected by contacting the inside of the T cell with a moiety that binds the expressed proteins in a specific manner and which allows sorting of the T cells by flow cytometry.
In one embodiment when binding the TCR me agent is bound to an MHC class n molecule (typically HLA-DQ2 or -DQ8), which is typically present on the surface of an antigen presenting cell (APC). However as mentioned herein other agents can bind a TCR without the need to also bind an MHC molecule.
Generally the T cells which are contacted in the method are taken from the individual in a blood sample, although other types of samples which contain T cells can be used. The sample may be added directly to the assay or may be processed first Typically the processing may comprise diluting of the sample, for example with water or buffer. Typically the sample is diluted from.l .5 to 100 fold, for example 2 to 50 or 5 to 10 fold.

The processing may comprise separation of components of the sample. Typically mononuclear cells (MCs) are separated from the samples. The MCs will comprise the T cells and APCs. Thus in the method the APCs present in the separated MCs can present the peptide to the T cells. In another embodiment only T cells, such as only CD4 T cells, can be purified from the sample. PBMCs, MCs and T cells can be, separated from the sample using techniques known in the art, such as those described in Lalvani et al (1997) J. Exp. Med. 186, p859-865.
La one embodiment, the T cells used in the assay are in the form of unprocessed or diluted samples, or are freshly isolated T cells (such as in the form of freshly isolated MCs or PBMCs) which are used directly ex. vivo, i.e. they are hot cultured before being used in the method. Thus the T cells have not been restimulated in an antigen specific manner in vitro. However the T cells can be cultured before use, for example in the presence of one or more of the agents, and generally also exogenous growth promoting cytokines. During culturmg the agent(s) are typically present on the surface of APCs, such as the APC used in the method. Pre-culturing of the T cells may lead to an increase in the sensitivity of the method. Thus the T cells can be converted into cell lines, such as short term ceH lines (for example as described in Ota et al (1990) Nature 346, pi 83-187).
The APC that is typically present in the"method may be from the same individual as the T cell or from a different host The APC may "be a naturally occurring APC or an artificial APC. The APC is a ceH that is capable of presenting the peptide to a T cell. It is typically a B cell, dendritic cell or macrophage. It is typically separated from the same sample as the T cell and is typically co-purified with the T cell. Thus me APC may be present in MCs or PBMCs: The APC is . typically a freshly isolated ex vivo cell or a cultured'cell. It may be in the form of a cell line, such as a short term or immortalised cell line. The APC may express empty MHC class E molecules on its surface.
In the method one or more (different) agents may be used. Typically the T cells derived from the sample can be placed into an assay with all the agents which it is intended to test or the T cells can be divided and placed into separate assays each of which contain one or more of the agents.

The invention also provides the agents such as two or more of any of the agents mentioned herein (e.g. the combinations of agents which are present in the composition agent discussed above) for simultaneous separate or sequential use (eg. for z>i vivo use).
In one embodiment agent per se is added directly to an assay comprising 1 ceus and APCs. As discussed above the T cells and APCs in such an assay could be in the form of MCs. When agents that can be recognised by the T cell without the need for presentation by APCs are used then APCs are not required. Analogues which mimic the original (i) or (ii) bound to a MHC molecule are an example of such an agent.
. In one embodiment the agent is provided to the APC in the absence of the T cell. The APC is-then provided to the T cell, typically after being allowed to present the agent on its surface. The peptide may have been taken up inside the APC and presented, or simply be taken up onto the surface without entering inside the APC.
The duration for which the agent is contacted with the T cells will vary depending on the method used for determining recognition of me peptide. Typically 10s to 107, preferably 5x105 to 106 PBMCs are added to each assay. In the case where agent is added directly to the assay its concentration is from 10"' to 103ug/ml, preferably 0.5 to 50u.g/ml or 1 to 10ug/ml.
Typically the length of time for which the T cells are incubated with the agent is from 4 to 24 hours, preferably 6 to 16 hours. "When using ex vivo PBMCs it has been found mat 0.3x106 PBMCs can be incubated in lOug/ml of peptide for 12 hours at37°C. ' ;
The determination of the recognition of the agent by the T cells may be done by measuring the binding of the agent to the T cells (this can be carried out using any suitable binding assay format discussed herein). Typically T cells which bind the agent can be sorted based on this binding, for example using a FACS machine. The presence of T cells that recognise the agent will be deemed to occur if me frequency of cells sorted using the agent is above a "control" value. The frequency of antigen-experienced T cells is generally 1 in 106 to 1 in 103, and therefore whether or not the sorted cells are antigen-experienced T cells can be determined.

The determination of the recognition of the agent by the T cells may be measured in vivo. Typically the agent is administered to the host and then a response which indicates recognition of the agent may be measured. The agent is typically administered intradermally or epidermally. The agent is typically administered by contacting with the outside of the skin, and may be retained at the site with the aid of a plaster or dressing. Alternatively the agent may be administered by needle, such as by injection, but can also be administered by other methods such as ballistics (e.g. the ballistics techniques which have been used to deliver nucleic acids). EP-A-0693119 describes techniques that can typically be used to administer the agent. Typically from 0.001 to 1000 ug, for example from 0.01 to 100 ug or 0.1 to 10 ug of agent is administered.
hi one embodiment a product can be administered which is capable of providing the agent in vivo. Thus a polynucleotide capable of expressing the agent can be administered, typically in any of the ways described above for the administration of the agent The polynucleotide typically has any of the characteristics of the polynucleotide provided by the invention which is discussed below. The agent is expressed from the polynucleotide in vivo. Typically from 0.001 to 1000 ug, for example from 0.01 to 100 ug or 0.1 to 10 ug of polynucleotide is administered.
Recognition of the agent administered to the slon is typically indicated by the occurrence of inflammation (e.g. induration, erythema or oedema) at the site of adtarnistration. This is generally measured by visual examination of the site.
The method of diagnosis based on the detection of an antibody that binds the . agent is typically carried out by contacting a sample -from the individual (such as any of the samples mentioned here, optionally processed in any manner mentioned herein) with the agent and determining whether an antibody hi the sample binds the agent, such a binding indicating that the individual has, or is susceptible to coeliac disease. Any suitable format of binding assay may be used, such as any such format mentioned herein.
Therapy

The identification of the immunodorninant epitope and other epitopes described herein allows therapeutic products to be made which target the T cells which recognise this epitope (such T cells being ones which participate in the immune response against gliadin). These findings also allow the prevention or treatment of coeliac disease by suppressing (by tolerisation) an antibody or T cell response to the epitope(s).
Certain agents of the invention bind the TCR that recognises the epitope of the invention (as measured using any of the binding assays discussed above) and cause tolerisation of the T cell that carries the TCR. Such agents, optionally in association with a carrier, can therefore be used to prevent or treat coeb'ac disease.
Generally tolerisation can be caused by fee same peptides which can (after being recognised by the TCR) cause antigen specific functional activity of the T cell (such as any such activity mentioned herein, e.g. secretion of cytokines). Such agents cause tolerisation when they are presented to the rmrmmft system in a 'tolerising' context.
Tolerisation leads to a decrease in the recognition of a T cell or antibody epitope by the immune system. In the case of a T cell epitope mis can be caused by the deletion or energising of T cells that recognise Ihe epitope. Thus T cell activity (for example as measured in suitable assays mentioned herein) in response to the epitope is decreased. Tolerisation of an antibody response means that a decreased amount of specific antibody to the epitope is produced when me epitope is administered.
Methods of presenting antigens-to the immune system in such a context are known and are described for example in Yoshida et aL din. Irnrminol. ImmtmopamoL 82,207-215 (1997), Thurau et aL Clin. Exp. Jmmunol. 109,370-6 (1997), and Weiner et al. Res. Immunol. 148, 528-33 (1997). In particular certain routes of administration can cause tolerisation, such as oral, nasal or intraperitoneal. Tolerisation may also be accomplished via dendritic cells and tetramers presenting peptide. Particular products which cause tolerisation may be administered (e.g. in a composition that also comprises the agent) to the individual. Such products include cytokines, such as cytokines that favour a Th2 response (e.g. IL-4, TGF-J3 or IL-10). Products or agent may be art-ministered at a dose that causes tolerisation.

The invention provides a protein that comprises a sequence able to act as an antagonist of the T cell (which T cell recognises the agent). Such proteins and such antagonists can also be used to prevent or treat coeliac disease. The antagonist will cause a decrease in the T cell response. In one embodiment, the antagonist binds the TCR of fee T cell (generally in the form of a complex with HLA-DQ2 or -DQ8) but instead of causing normal functional activation causing an abnormal signal to be passed through the TCR intracellular signalling cascade, which causes the T cell to have decreased function activity (e.g. in response to recognition of an epitope, typically as measured by any-suitable assay mentioned herein).
In one embodiment the antagonist competes with epitope to bind a component of MHC processing and presentation pathway, such as an MHC molecule (typically HLA-DQ2 or -DQ8). Thus the antagonist may bind HLA-DQ2 or -DQ8 (and thus be a peptide presented by this MHC molecule), such as peptide TP (Table 10) or a homologue thereof
Methods of causing antagonism are known in the art. In one embodiment the antagonist is a homologue of the epitopes mentioned above and may have any of the sequence, binding or other properties of the agent (particularly analogues). The antagonists typically differ from any of the above epitopes (which are capable of causing a normal antigen specific function in the T cell) by 1,2,3,4 or more mutations (each of which may be a substitution, insertion or deletion). Such antagonists are termed "altered peptide Ugands" or "APL" hi the art The mutations are typically at the amino acid positions that contact the TCR.
The antagonist may differ from the epitope by a substitution within the sequence that is equivalent to the sequence represented by amino acids 65 to 67 of A-gliadin (such antagonists are shown in Table 9). Thus preferably the antagonist has a substitution at the equivalent of position 64, 65 or 67. Preferably the substitution is 64W, 67W367Mor65T.
Since the T cell immune response to the epitope of the invention in an individual is polyclonal, more than one antagonist may need to be administered to cause antagonism of T cells of the response which have different TCRs. Therefore the antagonists may be administered in a composition which comprises at least 2,4, 6 or more different antagonists, which each antagonise different T cells.

The invention also provides a method of identifying an antagonist of a T cell (which recognises the agent), comprising contacting a candidate substance with the T cell and detecting whether the substance causes a decrease in the ability of the T cell to undergo an antigen specific response (e.g. using any suitable assay mentioned herein), the detecting of any such decrease in said ability indicating that the substance is an antagonist.
In one embodiment, the antagonists (including combinations of antagonists to a particular epitope) or tolerising (T cell and antibody tolerising) agents are present in a composition comprising at least 2,4, 6 or more antagonists or agents which antagonise or tolerise to different epitopes of the invention, for example to the combinations of epitopes discussed above in relation to the agents which are a product comprising more than one substance.
Testing whether a composition is capable of causing coeliac disease
As mentioned above the invention provides a method of determining whether a composition is capable of causing coeliac disease comprising detecting the presence of a protein sequence which is capable of being modified by a transglutaminase to as sequence comprising the agent or epitope of the invention (such transglutaminase activity may be a hitman intestinal transglutaminase activity). Typically mis is performed by using a binding assay in which a moiety which binds to the sequence in a specific manner is contacted with the composition and the formation of sequence/moiety complex is detected and used to ascertain the presence of the agent Such a moiety may be any suitable substance (or type of substance) mentioned herein, and is typically a specific antibody. Any suitable format of binding assay can be used (such as those mentioned herein).
In one embodiment, the composition is contacted with at least 2, 5,10 or more antibodies which are specific for epitopes of the invention from different gliadins, for example a panel of antibodies capable of recognising the combinations of epitopes discussed above in relation to agents of the invention which are a product comprising more than one substance.
The composition typically comprises material from a plant that expresses a gliadin which is capable of causing coeliac disease (for example any of the gliadins

or plants mentioned herein). Such material may be a plant part, such as a harvested product (e.g. seed). The material may be processed products of the plant material (e.g. any such product mentioned herein), such as a flour or food that comprises the gliadin. The processing of food material and testing in suitable binding assays is routine, for example as mentioned in Kricka LJ, J. Biolumin. Chemilumin. 13,189-93 (1998).
Binding assays
The determination of binding between any two substances mentioned herein
may be done by measuring a characteristic of either or both substances that changes
i upon binding, such as a spectroscopic change.
The binding assay format may be a 'band shift' system. This involves determining whether the presence of one substance (such as a candidate substance) advances or retards the progress of the other substance during gel electrophoresis.
The format may be a competitive binding method which determines whether the one substance is able to inhibit the binding of the other substance to an agent which is known to bind the other substance, such as a specific antibody.
Mutant gliadin proteins
The invention provides a gliadin protein in which an epitope sequence of the invention, or sequence which can be modified by a transghitaminase to provide such a sequence has been mutated so that it no longer causes, or is recognised by, a T cell response that recognises the epitope. In this context the term recognition refers to the TCR binding the epitope in such a way that normal (not antagonistic) antigen-specific functional activity of the T cell occurs. .
Methods of identifying equivalent epitopes in other gliadins are discussed above. The wild type of the mutated gliadin is one which causes coeliac disease. Such a gliadin may have homology wife SEQ ID NO:35 for example to the degree mentioned above (in relation to the analogue) across all of SEQ ID NO:3 or across 15,30,60,100 or 200 contiguous amino acids of SEQ ID NO:3. Likewise, for other non-A-gliadins, homology will be present between the mutant and the native form of that gliadin. The sequences of other natural gliadin proteins are known in the art

The mutated gliadin will not cause coeliac disease or will cause decreased symptoms of coeliac disease. Typically the mutation decreases the ability of the epitope to induce a T cell response. The mutated epitope may have a decreased binding to HLA-DQ2 or -DQ8, a decreased ability to be presented by an APC or a decreased ability to bind to or to be recognised (i.e. cause antigen-specific functional activity) by T cells that recognise the agent The mutated gliadin or epitope will therefore show no or reduced recognition in any of the assays mentioned herein in relation to the diagnostic aspects of the invention.
The mutation may be one-or more deletions, additions or substitutions of length 1 to 3,4 to 6,6 to 10, 11 to 15 or more in the epitope, for example across sequence SEQ ID NO:2 or across any of SEQIDNOS: 18-22, 31-36, 39-44, and 46; or across equivalents thereof. Preferably the mutant gliadin has at least one mutation. in the sequence SEQ ID NO:1. A preferred mutation is at position 65 in A-gliadin (or in an equivalent position in other gliadins). Typically the naturally occurring glutamine at this position is substituted to any of the amino acids shown in Table 3, preferably to histidiue, tyrosine, tryptophan, lysrne, proline, or arginine.
The invention thus also provides use of a mutation (such any of the mutations in airy of the sequences discussed herein) in an epitope of a gHadin protein, which epitope is an epitope of ihe invention, to decrease the ability of the gliadin protein to cause coeliac disease.
In one embodiment the mutated sequence is able to act as an antagonist Thus the invention provides a protein that comprises a sequence which is able to bind to a T cell receptor, which T cell receptor recognises an agent of the invention, and which sequence is able to cause antagonism of a T cell that carries such a T cell receptor.
The invention also provides proteins which are fragments of the above mutant gliadin proteins, which are at least 15 amino acids long (e.g. at least 30, 60,100, 150, 200, or 250 amino acids long) and which, comprise the mutations discussed above which decrease the ability of the gliadin to be recognised. Any of the mutant proteins (including fragments) mentioned herein may also be present in the form of fusion proteins, for example with other gliadins or with non-gliadin proteins.

