WO1994026774A1 - Nouveaux traitements de maladies allergiques - Google Patents

Nouveaux traitements de maladies allergiques Download PDF

Info

Publication number
WO1994026774A1
WO1994026774A1 PCT/US1994/005632 US9405632W WO9426774A1 WO 1994026774 A1 WO1994026774 A1 WO 1994026774A1 US 9405632 W US9405632 W US 9405632W WO 9426774 A1 WO9426774 A1 WO 9426774A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
peptides
cell
mhc
antigen
Prior art date
Application number
PCT/US1994/005632
Other languages
English (en)
Inventor
Jeff Alexander
Alessandra Franco
Howard M. Grey
Alessandro D. Sette
Original Assignee
Cytel Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cytel Corporation filed Critical Cytel Corporation
Priority to AU69166/94A priority Critical patent/AU6916694A/en
Publication of WO1994026774A1 publication Critical patent/WO1994026774A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to allergic diseases such as celiac disease and to materials useful in the diagnosis and treatment of these diseases.
  • it relates to novel peptides recognized by pathogenic T cells associated with the disease, e.g, celiac disease.
  • Celiac disease also called non-tropical sprue, is a disorder characterized by malabsorption, abnormal small bowel structure and intolerance to gluten, a protein found in wheat and wheat products.
  • the incidence of celiac disease is difficult to estimate because the severity of the disease varies greatly and individuals with the disease may have no overt symptoms. Seventy percent of the cases reported are women.
  • Celiac patients have an increased frequency of certain MHC haplotypes, particularly of the HLA-B8 and HLA-Dw3 types.
  • the HLA-B8 haplotype has been found in 85-90% of celiac patients as compared with 20 to 25% in normal subjects.
  • gluten which comprises gliadins and glutenins
  • glutenins is a major component of the wheat endosperm.
  • the alcohol soluble gliadin fraction of gluten has been clearly demonstrated to activate celiac disease.
  • the alcohol soluble proteins associated with the activation of disease are termed secalins, hordeins, and avenins, respectively.
  • SUBS ⁇ TUTESHEET BBLE2
  • BBLE2 SUBS ⁇ TUTESHEET
  • Diagnostic procedures typically require gut biopsies.
  • the mainstay of treatment is imposition of gluten- free diet.
  • wheat is frequently used as an extender in many processed foods, such as salad dressings, canned vegetables and soups, ice cream, and candy bars.
  • the invasive diagnostic procedures and inadequate treatments presently available demonstrate the urgent need for agents to treat celiac disease.
  • Such agents should also be economical to produce and possess favorable pharmacologic properties for example a relatively long half-life, thereby facilitating lower dosages and/or less frequent administration.
  • the present invention relates to peptides useful for diagnosing or treating celiac disease.
  • the peptides bind an MHC molecule on an antigen presenting cell associated with celiac disease and thereby inhibit activation of pathogenic T cells.
  • the peptides inhibit binding of an antigenic peptide to the MHC molecule.
  • the peptides may be T cell antagonist peptides and bind to the MHC molecule to form an inhibitory complex.
  • the peptides typically consist of between about 5 and about 25 amino acids and may comprise one or more amino acid or peptide bond mimetic.
  • the peptides may also comprise a D-amino acid.
  • the present invention also relates to the discovery that antigen presenting cells expressing DR7 or DR53 MHC glycoproteins are involved in antigen presentation in celiac disease.
  • the invention further relates to pharmaceutical compositions comprising a pharmaceutically acceptable carrier and the peptides described above. Methods of treating celiac disease comprising administering to a patient a therapeutically effective dose of the pharmaceutical compositions are also provided.
  • Fig. 1 presents data showing that pulsing target T cells with T cell receptor antagonist peptides does not inhibit the proliferative response, while incubation of antigen presenting cells with the peptide induces dose-dependent inhibition of T cell proliferation.
  • Fig. 2 presents data showing that preincubation of T cells with antigenic peptides results in induction of T cell unresponsiveness (anergy) but preincubation with T cell antagonist peptides does not.
  • Fig. 3 demonstrates that T cell antagonist peptides (unlike antigenic peptides) do not induce IL-2 production in T cells as measured by growth of the IL-2 dependent HT-2 cell line.
  • Fig. 4 demonstrates that MHC complexes comprising T cell antagonist peptides do not induce detectable increases in inositol phosphate production.
  • Fig. 5A presents data showing that peptides unrelated to the antigenic peptide act as inhibitors of antigen presentation only in the classical MHC competition assay, but are devoid of any activity in a prepulse assay.
  • Figs. 5B and C5 show that in prepulse assays T cell antagonist peptides act as potent inhibitors of target T cell proliferation but have no effect on the proliferation of non- target T cells.
  • Fig. 6 shows proliferative dose response of JR and RCL, alpha gliadin T cell lines. T cell lines were testes in the proliferation assay as described in the Materials and
  • JR open circle
  • RCL closed circle
  • Fig. 7 shows both JR and RCL lines are DR-restricted.
  • mAb inhibition studies were performed using 10 ⁇ g/ml of purified anti-class II mabs specific for various isotypes [LB3.1 (closed square) anti-DR; B27.ll (lined square) anti-DP; and 11B.5 (half-tone square) anti-DQ] .
  • the rabies glycoprotein 285-299 DR7-restricted clone C25 and the alloreactive DQ2.7-restricted T cell clone were also used as controls.
  • Figs. 8A and 8B show genetic restriction of JR and RCL T cell lines.
  • JR (Fig. 8A) and RCL (Fig. 8B) T cell lines were tested for their capacity to proliferate in response to autologous or homozygous EBV lines or DR-transfected fibroblasts, in presence (closed bars) or absence (open bars) of 100 ⁇ g/ml purified whole alpha gliadin.
  • Figs. 9A-9C show differential antigen-processing capacity of EBV lines and DR-transfected fibroblasts. Proliferation of the alpha gliadin-specific clones (Figs. 9A and 9B) or the PPD-specific line VIII (Fig. 9C) was measured in response to a dose range of antigen, utilizing either irradiated EBV lines (closed circles) or irradiated DR7-transfected fibroblasts (closed squares) as APCS.
  • Fig. 10A and 10B show proliferation of RCL cloned T cells in response to a dose range of alpha gliadin peptides presented by EBV lines or DR-transfected fibroblasts.
  • Proliferation of RCL T cells in response to a dose range of alpha gliadin (AGL) 1-20 (10A) or alpha gliadin (AGL) 1-8 (10B) was measured.
  • the APC types used were: irradiated DR-transfected fibroblasts (closed squares) ; irradiated homozygous EBV lines (closed circles) ; and fixed homozygous EBV lines (open circles) .
  • compositions of the invention comprise peptides recognized by T cells associated with the disease.
  • the peptides can be used, linked to the appropriate molecule or carrier, for the diagnosis (e.g., using intradermal reaction skin tests) of the disease.
  • the peptides can be used for treatment by 1) vaccination 20 induction of tolerance, or 3 inhibition by substituting novel peptides for the native peptide to inhibit the immune response.
  • the sequence of the peptides is preferably based on an antigen derived from wheat gluten, preferably from the gliadin fraction.
  • the peptides and analogs of interest can be tested for the ability to inhibit T cell activation using the protocols described below.
  • each residue is generally represented by standard three letter or single letter designations.
  • the L-form of an amino acid residue is represented by a capital single letter or a capital first letter of a three-letter symbol
  • the D-form for those amino acids having D-forms is represented by a lower case single letter or a lower case three letter symbol.
  • Glycine has no asymmetric carbon atom and is simply referred to as "Gly" or G.
  • the invention provides the ability to inhibit antigen specific immune responses associated with particular T cell clones.
  • This invention includes the discovery of the antigenic (or autoantigenic) peptides recognized by pathogenic T cells when the antigenic peptide is unknown.