The equivalent wild type protein to the mutated gliadin protein is typically from a graminaceous monocotyledon, such as a plant of genus Triticum, e.g. wheat, rye, barley, oats or triticale. The protein is typically an a, 0$, p, y or m gliadin. The gliadin may be an A-gliadin.
Kits
The invention also provides a kit for carrying out the method comprising one . or more agents and optionally a means to detect the recognition of the agent by the.T cell. Typically the different agents are provided for simultaneous, separate or sequential use. Typically the means to detect recognition allows or aids detection based on the techniques discussed above.
Thus the means may allow detection of a substance secreted by the T cells after recognition. The kit may thus additionally include a specific binding moiety for the substance, such as an antibody. The moiety is typically specific for IFN-y. The moiety is typically immobilised on a solid support. This means that after binding the moiety the substance will remain in the vicinity of the T cell which secreted it Thus "spots" of substance/moiety complex are formed on the support, each spot representing a T cell which is secreting the substance. Quantifying me spots, and typically comparing against a control, allows determination of recognition of the agent
The kit may also comprise a means to detect the substance/moiety complex. A detectable change may occur in the moiety itself after binding the substance, such as a colour change. Alternatively a second moiety directly or indirectly labelled for detection may be allowed to bind the substance/moiety complex to allow the determination of the spots. As discussed above the second moiety may be specific for the substance, but binds a different site on the substance than the first moiety.
The immobilised support may be a plate with wells, such as a microtitre plate. Each assay can therefore be carried out in a separate well in the plate.
The kit may additionally comprise medium for the T cells, detection moieties or washing buffers to be used in the detection steps. The kit may additionally comprise reagents suitable for the separation from the sample, such as the separation of PBMCs or T cells from the sample. The kit may be designed to allow detection of

the T cells directly in the sample without requiring any separation of the components of the sample.
The kit may comprise an instrument which allows administration of the agent, such as intradennal or epidermal administration. Typically such an instrument comprises plaster, dressing or one or more needles. The instrument may allow ballistic delivery of the agent The agent in the kit may be in the form of a pharmaceutical composition.
The kit may also comprise controls, such as positive or negative controls. The positive control may allow the detection system to be tested. Thus the positive control typically mimics recognition of the agent in any of the above methods. Typically in the kits designed to determine recognition in vitro the positive control is a cytokine. In the kit designed to detect in vivo recognition of the agent the positive control maybe antigen to which most individuals should response.
The kit may also comprise a means to take a sample containing T cells from the host, such as a blood sample. The kit may comprise a means to separate mononuclear cells or T cells from a sample from me host
Polynucleotides,-cells, transgenic mammals and antibodies
The invention also provides a polynucleotide which is capable of expression to provide the agent or mutant gliadin proteins. Typically the polymicleotide is DNA or KNA, and is single or double stranded- The polynucleotide win preferably comprise at least 50 bases or base pairs, for example 50 to 100,100 to 500, 500 to 1000 or 1000 to 2000 or more bases or base pairs. The polynucleotide therefore
V'
comprises a sequence which encodes the sequence of SEQ ID NO: 1 or 2 or any of the other agents mentioned herein. To the 5' and 3' of this coding sequence the polynucleotide of the invention has sequence or codons which are different from the sequence or codons 5' and 3" to these sequences in the corresponding gliadin gene.
5' and/or 3' to the sequence encoding the peptide the polynucleotide has coding or non-coding sequence. Sequence 5' and/or 3' to the coding sequence may comprise sequences which aid expression, such as transcription and/or translation, of the sequence encoding the agent. The polynucleotide may be capable of expressing the agent prokaryotic or eukaryotic cell. In one embodiment the polynucleotide is

capable of expressing the agent in a mammalian cell, such as a human, primate or rodent (e.g. mouse or rat) cell.
A polynucleotide of the invention may hybridise selectively to a polynucleotide that encodes SEQ ID NO:3 at a level significantly above background. Selective hybridisation is typically achieved using conditions of medium to high stringency (for example 0.03M sodium chloride and 0.03M sodium citrate at from about 50°C to about 60°C). However, such hybridisation may be carried out under any suitable conditions known in the art (see Sambrook et al (1989), Molecular Cloning: A Laboratory Manual). :For example, if high stringency is required, suitable conditions include 0.2 x SSC at 60°C. If lower stringency is required, suitable conditions include 2 x SSC at 60°C.
Agents or proteins of the invention may be encoded by the polynucleotides described herein.
The polynucleotide may form or be incorporated into a replicable vector. Such a vector is able to replicate in a suitable cell. The vector may be an expression vector. In such a vector the polynucleotide of the invention is operably linked to a control sequence which is capable of providing for the expression of the polynucleotide. The vector may contain a selectable marker, such as the ampiciUin resistance gene. •
The polynucleotide or vector may be present in a cell. Such a cell may have been transformed by the polynucleotide or vector. The cell may express the agent The cell will be chosen to be compatible with the said vector and may for example be a prokaryotic (bacterial), yeast, insect or mammalian cell. The polynucleotide or' vector may be introduced into host cells using conventional techniques including calcium phosphate precipitation, DEAE-dextran transfection, or electroporation.
The invention provides processes for the production of the proteins of the invention by recombinant means. This may comprise (a) cultivating a transformed cell as defined above under conditions that allow the expression of the protein; and preferably (b) recovering the expressed polypeptide. Optionally, the polypeptide may be isolated and/or purified, by techniques known in the art.
The invention also provides TCRs which recognise (or bind) the agent, or fragments thereof which are capable of such recognition (or binding). These can be

present in the any form mentioned herein (e.g. purity) discussed herein in relation to the protein of the invention. The invention also provides T cells which express such TCRs which can be present in any form (e.g. purity) discussed herein for the cells of the invention.
The invention also provides monoclonal or polyclonal antibodies which specifically recognise the agents (such as any of the epitopes of the invention) and which recognise the mutant gliadin proteins (and typically which do not recognise the equivalent wild-type gliadins) of the invention, and methods of making such antibodies. Antibodies of the invention bind specifically to these substances of the invention.-
For the purposes of this invention, the term "antibody" includes antibody fragments such as Fv, F(ab) and F(ab)2 fragments, as well as single-chain antibodies.
A method for producing a polyclonal antibody comprises immunising a suitable host animal., for example an experimental nnfriml with me irmmmogen and isolating immunoglobulins from the serum. The animal may therefore be inoculated with the immunogen, blood subsequently removed from the animal and the IgG fraction purified, A method for producing a monoclonal antibody comprises immortalising cells which produce the desired antibody. Hybridoma cells may be produced by fusing spleen cells from an inoculated experimental grrirnal wifli tumour cells (Kohler and Milstein (1975) Nature 256,495-497).
An immortalized cell producing the desired antibody may be selected by a conventional procedure. The hybridomas may be grown in culture or injected intraperitoneaHy for formation of ascites fluid or into the blood stream of an allogenic host or hnmunocompromised host Human antibody may be prepared by in vitro immunisation of human lymphocytes, followed by transformation of the lymphocytes with Epstein-Barr virus.
For the production of both monoclonal and polyclonal antibodies, the experimental animal is suitably a goat, rabbit, rat or mouse. If desired, the unmunogen may be administered as a conjugate in which the immunogen is coupled, for example via a side chain of one of the amino acid residues, to a suitable carrier. The carrier molecule is typically a physiologically acceptable carrier. The antibody obtained may be isolated and, if desired, purified.

The polynucleotide, agent, protein 01 antibody of the invention, may cany a detectable label. Detectable labels which allow detection of the secreted substance by visual inspection, optionally with the aid of an optical magnifying means, are preferred. Such a system is typically based on an enzyme label which causes colour change in a substrate, for example alkaline phosphatase causing a colour change in a substrate. Such substrates are commercially available, e.g. from BioRad. Other suitable labels include other enzymes such as peroxidase; or protein labels, such as biotin; or radioisotopes, such as P or S. The above labels may be detected using known techniques.
Polynucleotides, agents, proteins, antibodies or cells of the invention may be in substantially purified form. They may be in substantially isolated form, in which case they will generally comprise at least 80% e.g. at least 90,95, 97 or 99% of the polynucleotide, peptide, antibody,-cells or dry mass in fhe preparation. The polynucleotide, agent, protein or antibody is typically substantially free of other cellular components. The polynucleotide, agent, protein or antibody may be used in such a substantially isolated, purified" or free form in the method or be present in such forms in fhe kit
The invention also provides a transgenic non-human mammal which expresses a TCR of the invention. This may be any of the mammals discussed herein (e.g. in relation to the production of the antibody). Preferably the mammal has, or is susceptible, to coeliac disease. The -mammal may also express HLA-DQ2 or -DQ8 or HLA-DR3-DQ2 and/or may be given a diet comprising a gliadin which cause coeliac disease (e.g. any of the gliadin proteins mentioned herein). Thus the mamma? may act as an animal model for coeliac disease.
The invention also provides a method of identifying a product which is therapeutic for coeliac disease comprising administering a candidate substance to a mammal of the invention which has, or which is susceptible to, coeliac disease and determining whether substance prevents or treats coeliac disease in the mammal., fas prevention or treatment of coeliac disease indicating that the substance is a therapeutic product Such a product may be used to treat or prevent coeliac disease.
The invention provides therapeutic including prophylactic) agents or diagnostic substances (the agents, proteins andpotynucleotides of the invention).

These substances are formulated for clinical administration by mixing them with a pharmaceutically acceptable carrier or diluent. For example they can be formulated for topical, parenteral, intravenous, intramuscular,, subcutaneous, intraocular, intradermal, epidermal or transdennal administration. The substances may be mixed with any vehicle which is pharmaceutically acceptable and appropriate for the desired route of administration. The pharmaceutioally carrier or diluent for injection may be, for example, a sterile or isotonic solution such as Water for Injection or physiological saline, or a carrier particle for ballistic delivery.
The dose of the substances may be adjusted according to various parameters, especially according to the agent used; the age, weight and condition of the patient to be treated; the mode of administration used; the severity of the condition to be treated; and the required clinical regimen. As a guide, the amount of substance administered by injection is suitably from 0.01 mg/kg to 30 mg/kg, preferably from 0.1 mg/kg to 10 mg/kg.
The routes of administration and dosages described are intended only as a guide since a sHUed practitioner wfll be able to determine readily the optimum route of administration and dosage for any particular patient and condition.
The substances of the invention may thus be used in a method of treatment of the human or animal body, or in a diagnostic method practised on the human body. In particular they may be used in a method of treating or preventing coeliac disease. The invention also provide the agents for use in a method of manufacture of a medicament for treating or preventing coeliac disease. Thus the invention provides a method of preventing or treating coeliac disease comprising administering to a human hi need thereof a substance of the invention (typically a non-toxic effective amount thereof).
The agent of the invention can be made using standard synthetic chemistry techniques, such as by use of an automated synthesizer. The agent may be made from a longer polypeptide e.g. a fusion protein, which polypeptide typically comprises the sequence of the peptide. The peptide may be derived from the polypeptide by for example hydrolysing me polypeptide, such as using a protease; or by physically breaking the polypeptide. The polynucleotide of the invention can be made using standard techniques, such as by using a synthesiser.

Plant cells and plants that express mutant gliadin proteins or express proteins comprising sequences which can act as antagonists
The cell of the invention may be a plant cell, such as a cell of a graminaceous monocotyledonous species. The species may be one whose wild-type form expresses gliadins, such as any of the gliadin proteins mentioned herein (including gliadins with any degree of homology to SEQ ID NO:3 mentioned herein). Such a gliadin may cause coeliac disease in humans. The cell may be of wheat, maize, oats, rye, rice, barley, triticale, sorghum, or sugar cane. Typically the cell is of the Txiticum genus, such as aestivum, spelta, polonicum or monococcum.
The plant cell of the invention is typically one which does not express a wild-type gliadin (such.as any of the gliadins mentioned herein which may cause coeliac disease), or one which does not express a gliadin comprising a sequence that can. be recognised by a T cell that recognises the agent Thus if the wild-type plant cell did express such a gliadin then it may be engineered to prevejnt or reduce the expression of such a gliadin or to change the amino acid sequence of the gliadin so mat it no longer causes coeliac disease (typically by no longer expressing the epitope of the invention).
This can be done for example by introducing mutations into 1,2,3 or more or all of such gliadin genes hi the cell, for example into coding or non-coding (e.g. promoter regions). Such mutations can be any of the type or length of mutations discussed herein (e.g., in relation to homologous proteins). The mutations can be introduced in a directed manner (e.g., using site directed mutagenesis or homologous recombination techniques) or in a random manner (e.g. using a mutagen, and men typically selecting for mutagenised cells which no longer express the gliadin (or a gh'adin sequence which causes coeliac disease)).
In the case of plants or plant cells that express a protein that comprises a sequence able to act as an antagonist such a plant or plant cell may express a wild-type gliadin protein (e.g. one which causes coeliac disease). Preferably though the presence of the antagonist sequence will cause reduced coeliac disease symptoms (such as no symptoms) in an individual who ingests a food comprising protein firoi the plant or plant cell.