  • the two major classes of immune response are mediated by different classes of lymphocytes, B cells, which produce antibodies, and T cells, which are responsible for cell-mediated immunity.
  • B cells which produce antibodies
  • T cells which are responsible for cell-mediated immunity.
  • T cells see antigen only when bound to MHC glycoproteins on antigen presenting cells.
  • MHC molecules are heterodimeric glycoproteins expressed on cells of higher vertebrates and play a role in immune responses. MHC glycoproteins are divided into two groups, class I and class II, which differ structurally and functionally from each other. In general, the major function of MHC molecules is to bind antigenic peptides and display them on the surface of cells. These peptides result from an antigen presenting cell (APC) processing an antigen into peptide fragments, which can be as short as 8 to 20 amino acids.
  • APC antigen presenting cell
  • Class I MHC molecules are expressed on almost all nucleated cells and are recognized by cytotoxic T lymphocytes, which then destroy the antigen-bearing cells.
  • Class II MHC molecules are expressed primarily on cells involved in initiating and sustaining immune responses, such as T lymphocytes, B lymphocytes, macrophages, etc.
  • Class II MHC molecules are recognized by helper T lymphocytes and induce proliferation of helper T lymphocytes and amplification of the immune response to the particular antigenic peptide that is displayed.
  • Engagement of the T cell receptor induces a series of molecular events characteristic of cell activation, such as, increase in tyrosine phosphorylation, Ca ++ influx, PI turnover, synthesis of cytokines and cytokine receptors, and cell division (see.
  • the present invention provides novel treatments for any condition involving unwanted T cell reactivity, such as foreign infectious diseases that can cause immunopathology (e.g.. lyme disease, hepatitis, LCMV, post-streptococcal myocarditis, or glo erulonephritis) .
  • foreign infectious diseases e.g.. lyme disease, hepatitis, LCMV, post-streptococcal myocarditis, or glo erulonephritis
  • food hypersensitivities such as celiac disease and Crohn disease as well as other allergic diseases associated with particular histocompatibility haplotypes.
  • allergic diseases suitable for treatment using the methods of the present invention see. O'Hehire, et al. Ann. Rev. Immunol. 9:67-95 (1991), which is incorporated herein by reference.
  • the treatment of celiac disease is disclosed in detail, it will be understood that the same general approach can be used for other allergic diseases
  • the antigen associated with celiac disease is the gliadin fraction of gluten.
  • Gliadins are single polypeptide chains that range in molecular weight from 30,000 to 75,000 daltons. They have a low charge and a high glutamine and proline content. These proteins have been categorized into four major electrophoretic fractions: alpha gliadins, beta gliadins, gamma gliadins and omega gliadins. When alpha gliadins from suitable wheat varieties are ultra- centrifuged, a precipitate of aggregatable alpha gliadins termed alpha gliadin is formed. This major alpha gliadin fraction is known to activate disease.
  • alpha gliadin The complete primary amino acid sequence of alpha gliadin has been determined from amino acid sequencing and this and other gliadin sequences have been deduced from sequences of cDNA clones. Bartels et al. , Nucleic Acids Res. 11:2961 (1983); Kasarda et al., Proc. Nat'l. Acad. Sci. (USA) 81:4712 (1984); and Rafalski et al. EMBO J. 3:1409 (1984), which are incorporated herein by reference.
  • Peptide fragments from gliadins capable of binding the appropriate MHC molecule can be identified using a number of methods well known to those of skill in the art. For instance, PCT application No. WO 92/02543 (which is incorporated herein by reference) describes assays for measuring binding to a desired MHC molecule. The ability of the peptide to activate the appropriate T cell can determined using in vitro assays using antigen presenting cells expressing a preselected MHC molecule. Suitable assays for this purpose are described in the Example section below.
  • antigenic peptides bind the appropriate MHC glycoprotein to form, what are referred to herein as, "MHC-antigenic peptide complexes", which then induce activation of the target T cells.
  • antigenic peptides associated with celiac disease comprise that portion of an antigen (e.g. , alpha gliadin) that binds the appropriate MHC glycoprotein (e.g. DR7) and induces activation of a T cell associated with celiac disease.
  • an antigen e.g. , alpha gliadin
  • MHC glycoprotein e.g. DR7
  • T cell clones associated with celiac disease may be obtained, for instance, from peripheral blood lymphocytes derived from healthy volunteers and celiac disease patients. Clones which proliferate in the presence of peptide fragments of the antigen can then be identified. Analogs of the antigenic peptides identified above are then screened for the ability to bind the appropriate MHC molecule and inhibit T cell proliferation.
  • Peptides identified in these assays have immunosuppressive activity in that they can compete with the antigenic peptide for MHC binding.
  • the present invention provides further assays in which the peptides are also tested for their ability to competitively inhibit binding of MHC-antigenic peptide complexes that induce T cell proliferation.
  • modified antigenic proteins e.g.. alpha gliadin
  • live antigen presenting cells which process the protein and present the blocking peptide bound to the appropriate MHC glycoprotein (e.g.. DR7) .
  • the proteins need not be antigenic proteins, but may be other unrelated proteins comprising sequences recognized by T cells associated with the disease. These proteins can also be screened for the ability to block celiac disease in celiac patients.
  • the modified gliadin proteins identified in this manner can block T cell activation by either inhibiting binding of antigenic peptides to MHC molecules or by competitively inhibiting binding of MHC-antigenic peptide complexes to T cells.
  • deletion mutants of the protein antigen are screened in celiac patients for the ability to abolish pathogenicity.
  • amino acid sequences responsible for induction of the T cell activation are identified.
  • the peptides or protein antigens identified by these assays comprise sequences necessary for recognition by the appropriate MHC allele (the agretope) and thus inhibit binding of the natural antigen to the MHC molecule.
  • the peptides may also comprise sequences necessary for recognition by the appropriate T cell receptor (the epitope) .
  • the epitope the T cell receptor
  • multiple changes are introduced into the peptides so that they comprise sequences recognized by more than one T cell clone.
  • a single peptide inhibits polyclonal responses to an antigen.
  • the sequences on the peptides recognized by the T cell receptors are sufficiently different from the wild- type epitope, so that, although binding occurs, T cell activation (e.g., proliferation) does not occur.
  • T cell antagonism The ability of the peptides, once bound to MHC glycoproteins, to bind T cell receptors without inducing T cell activation is referred to here as T cell antagonism. Such peptides are referred to as "T cell antagonist peptides.”
  • antagonists are compounds which reverse or inhibit the physiological effect of a ligand or exclude binding of the ligand to a receptor.
  • An antagonist competes directly or indirectly with the ligand (e.g.. MHC- antigenic peptide complex) for the receptor (e.g.. a T cell receptor) and, thus, reduces the proportion of ligand molecules bound to the receptor.
  • an antagonist is the topographical equivalent of the natural ligand and will compete directly with the ligand for the binding site on the receptor.
  • a compound is referred to here as a "mimetic.”
  • a ligand mimetic is a molecule (or complex of molecules) that conformationally and functionally serves as substitute for the natural ligand recognized by a receptor.
  • “Inhibitory complexes” of the present invention comprise T cell antagonist peptides bound to the appropriate MHC molecule. These complexes competitively inhibit binding by MHC-antigenic peptide complexes.
  • competitive inhibition refers to the ability of complexes comprising peptides of the present invention to compete directly or indirectly with MHC-antigenic peptide complexes.