The polynucleotide which is present in (or which was transformed into) the plant cell will generally comprise promoter capable of expressing the mutant gliadin protein the plant cell. Depending on the pattern of expression desired, the promoter may be constitutive, tissue- or stage-specific; and/or inducible. For example, strong constitutive expression in plants can be obtained with the CAMV 35S, Rubisco ssu, or histone promoters. Also, tissue-specific or stage-specific promoters may be used to target expression of protein of the invention to particular tissues in a transgenic plant or to particular stages in its development Thus, for example seed-specific, root-specific, leaf-specific, flower-specific etc promoters may be used. Seed-specific promoters include those described by Dalta et al (Biotechnology Ann. Rev. (1997), 3, pp.269-296). Particular examples of seed-specific promoters are napin promoters (EP-A-0 255, 378), phaseoUn promoters, glutenine promoters, helianthenrne promoters (WO92/17580), albumin promoters (WO98/45460), oleosin promoters (W098/45461) and ATS1 and ATS3 promoters (PCT/US98/06798).
The cell may be in any form. For example, it may be an isolated cell, e.g. a protoplast, or it may be part of a plant tissue, e.g. a callus, or a tissue excised from a plant, or it may be part of a whole plant The cell may be of any type (e.g. of any ' type of plant part). For example, an undifferentiated cell, such as a callus cell; or a differentiated cell, such as a cell of a type found in embryos, pollen, toots, shoots or leaves. Plant parts include roots; shoots; leaves; and parts involved .in reproduction, such as pollen, ova, stamens, anthers, petals, sepals and other flower parts.
The invention provides a method of obtaining a transgenic plant cell comprising tansforming a plant cell with a polynucleotide or vector of the invention to give a tra-nsggnic plant celL Any suitable transformation method may be used (in the case of wheat the techniques disclosed in Vasfl V et al, Biotechnology 10,667-674 (1992) may be used). Preferred transformation techniques include electroporation of plant protoplasts and particle bombardment Transformation may thus give rise to a chimeric tissue or plant in which some cells are transgenic and some are not.
The cell of the invention or thus obtained cell may be regenerated into a transgenic plant by techniques known in the art These may involve the use of plant growth substances such as auxins, giberellins and/or c^kinins to stimulate the

growth and/or division of the transgenic cell. Similarly, techniques such as somatic embryogenesis and meristem culture may be used. Regeneration techniques are well known in the art and examples can be found in, e.g. US 4,459,355, US 4,536,475, US 5,464,763, US 5,177,010, US 5,187,073, EP 267,159, EP 604,662, EP 672, 752, US 4,945,050, US 5,036,006, US 5,100,792, US 5,371,014, US 5,478,744, US 5,179,022, US' 5,565,346, US 5,484,956, US 5,508,468, US 5,538,877, US 5,554,798, US 5,489,520, US 5,510,318, US-5,204,253, US 5,405,765, EP 442,174, EP 486,233, EP 486,234, EP 539,563, EP 674,725, WO91/02071 and WO 95/06128.
In many such techniques, "one step is the formation of a callus, Le. a plant tissue comprising expanding and/or dividing cells. Such calli are a further aspect of the invention as are other types of plant cell cultures and plant parts. Thus, for example, the invention provides transgenic plant tissues and parts, including embryos, meristems, seeds, shoots, roots, stems, leaves and flower parts. These may be cbimeric in the sense mat some of their cells are cells of the invention and some are not Transgenic plant parts and tissues, plants and seeds of the invention may be of any of the plant species mentioned herein.
Regeneration procedures will typically involve the selection of transformed cells by means of marker genes.
The regeneration step gives rise to a first generation transgenic plant The invention also provides methods of obtaining transgenic plants of farmer generations from this first generation plant These are known as progeny transgenic plants. Progeny plants of second, third, fourth, fifth, sixth and further generations may be obtained from the first generation transgenic plant "by any means known in fee art
Thus, the invention provides a method of obtaining atransgenic progeny plant comprising obtaining a second-generation ttansgenic progeny plant from a first-generation transgenic plant of the invention, and optionally obtaining transgenic plants of one or more further generations from the second-generation progeny plant thus obtained.
Progeny plants may be produced from their predecessors of earlier generations by any known technique. In particular, progeny plants may be produced by:

obtaining a transgenic seed from a transgenic plant of the invention belonging to a previous generation, then obtaining a transgenic progeny plant of the invention belonging to a new generation by growing up the tcansgenic seed; and/or
propagating clonally a transgenic plant of the invention belonging to a previous generation to give a transgenic progeny plant of the invention belonging to a new generation; and/or
crossing a first-generation transgenic plant of the invention belonging to a previous generation with another compatible plant to give a transgenic progeny plant of the invention belonging to-a new generation; and optionally
obtaining transgenic progeny plants of one or more further generations from the progeny plant thus obtained.
These techniques may be used in any combination. For example, clonal propagation and sexual propagation may be used at different points in a process mat gives rise to a transgenic plant suitable for cultivation. In particular, repetitive back-crossing with a plant taxon with agronomically desirable characteristicB may be undertaken. Further steps of removing cells from a plant and regenerating new plants therefrom may also be carried-out
Also, further desirable characteristics may be introduced by transforming the cells, plant tissues, plants or seeds, at any suitable stage in the above process, to introduce desirable coding sequences other than the polynucleotides of the invention. This may be carried out by the techniques described herein for the mtromiction of polynucleotides of the invention.
For example, farther transgenes may "be selected from those coding for other herbicide resistance traits, e.g. tolerance to: Glyphosate (e.g. using an EPSP synthase gene (e.g. EP-A-0 293,358) or a glyphosate oxidoreductase (WO 92/000377) gene); or tolerance to fosametin; a dihalobenzonitrile; glufosinate, e.g. using a phosphinothrycin acetyi transferase (PAT) or glutamme synthase gene (cf. EP-A-0 242,236); asulam, e.g. using a dihydropteroate synthase gene (EP-A-0 369,367); or a sulphonylurea, e.g. using an ALS gene); diphenyl ethers such as acifluorfen or oxyfhiorfen, e.g. using aprotoporphyrogen oxidase gene); an oxadiazole such as oxadiazon; a cyclic imide such as chlorophthalim; a phenyl pyrazole such as TNP, or a phenopylate or carbamate analogue thereof.

Similarly, genes for beneficial properties other than herbicide tolerance may be introduced. For example, genes for insect resistance may be introduced, notably genes encoding Bacillus thuringiensis (Bf) toxins. Likewise, genes for disease resistance may be introduced, e.g. as in WO91/02701 or WO95/06128.
Typically, a protein of the invention is expressed in a plani of the invention. Depending on the promoter used, this expression may be constitutive or inducible, Similarly, it may be tissue- or stage-specific, i.e. directed towards a particular plant tissue (such as any of the tissues mentioned herein) or stage in plant development
The invention also provides methods of obtaining crop products by harvesting, and optionally processing further, transgenic plants of the invention. By crop product is meant any useful product obtainable from a crop plant
Products that contain mutant gliadin proteins or proteins that comprise sequence capable of acting as an antagonist
The invention provides a product that comprises the mutant gliadin proteins or protein that comprises sequence capable of acting as an antagonist This is typically derived from or comprise plant parts from plants mentioned herein which express such proteins. Such a product may be obtainable directly by harvesting or indirectly, by harvesting and further processing the plant of the invention. Directly obtainable products include grains. Alternatively, such a product may be obtainable indirectly, by harvesting and further processing. Examples of products obtainable by further processing are flour or distilled alcoholic beverages; food products made from directly obtained or further processed material, e.g. baked products (e.g. bread) made from flour. Typically such food products, which are ingestible and digestible (i.e. non-toxic and of nutrient value) by human individuals.
In the case of food products that comprise the protein which comprises an antagonist sequence the food product may also comprise wild-type gliadin, but-preferably the antagonist is able to cause a reduction (e.g. completely) in the coeliac disease symptoms after such food is ingested.
The invention is illustrated by the following nonlimiting Examples: Example 1

We carried out epitope mapping in Coeliac disease by using a set of 51 synthetic 15-mer peptides that span the complete sequence of a fully characterized a-gliadin, "A-gliadin" (see Table 1). A-Gliadin peptides were also individually treated with tTG to generate products that might mimic those produced in'vivo3. We also sought to study Coeliac disease patients at the point of initiation of disease relapse to avoid the possibility that epitope "spreading" or "exhaustion" may have occurred, as described in experimental infectious and autoimmune diseases.
Clinical and A-gliadin specific T'cell responses with 3 and 10 day bread challenge In a pilot study, two subjects with Coeliac disease in remission, defined by absence of serum anti-endomysial antibody (EMA), on a gluten free diet were fed four slices of standard gluten-containing white bread daily in addition to their usual gluten free diet. Subject 1 ceased bread because of abdominal pain, mouth ulcers and mild diarrhoea after three days, but Subject 2 continued for 10 days wirh-only mild nausea at one week. The EMA became positive in Subject 2 one week after the bread challenge, indicating the bread used had caused a relapse of Coeliac disease. But in Subject 1, EMA remained negative up to two months after bread challenge. In both subjects, symptoms that appeared with bread challenge resolved within two days after returning to gluten free diet.
PBMC responses in IFNy ELISPOT assays to A-gliadin peptides were not found before or during bread challenge. But from the day after bread -withdrawal (Day-4) in Subject 1 a single pool of 5 overlapping peptides spanning A-gliadin 51-85 (Pool 3) treated with tTG showed potent IFNy responses (see Figure la). In Subject 1, the PBMC IFNy response to A-gliadin peptide remained targeted to Pool 3 alone and was maximal on Day 8. The dynamics and magnitude of the response to Pool 3 was similar to that elicited by a-chymotrypsin digested gliadin. PBMC IFNy responses to tTG-treated Pool 3 were consistently 5 to 12-fold greater than Pool 3 not treated with tTG, and responses to a-chymotrypsin digested gliadin were 3 to 10-fold greater if treated with tTG. In Subject 2, Pool 3 treated with tTG was also the only immunogenic set of A-gliadin peptides on Day 8, but this response was weaker than Subject 1, was not seen on Day 4 and by Day 11 the response to Pool 3 had diminished and other tTG-treated pools of A-gliadin peptides elicited stronger IFNa

responses (see Figure Ib).
The pilot study indicated that the initial T cell response in these Coeliac disease subjects was against a single tTG-treated A-gliadin pool of five peptides and was readily measured in peripheral blood. But if antigen exposure is continued for ten days instead of three, T eel] responses to other A-gliadin peptides appear, consistent with'epitope spreading.
Coeliac disease-specific IFN-g induction by tTG-treated A-gliadin peptides
In five out of six further Goeliac disease subjects on gluten free diet (see Table 1), bread challenge for three days identified tTG-treated peptides in Pool 3, and in particular, peptides corresponding to 56-70 (12) and 60-75 (13) as the sole A-gliadin components eliciting IFNy from PBMC (see Figure 2). EL-10 ELISPOT assays run in parallel to IFN-y' ELISPOT showed no IL-10 response to tTG-treated peptides 12 or 13. In one subject, there were no IFNy responses to any A-gliadin peptide or a-chymotrypsin digested gliadin before, during or up to four days after bread challenge. In none of these Coeliac disease subjects didEMA status change from baseline when measured for up to two months after bread challenge.
PBMC from four healthy, EMA-negative subjects with the HLA-DQ aBeles al *0501, pi *0201 (ages 28-52,2 females) who had been challenged for tiiree days with bread after following a gluten free diet for one month, showed no IFNy responses above the negative control to any of the A-gHadin peptides with or without tTG treatment Thus, induction of IFN-y in PBMC to tTG-treated Pool 3 and A- • gliadin peptides 56-70 (12) and 60-75 (13) were Coeliac disease specific (7/8 vs. 0/4,. p Fine mapping of the minimal A-gliadin T cell epitope
tTG-treated peptides representing truncations of A-gliadin 56-75 revealed that the same core peptide sequence QPQLP (SEQ ID NO:9) was essential for antigenicity in all of the five Coeliac disease subjects assessed (see Figure 3). PBMC TFN-y responses to tTG-treated peptides spanning this core sequence beginning with the 7-mer PQPQLPY (SEQ ID NO:4) and increasing in length, 'indicated mat the tTG-treated 17-mer QLQPFPQPQLPYPQPQS (SEQ ID NO.10). (A-gliadin 57-73)

possessed optimal activity in the IFN-y ELISPOT (see Figure 4).
Deamidation ofQ65 by tTG generates the immunodominant T cell epitope in A-gliadin
HPLC analysis demonstrated that tTG treatment of A-gliadin 56-75 generated a single product that eluted marginally later than the parent peptide. Amino acid sequencing indicated that out of the six glutamine (Q) residues contained in A-gliadin 56-75, Q65 was preferentially deamidatedby tTG (see Figure 5). Bioactivity of peptides corresponding to serial expansions from the core A-gliadin 62-68 sequence in which glutamate (E) replaced Q65, was equivalent to the same peptides with Q65 after tTG-treatment (see Figure 4a). Replacement of Q57 and Q72 by E together or alone, with E65 did not enhance antigenicity of the 17-mer in the three Coeliac disease subjects studied (see Figure 6). Q57 and Q72 were investigated because glutamine residues followed by proline in gliadin peptides are not deamidated by tTG in vitro (W. Vader et al, Proceedings 8th International .Symposium Coeliac Disease). Therefore, the irnmunodominant T cell epitope was defined as QLQPFPQPELPYPQPQS (SEQ ID N0:2).
Immunodominant T cell epitope response is DQ2-restricted and CD4 dependent
In two Coeliac disease subjects homozygous forHLA-DQ al*0501, P 1*0201, anti-DQ monoclonal antibody blocked the ELISPOT EFNy response to tTG-treated A-gliadin 56-75, but anti-DP and -DR antibody did not (see Figure 7). Anti-CD4 and anti-CDS magnetic bead depletion of PBMC from two Coeliac disease subjects indicated the IFNy response to tTG-treated A-gliadin 56-75 is CD4 T cell-mediated.
Discussion
In this study we describe a rather simple dietary antigen challenge using standard white bread to elicit a transient population of CD4 T cells in peripheral blood of Coeliac disease subjects responsive to a tTG-treated A-gliadin 17-mer with the sequence: QLQPFPQPELPYPQPQS (SEQ ID NO:2) (residues 57-73). The immune response to A-gliadin 56-75 (Q-E65) is restricted to the Coeliac disease-

associated HLA allele, DQ a 1*0501, P 1*0201. Tissue transglutaminase action in vitro selectively deamidates Q65. Elicited peripheral blood IFNg responses to synthetic A-gliadin peptides with the substitution Q-+E65 is equivalent to tTG-treated Q65 A-gliadin peptides; both stimulate up to 10-fold more T cells in ihe IFNg ELISPOT than unmodified Q65 A-gliadin peptides.
We have deliberately defined this Coeliac disease-specific T cell epitope using in vivo antigen challenge and short-term ex vivo immune assays to avoid the possibility of methodological artifacts that may occur with the use of T cell clones in epitope mapping. Our findings indicate that peripheral blood T cell responses to ingestion of gluten are rapid but short-lived and can be utilized for epitope mapping. In vivo antigen challenge has also shown there is a temporal hierarchy of immune responses to A-gliadin peptides; A-gliadin 57-73 modified by tTG not only elicits the strongest IFNg response in PBMC but it is also the first IFNg response to appear.
Because we have assessed only peptides spanning A-gliadin, there may be other epitopes in other gHadins of equal or greater importance in me pamogenesis of Coeliac disease. Indeed, the peptide sequence at Hie cote of the epitope in A-gliadin that we have identified PQPQLPY (SEQ ID NO:4) is shared by several other gliadins (SwissProt and Trembl accession numbers: P02863, Q41528, Q41531, Q41533, Q9ZP09, P04722, P04724, P18573). However, A-gliadin peptides that have previously been shown to possess bioactiviry in biopsy challenge and in vivo studies (for example: 31-43,44-55, and 206-217)4>5 did not elicit IFNg responses in PBMC following three day bread challenge in Coeliac disease subjects. These peptides may be "secondary". T cell epitopes that arise with spreading of the immune response.
Example 2
The effect on T cell recognition of substitutions in the immunodominant epitope
The effect of substituting the glutamate at position 65 hi the 57-73 A-gliadin epitope was determined by measuring peripheral blood responses, against the substituted epitopes in an IFNy ELISPOT assay using synthetic peptides (at 50 (.ig/ml). The responses were measured hi 3 Coeliac disease subjects 6 days after commencing gluten challenge (4 slices bread daily for 3 days). Results are shown in table 3 and Figure 8. As can be seen substitution of the glutamate to histidine,

tyrosine, tryptophan, lysine, proline or arginine stimulated a response whose magnitude was less than 10% of the magnitude of the response to the immunodominant epitope. Thus mutation of A-gliadin at this position could be used to produce a mutant gliadin with reduce or absent immunoreactivity.
Example 3
Testing the immunoreactivity of equivalent peptides from other naturally occurring gliadins
The immunoreactivity of-equivalent peptides form other naturally occurring wheat gliadins was assessed using synthetic peptides corresponding to the naturally occurring sequences which were then treated with transglutaminase. "These peptides were tested in an ELISPOT in the same manner and with PBMCs from the same subjects as described in Example 2. At least five of the peptides show irmnunoreactivity comparable to the A-gliadin 57-73 E65 peptide (after transglutaminase treatment) indicating that other gliadin proteins in wheat axe also likely to induce this Coeliac disease-specific immune response (Table 4 and Figure 9)-
Methods
Subjects: Patients used in the study attended a Coeliac Clinic in Oxford, United Kingdom. Coeliac disease was diagnosed on the basis of typical small intestinal histology, and normalization of symptoms and small intestinal histology with gluten free diet.
Tissue typing: L issue cyping was performed using DNA extracted from EDTA-anticoagulated peripheral blood. HLA-DQA and DQB genotyping was performed by PCR using sequence-specific primer mixes64.
Anti-endomysial antibody assay: EMA were detected by indirect immunofluorescence using patient serum diluted 1:5 with monkey oesophagus, followed by FITC-conjugated goat anti-human IgA. IgA was quantitated prior to EMA, none of the subjects were IgA deficient

Antigen Challenge: Coeliac disease subjects following a gluten fiee diet, consumed 4 slices of gluten-containing bread (50g/slice, Sainsbuiy's "standard white sandwich bread") daily for 3 or 10 days. EMA was assessed the week before and up to two months after commencing the bread challenge. Healthy subjects who had followed a gluten free diet'for four weeks, consumed their usual diet including four slices of gluten-containing bread for three days, then returned to gluten free diet for a further six days.
IFNy aridlLlOEHSPOT: PBMC were prepared from 50-100 ml of venous blood by Ficoll-Hypaque densiiy centrifugation. After three washes, PBMC were resuspended in complete RPMI containing 10% heat inactivated human AB serum. ELISPOT assays for single cell secretion of IFNy and IL-10 were performed using commercial kits (Mabtech; Stockholm, Sweden) with 96-wellplates (MAIP-S-45; Millipore, Bedford, MA) according to the manufacturers instructions (as described elsewhere9) with 2-5x10s (IFNy) or 0.4-lxlO5 (EHO) PBMC in each well PeptLdes were assessed in duplicate wells, andMycobacterium tuberculosis purified protein derivative (PPD RT49) (Serum Institute; Copenhagen, Denmark) (20 ug/ml) was included as a positive control in all assays.
Peptides: Synthetic peptides were purchased from Research Genetics (Huntsville, Alabama) Mass-spectroscopy and HPLC verified peptides' authenticity and >70% purity. Digestion of gliadin (Sigma; G-3375) (100 mg/ml) with a-chymotrypsin (Sigma; C-3,142) 2&0:1 (w/w) was performed at room temperatare in 0.1 M NEUHCQs with 2M urea and was halted after 24 h by heating to 98°C for 10 minutes. After centrifugation (13,000g, 10 minutes), the gliadin digest supernatant was filter-sterilized (0.2 mm). Digestion of gliadin was verified by SDS-PAGE and protein concentration assessed. a-Chymotrypsin-digested gliadin (640 jig/ml) and synthetic gliadin peptides (15-mers: 160 fig/ml, other peptides: 0.1 mM) were individually treated with tTG (Sigma; T-5398) (50 ng/ml) hi PBS + CaCl2 1 mM for 2 h at 37°C. Peptides and peptide pools were aliquotted into sterile 96-well plates and stored frozen at -20°C until use.

Amino acid sequencing ofpeptides: Reverse phase HPLC was used to purify the peptide resulting from tTG treatment of A-gliadin 56-75. A single product was identified and subjected to amino acid sequencing (automated sequencer Model 494 A, Applied Biosystems, Foster City, California). The sequence of unmodified G56-75 was confirmed as: LQLQPFPQPQLPYPQPQSFP (SEQ ID NO:5), and tTG treated G56-75 was identified as: LQLQPFPQPELPYPQPQSFP (SEQ ID NO:11). Deamidation of glutamyl residues was defined as the amount (pmol) of glutamate recovered expressed as a percentofthe combined amount of glutamine and glutamate recovered in cycles 2,4, 8,10,15 and 17 of the amino acid sequencing. Deamidation attributable to tTG was defined as (% deamidation of glutamine in the tTG treated peptide - % deamidation in "the untreated peptide) / (100 - % deamidation in the untreated peptide).
CD4/CD8 and HLA Class H Restriction: Anti-CD4 or anti-CD8 coated magnetic beads (Dynal, Oslo, Norway) were washed four times with. RPMI men incubated with PBMC in complete RPMI containing 10% heat inactivated human AB serum (5x106 cells/ml) for 30 minutes on ice. Beads were removed using a magnet and cells remaining counted. In vivo HLA-class E restriction of the Tnrnmmft response to tTG-treated A-gliadin 56-75 was established by incubating PBMC (5xl06 cells/ml) with anti-HLA-DR (L243), -DQ (L2), and -DP (B7.21) monoclonal antibodies (10 ug/ml) at room temperature for one hour prior to the addition of peptide.
Example 4
Mucosal integrin expression by gliadm -specific peripheral blood lymphocytes
Interaction between endothelial and lymphocyte adressins facilitates homing of organ-specific lymphocytes. Many adressins are known. The heterodimer 04(37 is specific for lamina propria gut and other mucosal lymphocytes, and a pV is specific and intra-epithelial lymphocytes in the gut and skin- Approximately 30% of peripheral blood CD4 T cells express 04^7 and are presumed to be in transit to a mucosal site, while 5% of peripheral blood T cells express cEp7. Jmmunomagnetic beads coated with antibody specific for aE or P 7 deplete PBMC of cells expressing aE^7 or aEpv and 04^7, respectively. In combination with ELISpot assay,

immunomagnetic bead depletion allows determination of gliadin-specific T cell addressin expression that may identify these cells as homing to a mucosal surface. Interestingly, gluten challenge hi vivo is associated with rapid influx of CD4 T cells to foe small intestinal lamina propria (not intra-epithelial sites), where over 90% lymphocytes express 04^7.
Immunomagnetic beads were prepared and used to deplete PBMC from coeliac subjects on day 6 or 7 after commencing 3 day gluten challenge. FACS analysis demonstrated a E beads depleted approximately 50% of positive CD4 T cells, while p7 beads depleted all-£7 positive CD4 T cells. Depletion of PBMC using CD4- or P 7-beads, but not CDS- or a E -beads, abolished responses in the interferon gamma ELISpot. tTG gliadin and PPD responses were abolished by CD4 depletion, but consistently affected by rutegrin-specific bead depletion.
Thus A-gliadin 57-73 QB65-specific T cells induced after gluten challenge in coeliac disease express the mtegrin, 04^7, present on lamina propria CD4 T cells in the small intestine.
Examples '
Optimal T cell Epitope Length
Previous data testing peptides from 7 to 17 amino acids in length spanning
the
core of the dominant T cell epitope in A-gliadin indicated that 1fae 17mer, A-gliadin 57-73 QE65 (SEQ ID NO:2) induced maximal responses hi 1he interferon gamma Ehspot using peripheral blood mononuclear cells (PBMC) from coeliac volunteers 6 days after commencing a 3-day gluten challenge.
Peptides representing expansions form the core sequence of the dominant T cell epitope hi A-gliadin were assessed in the IFN gamma ELISPOT using peripheral blood moaonuclear cells (PBMC) from coeliac volunteers in 6 days after commencing a 3-day gluten challenge (n=4). Peptide 13: A-gliadin 59-71 QE65 (13mer), peptide 15: 58-72 QE65 (15mer), „., peptide 27: 52-78 SE65 (27mer).
As shown hi Figure 11 expansion of the A-gliadin 57-73 QE65 sequence does not substantially enhance response in the IFNgamma Elispot Subsequent Examples

characterise the agonist and antagonist activity of A-gliadin 57-73 QE65 using 17mer peptides.
Example 6
Comparison of A-gliadin 57-73 'QE65 with other DQ2-restricted T cell epitopes in coeliac disease
Dose response studies were performed using peptides corresponding to unmodified and transglutaminase-rreated peptides corresponding to T cell epitopes of gluten-specific T cell clones and iiries from intestinal biopsies of coeliac subjects. Responses to peptides were expressed as percent of response to A-gliadin 57-73 QE65.' All subjects were HLA-DQ2+ (none were DQ84-).
The studies indicate that A-gliadin 57-73 QE65 is the most potent gtiadin peptide for induction of interferon gamma in the ELJSpot assay using coeliac PBMC after gluten challenge (see Figure 12a-h, and Tables 5 and 6). The second and third epitopes are suboptimal fragments of larger peptides Le. A-gliadin 57-73 QE65 and GDA4_WHEAT P04724-84-100 QE92. The epitope is only modestly bioactive (approximately 1/20* as active as A-gliadin 57-73 QE65 after blank is subtracted).
A-gliadin 57-73 QE65 is more potent man other known T cell epitopes in coeliac disease. There are 16 polymorphisms of A-gliadin 57-73 (including the sequence PQLPY (SEQ ID NO: 12)) amongst sequenced gliadin genes, their bioactiviry is assessed next
Example 7
Comparison of gliadin- and A-gliadin 57-73 QE65-spedfic responses in peripheral
blood
The relative contribution of the dominant epitope, A-gliadin 57-73 QE65, to the total T cell response to gliadin in coeliac disease is a critical issue. Pepsin-"trypsin and chymotrypsin-digested gliadin have been traditionally used as antigen for development of T cell lines and clones in coeliac disease. However, it is possible mat these proteases may cleave through certain peptide epitopes. Indeed, chymotrypsin digestion of recombinant a9-gliadin generates the peptide QLQPFPQPELPY (SEQ ID NO: 13), that is a truncation of fte optimal epitope

sequence QLQPFPQPELPYPQPQS (SEQ ID N0:2) (see above). Transglutaminase-treatment substantially increases the potency of chymotrypsin-digested gliadin in proliferation assays of gliadin-specific T cell clones and lines. Hence, transglutaminase-treated chymotrypsin-digested gliadin (tTG gliadin) may not be an ideal antigen, but responses against this mixture may approximate the "total" number of peripheral blood lymphocyte specific for gliadin. Comparison of responses against A-gliadin 57-73 QE65 and tTG gliadin in the ELISpot assay gives an indication of the contribution of this dominant epitope to the overall immune response to gliadtn in coeliac'disease, and also be a measure of epitope spreading.
PBMC collected on day 6 or 7 after commencing gluten challenge in 4 coeliac subjects were assessed in dose response studies using chymotrypsin-digested gliadin +/- tTG treatment and compared with ELISpot responses to an optimal concentration of A-gliadin 57-73 QE65 (25mcg/ml). TTG treatment of guadin enhanced PBMC responses in the ELISpot approximately 10-fold (tTG was comparable to blank when assessed alone) (see Figure 13a-c). In the four coeliac subjects studied, A-gliadin 57-73 QE65 (25 meg/ml) elicited responses between 14 and 115% those of tTG gliadin (500 meg/ml), and the greater the response to A-gliadin 57-73 QE65 me greater proportion it represented of the tTG gliadin response.
Relatively limited data suggest that A-gUadin 57-73 QE65 responses are comparable to tTG gliadin in some subjects. Epitope spreading associated with more evolved anti-gKadin T cell responses may account for the smaller contribution of A- • giia/tin 57-73 QE65 to "total" gliadin responses in peripheral blood in some individuals. Epitope spreading may be maintained in individuals with less strictly gluten free diets.
Example 8
Definition of gliadin peptides coeliac disease: polymorphisms ofA-gliadin 57-73
Overlapping 15mer peptides spanning the complete sequence of A-gliadin were assessed in order to identify the immunodominant sequence in coeliac disease. A-gliadin was the first fully sequenced alpha gliadin protein and gene, but is one of approximately 30-50 related alpha gliadin proteins in wheat Twenty five distinct

lpha-gliadin genes have been identified by searching protein data bases, Swiss-Prot and TREMBL describing a further 8 alpha-gliadins. Contained within these 25 alpha-gliadins, there are 16 distinct polymorphisms of the sequence corresponding to A-gliadin 57-73 (see Table 7).
Synthetic peptides corresponding to these 16 polymorphisms, in an unmodified form, after treatment with transglutaminase in vitro, as well as with •glutamate substituted at position 10 (equivalent to QE65 in A-gliadin 57-73) were assessed using PBMC from coeliac subjects, normally following a gluten free diet, day 6 or 7 after gluten challenge in interferon gamma ELISpot assays. Glutamate-substituted peptides were compared at three concentrations (2.5,25 and 250 mcg/ml), unmodified peptide and transglutaminase-treatedpeptides were assessed at 25 mcg/ml only. Bioactivity was expressed as % of response associated with A-gliadin 57-73 QB65 25 mcg/ml in individual subjects (n=4). (See Fig 14).
Bioactivity of "wild-type" peptides was substantially increased (>5-fold) by treatment with transglutaTninase. Transglutaminase treatment of wild-type peptides resulted in bioactivity similar to that of the same peptides substituted with ghltamate at position 10. Bioactivities of five glutamate-substituted peptides (B, C, K, L, M), were >70% that of A-gliadin 57-73 QE65 (A), but none was significantly more bioactive than. A-gliadin 57-73 QE65. PBMC responses to glutamate-substituted peptides at concentrations of 2.5 and 250 mcg/ml were comparable to those at 25 mcg/ml Six glutamate-substituted gliadin peptides (H, 1, J, N, O, P) were At least six gliadin-derived peptides are equivalent in potency to A-gliadin 57-73 QE65 after modification by transglutaminase.' Relatively non-bioactive polymorphisms of A-gliadin 57-73 also exist These data indicate that transglutaminase modification of peptides from several gliadins of Triticum aestivum, T. itartu and T. spelta may be capable of geneiating the immunodominant T cell epitope in coeliac disease.
Genetic modification of wheat to generate non-coeliac-toxic wheat may likely require removal or modification of multiple gliadin genes. Generation of wheat containing gliadins or other proteins or peptides incorporating sequences defining altered peptide ligand antagonists of A-gliadin 57-73 is an alternative strategy to

generate genetically modified wheat that is therapeutic rather than "non-toxic" in coeliac disease.
Example 9
Definition of Core Epitope Sequence:
Comparison of peptides corresponding to truncations of A-gliadin 56-75 from the N- and C-terminal indicated that the core sequence of the T cell epitope is .PELPY (A-gliadin 64-68). Attempts to define non-agonists and antagonists will focus on variants of A-gliadin that are substituted at residues that substantially contribute to its bioactivity.
Peptides corresponding to A-gliadin 57-73 QE65 with alanine (Figure 15) or lysine (Figure 16) substituted for residues 57 to 73 were compared in the IFN gamma ELISPOT using peripheral blood mononuclear cells (PBMC) from coeliac volunteers 6 days after commencing a 3-day gluten challenge (n=8). (BL is blank, E is A-gliadin 57-73 QE65: QLQPFPQPELPYPQPQS (SEQ ID NO2)).
It was found that residues corresponding to A-gliadin 60-70 QE65 (PFPQPELPYPQ (SEQ ID NO: 14)) contribute substantially to me bioactivity in A-gliadin 57-73 QE65. Variants of A-gliadin 57-73 QE65 substituted at positions 60-70 are assessed in a 2-step procedure. Initially, A-gliadm 57-73 QE65 substituted at positions 60-70 using 10 different amino acids with contrasting properties are assessed. A second group of A-gliadin 57-73 QE65 variants (substituted with, all other naturally occurring amino acids except cysteine at positions mat prove are sensitive to modification) are assessed in a second round.
Example 10
Agonist activity of substituted variants of A-gliadin 57-73 QE65
A-gliadin 60-70 QE65 is the core sequence of the dominant T cell epitope in A-gliadin. Antagonist and non-agonist peptide variants of this epitope are most likely generated by modification of this core sequence. Initially, A-gliadin 57-73 QE65 substituted at positions 60-70 using 10 different amino acids with contrasting properties will be assessed in the IFNgamma ELISPOT using PBMC from coeliac subjects 6 days after starting 3 day gluten challenge. A second group of A-gliadin