  • the antagonistic MHC-peptide complexes of the invention act as classical competitive inhibitors in that their inhibitory effect can be overcome by a sufficiently high concentration of the complexes which induce proliferation.
  • the data presented below shows that in the case of the T cell receptor (as in the case of many other membrane receptors) , besides binding followed by activation (natural ligands or agonists) and no binding (obviously not followed by any activation; non-ligands) , an intermediate state is attainable.
  • This state entails engagement of the receptor by an agonist which does not trigger the biological response of the ligand.
  • the antagonist analog comprises the MHC-peptide complex.
  • Peptides analogs of an antigenic peptide may be determined by, e.g. synthesizing overlapping peptides, and/or employing N-terminal or C-terminal deletions (truncations) or additions. Such modifications may be made using well known peptide synthesis procedures, as described in e.g., Merrifield, Science 232:341-347 (1986), Barany and Merrifield, The Peptides. Gross and Meienhofer, eds. (N.Y., Academic Press), pp. 1-284 (1979) ; and Stewart and Young, Solid Phase Peptide Synthesis. (Rockford, 111., Pierce), 2d Ed. (1984), incorporated by reference herein. Mutant protein antigens which are processed by antigen presenting cells to form blocking peptides are recombinantly produced using suitable expression systems well known to those of skill in the art.
  • Peptides having the desired activity may be modified as necessary to provide certain desired attributes, e.g., improved pharmacological characteristics, while increasing or at least retaining substantially all of the biological activity of the unmodified peptide to inhibit celiac disease.
  • the peptides can be modified by extending, decreasing or substituting in the compound's amino acid sequence, e.g., by the addition or deletion of amino acids on either the amino terminal or carboxy terminal end, or both, of the sequences disclosed above.
  • the peptides of the invention can be modified by altering the order or composition of certain residues, it being readily appreciated that certain amino acid residues essential for biological activity, e.g., those at critical contact sites, may generally not be altered without an adverse effect on biological activity.
  • the non-critical amino acids need not be limited to those naturally occurring in proteins, such as L- ⁇ - amino acids, or their D-isomers, but may include non-protein amino acids as well, such as ⁇ - ⁇ -tf-amino acids, as well as many derivatives of L- ⁇ -amino acids.
  • a series of peptides with single amino acid substitutions are employed to determine the effect of electrostatic charge, hydrophobicity, etc. on binding.
  • a series of positively charged (e.g., Lys or Arg) or negatively charged (e.g., Glu) amino acid substitutions are made along the length of the peptide revealing different patterns of sensitivity towards various DR molecules and T cell receptors.
  • multiple substitutions using small, relatively neutral moieties such as Ala, Gly, Pro, or similar residues may be employed.
  • the substitutions may be homo-oligomers or hetero-oligomers.
  • SUBSTITUTE SHEB' LE 26 substitutions should employ amino acid residues or other molecular fragments chosen to avoid, for example, stearic and charge interference which might disrupt binding.
  • D-amino acids The effect of single amino acid substitutions may also be probed using D-amino acids.
  • Modifications of peptides with various amino acid mimetics or D-amino acids, for instance at the N- or C- termini, are particularly useful in increasing the stability of the peptide in vivo. Stability can be assayed in a number of ways. For instance, peptidases and various biological media, such as human plasma and serum, have been used to test stability. See, e.g.. Verhoef et al., Eur. J. Drug Metab. Pharmacokin. 11:291-302 (1986); Walter et al., Proc. Soc. Exp. Biol. Med.
  • the peptides may also comprise isosteres of two or more residues in the antigenic peptide.
  • An isostere as defined here is a sequence of two or more residues that can be substituted for a second sequence because the stearic conformation of the first sequence fits a binding site specific for the second sequence.
  • the term specifically includes peptide backbone modifications well known to those skilled in the art. Such modifications include modifications of the amide nitrogen, the ⁇ -carbon, amide carbonyl, complete replacement of the amide bond, extensions, deletions or backbone crosslinks. See, generally. Spatola, , Chemistry and Biochemistry of Amino Acids. Peptides and Proteins. Vol. VII (Weinstein ed. , 1983), which is incorporated herein by reference) .
  • the peptides of the present invention which bind to MHC molecules and inhibit or block MHC restricted antigen- specific T cell activation will generally comprise at least 5 amino acid residues or the conformational equivalent thereof, more usually at least 6, more often at least about 8 and frequently at least about 13 residues long, and usually will not exceed up to about 28 residues or the equivalent thereof in length, more typically 14 residues or less, and preferably no more than about 18 to 25 amino acid residues or the equivalent thereof in length.
  • the approximate length of the peptide should generally not exceed that which may be accommodated by the binding domain of the selected MHC molecule.
  • the biological activity of the peptide i.e.. the ability to inhibit antigen-specific T cell activation (typically, proliferation of T helper cells) is assayed in a variety of systems.
  • the assays are designed to detect the ability of the peptides to form MHC-peptide complexes which competitively inhibit selective binding of MHC-antigenic peptide complexes to T cell receptors on target T cells.
  • Selective binding refers to specific recognition by one molecule (e.g.. the T cell receptor) of another molecule or complex of molecules (e.g. , the MHC-peptide complex) by the spatial or polar organization of a recognition determinant on the second molecule or complex of molecules.
  • the assays are carried out by incubating the peptide with an antigen presenting cell of known MHC expression, and a target T cell clone of known antigen specificity (i.e. , T cells specific for an antigen associated with celiac disease) and MHC restriction, and the antigenic peptide itself.
  • the assay culture is incubated for a sufficient time for T cell proliferation, such as four days, and proliferation is then measured using standard procedures, such as pulsing with tritiated thymidine during the last 18 hours of incubation. If T cell hybridomas are used in the assay, interleukin-2 production is detected. The percent inhibition, compared to the controls which received no inhibitor, is then calculated.
  • a non-target T cell is one with the same MHC restriction as the target T cell, but which recognizes an a different antigen, antigenic peptide, or epitope.
  • Peptides which inhibit target T cell activation to a greater extent than they inhibit non-target T cell activation are selected.
  • Peptides identified by this method comprise
  • a second exemplary assay involves pre-pulsing antigen presenting cells with a suboptimal dose of antigenic peptides.
  • a suboptimal dose is selected such that less than about 1% of the available MHC glycoproteins are occupied by antigenic peptides.
  • the pre-pulsed antigen presenting cells are then incubated with the peptides to be tested and the appropriate target T cell clones. Inhibition of T cell activation is then measured as described above. Since the antigen presenting cells are pre-pulsed with antigenic peptides, competition for T cell receptors binding and not MHC binding is detected in the assay.
  • the capacity of peptides to inhibit antigen presentation in .in vitro assays, such as those described above, is correlated with the capacity of the peptide to inhibit an immune response in vivo.
  • In vivo activity may be determined in animal models, for example, by administering an antigen known to be restricted to the particular MHC molecule recognized by a target T cell, and the i munosuppressive peptide. T lymphocytes are subsequently removed from the animal and cultured with a dose range of antigen. Inhibition of stimulation is measured by conventional means, e.g., pulsing with tritiated thymidine and comparing to appropriate controls. Certain experimental details will of course be apparent to the skilled artisan.
  • the peptides identified using the assays described above are suitable for treating celiac and other diseases.
  • the peptides can be used in a number of applications. For instance, they may be linked to an appropriate vehicle such as a protein or lipid molecule and administered by intravascular or subcutaneous injection to induce an inflammatory response.
  • peptides which are T cell antagonists offer a number of advantages. For instance, very specific inhibition of an immune response is possible because T cell antagonism is effective against only T cells recognizing the particular antigen involved in the celiac disease. In addition, relatively low doses of peptides are required to inhibit an immune response because inhibition using these peptides is extremely efficient. It is known that as little as about 0.01% to about 0.1% of the MHC glycoproteins on an antigen presenting cell need to be occupied by antigenic peptides to trigger T cell activation (Harding et al..
  • the peptides of the present invention and pharmaceutical compositions thereof are particularly useful for administration to vertebrates, particularly humans and other mammals, to treat celiac disease.
  • the dose of the immunosuppressive peptides of the invention for treatment of celiac disease will vary according to, e.g., the peptide composition, the manner of administration, the particular disease being treated and its severity, the overall health and condition of the patient, and the judgment of the prescribing physician. Administration should begin at the first sign of symptoms or shortly after diagnosis, and continue at least until symptoms are substantially abated and for a period thereafter. In established cases loading doses followed by maintenance doses may be required.
  • compositions are intended for parenteral, topical, oral or local administration, such as by aerosol or transdermally, for prophylactic and/or therapeutic treatment.
  • the compositions are suitable for use in a variety of drug delivery systems.
  • oral administration is generally preferred.
  • the pharmaceutical compositions may be in the form of a yogurt or bacterial broth comprising bacteria that express and secrete the desired protein or peptide. The bacteria colonize the gut and deliver the immunosuppressant peptides or proteins directly to cells involved in the pathology of celiac disease.
  • a pharmaceutically acceptable nontoxic composition is generally formed by incorporating any of the normally employed excipients, and generally 10-95% of active ingredient, that is, one or more peptides of the invention, preferably 25%-75%.
  • conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • compositions for parenteral administration which comprise a solution of the immunosuppressive peptide molecules dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
  • an acceptable carrier preferably an aqueous carrier.
  • aqueous carriers may be used, e.g., water, buffered water, 0.4% saline, 0.3% glycine, hyaluronic acid and the like.
  • compositions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • the concentration of immunosuppressive peptides, which may be combined to form a peptide "cocktail" under certain circumstances for increased efficacy, in the pharmaceutical formulations can vary widely, i.e., from less than about .01%, usually at or at least about 5% to as much as 50 to 75% by weight and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • a typical pharmaceutical composition for intravenous infusion could be made up to contain 250 ml of sterile Ringer's solution, and 10 mg of peptide.
  • Actual methods for preparing parenterally administrable compounds will be known or apparent to those skilled in the art and are described in more detail in for example, Remington's Pharmaceutical Sciences. 17th ed. , Mack Publishing Company, Easton, PA (1985) , which is incorporated herein by reference.
  • conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more peptides of the invention, and more preferably at a concentration of 25%-75%.
  • the immunosuppressant peptides are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of peptides are 0.01%-20% by weight, preferably 1%-10%.
  • the surfactant must, of course, be nontoxic, and preferably soluble in the propellant.
  • esters or partial esters of fatty acids containing from 6 to 22 carbon atoms such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride such as, for example, ethylene glycol, glycerol, erythritol, arbitol, mannitol, sorbitol, the hexitol anhydrides derived from sorbitol, and the polyoxyethylene and polyoxypropylene derivatives of these esters.
  • an aliphatic polyhydric alcohol or its cyclic anhydride such as, for example, ethylene glycol, glycerol, erythritol, arbitol, mannitol, sorbitol, the hexitol anhydrides derived from sorbitol, and the polyoxyethylene
  • the surfactant may constitute 0.1%-20% by weight of the composition, preferably 0.25-5%.
  • the balance of the composition is ordinarily propellant.
  • Liquified propellants are typically gases at ambient conditions, and are condensed under pressure.
  • suitable liquified propellants are the lower alkanes containing up to 5 carbons, such as butane and propane; and preferably fluorinated or fluorochlorinated alkanes. Mixtures of the above may also be employed.
  • a container equipped with a suitable valve is filled with the appropriate propellant, containing the finely divided peptide(s) and surfactant.
  • compositions containing the immunosuppressive peptides can be administered for prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already suffering from a disease, as described above, in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications.
  • An amount adequate to accomplish this is defined as "therapeutically effective dose.” Amounts effective for this use will depend on the severity of the disease and the weight and general state of the patient, but generally range from about 1 ⁇ g to about 2,000 mg of peptide per day for a 70 kg patient, with dosages of from about 0.5 mg to about 1,000 mg of peptide per day being more commonly used.
  • the materials of the present invention may generally be employed in serious disease states, that is, life-threatening or potentially life threatening situations.
  • the relative nontoxic nature and the general lack of immunogeneticity of peptides it is possible and may be felt desirable by the treating physician to administer substantial excesses of these immunosuppressive compositions.
  • compositions containing the peptides of the invention are administered to a patient
  • the present invention is directed to vaccines which contain as an active ingredient an immunogenically effective amount of a peptide as described herein.
  • the peptide(s) may be introduced into a host, including humans, linked to its own carrier or as a homopolymer or heteropolymer of active peptide units.
  • Such a polymer has the advantage of increased immunological reaction and, where different peptides are used to make up the polymer, the additional ability to induce antibodies that react with different antigenic determinants of the antigen.
  • Useful carriers are well known in the art, and include, e.g., thyroglobulin, albumins such as human serum albumin, tetanus toxoid, polyamino acids such as poly(lysine:glutamic acid) , influenza, hepatitis B virus core protein, hepatitis B virus recombinant vaccine and the like.
  • the vaccines can also contain a physiologically tolerable (acceptable) diluent such as water, phosphate buffered saline, or saline, and further typically include an adjuvant.
  • Adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide, or alum are materials well known in the art.
  • the immune system of the host Upon immunization with a peptide composition as described herein, via injection, aerosol, oral, transdermal or other route, the immune system of the host responds to the vaccine by producing an immune response specific for the desired antigen, and the host becomes at least partially immune to later infection, or resistant to developing chronic infection.
  • Vaccine compositions containing the peptides of the invention are administered to a patient susceptible to or otherwise at risk to elicit an immune response against the antigen and thus enhance the patient's own immune response capabilities.
  • Such an amount is defined to be an "immunogenically effective dose.”