57-73 QE65 variants (substituted with all other naturally occurring amino acids except cysteine) at positions 61-70 were also assessed. Both groups of peptides (all at 50 meg/ml, in duplicate) were assessed using PBMC from 8 subjects and compared to the unmodified peptide (20 replicates per assay). Previous studies indicate that the optimal concentration for A-gliadin 57-73 QE65'in this assay is between 10 and 100 meg/ml.
Results are expressed as mean response in spot forming cells (95% confidence interval) as % A-G 57-73 QE65 mean response in each individual. Unpaired t-tests will be used to compare ELISPOT responses of modified peptides with A-G 57-73 QE65. Super-agonists were defined as having a greater response than A-G 57-73 QE65 at a. level of significance of pO.Ol; partial agonists as having a response less than A-G 57-73 QE65 at a level of significance of pO.Ol, and non-agonists as being'not significantly different (p>0.01) from blank (buffer without peptide). Peptides with agonist activity 30% or less that of A-gliadin 57-73 QE65 were considered "suitable" partial or non-agonists to assess for antagonistic activity (see Table 8 and Figures 17-27).
The EFNgamma ELISPOT response of PBMC to A-gliadin 57-73 QE65 is highly specific at & molecular level Proline-at position 64 (P64), glutamate at 65 (E65) .and leucine at position 66 (L66), and to a lesser extent Q63, P67, Y68 and P69 are particularly sensitive to modification. The substitutions Y61 and Y70 both generate super-agonists with 30% greater bioactivity than the parent peptide, probably by enhancing binding to HLA-DQ2 since the motif for this HLA molecule indicates a preference for bulky hydrophobic resides at positions 1 and 9. Eighteen non-agonist peptides were identified. Bioactivities of the variants (50 meg/ml): ?65, K64, K65 and Y65 (bioactivity 7-8%) were comparable to blank (7%). In total, 57 mutated variants of A-gliadin 57-73 QE65 were 30% or less bioactive than A-gliadin 57-73 QE65.
The molecular specificity of the peripheral blood lymphocyte (PBL) T cell response to the dominant epitope, A-gliadin 57-73 QE65, is consistently reproducible amongst HLA-DQ2+ coeliac subjects, and is highly specific to a restricted number of amino acids in the core 7 amino acids. Certain smgle-amino acid variants of A-gliadin 57-73 QE65 are consistently non-agonists in all HLA-DQ2+ coeliac subjects.

Example 11
Antagonist activity of substituted variants
The homogeneity of the PBL T cell response to A-gliadin 57-73 QE65 in HLA-DQ2+ cpeliac disease suggests that altered peptide ligands (APL) capable of. antagonism in PBMC ex vivo may exist, even though the PBL T cell response is ' likely to be poly- or oligo-clonaL APL antagonists are generally weak agonists. Fifty-seven single amino acid-substituted variants of A-gliadin 57-73 QE65 witti agonist activity 30% or less have'been identified and are suitable candidates as APL antagonists. In addition, certain weakly bioactive naturally occurring polymorphisms of A-gliadin 57-73 QE65 have also been identified (see below) and may be "naturally occurring" APL antagonists. It has also been suggested that competition for binding MHC may also antagonise antigen-specific T cell immune. Hence, non-gHarlin peptides mat do not induce IFNgamma responses in coenac PBMC after gluten challenge but are known to bind to HLA-DQ2 may be capable of reducing T cell responses elicited by A-gliadin 57-73 QE65. Two peptides mat bind avidly to HLA-DQ2 are HLA class I a 46-60 (HLA la) (PRAPWIEQEGPEYW (SEQ ID NO:15)) and tibyroidperoxidase (tp) 632-645Y (IDVWLGGLLAENFLPY.(SEQ ID NO:16)).
Simultaneous addition of peptide (50}ig/ml) or buffer and A-gliadin 57-73 QE65 (lOug/ml) in IFNgamma ELISPOT using PBMC from coeliac volunteers 6 days after commencing 3 day gluten challenge (n=5). Results were expressed as response with peptide plus A-G 57-73 QE65 (mean of duplicates) as % response with buffer plus A-G 57-73 QE65 (mean of 20 replicates). (See Table 9).
Four single amino acid-substituted variants of A-gliadin 57-73 QE65 reduce flie interferon gamma PBMC ELISPOT response to A-gliadia 57-73 QE65 (p
QE90 (PQPQPFPPELPYPQPQS (SEQ ED NO: 17)) reduces responses to A-gliadin 57-73 QE65 by 19% (p Interferon gamma responses of PBMC to A-gliadin 57-73 QE65 inELISPOT assays are reduced by co-administration of certain single-amino-acid A-gliadin 57-73 QE65 variants,-a polymorphism of A-gliadin 57-73 QE65, and an unrelated peptide known to bind'HLA-DQ2 in five-fold excess. These finding suggest that altered peptide ligand antagonists of A-gliadin 57-73 QE65 exist. Not only putative APL antagonists but also certain peptides that bind HLA-DQ2 effectively reduce PBL T cell responses to A-gliadin 57-73"QE65.
These findings support two strategies to interrupt the T cell response to the dominant A-gliadin epitope in HLA-DQ2+ coeliac disease.
1. Optimisation of APL antagonists by substituting amino acids at more than
one position (64-67) for use as "traditional" peptide pharmaceuticals or for
specific genetic modification of gliadin genes in wheat
2. Use of high affinity HLA-DQ2 binding peptides to competitively inhibit
presentation of A-gliadin 57-73 QE65 in association with HLA-DQ2.
These two approaches may be mutually compatible. Super-agonists were generated by replacing F61 and Q70 with tyrosine residues. It is likely these super-agonists resulted from improved binding to HLA-DQ2 rather than enhanced contact with the T cell receptor. By combining these modifications with, other substitutions that generate modestly effective APL antagonists might substantially enhance the inhibitory effect of substituted A-gliadin 57-73 QE65 variants.
Example 12
Development ofinterferon gamma ELISpot using PBMC and A-gliadin 57-73 QE65 and P04724 84-100 QE92 as a diagnostic for coeliac disease: Definition of immune-responsiveness in newly diagnosed coeliac disease
Induction of responsiveness to the dominant A-gliadin T cell epitope in
PBMC measured in the interferon gamma ELISpot follows gluten challenge in almost all DQ2+ coeliac subjects following a long term strict gluten free diet (GFD) but not hi healthy DQ2+ subjects after 4 weeks following a strict GFD. A-gliadin

57-73 QE65 responses are not measurable in PBMC of ooeliac subjects before gluten challenge and pilot data have suggested these responses could not be measured in PBMC of untreated coeliacs. These data suggest that in coeliac disease immune-responsiveness to A-gliadin 57-73 QE65 is restored following antigen exclusion (GFD). If a diagnostic test is to be developed using the ELISpot assay and PBMC, it is desirable to define the duration of GFD required before gluten challenge is capable of inducing responses to A-gliadin 57-73 QE65 and other immunoreactive gliadin peptides in blood.
Newly diagnosed DQ2+'coeliac subjects were recruited from the gastroenterology outpatient service. PBMC were prepared and tested in interferon gamma ELISpot assays before subjects commenced GFD, and at one or two weeks after commencing GFD. In addition, gluten'challenge-(3 days consuming 4 slices standard white bread, 200g/day) was performed at one or two weeks after starting GFD. PBMC were prepared and assayed on day six are after commencing gluten challenge. A-gliadin 57-73 QE65 (A), P04724 84-100 QE92 (B) (alone and combined) and A-gliadin 57-73 QP65 (P65) (non-bioactive variant, see above) (all 25 meg/ml) were assessed.
All but one newly diagnosed coeliac patient was DQ24- (one was DQ8+) (n=l 1). PBMC from newly diagnosed coeliacs that were untreated, or after 1 or 2 weeks following GFD did not show responses to A-gliadin 57-73 QE65 and P04724 84-100 QE92 (alone or combined) mat were not significantly different from blank or A-gliadin 57-73 QP65 (n=9) (see Figure 28). Gluten challenge in coeliacs who had followed GFD for only one week did not substantially enhance responses to A-gliadin 57-73 QE65 or P04724 84-10.0 QE92 (alone or combined). But gluten challenge 2 weeks after commencing GFD did induce responses to A-gJiadin 57-73 QE65 and P04724 84-100 QE92 (alone or combined) that were significantly greater than the non-bioactive variant A-gliadin 57-73 QP65 and blank. Although these responses after gluten challenge at 2 weeks were substantial they appear to be less man in subjects >2 months after commencing GFD. Responses to A-gliadin 57-73 QE65 alone were equivalent or greater than responses to P04724 84-100 QE92 alone or when mixed with A-gliadin 57-73 QE65. None of the subjects experienced troubling symptoms with gluten challenge.

Immune responsiveness (as measured in PBMC after gluten challenge) to A-gliadin is partially restored 2 weeks after commencing GFD, implying that "immune unresponsrveness" to this dominant T cell epitope prevails in untreated coeliac disease and for at least one week after starting GFD. The optimal timing of a diagnostic test for coeliac disease using gluten challenge and measurement of responses to A-gliadin 57-73 QE65 in fhe ELISpot assay is at least 2 weeks after commencing a GFD.
Interferon gamma-secreting T cells specific to A-gliadin 57-73 QE65 cannot be measured in the peripheral blood in untreated coeliacs, and can only be induced by gluten challenge after at least 2 weeks GFD (antigen exclusion). Therefore, timing of a diagnostic test using this methodology is crucial and further studies are needed for its. optimization. These finding are consistent with functional anergy of T cells specific for the dominant epitope, A-gliadin 57-73 QE65, reversed by antigen - exclusion (GFD). This phenomenon has not been previously demonstrated in a human disease, and supports the possibility that T cell anergy may be inducible with peptide therapy in coeliac disease.
Example 13
Comprehensive Mapping of Wheat Gljadin T Cell Epitopes
Antigen challenge induces antigen-specific T cells in peripheral blood. In coeliac disease, gluten is the antigen that maintains this immune-mediated disease. • Gluten challenge in coeliac disease being treated with a gluten free diet leads to the appearance of gluten-specific T cells in peripheral blood, so enabling determination of me molecular specificity of gluten T cell epftopes. As described above, we have identified a single dominant T cell epitope in a model gluten protein, A-gliadin (57-73 deamidated at Q65). In this Example, gluten challenge in coeliac patients was used to test all potential 12 amino acid sequences in every known wheat gh'adin protein derived from 111 entries in Genbank. hi total, 652 20mer peptides were tested in HLA-DQ2 and HLA-DQ8 associated coeuac disease. Seven of the 9 coeliac subjects with the classical HLA-DQ2 complex (HLA-DQA1*05, HLA-DQB1*02) present in over 90% of coeh'acs had an inducible A-gliadin 57-73 QE65-and gh'adiD-specific T ceH response in peripheral blood. A-gliadin 57-73 was the

only significant a-gliadin T cell epitope, as well as the most potent gliadin T cell epitope, in HLA-DQ2-associated coeliac disease. In addition, there were as many as 5 families of structurally related peptides that were between 10 and 70% as potent as A-gliadin 57-73 in the interferon-y ELISpot assay. These new T cell epitopes were derived from y- and o-gliadins and included common sequences that were structurally very similar, but not identical to the core sequence of A-gliadin 57-73 (core sequence: FPQPQLPYP (SEQ ID NO:18)),'for example: FPQPQQPFP (SEQ ID NO: 19) and PQQPQQPFP (SEQ ID NO:20). Although no homologues of A-gliadin 57-73 have been found irrrye or barley, the other two cereals toxic in coeliac disease, the newly defined T cell epitopes in 7- and a>-gliadins have exact matches in rye and barley storage proteins (secauns and hordeins, respectively).
Coeliac disease not associated with HLA-DQ2 is almost always associated with HLA-DQ8. None of the seven ELA-DQ8+ coeliac subjects had inducible A-gliadin 57-73-specific T cell responses following gluten challenge, unless they also possessed the complete HLA-DQ2 complex. Two of 4 HLA-DQ8+ coeliac subjects who did not possess the complete HLA-DQ2 complex, had induoible gliadin peptide-specific T cell responses following gluten challenge. In one HLA-DQ8 subject, a novel dominant T cell epitope was identified with the core sequence LQPQNPSQQQPQ (SEQ ID N0:21). The transglutaminase-deamidated version of this peptide was more potent than the non-deamidated peptide. Previous studies suggest mat the teansglutarninase-deamidated peptide would have the sequence LQPENPSQEQPE (SEQ ID NO:22); but further studies are required to confirm this sequence. Amongst the healthy HLA-DQ2 (10) and HLA-DQ8 (1) subjects who followed a gluten free diet for a month, gliadin peptide-specific T cell responses were uncommon, seldom changed with gluten'challenge, and were never potent T cell epitopes revealed with gluten challenge in coeliac subjects. In conclusion, there are unlikely to be more than six important T cell epitopes in HLA-DQ2-associated coeliac disease, of which A-gliadin 57-73 is the most potent HLA-DQ2- and HLA-DQ8-associated coeliac disease do not share the same T cell specificity.
We have shown that short-term gluten challenge of individuals with coeliac disease following a gluten free diet induces gliadin-specific T cells in peripheral blood. The frequency of these T cells is maximal in peripheral blood on day 6 and

then rapidly wanes over the following week. Peripheral blood gliadin-specific T cells express the integrin a4p7 that is associated with homing to the gut lamina propria. We exploited this human antigen-challenge design to map T cell epitopes relevant to coeliac disease in the archetypal gluten a-gliadin protein, A-gliadin. Using 15mer peptides overlapping by 10 amino acids with and without deamidation by transglutaminase (tTG), we demonstrated that T cells induced in peripheral blood initially target only one A-gliadin peptide, residues 57-73 in which glutamine at position 65 is deamidated. The epitope is HLA-DQ2-restricted, consistent with the intimate association of coeliac disease with HLA.-DQ2.
Coeliac disease is reactivated by wheat, rye and barley exposure. The aJ$-gliadin fraction of wheat gluten is consistently toxic in coeliac disease, and most studies have focused on these proteins. The gene cluster coding for a/p"-gliadins is located on wheat chromosome 6C. There are no homologues of o/p-gliadins in rye or barley. However, all three of the wheat gliadin subtypes (o/P.y, and to) are toxic in coeliac disease. The 7- and ff>-gliadin genes are located on chromosome LA in wheat, and are homologous to the secalins and hordeins in rye and barley.
There are now genes identified for 61 a-gliadins in wheat (Triticnm aestivum). The a-gliadin sequences are closely homologous, but the dominant epitope in A-gliadin derives from the most polymorphic region in the a-gliadin sequence. Anderson et al (1997) have estimated mat there are a total of about 150 distinct a-gliadin genes in T. aestivum, but many are psuedogenes. Hence^ it is unlikely that T-cell epitopes relevant to coeliac disease are not included within known a-gliadin sequences.
Our work has identified a group of deamidated a-gliadin peptides almost identical to A-gliadin 57-73 as potent T cell epitopes specific to coeliac disease. Over 90% of coeliac patients are HLA-DQ2+, and so far, we have only assessed HLA-DQ2+ coeliac subjects after gluten challenge. However, coeliac patients who do not express HLA-DQ2 nearly all carry HLA-DQ8. Hence, it is critical to know whether A-gliadin 57-73 and its homologues in other wheat, rye and barley gluten proteins are the only T-cell epitopes recognized by T cells induced by gluten challenge in both HLA-DQ2+ and HLA-DQ8+ coeliac disease. If lids were the case, design of peptide therapeutics for coeliac disease might only require one peptide.

Homologues ofA-gliadin 57-73 as T-cell epitopes
Initial searches of SwissProt and Trembl gene databases for cereal genes coding for the core sequence of A-gliadin 57-73 (PQLPY ) only revealed a-gliadins. However, our fine-mapping studies of the A-gliadin 57-73 QE65 epitopeTevealed a limited number of permissive point substitutions in the core region (PQLP) (note Q65 is actually deamidated in the epitope). Hence, we extended our search to genes in SwissProt or Trembl databases encoding for peptides with the sequence XXXXXXXPQ[ILMPJ[PST]XXXXXX (SEQ ID N0:23). Homologues were identified amongst y-gliadins, glutenins, hordeins and secalins (see Table 12). A further homologue was identified in co-gliadin by visual search of the three o>-gliadin entries in GeribariL
These homologues of A-gliadin 57-73 were assessed after deamidation by tTG (or synthesis of the glutamate(QE)-substituted variant in four close homologues) using the IFNy ELISpot assay with peripheral blood mononuclear cells after gluten challenge in coeliac subjects. The m-gliadin sequence (AAG17702 141-157) was the only bioactive peptide, approximately half as potent as A-gliadin 57-73 (see Table 12, and Figure 29). Hence, searches for homologues of the dominant A-gliadin epitope failed to account for the toxicity of y-gliadin, secalins, and nordeins. Methods Design of a set of peptides spanning all possible -wheat gliadin T-cell epitopes
In order to identify all possible T cell epitopes coded by the known wheat (Triticum aestivum) gliadin genes or gene fragments (61 o/p\ 47 y-, and 3 oo-gliadin entries in Genbank), gene-derived protein sequences were aligned using the CustaTW software (MegAlign.) and arranged into phylogenetic groupings (see Table 22). Many entries represented truncations of longer sequences, and many gene segments were identical except for the length of polyglutamine repeats or rare substitutions. Hence, it was possible to rationalize all potential unique 12 amino acid sequences encoded by known wheat genes to be included in a set of 652 ZOmer peptides. (Signal peptide sequences were not included). Peptide sequences are listed in Table 23. Comprehensive epitope mapping

Healthy controls (HLA-DQ2+ n=10, and HLA-DQ8+ n=l) who had followed a gluten free diet for 4 weeks, and coeliac subjects (six HLA-DQ2, four complex heterozygotes HLA-DQ2/8, and three HLA-DQ8/X) (see Table 13) following long-term gluten free diet were studied before and on day 6 and 7 after 3-day gluten challenge (four 50g slices of standard white bread- Sainsbury's sandwich bread, each day). Peripheral blood (a total of 300ml over seven days) was collected and peripheral blood mononuclear cells (PBMC) were separated by Lymphoprep density gradient. PBMC were incubated with pools of 6 or 8 20mer peptides, or single peptides with or without deatnidation by tTG hi overnight interferon gamma (ENy) ELISpot assays.
Peptides were synthesized hi batches of 96 as Pepsets (Mimotopes Inc., Melbourne Australia). Approximately 0.6 micromole of each of 652 20mers-was provided. Two marker 20mer peptides were included in each set of 96 (VLQQENIAHGSSQVLQESTY -peptide 161 (SEQ ID NO24), and -IKDFHVYFRESRDALWK.GPG (SEQ ID NO'25)) and were characterized by reverse phase-HPLC and amino acid sequence analyBis. Average purities of these marker peptides were 50% and 19%, respectively. Peptides were initially dissolved hi acetonitrile (10%) and'Hepes lOOmM to lOmg/mL
The fi™l concentration of individual peptides in pools (or alone) incubated with PBMC for the IFNy ELISpot assays was 20 ug/ml. Five-times concentrated solutions of peptides and pools in PBS with calcium chloride ImM were aliquotted and stored in 96-well plates according to the template later used in ELISpot assays. Deamidated peptides and pools of peptides were prepared by incubation with guinea pig tissue tTG (Sigma T5398) in the ratio 100:32-ug/ml for two hours at 37°C. Peptides solutions were stored at—20°C and freshly thawed prior to use.
GKadin (Sigma G3375) (100 mg/ml) hi endotoxin-free water and 2M urea was boiled for 10 minutes, cooled to room temperature and incubated with filter (0.2 um)-sterilised pepsin (Sigma P6887) (2 mg/ml) in HC10.02M or chymotrypsin (C3142) (4mg/ml) hi ammonium bicarbonate (0.2M). After incubation for 4 hours, pepsin-digested gliaflin was neutralized with sodium hydroxide, and men both pepsin- and chymotrypsin-digested gliadin were boiled for 15 minutes. Identical incubations with protease in which gliadin was omitted were also performed.

Samples were centrifuged at 15 OOOg, then protein concentrations were estimated in supernatants by the BCA method (Pierce, USA.). Before final use in IFNy ELISpot assays, aliquots of gliadin-protease were incubated with tTG in the ratio 2500:64 ug/ml.
IFNy ELISpot assays (Mabtech, Sweden) were performed in 96-well plates (MAIP S-45, Millipore) in which each well contained 25ul of peptide solution and lOOu-1 of PBMC (2-8xl05/weU) in RPMI containing 10% heat inactivated human AB serum. Deamidated peptide pools were assessed in one 96-well ELISpot plate, and peptides pools without deamidatiDn in a second plate (with an identical layout) on both day 0 and day 6. All wells in the plate containing deamidated peptides included tTG (64 ug/ml). In each ELISpot plate there were 83 wells with'peptide pools (one unique pool in each well), and a series of wells for "control" peptides (peptides all >90% purity, characterized by MS and HPLC, Research Genetics): P04722 77-93 (QLQPFPQPQLPYPQPQP (SEQ ID NO:26)), P04722 77-93 QE85 (in-dupEcate) (QLQPFPQPELPYPQPQP (SEQ ID NO:27)), P02863 77-93 (QLQPFPQPQLPYSQPQP (SEQ ID N0:28)), P02863 77-93 QE85 (QLQPFPQPELPYSQPQP (SEQ ID NO:29)), and chymotrypsiiL-digested gliadin (500 )ig/ml), pepsin-digested gliadin (500 ug/ml), chymotrypsin (20 pg/ml) alone, pepsin (10 ug/ml) alone, and blank (PBS+/-tTG) (in triplicate).
After development and drying, IFNy ELISpot plates were assessed using the MAIP automated ELISpot.plate counter. In HLA-DQ2 healthy and coeliac subjects, induction of spot forming cells (sfc) by peptide pools in the IFNy ELISpot assay was tested using a one-tailed Wilcoxon Matched-Pairs Sigried-Ranks test (using SPSS software) applied to spot forming cells (sfc) per million PBMC minus "blank on day 6 versus day 0 ("net response"). Significant induction of an IFNy response to peptide pools in PBMC by in vivo gluten challenge was defined as a median "net response" of at least 10 sfc/miUion PBMC and p For IFNy ELISpot assays of individual peptides, bioacrrviry was expressed as a percent of response to P04722 77-93 QE85 assessed in the same ELISpot plate. Median response to blank (PBS alone) was 0.2 (range 0-5) sfc per well, and the

positive control (P04722 77-93 QE85) 76.5 (range: 25-282) sfc per well using a median of 0.36 million (range: 0.3-0.72) PBMC. Hence, median response to blank expressed as a percentage of P04722 77-93 QE65 was 0.2% (range: 0-6.7). Individual peptides with mean bioactivity greater thanlO% that of P04722 QE85 5 were analyzed for common structural motifs. Results Healthy HLA-DQ2 subjects
None of the healthy HLA-DQ2+ subjects following a gluten free diet for a month had IFNy EUSpot responses to homologues of A-gliadin 57-73'before or after
gluten challenge. However, in 9/10 healthy subjects, gluten challenge was associated with a significant increase in IFNy responses to both peptic- and chvmotryptic-digests of gliadin, from a median of 0-4 sfc/million on day 0 to a median of 16-29 sfc/nnHion (see Table 14). Gliadin responses in healthy subjects were unaffected by deamidation (see Table 15). Amongst healthy subjects, there was no consistent
induction of IFNy responses to specific gliadin peptide pools with gluten challenge (see Figure 30, and Table 16). IFNy ELISpot responses were occasionally found, but these were weak, and not altered by deamidation. Many of the strongest responses to pools were also present on day 0 (see Table 17, subjects H2, H8 and H9). Four healthy subjects did show definite responses to pool 50,and the two with strongest
responses on day 6 also had responses on day 0. ID both subjects,-the post-challenge' responses to pool 50 responses were due to peptide 390 (QQTYPQRPQQPFPQTQQPQQ (SEQ ID NO:30)). HLA-DQ2 coeliac subjects
Following gluten challenge in HLA-DQ2+ coeliac subjects, median IFNy
ELISpot responses to P04722 77-93 E85 rose from a median of 0 to 133 sic/million (see Table 4). One of the six coeliac subjects (C06) did not respond to P04722 77-93 QE85 (2 sfc/million.) and had only weak responses to gliadin peptide pools (maximum: Pool 50+tTG 27 sfc/million). Consistent with earlier work, bioactivity of wild-type P04722 increased 6.5 times with deamidation by tTG (see Table 15).
Interferon-garnrria responses to gliadin-digests were present at baseline, but were substantially increased by gluten challenge from a median of 20 up to 92 sfc/million for chymotryptic-gtiadin, and from 44 up to 176 sfc/million for peptide-gliadin.

DeamidatioB of gliadin increased bioactivity by a median of 3.2 times for chymotryptic-gliadin and 1.9 times for peptic-gliadin (see Table 15). (Note that the acidity required for, digestion by pepsin is likely to result in partial deamidation of gliadin.)
In contrast to healthy subjects, gluten challenge induced IFNy ELISpot
responses to 22 of the 83 tTG-treated pools including peptides from a-, y~ and o>-gliadins (see Figure 31, and Table 17). Bioactivity of pools was highly consistent between subjects (see Table 18). IFNy ELISpot responses elicited by peptide pools were almost always increased bydeamidation (see Table 17). But enhancement of
bioactivity of pools by deamidation was not as marked as for P04722 77-73 Q85, even for pools including hoinologues of A-gliadin 57-73.. This suggests mat Pepset peptides were partially deamidated during synthesis or in preparation, for example the Pepset peptides are delivered as salts of trifluoracetic acid (TFA) after lyophilisation from a TFA solution.
' One hundred and seventy individual tTG-deamidated peptides from 21 of the
most bioactive pools were separately assessed. Seventy-two deamidated peptides were greater than 10% as bioactive as P04722 77-93 QE85 at an equivalent concentration (20 ug/ml) (see Table 19). The five most potent peptides (85-94% bioactivity of P04722 QE85) were previously identified a-gliadin homologues A-
gliadin 57-73. Fifty of the biqactive peptides were not homologues of A-gliadin 57-73, but could be divided into six -families of structurally related sequences (see Jable 20). The most bioactive sequence of each of the peptide femih'es were: POOPOOPOOPFPOPOOPFPW (SEQ ID NO:31) (peptide 626, median 72% bioactivity of P04722 QE85), OOPOOPFPOPOOPOLPFPOO (SEQ E> NO32)
(343,34%), OAFPOPOOTFPHOPOOOFPO (SEQ ID N0:33) (355,27%), TOOPOOPFPOOPOOPFPOTO (SEQ ED NO:34) (396,23%), PIOPOOPFPOOPQQPQQPFP (SEQ ID NO:35*> (625,22%). POOSFSYOQOPFPOOPYPOO (SEQ ID NO:36) (618,18%) (core sequences are underlined). All of these sequences include glutamine residues predicted to be
susceptible to deamidation by transglutaminase (e.g. QXP, QXPF (SEQ ID N0:37), QXX[FY] (SEQ ID NO:38)) (see Vader et al 2002). Some bioactive peptides contain two core sequences from different families.

Consistent with the possibility that different T-cell populations respond to peptides with distinct core sequences, bioactivity of peptides from different families appear to be additive. For example, median bioactivity of tTG-treated Pool 81 was 141% of P04722 QE85, while bioactivity of individual peptides was in rank order. 5 Peptide 631 (homologue of A-gliadro 57-73) 61%, 636 (homologue of 626) 51%, and 635 19%, 629 16%, and 634 13% (all homplogues of 396).
Although likely to be an oversimplification, the contribution of each "peptide family" to the summed IFNy ELISpot response to gliadin peptides was compared in the HLA-DQ2-1- coeliac subjects (see Figure 32). Accordingly, Ihe contribution of
P04722 77-73 E85 to the summed response to gliadin peptides is between 1/5 and
2/3. ' •
Using the peptide homology search programme, WWW PepPepSeaich, which can be accessed through the world wide web of the internet at, for example, "chrg.in£e1hz.ch/subsection3_l_5.html.", and by direct comparison with Geribank
sequences forrye secalins, exact matches werefound for the core sequences
QQPFPQPQQPFP (SEQ ID NO:39) in barley hordeins (HOR8) and rye secalins (A23277, CAA26449, AAG35598), QQPFPQQPQQPFP (SEQ ID N0:40) in barley hordeins (HOG1 and HOR8), and for PIQPQQPFPQQP (SEQ ID NO:41) also in barley hordeins (HOR8).
HLA~DQ8-associated coeliac disease
Seven HLA-DQ8+ coeliac subjects were studied before and after gluten challenge. Five of these HLA-DQ8+ (HLA-DQAO*0301-3, HLA-DQBO*0302) subjects also carried one or both of the coeliac disease-associated HLA-DQ2
complex pQAO*OS, DQBO*02). Two of the three subjects with both coeliac-associated HLA-DQ complexes had potent responses to gliadin peptide pools (and individual peptides including P04722 77-93 E85) that were qualitatively and quantitatively identical to HLA-DQ2 coeliac subjects (see Figures 33 and 34, and Table 18). Deamidated peptide pool 74 was biqactive in both HLA-DQ2/8 subjects,
"but only in one of the 6 HLA-DQ2/X subjects. Pretreatment of pool 74 with tTG enhances bioactivity between 3.8 and 22-times, and bioactivity of tTG-treated pool 74 in the three responders is equivalent to between 78% and 350% the bioactivity of