  • the precise amounts again depend on the patient's state of health and weight, the mode of administration, the nature of the formulation, etc., but generally range from about 1.0 ⁇ g to about 5000 ⁇ g per 70 kilogram patient, more commonly from about
  • SUBSUME SHEET 8 10 ⁇ g to about 500 ⁇ g mg per 70 kg of body weight.
  • peptide vaccines of the invention may be desirable to combine with vaccines which induce neutralizing antibody responses to the virus of interest, particularly to viral envelope antigens.
  • the peptides of the invention can also be expressed by attenuated viral hosts, such as vaccinia or fowlpox.
  • attenuated viral hosts such as vaccinia or fowlpox.
  • This approach involves the use of vaccinia virus as a vector to express nucleotide sequences that encode the peptides of the invention.
  • the recombinant vaccinia virus Upon introduction into an acutely or chronically infected host or into a non-infected host, the recombinant vaccinia virus expresses the immunogenic peptide, and thereby elicits a host response.
  • Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Patent No. 4,722,848, incorporated herein by reference.
  • BCG Bacillus Calmette Guerin
  • BCG vectors are described in Stover et al. (Nature 351:456-460 (1991)) which is incorporated herein by reference.
  • Other vectors useful for therapeutic administration or immunization of the peptides of the invention e.g., Salmonella typhi vectors and the like, will be apparent to those skilled in the art from the description herein.
  • the peptides may also find use as diagnostic reagents. For example, in the case of celiac disease a subcutaneous or intradermal injection of the peptides gives a local inflammatory reaction within 24-48 hours. The concentration required is generally in the range of from about O.Olmg to 5 mg.
  • the peptide can be branched (e.g., multiple linking of the same peptide) or linked to an appropriate protein carrier such as albumin or attached to a lipid molecule (e.g., palmytol)
  • Example 1 This example details how T cell antagonist peptides are identified. Briefly, peptide fragments from the appropriate antigen are prepared and screened for the ability to induce T cell activation. Those fragments which induce T cell activation are termed antigenic peptides. Next, analogs of the antigenic peptides are prepared and tested for the ability to bind the MHC molecule and form inhibitory complexes. The exemplified case described in detail below demonstrates the identification of T cell antagonist peptides derived from hemagglutinin of the influenza virus.
  • Antigenic peptides capable of inducing T cell proliferation in DR-1 restricted T cell clone were identified as follows.
  • Cells were lysed at a concentration of 10 8 cells/ml in 50 mM Tris-HCl, pH 8.5, containing 2% Renex, 150 mM NaCl, 5 mM EDTA, and 2 mM PMSF. The lysates were cleared by centrifugation at 10,000 x g for 20 min.
  • DR molecules were purified using the mAb LB3.1 covalently coupled to protein A-Sepharose CL-4B, as described in O'Sullivan et al. J. Immunol. 145:1799-1808 (1990), which is incorporated herein by reference) .
  • cell lysates were passed sequentially through the following columns: Sepharose CL-4B, protein A-Sepharose, W6/32-protein A-Sepharose, and LB3.1-protein A-Sepharose washed with 10-column volumes of 10 mM Tris-HCl, pH 8.0, 0.1% Renex (5 ml/h) ; 2-column volumes of PBS, and 2-column volumes of PBS-1% octylglucoside.
  • the LB3.1 column was eluted with 0.05 M diethylamine in 0.15 M NaCl/1%
  • protease inhibitors 5 nM 125 I-radiolabeled Y(HA 307-319) peptide for 48 hr in PBS containing 5% DMSO 0.05% NP-40 in the presence of a protease inhibitor mixture.
  • the final concentrations of protease inhibitors were: 1 mM PMSF, 1.3 mM 1.10 phenanthroline, 73 ⁇ M pepstatin A, 8 mM EDTA, 6 mM N-ethylmaleimide, and 200 ⁇ M N ⁇ -p- tosyl-L-lysine chloromethyl ketone.
  • the DR-peptide complexes were separated from free peptide by gel filtration on Sephadex G50 or TSK columns.
  • peptide inhibitors were added to DR molecules at the same time that radiolabeled peptides were added.
  • Peptide inhibitors were typically tested at concentrations ranging from 120 ⁇ g/ml to 1.2 ng/ml. The data were then plotted and the dose yielding 50% inhibition measured.
  • Peptides were synthesized on an Applied Biosystems (Foster City, CA) 430A peptide synthesizer and radiolabeled as previously described in O'Sullivan et al., supra. Briefly, after removal of the ⁇ -amino-tert-butyloxycarbonyl protecting group, the phenylacetamidomethyl resin peptide was coupled with a fourfold excess of preformed symmetrical anhydride (hydroxybenzyltriazole esters for arginine, histidine, asparagine, and glutamine) for 1 hr in dimethyIformamide. For arginine, asparagine, glutamine, and histidine residues, the coupling step was repeated in order to obtain a high coupling efficiency.
  • symmetrical anhydride hydroxybenzyltriazole esters for arginine, histidine, asparagine, and glutamine
  • Peptides were cleaved by treatment with hydrogen fluoride in the presence of the appropriate scavengers purified by reversed phase HPLC.
  • the purity of the peptides was substantiated by amino acid sequence and/or composition analysis. They were routinely greater than 95% pure after HPLC.
  • PBMC peripheral blood mononuclear cells
  • the media formulation used was RPMI 1640 supplemented with 1 mM Sodium pyruvate, 0.1 mM nonessential amino acids, 0.2 mM L-glutamine, 1000 Units/ml penicillin, 100 mg/ml Streptomycin sulphate, and 5% Human AB serum (RPMI-HS) .
  • rIL-2 (Sandoz, Basel, Switzerland) was added to 10 ng/ml final concentrations, and proliferating cultures were further expanded in rIL-2 and screened for Ag specificity.
  • Clone l was obtained by cloning an HA 307-319-specific T cell line by limiting dilution, as described in Krieger et al., J. Immunol. 146:2331-2340 (1991), which is incorporated herein by reference.
  • Clone 1 was kept in culture by periodic (-20 days) restimulation with PHA (l ⁇ g/ml) and irradiated allogenic PBMC.
  • mitomycin C-treated or fixed LG-2 APC (4 x 10 4 ) were cultured in RPMI-HS in round-bottomed 96-well microtiter plates in the presence of suboptimal Ag dose (20 nM for both HA 307-319 and TT 830-843) and varying concentrations of inhibitor peptide (70 ⁇ M to 7nM) .
  • suboptimal Ag dose (20 nM for both HA 307-319 and TT 830-843
  • concentrations of inhibitor peptide 70 ⁇ M to 7nM
  • T cells (2 x 10 4 ) were added to cultures, and after an additional 24 hr., 3 H-Thymidine (1 ⁇ Ci/well; ICN, Irvine, CA) was added.
  • IL-2 assays 50 ⁇ l of the supernatant of Clone 1 T cell cultures (2 x 10 4 cells/well) , stimulated for 24 hr with individual peptides in the presence of LG-2 APC (4 x 10 4 cells/well) , were harvested and then assayed for IL-2 content by adding them to the wells of a flat-bottomed 96-well microtiter plate containing 7 x 10 3 cells/well of the IL-2- dependent T cell line HT-2 (final supernatant dilution 1:2). After 18 hr., 3 H-Thymidine was added to HT-2 cultures, and proliferation was measured after 8 hr.
  • HA Analogs are Highly Efficient Inhibitors of an HA-Specific T Cell Clone
  • HA 307-319 and TT 830-843 peptides were radiolabeled and then assayed for their capacity to be inhibited by either HA or TT analogs. No significant difference was found between the relative capacity of either HA or TT peptides to be inhibited by either TT or HA analogs, thus formally ruling out the possibility that the observed antigen-specific component of inhibition of antigen presentation might be reflective of differential inhibitory capacity at the level of the DRl molecule.
  • the T cells were co-cultured with LG-2 APC that had been prepulsed with or without various concentrations of HA 307-319 or 601.20 as indicated. After 1 hr, the cultures were organic extracted, and the aqueous layer was analyzed for total inositol phosphates. The data were expressed as the amount of radioactivity recovered from the IP fraction divided by the total amount of radioactivity incorporated by the T cells (X100) . The values obtained were the average and standard error of duplicate samples. The results ( Figure 4) reveal a dose-dependent increase in IP formation when T cells were stimulated with APC prepulsed with antigenic peptide HA 307- 319.
  • antigen analogs can act simultaneously as: 1) MHC blocker ⁇ , competing for the peptide binding site of DRl molecules, and 2) as illustrated above, could also act by generating analog-MHC complexes capable of competing with antigen-MHC complexes for binding to the T cell receptor.