P04722 77-93 E85. Currently, it is not known which peptides are bioactive in Pool 74 in subject C02, C07, and COS.
Two of the four HLA-DQ8 coeliac subjects that lacked both or one of the HLA-DQ2 alleles associated with coeliac disease showed very weak IFNy ELlSpot 5 responses to gliadin peptide pools, but the other two did respond to both protease-digested gliadin and specific peptide pools. Subject C12 (HLA-DQ7/8) responded vigorously to deamidated Pools 1-3 (see Figure 3 5). Assessment of individual peptides in these pools identified a series of closely related bioactive peptides including the core sequence LQPQNPSQ£)QP£> (SEQ ID NO:42) (see Table 20).
Previous work (by us) has demonstrated that three glutamine residues in mis sequence are susceptible to tTG-mediated deamidation (underlined). Homology searches using WWW PepPepSearch have identified close matches to LQPQNPSQQQPQ (SEQ ID N0:43) only in wheat a-gliadins.
The fourth HLA-DQ8 subject (Cl 1) had inducible IFNy ELISpot responses
to tTOT-treated Pool 33 (see Figure 36). Pools 32 and 33 include polymorphisms of a previously defined HLA-DQ8 restricted gliadin epitope
(QQYPSGQGSFQPSQQNPQ (SEQ ID NO:44)) active after deamidation by tTG (underlined Gin are deamidated and convey bioactivity) (van der Wai et al 1998). Currently, it is not known which peptides are bioactive in Pool 33 in subject Cl 1.
Comprehensive T cell epitope mapping in HLA-DQ2-associated coeliac
disease using in vivo gluten challenge and a set of 652 peptides spanning all known 12 amino acid sequences in wheat gliadin has thus identified at least 72 peptides at 10% as bioactive as the known a-gliadin epitope, A-gliadin 57-73 E65. However, mese bioactive peptides can be reduced to a set of perhaps as few as 5 distinct but
closely related families of peptides. Almost all mese peptides are rich in proline, glutamine, phenylalanine, and/or tyrosine and include the sequence PQ(QL)P(FY)P (SEQ ID N0:45). This sequence facilitates deamidation of Q in position 2 by tTG. By analogy with deamidation of A-gliadin 57-68 (Arentz-Hansen 2000), the enhanced bioactivity of these .peptides generally found with deamidationby tTG may
be due to increased affinity of binding for HT..A-DQ2.
Cross-reactivity amongst T cells in vivo recognizing more man one of these bioactive gliadin peptides is possible. However, if each set of related peptides does

activate a distinct T cell population in vivo, the epitope corresponding to A-gliadin 57-73 E6 5 is the most potent and is generally recognized by at least 40% of the peripheral blood T cells that secrete IFNy in response to gliadin after gluten challenge.
No gliadin-peptide specific responses were found in HLA-DQ2/8 coeliac disease that differed qualitatively from those in HLA-DQ2/X-associated coeliac disease. However, peripheral blood T cells in BLA.-DQ8+ coeliac subjects without both HLA-DQ2 alleles did not recognize A-gliadin. 57-73 E65 homologues. Two different epitopes were dominant in two HLA-DQ8+ coeliacs. The dominant epitope in one of these HLA-DQ8+ individuals has not been identified previously (LQPQNPSQQQPQ (SEQ ID NO:46)).
Given the teaching herein, design of an nnmunotherapy for coeliac disease utilizing all the commonly recognised T cell epitopes is practical and may include fewer than six distinct peptides. Epitopes in wheat y- and m-gliadrns are also present in barley hordeins and rye secalins. .
Example 14
Several ELJSpot assays were performed as previously described and yielded the following results and/or conclusions: Examination of multiple a-gliadin polymorphisms with PQLP7
Potent agonists of A-gliadin 57-73QE (G01) include
QLQPFPQPELPYPQPQS (GDI), PQL-Y - P (G10), andPQPQPFL-- (G12). Less potent include - L - P (G04), -R - p (G05), and - S - P (G06). Less potent yet
include - L -- S - P (GOT), - S - S - P (GOB), „ - S-S - P (G09), andPQPQPFP - (G13). Dashes indicate identity with the GO 1 sequence in the particular position.
Gluten challenge induces A-gUadin 57-73 QE65 T cells only after two weeks of gluten-free diet in newly diagnosed coeliac disease
Additional analyses indicated th'atlTG-deamidated gliadin responses change after two weeks of gluten-free diet in newly diagnosed coeliac disease. Other

analyses indicated that deamidated gliadin-specific T cells are CCM^cuipV FT A-DQ2 restricted.
Optimal epitope (clones versus gluten challenge)
A "dominant" epitope is defined by yIFN ELISpot after gluten challenge. QLQPFPQPELPYPQPQS (100% ELISpot response). Epitopes defined by intestinal T cell clones: QLQPFPQPELPY (27%), PQPELPYPQPELPY (52%), and QQLPQPEQPQQSFPEQEKPF (9%).
Dominance among individual peptide responses
Dominance depends on wheat or rye. For wheat, dominant peptides include peptide numbers 89,90 and 91 (referring to sequence numbers in Table 23). For rye, dominant peptides include peptide numbers 368,369,370,371, and 372 (referring to sequence numbers in Table 23). Some peptides, including 635 and 636 (referring to sequence numbers in Table 23) showed activity in both rye and wheat
In vivo gluten challenge allows T cell epitope hierarchy to be defined for coeliac disease
The epitope hierarchy is consistent among ELA-DQ2* coeliacs but different for HLA-DQ84 coeliacs. The hierarchy depends on what cereal is consumed. Deamidation generates almost all gliadin epitopes. HT.A-DQ2, DQ8, andDR4 present deamidated peptides. HLA-DQ2/8-associated coeliac disease preferentially present DQ2-associated gliadin epitopes. Gliadin epitopes are sufficiently restricted to justify development of epitope-hased therapeutics. '
Other analyses indicated the following: HLA-DR3-DQ2 (85-95%) and HLA-DR4-DQ8 (5-15%).
Other analyses indicated the following:
HLA-DQ HLA-DQA1 HLA-DQB1 Duodenal Gluten EMAon
allele allele histology tree gluten
(onGFD)

(Table Removed) Another analysis was carried out-to determine the bioactrvity of individual tTG-deamidated peptides in pools 1-3 in subject C12. The results are as follows (sequence numbers refer to the peptides listed in Table 23): Sequence 8 (100%), Sequence 5 (85%), Sequence 6 (82%), Sequence 3 (77%), Sequence 1 (67%), Sequence 2 (59%), Sequence 9 (49%), Sequence 7 (49%), Sequence 10 (33%), Sequence 4 (15%), Sequence 12 (8%), Sequence 11 (0%), Sequence 23 (26%), Sequence 14 (18%), Sequence 15 (18%), Sequence. 17 (18%), Sequence 16 (13%), Sequence 14 (8%), Sequence 22 (5%), Sequence 18 (3%), Sequence 19 (3%), Sequence 20 (0%), Sequence 21 (0%). The predicted deamidated sequence is LQPENPSQEQPE.
Individual ELLSpot responses by PBMC (Spot forming cells determined by EHSpat
Reader)
(Table Removed) To deal with data from 652 peptides in 29 subjects, or to determine when a particular response is a true positive peptide-specific T-cell response, or to determine when a response to a peptide is due to cross-reactivity with another structurally related peptide, expression of a particular peptide response can be as a percentage of a "dominanf' peptide response. Alternately, the expression can be a "relatedness" as correlation' coefficients between peptide responses, or via bioinformatics.
Additional epitqpes
A representative result is as follows:
Combination of peptides with P04722E (all 20mcg/ml) (n=4)
(Table Removed)
Multiple epitopes:
P04724E: QLQPFPQPQLPYPQPQL
6264-tTG: PQQPQQPQQPFPQPQQPFPW
Minimal epitope: QPQQPFPQPQQPFPW
Immunomagnetic depletion of PBMC by beads coated with anti-CD4 and by anti-integrin (J? depleted IFNy ELISpot responses, while immunomagnetic depletion of PBMC by beads coated with anti-CD8 or anti-alphaE integrin. Thus, the PBMC secreting IFNy are CD4+ and cupYh associated with homing to the lamina propria in the gut
Blocked by anti-DQ antibody but not by anti-DR antibody in heterozygotes and homozygotes for HLA-DQ2. This may imply multiple epitopes within one sequence.
Tcell epitopes in coeliac disease
Other investigators have characterized certain intestinal T cell clone epitopes. See, e.g.; Vader et aL, Gastroenterology 2002,1-22:1729-37; Arentz-Hansen et aL, Gastroenterology 2002,123:803-809. These are examples of epitopes whose relevance is at best unclear because of the in vitro techniques used to clone T cells.
Intestinal versus peripheral blood clones
Intestinal: 1) intestinal biopsies, 2) T cell clones raised against peptic-iryptic digest
of gluten, 3) all HLA-DQ2 restricted, 4) clones respond to gliadin deamidated by
transglutaminase.
Peripheral blood: 1) T cell clones raised against gluten are HLA-DR. DQ and DP
restricted. Result Intestinal T cell clones can be exclusively used to map coeliac
disease associated epitopes
GDA_9Wheat 307 aa Definition Alpha/Beto-Gliadin MM1 Precursor (Prolamin) Accession P1'8573 — Genbank (which is incorporated herein by reference in its entirety)
Intestinal Tcell clone epitopes

A definition of intestinal T cell clone epitopes can be found in, for example, Areatz-Hansen et al., J Exp Med. 2000,191:603-12. Also disclosed therein are gliadin epitopes for intestinal T cell clones. Deamidated QLQPFPQPQLPY is an epitope, with a deamidated sequence of QLQPFPQPELPY. There is anHLA-DQ2 restriction.. A homology search shows other bioactive rAlpha-gliadins include PQPQLPY singly or duplicated. A majority of T cell clones respond to either/or DQ2-OI: QLQPFPQPELPY DQ2-oII: PQPELPYPQPELPY
Dominant gliadin T cell epitopes-
All deamidated by transglutarninase.
Peripheral blood day 6 after gluten challenge: A-gliadin 57-73:
QLQPFPQPELPYPQPQS
Intestinal T cell clones: DQ2-oI: QLQPFPQPELPY DQ2-o3I: PQPELPYPQPELPY
Intestinal 1-cell Clone Epitope Mapping
a-Gliadins Al PFPQPQLPY
. A2 ' PQPQLPYPQ .
A3 PYPQPQLPY
Glia-20 PQQPYPQPQPQ
r-Gliadins Gl PQQSFPQQQ
G2 HPQQPAQ
G3 - FPQQPQQPYPQQP
G4 FSQPQQQFPQPQ
G5 LQPQQPFPQQPQQPYPQQPQ
Glu-21 QSEQSQQPFPQQF
Glu-5 Q(TL)PQQPQQF
Glutenin Glt-156 PFSQQQQSPF
Glt-17 PFSQQQQQ

Gluten exposure and induction ofIFNy-secreting A-Gliadin 57-7SQE65-specific T cells in peripheral blood
Untreated coeliac disease, followed by gluten free diet for 1, 2, or 8 weeks, followed by gluten exposure (3 days bread 200g/day), followed by gluten free diet
Result 1: Duration of gluten free diet and IFNy ELISpot responses on day 0 and day 6 of gluten challenge: A-gliadin 57-73 QE65 (results expressed as IFNy specific spots/million PPBMC)
Day 0: none (5), 1 week (1), 2 weeks (2), 8 weeks (1)
Day 6: none (0), 1 week (4), 2 weeks (28), 8 weeks (48)
Result 2: Duration of gluten free diet and IFNy ELISpot responses on day 0 and day 6 of gluten challenge: tTG-gliadin (results expressed as IFNy specific spots/million PPBMC)
Day 0: none (45), 1 week (62), 2 weeks (5), 8 weeks (5)
Day 6: none (0), 1 week (67), 2 weeks (40), 8 weeks (60)
Result 3: Duration of gluten free diet and IFNy ELISpot responses on day 0 and day 6 of gluten challenge: A-gliadin 57-73 P65 (results expressed as IFNy specific spots/miDion PPBMC)
Day 0: none (1), 1 week (2), 2 weeks (1), 8 weeks (1)
Day 6: none'(0), 1 week (0), 2 weeks (0), 8 weeks (0)
Result 4: Duration of gluten free diet and IFNy ELISpot responses on day 0
and day 6 of gluten challenge: PPD (results expressed as IFNy specific spots/million
PPBMC)' •
Day 0: none (90), 1 week (88), 2 weeks (210), 8 weeks (150) .
Day 6: none (0), 1 week (100), 2 weeks (210), 8 weeks (100)
Result 5: Duration.of gluten free diet and'IFNy ELISpot responses on day 0 and day 6 of gluten challenge: tTG (results expressed as IFNy specific spots/million PPBMC)
Day 0: none (5), 1 week (4), 2 weeks (3), 8 weeks (2)
Day 6: none (0), 1 week (4), 2 weeks (1), 8 weeks (2)
Gluten challenge in HLA-DQ2 coeliac disease on long term gluten

Characterization of anti-gliadin T cell response was carried out in peripheral blood on day 6-8 after 3-day gluten challenge.
Result 1: PBMC Day 6 Long-term gluten free diet (preincubation with anti-HLA-DR and -DQ antibody) (expressed as % inhibition)
DR-: tTG-gliadin 100 meg/ml (105), A-gliadin 57-73 QE65 50 meg/ml (90), PPD 5 meg/ml '(30)
DQ-: tTG-gliadin 100 meg/ml (5), A-gliadin 57-73 QE65 50 meg/ml (22), PPD 5 meg/ml (78).
Result 2: PBMC Day 6 Long-term gluten free diet (expressed as % CD8-depleted PBMC response)
B7 depletion: tTG-gliadin n=6 (7), A-gliadin 57-73 ir=9 (6), PPD n=8 (62)
AE depletion: tTG-gliadin u=6 (120), A-gliadin 57-73 n=9 (80), PPD n=8 (110).
CD4 depletion: tTG-gliadin n=6 (10), A-gliadin 57-73 n=9 (9), PPD n=8 (10).
Therapeutic peptides include, but are not limited to QLQPFPQPQLPYPQPQS (AG01) QLQPFPQPQLPYPQPQP (AG02) QLQPFPQPQLPYPQPQL (AG03) QLQPFPQPQLPYLQPQP (AG04) QLQPFPRPQLPYPQPQP (AG05) QLQPEPQPQLPYSQPQP (AG06) QLQPFLQPQLPYSQPQP (AG07) QLQPFSQPQLPYSQPQP (AG08) QLQPFPQPQLSYSQPQP (AG09) PQLPYPQPQLPYPQPQP (AGIO) PQLPYPQPQLPYPQPQL (AG11) PQPQPFLPQLPYPQPQS (AG12) PQPQPFPPQLPYPQPQS (AG13) PQPQPFPPQLPYPQYQP (AG14) PQPQPFPPQLPYPQPPP (AGO 15)

Briefly after oral antigen challenge, specificities of peripheral blood T cells reflect those of intestinal T cell clones. In peripheral blood, epitopes of intestinal T cell clones are sab-optimal compared to A-gliadin 57-73 QE65, which is an optimal a-gliadin epitope.
Example 15
ELISpot assays were also carried out for mapping purposes as follows. Fine-mapping the dominant DQ-8 associated epitope

Sequence / sfc VPQLQPQNPSQ'QQPQEQV / 76
VPQLQPENPSQQQPQEQV / 3 VPQLQP1NPSQQQPQEQV / 76
VPQLQPQNPSQEQPQEQV /100 VPQLQPQNPSQRQPQEQV/ 1

tTG-treated sequence / sfc RWPVPQLQPQKPSQQ / 60 WPVPQLQPQNPSQQQ / 90 PVPQLQPQNPSQQQP /130 VPQLQPQNPSQQQPQ /140 PQLQPQNPSQQQPQE / 59 QLQPQNPSQQQPQEQ / 95 LQPQNPSQQQPQEQV / 30 QPQNPSQQQPQEQVP / 4

VPQLQPQNPSQQQPEEQV / 71 VPQLQPQNPSQQQPREQV / 27 DQ8 Gliadin Epitope
GDA09202Q/6
GDA09202Q+tTG/17
BI + tTG/0
BI/0
VPQLQPQNPSQEQPEEQV / 81 GDA09 202E / 83 VPQLQPENPSQQQPEEQV / 2 VPQLQPENPSQEQPQEQV / 6 VPQLQPENPSQEQPEEQV / 5
Fine-mapping dominant epitope (2) Pool 33 (deamidated) / sfc A2b3 301 qqyp sgqg ffqp sqqn pqaq / 2 A2b5 301 qqyp sgqg ffqp fqqn pqaq / 1 A3al 301 qqyp sgqg ffqp sqqn pqaq / 0 A3bl 301 qqyp ssqv sfqp sqlnpqaq / 0

A3b2 30] qqyp ssqg sfqp sqqn pqaq / 2 A4a 301 eqyp sgqv sfqs sqqn pqaq / 28 Albl 309 sfrp sqqn plaq gsvq pqql / 2 Alal 309 sfcp sqqn pqaq gsvq pqql / 2
Example 16
Bioactivity ofgliadin epitopes in IFNy-ELISpot (25 meg/ml, n~6) (expressed as%A-
gliadin 57-73 QE65 response)
DQ2rAH: wild type (WT) (4), WT + tTG (52), Glu-substituted (52) DQ2-A1: wild type (WT) (2), WT + tTG (22), Glu-substitnted (28) GDA09: wild type (WT) (1), WT + tTG (7), Glu-substituted (8) A-G31-49: wild type (WT) (2), WT 4- tTG (3), Glu-substituted (0)
Dose response ofA-Gliadin 57-73 QE65 (G01E) (n=8) (expressed as %GQ1E maximal response)
0.025 meg/ml (1), 0.05 meg/ml (8), 0.1 meg/ml (10), 025 meg/ml (22), 0.5 meg/ml (38), 1 meg/ml (43), 2.5 meg/ml (52), 5 meg/ml (70), 10 meg/ml (81), 25 meg/ml (95), 50 meg/ml (90), 100 meg/ml (85).
IFNy ELISpot response to giiadin epitopes alone or mixed witii A-gliadin 57-75 (GO IE) (all 50 meg/ml, tTG-gliadin 100 meg/ml, PPD 5 meg/ml; n=9) (expressed as % GO IE response)
Alone: DQ2-A1 (20), DQ2-A2 (55), Omega Gl (50), fTG Giiadin (80), PPD (220), DQ2 binder (0)
G01E+: DQ2-A1 (90), DQ2-A2.(95), Omega Gl (100), tTG Giiadin (120), PPD (280), DQ2 binder (80)
Effect ofalanine and lysine substitution of A-gliadin 57-73 QE65 on IFNy ELISpot responses in individual coeliac subjects (n=8) Epitope sequence: QLQPFPQPELPYPQPQS
Alanine substitution at positions 57-59 and 72-73 showed litfle to no . decrease in % A-gliadin 57-73 QE65 response. Alanine substitution at positions 60-62 and 68-71 showed moderate decrease in % A-gliadin 57-73 QE65 response.

Alanine substitution at positions 63-67 showed most decrease in % A-gliadin 57-73 QE65 response.
Effect of lysine substitution of A-gliadin 57-73 QE65 on IFNy ELISpot responses in individual coeliac subjects (n=8); Epitope sequence: QLQPFPQPELPYPQPQS
Lysine substitution at positions 57-59 and 71-73 showed little to no decrease in % A-gliadin 57-73 QE65 response. Lysine substitution at positions 60-61 and 69-70 showed moderate decrease in % A-gliadin 57-73 QE65 response. Lysine substitution at positions 62-68 showed most decrease in % A-gliadin 57-73 QE65 response.
Example 17
Table 24 shows the results of analyses examining the 652 pep-tides with several patients challenged with wheat or rye.
References
1. Molberg O, et aL Nature Med. 4, 713-717 (1998).
2. Quaisten E, et aL Eur. J. Imrnunol. 29,2506-2514 (1999).
.3. Greenberg CS et al FASEB 5,3071-3077 (1991).

4. MantzarisG, Jewell D. Scand. J. Gasrroenterol. 26,392-398 (1991).
5. Mauri L,etal Scand. J. Gastroenterol. 31,247-253(1996).
6. BunceM,etal. Tissue Antigens 46,355-367 (1995).
7. OlerupO,etal. Tissue antigens 41,119-134 (1993).
8. MulligaanCG.etal. Tissue-Antigens. 50,688-92 (1997).
9. PlebanskiMetaL Eur. J. Immunol. 28,4345-4355 (1998).

10. Anderson DO, Greene FC. The alpha-gliadin gene family. IE. DNA and protein
sequence variation, subfamily structure, and origins of pseudogenes. Theor Appl
Genet (1997) 95:59-65.
11. Aientz-Hansen H, Korner R, Molberg O, Quarsten H, Van der Wai Y, Kooy
YMC, Lundin KEA, Koning F, RoepstorffP, SollidLM, McAdam SN. The
intestinal T cell response to alpha-gliadin in adult celiac disease is focused on a

single deamidated glutamine targeted by tissue transglutaminase. J Exp Med. 2000; 191:603-12.
12. Vader LW, de Ru A, van der Wai, Kooy YMC, Benckhuijsen W, Mearin ML,
Drijfhout TW, van Veelen P, Koning F. Specificity of tissue transglutaminase
explains cereal toxicity in celiac disease. J Exp Med 2002; 195:643-649.
13. van der Wai Y, Kooy Y, van Veelan P, Pena S, Mearin L, Papadopoulos G,
Koning F. Selective deamidation by tissue transglutaminase strongly enhances
gliadin-specific T cell reactivity. Jfcnrnunol. 1998; 161:1585-8.
14. van der Wai Y, Kooy Y, van Veelan P, Pena S, Mearin L, Molberg 0, Lundin
KEA, Sollid L, Mutis T, BencMiuijsen WE, Drijfhout JW, Koning F. Proc Natl
Acad Sci USA 1998; 95:10050-10054.
15. Vader W, Kooy Y, Van Veelen P et aL The gluten response in children
with celiac disease is directed toward multiple gliadin and glutenin
peptides. Gastroenterology 2002,122:1729-37
16. Arentz-HanseaH, Me Adam SN, Molberg O.etal. Celiac lesion T cells
recognize epitopes that cluster in regions of gliadin rich in proline
residues. Gastroenterology 2002,123:803-809.
Each of the PCT publications, U.S. patents, other patents, journal references, and any other publications cited or referred to herein is incorporated herein by reference in their entirety.

(Table Removed) Table 16. Healthy subjects: IFNy ELISpot Responses (>10 sfc/million PBMC and 4 x buffer only) to tTG-treated gliadin peptide Pools on Day 6 of gluten challenge (sfc/million PBMC) (italic: response also present on Day 0): Group 1 - HLA-DQ2 (DQAl*0501-5, DQB1*0201)
Group 2 - KLA-DQ8 (DQA1*0301, DQB1*0302) and absent or "incomplete" DQ2 (only DQAl*0501-5 or DQB1*0201)
(Table Removed) Table 17: tTG-deamidated gliadin peptide pools showing significant increase in IFN gamma responses between Day 0 and Bay 6 of gluten challenge in HLA-DQ2 coeliac subjects C01-6 (Day 6 -Day 0 response, and ratio of responses to tTG-deamidated pool and same pool without tTG treatment)
(Table Removed) Table 18. Coeliac subjects: lENy ELISpot Responses >10 sic/million PBMC and
>4 x buffer only to tTG-treated Pepset Pools on Day 6 of gluten challenge
(sfc/million PBMC) (italic: response also present on Day 0):
Group 1-HLA-DQ2 (DQAl*0501-5, DQBl*0201/2),
Group 2 - HLA-DQ2/8 (DQAl*0501-5, *0301, and DQBl*0201/2, *0302)5 and
Group 3 - HLA-DQ8 (DQA1*0301, DQB1*0302) and absent or "incomplete"
DQ2 (only DQAl*0501-5 or DQBl*0201/2) (Table Removed)




WE CLAIM :
1. An agent for the preparation of a medicament for treating or preventing coeliac disease, wherein the agent is a peptide comprising at least one T-cell epitope, wherein the peptide is not more than 50 amino acids in length, and the T-cell epitope comprises a transglutaminase-deamidated FPQPQQPFP sequence.
2. The agent of claim 1, wherein the peptide is 10-50 amino acids in length.
3. The agent of claims 1 or 2, wherein the peptide comprises a transglutaminase-deamidated QQPFPQPQQPFP sequence.
4. The agent of any one of claim 1-3, wherein the peptide consists of a transglutaminase-deamidated QQPFPQPQQPFPWQP sequence.
5. The agent of any one of claims 1-4, wherein a modification is present on the N terminus.
6. The agent of any one of claims 1-5, wherein a modification is present on the C terminus.
7. The agent of any of claims 5 or 6, wherein said modifications are natural post-translational modifications.
8. An agent as defined in claim 1 wherein the agent is HLA-DQ2-restricted.
9. An agent as defined in claim 1 wherein the agent is HLA-DQ8-restricted.
10. An agent as defined in claim 1 wherein the agent has one or more epitopes selected from the group consisting of a wheat epitope, a barley epitope, and a rye epitope.
11. A pharmaceutical composition comprising an agent according to any one of claims 1 to 10 and a pharmaceutically acceptable carrier or diluent.

12. A pharmaceutical composition according to claim 11 additionally comprising a
peptide comprising at least one epitope comprising a sequence selected from SEQ ID
NO:l and SEQ ID NO:2.
13. A protein typically a gluten, comprising a sequence which is able to bind to a T cell receptor, which T cell receptor recognises an agent as claimed in claim 1, and which sequence is able to cause antagonism of a T cell that carries such a T cell receptor.
14. A mutant gliadin protein whose wild-type sequence can be modified by a transglutaminase to a sequence which is an agent as defined in claim 1, which mutant gliadin protein comprises a mutation, in the sequence FPQPQQPFP, QQPFPQPQQPFP or QQPFPQPQQPFPWQP which prevents its modification by a transglutaminase to a sequence which is an agent as defined in claim 1; or a fragment of such a mutant gliadin protein which is at least 15 amino acids long and which comprises the mutation.
15. A polynucleotide preferably having at least 50 base pairs that comprises a coding sequence that encodes a protein or fragment as defined in claim 14 or 15 and additionally comprises one or more regulatory sequences operably linked to the coding sequence, which regulatory sequences are capable of securing the expression of the coding sequence in a cell.
16. A polynucleotide according to claim 15 wherein the regulatory sequence(s) allow expression of the coding sequence in a prokaryotic or mammalian cell.
17. A polynucleotide according to either of claims 16 or 17 which is a vector or which is in the form of a vector.
18. A kit for carrying out a method of diagnosing coeliac disease comprising an agent as defined in any of claims 1 to 10 and a means to detect the recognition of the peptide by the T cell.
19. A kit according to claim 18. wherein the means to detect recognition comprises an antibody to IFN-.

20. A kit according to claim 19 wherein the antibody is immobilised on a solid support and optionally the kit also comprises a means to detect the antibody/IFN- complex.

Documents:

1497-DELNP-2007-Abstract-(24-01-2012).pdf

1497-delnp-2007-abstract.pdf

1497-DELNP-2007-Claims-(22-08-2012).pdf

1497-DELNP-2007-Claims-(24-01-2012).pdf

1497-delnp-2007-Claims-(27-08-2012).pdf

1497-delnp-2007-claims.pdf

1497-DELNP-2007-Correspondence Others-(14-07-2011).pdf

1497-DELNP-2007-Correspondence Others-(15-07-2011).pdf

1497-DELNP-2007-Correspondence Others-(22-08-2012).pdf

1497-DELNP-2007-Correspondence Others-(24-01-2012).pdf

1497-delnp-2007-Correspondence Others-(27-07-2012).pdf

1497-delnp-2007-Correspondence Others-(27-08-2012).pdf

1497-DELNP-2007-Correspondence-Others-(08-07-2010).pdf

1497-DELNP-2007-Correspondence-Others-(13-01-2011).pdf

1497-delnp-2007-correspondence-others-1.pdf

1497-DELNP-2007-Correspondence-Others.pdf

1497-delnp-2007-description (complete).pdf

1497-delnp-2007-drawings.pdf

1497-DELNP-2007-Form-1-(24-01-2012).pdf

1497-delnp-2007-form-1.pdf

1497-delnp-2007-form-18.pdf

1497-DELNP-2007-Form-2-(24-01-2012).pdf

1497-delnp-2007-form-2.pdf

1497-DELNP-2007-Form-3-(08-07-2010).pdf

1497-DELNP-2007-Form-3-(13-01-2011).pdf

1497-DELNP-2007-Form-3-(14-07-2011).pdf

1497-delnp-2007-Form-3-(27-07-2012).pdf

1497-DELNP-2007-Form-3.pdf

1497-delnp-2007-form-5.pdf

1497-DELNP-2007-GPA-(22-08-2012).pdf

1497-delnp-2007-gpa.pdf

1497-DELNP-2007-Petition-137-(24-01-2012).pdf


Patent Number 254995
Indian Patent Application Number 1497/DELNP/2007
PG Journal Number 03/2013
Publication Date 18-Jan-2013
Grant Date 11-Jan-2013
Date of Filing 23-Feb-2007
Name of Patentee ISIS INNOVATION LIMITED
Applicant Address EWERT HOUSE, EWERT PLACE, SUMMERTOWN, OXFORD OX2 7SG,UK
Inventors:
# Inventor's Name Inventor's Address
1 ANDERSON, ROBERT PAUL AUTOIMMUNITY AND TRANSPLANTATION DIVISION, WALTER AND ELIZA HALL INSTITUTE, C/O ROYAL MELBOURNE HOSPITAL PO, GRATTAN STREET, PARKVILLE, VICTORIA 3050, AUSTRALIA
2 HILL, ADRIAN VIVIAN SINTON WELCOME TRUST CENTRE FOR HUMAN GENETICS, UNIVERSITY OF OXFORD, ROOSEVELT DRIVE, OXFORD OX3 7BN, UK
3 JEWELL, DEREK PARRY GASTROENTEROLOGY UNIT, GIBSON BUILDING, RADCLIFFE INFIRMARY, WOODSTOCK ROAD, OXFORD OX2 6HE,UK
PCT International Classification Number C12N
PCT International Application Number PCT/GB03/02450
PCT International Filing date 2003-06-05
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 0212885.8 2002-06-05 U.K.