  • a "prepulse” assay in which LG-2 cells are pulsed for two hours at 37° with suboptimal doses of the HA antigen, was defined. After removing unbound antigen by washing the cells, the HA analogs are added to the APC cultures. It should be noted that under these experimental conditions, only a small number of DR molecules will be occupied by the HA analog. As discussed above, suboptimal T cell responses are induced by as little as 50 to 300 complexes, corresponding to as little as 0.01-0.1% of
  • SUBSimiTE SHEET (RULE 26) total class II, Schwartz, supra. Since peptide-MHC complexes are, in general, very long-lived (in the case of HA 307-319/DRl, the t 1/2 at 30°C is in the order of a few days, the addition of HA analogs subsequent to the antigen pulse should not result in competition at the DRl level.
  • the "prepulse" assays were performed as follows. Paraformaldehyde-treated LG-2 APC (2.7 x 10 5 /ml) were incubated with a suboptimal concentration of HA 307-319 (35 nM) for two hr at 37°C, then washed and plated at 4 x 10 /well in the presence of varying concentrations of inhibitor peptides (100 ⁇ M-5 nM) and 2 x 10 4 T cells. After 48 hr, cultures were pulsed with 3 H-Thymidine, and proliferation was measured as described above.
  • Example 2 This example shows which class II restriction elements are responsible for antigen presentation in celiac disease.
  • a panel of synthetic peptides spanning through the entire alpha gliadin sequence epitopes which are recognized by alpha gliadin-specific T cells are identified.
  • the study population consisted of three normal individuals and two patients with celiac disease (CD) .
  • Donor 1 was a 40-year-old Caucasian male with an HLA-DR3,7 haplotype
  • Donor 2 JR
  • Donor 3 was a 43-year-old Caucasian male with an HLA-DR3,4 haplotype.
  • Patient 1 was a 74-year-old Caucasian male with a long history of CD, in clinical status of remission at the time of the study, with an HLA-DR5,7 haplotype.
  • Patient 2 was a 65-year-old female with active disease at the time of the study, bearing an HLA-DR7,11 haplotype. Both patients are predicted to express DQ2.3 heterodimer that has been shown to be strongly associated with CD. The healthy controls selected for this study are also predicted to be all DQ2.3-positive, on the basis of their DR and DQ haplotypes.
  • T cell lines and clones were washed extensively and incubated (5 x 10 4 ) in 200 ⁇ l of complete medium plates with in flat-bottomed wells (Falcon Labware, Oxnard, CA) for 72 hr in duplicate with 5 x 10 4 irradiated autologous or DR3 (Mat) , DR5 (Sweig) , or DR7 (Pitout) homozygous EBV-transformed B cell lines or murine DR-transfected fibroblasts (provided by R. Karr, Monsanto Co., St. Louis, MO), in the presence of different amounts of antigen. Wells containing APCs and T cells in the absence of antigen were used as controls.
  • APCs were fixed for 30 sec with glutaraldehyde at a final concentration of 0.025% (Sigma Chemical Co., St. Louis, MO), quenched by FCS, washed extensively, and plated using the same protocol as for irradiated APCS. Eighteen hours before harvesting cultures, l ⁇ i of 3 H thymidine (ICN, Irvine, CA) was added to each well. Radioactivity retained after harvesting (1295-001 cell harvester, LKB, Gaithersburg, MD) in the filters was measured in a liquid scintillation counter (1205 beta plate, LKB, Gaithersburg, MD) . Results were expressed in mean cpm+ SEM.
  • MHC isotype restriction was determined by inhibition of proliferative responses by a panel of purified monoclonal antibodies (mabs) (anti-DR antibody LB3.1, anti-DQ2 antibody 11B.5, and anti-DP antibody B27.21).
  • mAbs were purified by Protein A affinity chromatography and added at 10 ⁇ g/ml to duplicate wells containing autologous or homozygous, heterologous irradiated EBV-B cell lines. After 1 hr incubation at 37°C in 5% C0 2 , T cells from the gliadin-specific T cell lines were added together with antigen. Two previously characterized clones, DR- and DQ-restricted, respectively, were used as controls.
  • the DR-restricted clone specific for a Rabies viral glycoprotein peptide in the context of DR7 class H molecules, was kindly donated by E. Celis (Cytel Corp. , San Diego, CA) .
  • the anti-DQ2.7 alloreactive T cell clone was kindly donated by H. Serra (Cytel Corp., San Diego, CA) .
  • the cultures were incubated for 72 hrs, then pulsed with 1 ⁇ Ci of 3 H thymidine, and incorporation was measured by liquid scintillation counter, as described below.
  • Antigen-specific T cell lines and clones were derived from peripheral blood mononuclear cells (PBMCs) from celiac disease patients and normal donors. PBL were plated at 2 x 10 5
  • Gliadin-specific T cell lines were generated by repeated in vitro stimulation with intact purified alpha gliadin of peripheral blood mononuclear cells (PBMC) derived from two different celiac disease patients. Three normal healthy individuals were also studied. Both patients were DR5,7 heterozygous and would therefore be predicted to express the DQ2.3 heterodimer which has been shown to be strongly associated with celiac disease. At the time of bleeding, the patient (RCL) was in remission, while patient AA had active disease.
  • PBMC peripheral blood mononuclear cells
  • the three healthy controls selected for this study were afl also predicted to express the DQ2.3 heterodimer, on the basis of their DR and DQ haplotype [DR3,7 (JA) ; DR3,7 (JR) ; and DR3,4 (MC) ] .
  • APCS APCS. Lines were then restimulated every 15 days as described in Materials and Methods. Before every round of stimulation, specific antigen proliferation responses were assessed. The lines that showed high background proliferation (more than five times the proliferation level of unstimulated controls) were eliminated, while lines showing no detectable specific response, but with low background, were restimulated further. After the second round of stimulation, gliadin-specific lines
  • SUBSi i ⁇ SHEET B Il were obtained from one of the normal donors (JR) and one of the two celiac patients (RCL) . No specific lines were obtained after as many as four restimulations from the remaining two normal donors and the other celiac patient. The alpha gliadin-specific proliferative responses of the two lines obtained are shown in Figure 6. In order to avoid, as much as possible, selecting in vitro for particular specificities, the two lines were immediately (after the second round of stimulation) cloned. At the same time, the lines were also expanded further and characterized for their class II restriction.
  • the DQ-specific mAb 11.B5 was effective in inhibiting the proliferation of the DQ2.7-restricted alloreactive clone JS87, but was ineffective against the control DR-restricted clone C25, and no inhibition was detected in the case of either JR or RCL gliadin-specific line. Finally, no inhibition was detected for any of the lines and clones tested in the case of the anti-DP antibody B27.21. Taken together, these data indicate that the gliadin-specific T cell lines obtained are mostly or totally DR-restricted.
  • the specific lines described above were cloned by limiting dilution in the presence of PHA, rIL2 and irradiated allogeneic feeder cells.
  • seven alpha gliadin- specific clones were generated from the JR line (normal donor)
  • four alpha gliadin-specific clones were generated from the RCL line (celiac patient) .
  • These clones were tested against a panel of 25 synthetic peptides spanning the entire alpha gliadin sequence, 20 amino acids in length and overlapping by 10 amino acids, using irradiated homozygous DR7 EBV-B cells as APC.
  • the purpose of this experiment was to determine which residues of the alpha gliadin sequences were recognized by the different clones. It was found that all the clones derived from the normal donor recognized alpha gliadin 21-40 peptide.
  • alpha gliadin peptide 1-8 was defined as the minimal epitope.
  • this peptide was used to study the T cell responses induced by irradiated transfected fibroblasts and EBV cells, it was found that fibroblasts and the EBV cell line had similar antigen-presenting capacity (Figure 10A) , thus supporting the concept that the difference between these two cell types lay in their differing capacity to process alpha gliadin and the alpha gliadin 1-20 peptide.
  • Figure 10B aldehyde-fixed EBV cells were also capable of presenting the alpha gliadin 1-8 peptide efficiently

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Botany (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne de nouveaux peptides utilisés dans le traitement de maladies allergiques, telles que la maladie c÷liaque. Les peptides fixent les glycoprotéines du complexe majeur d'histocompatibilité (MHC) associées à la maladie c÷liaque et bloquent l'activation des lymphocytes T associés à cette maladie. Les peptides peuvent bloquer la formation de complexes peptidiques antigéniques de MHC. Dans une autre variante, les peptides peuvent être des peptides antagonistes des lymphocytes T et peuvent former des complexes inhibiteurs qui inhibent de manière compétitive la liaison entre des complexes peptidiques antigéniques de MHC qui induisent l'activation des lymphocytes T associés à la maladie c÷liaque. La présente invention concerne également des procédés d'identification de ces peptides.
PCT/US1994/005632 1993-05-19 1994-05-19 Nouveaux traitements de maladies allergiques WO1994026774A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU69166/94A AU6916694A (en) 1993-05-19 1994-05-19 Novel treatments for allergic diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US6429893A 1993-05-19 1993-05-19
US08/064,298 1993-05-19

Publications (1)

Publication Number Publication Date
WO1994026774A1 true WO1994026774A1 (fr) 1994-11-24

Family

ID=22054965

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/005632 WO1994026774A1 (fr) 1993-05-19 1994-05-19 Nouveaux traitements de maladies allergiques

Country Status (2)

Country Link
AU (1) AU6916694A (fr)
WO (1) WO1994026774A1 (fr)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997027217A1 (fr) * 1996-01-26 1997-07-31 Istituto Superiore Di Sanita' Peptides et leurs utilisations dans la therapie de maladies coeliaques
ES2126512A1 (es) * 1997-03-10 1999-03-16 Consejo Superior Investigacion Peptido sintetico de la gamma-3 avenina para diagnostico y seguimiento de la enfermedad celiaca.
WO2000005249A2 (fr) * 1998-07-23 2000-02-03 The President And Fellows Of Harvard College Peptides synthetiques et procedes d'utilisation de ceux-ci dans des therapies de maladies auto-immunes
EP1098902A1 (fr) * 1998-07-23 2001-05-16 Yeda Research And Development Co. Ltd. Traitement de maladies auto-immunes a l'aide du copolymere 1 et des copolymeres et peptides apparentes
EP1128839A1 (fr) * 1998-11-12 2001-09-05 Yeda Research And Development Company, Ltd. Compositions pharmaceutiques comprenant des copolymeres de peptides synthetiques et methodes de prevention et de traitement de la reaction du greffon contre l'hote (gvhd) et de la reaction de l'hote contre le greffon (hvgd)
US6800287B2 (en) 1998-09-25 2004-10-05 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US6800285B2 (en) 2000-06-20 2004-10-05 Moses Rodriguez Treatment of central nervous system diseases by antibodies against glatiramer acetate
US7022663B2 (en) 2000-02-18 2006-04-04 Yeda Research And Development Co., Ltd. Oral, nasal and pulmonary dosage formulations of copolymer 1
US7033582B2 (en) 2000-06-05 2006-04-25 Teva Pharmaceutical Industries, Ltd. Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
US7053043B1 (en) 1999-07-23 2006-05-30 Yeda Research And Development Co.Ltd. Pharmaceutical compositions comprising synthetic peptide copolymers and methods for preventing and treating GVHD and HVGD
US7202216B2 (en) 2002-05-14 2007-04-10 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for celiac sprue
US7265093B2 (en) 2002-05-14 2007-09-04 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for Celiac Sprue
US7303871B2 (en) 2002-02-14 2007-12-04 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for Celiac Sprue
US7320788B2 (en) 2002-02-14 2008-01-22 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for Celiac Sprue
US7425332B2 (en) 1998-07-23 2008-09-16 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with Copolymer 1 and related Copolymers
US7429374B2 (en) 2001-12-04 2008-09-30 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7462688B2 (en) 2002-05-14 2008-12-09 The Board Of Trustees Of The Leland Stanford Junior University Peptides for diagnostic and therapeutic methods for celiac sprue
US7534426B2 (en) 2004-04-26 2009-05-19 The Board Of Trustees Of The Leland Stanford Junior University Glutenase enzyme assays
US7563864B2 (en) 2004-04-26 2009-07-21 Celiac Sprue Research Foundation Prolyl endopeptidase mediated destruction of T cell epitopes in whole gluten
US7579313B2 (en) 2003-11-18 2009-08-25 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
US7605150B2 (en) 2002-05-14 2009-10-20 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for Celiac Sprue
US7628985B2 (en) 2004-04-26 2009-12-08 The Board Of Regents Of The Leland Stanford Junior University Therapeutic enzyme formulations and uses thereof in celiac sprue and/or dermatitis herpetoformis
WO2010086294A2 (fr) 2009-01-28 2010-08-05 Epimmune Inc. Polypeptides de liaison de pan-dr et leurs utilisations
US7776545B2 (en) 2002-11-20 2010-08-17 The Board Of Trustees Of The Leland Stanford Junior University Diagnostic method for Celiac Sprue
US8143210B2 (en) 2002-02-14 2012-03-27 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for celiac sprue
US8778338B2 (en) 2007-03-16 2014-07-15 The Board Of Trustees Of The Leland Stanford Junior University Combination enzyme therapy for digestion of dietary gluten

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J. IMMUNOLOGY, Volume 145, Number 6, issued 15 September 1990, A.G. LAMONT et al., "Inhibition of Experimental Autoimmune Encephalomyelitis Induction in SJL/J Mice by Using a Peptide with High Affinity for IA-s Molecules", see pages 1687-1693. *
THE LANCET, Volume 1, issued 06 March 1976, S. PENA et al., "HLA-DW3 Associated with Coeliac Disease", pages 506-508. *

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997027217A1 (fr) * 1996-01-26 1997-07-31 Istituto Superiore Di Sanita' Peptides et leurs utilisations dans la therapie de maladies coeliaques
ES2126512A1 (es) * 1997-03-10 1999-03-16 Consejo Superior Investigacion Peptido sintetico de la gamma-3 avenina para diagnostico y seguimiento de la enfermedad celiaca.
EP1098902A4 (fr) * 1998-07-23 2002-07-24 Yeda Res & Dev Traitement de maladies auto-immunes a l'aide du copolymere 1 et des copolymeres et peptides apparentes
WO2000005249A3 (fr) * 1998-07-23 2000-10-05 Harvard College Peptides synthetiques et procedes d'utilisation de ceux-ci dans des therapies de maladies auto-immunes
EP1098902A1 (fr) * 1998-07-23 2001-05-16 Yeda Research And Development Co. Ltd. Traitement de maladies auto-immunes a l'aide du copolymere 1 et des copolymeres et peptides apparentes
US7566767B2 (en) 1998-07-23 2009-07-28 President And Fellows Of Harvard College Synthetic peptides and methods of use for autoimmune disease therapies
US7425332B2 (en) 1998-07-23 2008-09-16 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with Copolymer 1 and related Copolymers
WO2000005249A2 (fr) * 1998-07-23 2000-02-03 The President And Fellows Of Harvard College Peptides synthetiques et procedes d'utilisation de ceux-ci dans des therapies de maladies auto-immunes
US7279172B2 (en) 1998-07-23 2007-10-09 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with copolymer 1 and related copolymers
US7163802B2 (en) 1998-09-25 2007-01-16 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US6800287B2 (en) 1998-09-25 2004-10-05 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US7615359B2 (en) 1998-09-25 2009-11-10 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US8399211B2 (en) 1998-09-25 2013-03-19 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US7074580B2 (en) 1998-09-25 2006-07-11 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
EP1128839A1 (fr) * 1998-11-12 2001-09-05 Yeda Research And Development Company, Ltd. Compositions pharmaceutiques comprenant des copolymeres de peptides synthetiques et methodes de prevention et de traitement de la reaction du greffon contre l'hote (gvhd) et de la reaction de l'hote contre le greffon (hvgd)
EP1128839A4 (fr) * 1998-11-12 2002-10-09 Yeda Res & Dev Compositions pharmaceutiques comprenant des copolymeres de peptides synthetiques et methodes de prevention et de traitement de la reaction du greffon contre l'hote (gvhd) et de la reaction de l'hote contre le greffon (hvgd)
US7053043B1 (en) 1999-07-23 2006-05-30 Yeda Research And Development Co.Ltd. Pharmaceutical compositions comprising synthetic peptide copolymers and methods for preventing and treating GVHD and HVGD
US7022663B2 (en) 2000-02-18 2006-04-04 Yeda Research And Development Co., Ltd. Oral, nasal and pulmonary dosage formulations of copolymer 1
US7033582B2 (en) 2000-06-05 2006-04-25 Teva Pharmaceutical Industries, Ltd. Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
US6800285B2 (en) 2000-06-20 2004-10-05 Moses Rodriguez Treatment of central nervous system diseases by antibodies against glatiramer acetate
US8389228B2 (en) 2001-12-04 2013-03-05 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7923215B2 (en) 2001-12-04 2011-04-12 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7429374B2 (en) 2001-12-04 2008-09-30 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7928056B2 (en) 2002-02-14 2011-04-19 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for Celiac Sprue
US7910541B2 (en) 2002-02-14 2011-03-22 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for celiac sprue
US8962545B2 (en) 2002-02-14 2015-02-24 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for celiac sprue
US8796201B2 (en) 2002-02-14 2014-08-05 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for celiac sprue
US8143210B2 (en) 2002-02-14 2012-03-27 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for celiac sprue
US7943312B2 (en) 2002-02-14 2011-05-17 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for celiac sprue
WO2003068170A3 (fr) * 2002-02-14 2008-07-03 Univ Leland Stanford Junior Traitement de denrees alimentaires aux enzymes contre l'enteropathie au gluten
US7303871B2 (en) 2002-02-14 2007-12-04 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for Celiac Sprue
US7320788B2 (en) 2002-02-14 2008-01-22 The Board Of Trustees Of The Leland Stanford Junior University Enzyme treatment of foodstuffs for Celiac Sprue
US7923532B2 (en) 2002-02-14 2011-04-12 The Board Of Trustees Of The Leland Stanford Junior University Methods for diagnosing celiac sprue and reagents useful therein
US7202216B2 (en) 2002-05-14 2007-04-10 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for celiac sprue
US7605150B2 (en) 2002-05-14 2009-10-20 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for Celiac Sprue
US7462688B2 (en) 2002-05-14 2008-12-09 The Board Of Trustees Of The Leland Stanford Junior University Peptides for diagnostic and therapeutic methods for celiac sprue
US7265093B2 (en) 2002-05-14 2007-09-04 The Board Of Trustees Of The Leland Stanford Junior University Drug therapy for Celiac Sprue
US8426145B2 (en) 2002-11-20 2013-04-23 The Board Of Trustees Of The Leland Stanford Junior University Diagnostic method for celiac sprue
US8071316B2 (en) 2002-11-20 2011-12-06 The Board Of Trustees Of The Leland Stanford Junior University Diagnostic method for celiac sprue
US7776545B2 (en) 2002-11-20 2010-08-17 The Board Of Trustees Of The Leland Stanford Junior University Diagnostic method for Celiac Sprue
US7579313B2 (en) 2003-11-18 2009-08-25 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
US8153593B2 (en) 2003-11-18 2012-04-10 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
US7628985B2 (en) 2004-04-26 2009-12-08 The Board Of Regents Of The Leland Stanford Junior University Therapeutic enzyme formulations and uses thereof in celiac sprue and/or dermatitis herpetoformis
US7534426B2 (en) 2004-04-26 2009-05-19 The Board Of Trustees Of The Leland Stanford Junior University Glutenase enzyme assays
US7563864B2 (en) 2004-04-26 2009-07-21 Celiac Sprue Research Foundation Prolyl endopeptidase mediated destruction of T cell epitopes in whole gluten
US8470782B2 (en) 2005-08-26 2013-06-25 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
US8871718B2 (en) 2005-08-26 2014-10-28 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
US8778338B2 (en) 2007-03-16 2014-07-15 The Board Of Trustees Of The Leland Stanford Junior University Combination enzyme therapy for digestion of dietary gluten
WO2010086294A2 (fr) 2009-01-28 2010-08-05 Epimmune Inc. Polypeptides de liaison de pan-dr et leurs utilisations

Also Published As

Publication number Publication date
AU6916694A (en) 1994-12-12

Similar Documents

Publication Publication Date Title
WO1994026774A1 (fr) Nouveaux traitements de maladies allergiques
AU698962B2 (en) Alteration of immune response using pan DR-binding peptides
EP1230268B1 (fr) Analogues heteroclites de classe i epitopes
US6514942B1 (en) Methods and compositions for stimulating T-lymphocytes
US5662907A (en) Induction of anti-tumor cytotoxic T lymphocytes in humans using synthetic peptide epitopes
EP0656788B1 (fr) Peptides de liaison de hla, et leurs utilisations
EP1917970B1 (fr) Peptides se fixant au Hla et leurs utilisations
US5679640A (en) Immunosuppressant peptides
US7157091B1 (en) MAGE-A1 peptides presented by HLA class II molecules
JPH01131124A (ja) 免疫系調節ペプチド
WO1995026979A1 (fr) Regulation de l'activite des lymphocytes t cytotoxiques par des peptides de cmh de classe i
CA2223714A1 (fr) Vaccination par peptides de molecules du cmh de classe ii destinee au traitement de maladie auto-immune
Sheil et al. Identification of an autologous insulin B chain peptide as a target antigen for H-2Kb-restricted cytotoxic T lymphocytes.
WO1999005174A1 (fr) Antigene ha-1
Walker et al. Characterization of streptococcal antigen-specific CD8+, MHC class I-restricted, T cell clones that down-regulate in vitro antibody synthesis.
MXPA02009698A (es) Identificacion de peptidos de subunidad alfa del receptor de acetilcolina, inmunodominantes, potenciales.
Matsushita et al. Combinatorial peptide library for the analysis of antigen recognition by T cells
CA2402685A1 (fr) Peptides derives de l'antigene g250 associe au carcinome des cellules renales qui stimulent a la fois les lymphocytes t cd-4+ et cd-8+
ZA200301405B (en) T cell receptor Vbeta-Dbeta-Jbeta sequence and methods for its detection.
AU4863900A (en) Vaccination with peptide of MHC class II molecules for treatment of autoimmune disease
AU4111999A (en) Induction of anti-tumor cytotoxic T lymphocytes in humans using synthetic peptide epitopes
NZ524291A (en) T cell receptor Vbeta-Dbeta-Jbeta sequence LGRAGLTY (Leu Gly Arg Ala Gly Leu Thr Tyr) and methods for its detection and uses in treating autoimmune diseases
AU2002300986A1 (en) Renal Cell Carcinoma-Antigen G250-derived Peptides that elicit both CD4+ and CD8+T-cell Responses

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA