(19)
(11)EP 2 768 850 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
27.11.2019 Bulletin 2019/48

(21)Application number: 12775541.1

(22)Date of filing:  19.10.2012
(51)International Patent Classification (IPC): 
C07K 14/35(2006.01)
A61K 38/00(2006.01)
(86)International application number:
PCT/GB2012/052586
(87)International publication number:
WO 2013/057499 (25.04.2013 Gazette  2013/17)

(54)

NOVEL PEPTIDES

NEUE PEPTIDEN

NOUVELLES PEPTIDES


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30)Priority: 21.10.2011 GB 201118201

(43)Date of publication of application:
27.08.2014 Bulletin 2014/35

(73)Proprietor: Immune Regulation Limited
London WC1V 6XX (GB)

(72)Inventors:
  • COATES, Anthony Robert Milnes
    London SW17 0RE (GB)
  • TORMAY, Peter
    London SW18 4LT (GB)
  • LIGHTFOOT, Andrew
    Ware Hertfordshire SG12 0RP (GB)

(74)Representative: D Young & Co LLP 
120 Holborn
London EC1N 2DY
London EC1N 2DY (GB)


(56)References cited: : 
WO-A1-2007/096388
WO-A2-2009/106819
WO-A1-2010/006059
US-B1- 6 887 481
  
      
    Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


    Description


    [0001] The present invention relates to novel peptides derivable from the polypeptide chaperonin 60.1 and to their use in medicine, such as for the prevention and/or treatment of inflammatory conditions.

    [0002] Heat shock polypeptides are a family of molecules found in all organisms, whose function is to aid the biological processing and stability of biological molecules (Zugel & Kauffman (1999) Role of heat shock polypeptides in protection from and pathogenesis of infectious diseases. Clin. Microbiol. Rev. (12)1: 19-39; Ranford et al. (2000) Chaperonins are cell signalling polypeptides: - the unfolding biology of molecular chaperones. Exp. Rev. Mol. Med., 15 September, www.ermn.cbcu.cam.ac.uk/).

    [0003] Heat shock polypeptides are located in every cellular compartment, and possess the ability to interact with a wide range of biological molecules. In particular, the heat shock polypeptides aid and influence polypeptide folding and polypeptide translocation at any time from assembly through to disassembly of the polypeptide and any complexes thereof. The helper nature of the heat shock polypeptides has led to them to also being known as molecular chaperones (Laskey et al. (1978) Nucleosomes are assembled by an acidic polypeptide, which binds histones and transfers them to DNA. Nature (275): 416-420).

    [0004] Heat shock polypeptides are synthesised by cells in response to environmental stress, which includes, but is not limited to temperature changes (both increases and decreases), and pathophysiological signals such as cytokines. In response to the environmental stress, heat shock polypeptides use their ability to process other polypeptides to protect such polypeptides from any denaturation that may occur due to the presence of the stress. This mechanism also serves to protect cells which contain the protein.

    [0005] Chaperonin polypeptides are a subgroup of heat shock polypeptides whose role in polypeptide folding is well known. There are two families of chaperonin polypeptide, the chaperonin 60 (approximately 60 kDa) and chaperonin 10 (approximately 10 kDa) families (Ranford, 2000). The best characterised chaperonins are those derived from E. coli, from which the characteristic structure of chaperonin 60 and chaperonin 10 has been established. The chaperonin complexes of most other organisms also substantially conform to this characteristic structure.

    [0006] The characteristic structure of chaperonins is a complex formed from two heptamer rings (composed of seven chaperonin 60 monomers) which face one another and are capped by a heptamer ring composed of chaperonin 10 monomers.

    [0007] Conventionally, chaperonins assist polypeptide folding when the target polypeptide enters the central core of the ringed heptamers, and on the subsequent release of energy from ATP the target polypeptide is released from the central core by a conformational change in the chaperonin structure (Ranson et al. (1998) Review Article: Chaperones. Biochem. J (333): 233-242).

    [0008] Mycobacterium tuberculosis (M. tuberculosis) produces Chaperonin 60.1 (Cpn60.1), a polypeptide that is named based on its amino acid sequence identity to other known chaperonins. Further M. tuberculosis chaperonin polypeptides are chaperonin 10 (Cpn10) and chaperonin 60.2 (Cpn60.2). Cpn60.2 exhibits 59.6% amino acid sequence identity and 65.6% nucleic acid sequence identity to Cpn60.1.

    [0009] International Patent Application, Publication Number WO02/040037 discloses pharmaceutical compositions comprising Cpn60.1 from M. tuberculosis (MtCpn60.1) and its encoding nucleic acid molecules. This application also discloses a number of specific peptide fragments derivable from the whole length polypeptide which possess similar biological activity. A variety of therapeutic uses for these molecules is also disclosed, including the treatment and/or prevention of autoimmune disorders, allergic conditions, conditions typified by a Th2-type immune response and conditions associated with eosinophilia.

    [0010] International Patent Application, Publication Number WO2009/106819 discloses a series of novel peptides derivable from MtCpn60.1 including a peptide (designated as "Peptide 4") having an amino acid sequence: DGSVVVNKVSELPAGHGLNVNTLSYGDLAAD. Peptide 4 exhibits anti-inflammatory activity and has been shown to significantly reduce the recruitment of eosinophils in an animal model of allergic airway inflammation.

    [0011] The present invention is based upon the unexpected finding that certain novel sub-fragments of Peptide 4 (DGSVVVNKVSELPAGHGLNVNTLSYGDLAAD) exhibit biological activity, in particular an ability to inhibit leukocyte diapedesis. The novel peptides of the present invention are particularly suited for development as pharmaceuticals owing to their comparatively short amino acid chain length which renders them convenient to prepare and isolate in high yield. They also indicated to possess improved biological stability in vivo relative to MtCpn60.1 and known peptide fragments thereof.

    [0012] Thus, in a first aspect, the invention provides an isolated or recombinant peptide molecule consisting of an amino acid sequence selected from the group:
    1. (i) DGSVVVNKVSELPAGH;
    2. (ii) GLNVNTLSYGDLAAD; and
    3. (iii) SELPAGHGLNVNTLS.


    [0013] By "functionally equivalent" is meant any described peptide and/or variant or fragment thereof which possesses a function (e.g. biological activity) that is identical or substantially similar to any function displayed by or attributed to one or more of the defined amino acid sequences (i) to (iii). For example, peptides consisting of the amino acid sequences defined in (i) to (iii) exhibit anti-inflammatory properties permitting their use in the prevention and/or treatment of a variety of diseases and disorders, including arthritis and pain. Functional equivalence in respect of a particular biological activity can be measured using conventional models and methods; for example, by measuring paw latency on a heated plate or measuring the release of inflammatory cytokines in vivo or in vitro.

    [0014] By "variant" is meant a described peptide having an amino acid sequence which has or 70% or more, such as 75%, 80%, 85%, 90% or 95% identity to a sequence defined in any of the sequence lists (i) to (vi) above. Thus the term "variant" refers to polypeptides and peptides differing from naturally occurring molecules by amino acid insertions, deletions, and substitutions, created using, e g., recombinant DNA techniques. Guidance in determining which amino acid residues may be replaced, added or deleted without abolishing activities of interest, may be found by comparing the sequence of the particular polypeptide with that of homologous peptides and minimizing the number of amino acid sequence changes made in regions of high homology (conserved regions) or by replacing amino acids with consensus sequence.

    [0015] Alternatively, described recombinant variants encoding these same or similar polypeptides may be synthesized or selected by making use of the "redundancy" in the genetic code. Various codon substitutions, such as the silent changes which produce various restriction sites, may be introduced to optimize cloning into a plasmid or viral vector or expression in a particular prokaryotic or eukaryotic system. Mutations in the polynucleotide sequence may be reflected in the polypeptide or domains of other peptides added to the polypeptide to modify the properties of any part of the polypeptide, to change characteristics such as ligand-binding affinities, interchain affinities, or degradation/turnover rate.

    [0016] Described , amino acid "substitutions" are the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, i.e., conservative amino acid replacements. "Conservative" amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Described "Insertions" or "deletions" are preferably in the range of about 1 to 10 amino acids, more preferably 1 to 5 amino acids, such as 1, 2, 3, 4 or 5 amino acids. The described variation may be experimentally determined by systematically making insertions, deletions, or substitutions of amino acids in a polypeptide molecule using recombinant DNA techniques and assaying the resulting recombinant variants for biological activity.

    [0017] It is also described that, where alteration of function is desired, insertions, deletions or non-conservative alterations can be engineered to produce altered polypeptides. Such alterations can, for example, alter one or more of the biological functions or biochemical characteristics of the polypeptides of the invention. For example, such alterations may change polypeptide characteristics such as ligand-binding affinities, interchain affinities, or degradation/turnover rate. Further, such alterations can be selected so as to generate polypeptides that are better suited for expression, scale up and the like in the host cells chosen for expression.

    [0018] The present invention further provides an isolated or recombinant nucleic acid molecule consisting of a polynucleotide sequence which encodes a peptide consisting of an amino acid sequence selected from the group:
    1. 1. DGSVVVNKVSELPAGH;
    2. 2. GLNVNTLSYGDLAAD; and
    3. 3. SELPAGHGLNVNTLS.


    [0019] The term "polynucleotide" refers to a heteropolymer of nucleotides or the sequence of these nucleotides. It also refers to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA) or to any DNA-like or RNA-like material. In the sequences herein A is adenine, C is cytosine, T is thymine, G is guanine and N is A, C, G or T (U). It is contemplated that where the polynucleotide is RNA, the T (thymine) in the sequences provided herein is substituted with U (uracil). Generally, nucleic acid segments provided by this invention may be assembled from fragments of the genome and short oligonucleotide linkers, or from a series of oligonucleotides, or from individual nucleotides, to provide a synthetic nucleic acid which is capable of being expressed in a recombinant transcriptional unit comprising regulatory elements derived from a microbial or viral operon, or a eukaryotic gene.

    [0020] The polynucleotides of the invention include naturally occurring or wholly or partially synthetic DNA, e.g., cDNA and genomic DNA, and RNA, e.g., mRNA. The polynucleotides may include the entire coding region of the cDNA or may represent a portion of the coding region of the cDNA.

    [0021] The present invention also provides genes corresponding to the cDNA sequences disclosed herein. The corresponding genes can be isolated in accordance with known methods using the sequence information disclosed herein. Such methods include the preparation of probes or primers from the disclosed sequence information for identification and/or amplification of genes in appropriate genomic libraries or other sources of genomic materials. Further 5' and 3' sequence can be obtained using methods known in the art. For example, full length cDNA or genomic DNA that corresponds to any of the polynucleotides of the invention can be obtained by screening appropriate cDNA or genomic DNA libraries under suitable hybridization conditions using any of the polynucleotides of the invention or a portion thereof as a probe. Alternatively, the polynucleotides of the invention may be used as the basis for suitable primer(s) that allow identification and/or amplification of genes in appropriate genomic DNA or cDNA libraries.

    [0022] The nucleic acid sequences of the invention can be assembled from ESTs and sequences (including cDNA and genomic sequences) obtained from one or more public databases, such as dbEST, gbpri, and UniGene. The EST sequences can provide identifying sequence information, representative fragment or segment information, or novel segment information for the full-length gene.

    [0023] MtCpn60.1 may be cloned and expressed using the methods described in T.H. Kong et al., Proc. Natl. Acad. Sci., 1993, 90, 2608-2612 and J.C. Lewthwaite et al, Infection and Immunity, 2001, 69(12), 7349-7355. MtCpn60.1 is also commercially available from Lionex (Germany).

    [0024] The peptides of the present invention may be prepared and/or isolated using conventional methods known in the art. For example, by solution or solid phase synthesis using traditional methods or using a solid phase automated synthesizer, for example as described in I. Coin, Nature Protocols, 2007, 2, 3247-3256. Preferably, the peptides of the present invention are prepared by Fmoc solid phase synthesis using methods analogous to those described in G.B. Fields and R.L. Noble, Int. J. Peptide Protein Res., 1990, 35(3), 161-214.

    [0025] According to a further aspect of the present invention there is provided a peptide or nucleic acid molecule as defined herein for use in medicine.

    [0026] In one embodiment, the invention provides the use of a peptide or nucleic acid molecule as defined herein for the modulation of diapedesis, i.e. the movement or passage of blood cells, preferably white blood cells, through intact capillary walls into surrounding body tissue, in a human subject.

    [0027] The peptide and nucleic acid molecules of the present invention are also indicated to be useful in the prevention and/or treatment of any condition, disease and/or disorder in a human subject which is associated with an increase in the flow of white blood cells across the endothelium. Examples of such conditions include, but are not limited to, acute and/or chronic inflammatory conditions such as ischemia-reperfusion injury (including coronary thrombosis and cerebral artery blockage), autoimmune diseases such as multiple sclerosis, allergic conditions such as asthma, chronic obstructive pulmonary disease and altitude sickness.

    [0028] Preferably, the peptide or nucleic acid molecule of the present invention is used for the treatment and/or prevention of acute and/or chronic inflammatory conditions, particularly non-allergic inflammation. Examples of inflammatory conditions which may be prevented and/or treated with the peptide or nucleic acid molecules of the present invention include conditions associated with eosinophila and/or neutrophilia. Preferred examples of acute and/or chronic inflammatory conditions include intimation associated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury including coronary thrombosis and cerebral artery blockage, shock lung syndrome, endotoxin lethality, arthritis (particularly rheumatoid arthritis or chronic inflammatory arthritis), complement-mediated hyperacute rejection, nephritis, cytokine or chemokine-induced lung injury, non-allergic asthma, inflammatory bowel disease and Crohn's disease.

    [0029] Also described is a method of preventing and/or treating an acute and/or chronic inflammatory condition which comprises administering to a mammal, including man, a peptide or nucleic acid molecule as defined herein.

    [0030] Further descried is the use of a peptide or nucleic acid molecule as defined herein in the manufacture of a medicament for the prevention and/or treatment of an acute and/or chronic inflammatory condition.

    [0031] In an alternative embodiment, the present invention provides the use of a peptide or nucleic acid molecule as defined herein for the prevention and/or treatment of chronic obstructive pulmonary disease.

    [0032] In an alternative embodiment, the present invention provides the use of a peptide or nucleic acid molecule as defined herein for the prevention and/or treatment of autoimmune disorders.

    [0033] Within the term "autoimmune disorders" as used herein are included conditions where it can be shown that the autoimmune process contributes to the pathogenesis of a disease. Such disorders are typically associated with a T helper lymphocyte-1 (Th-1) type immune response.

    [0034] Examples of autoimmune disorders which may be prevented and/or treated with the peptide or nucleic acid molecules of the present invention include autoimmune disorders, such as haemolytic anaemia, thrombocytopenia, pernicious anaemia, Addison's disease, autoimmune diabetes, insulin dependent diabetes mellitus, myasthenia gravis, rheumatoid arthritis, systemic lupus erythematosus, atherosclerosis, autoimmune encephalitis, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease. Preferred autoimmune disorders include rheumatoid arthritis and multiple sclerosis.

    [0035] The immunosuppressive effects of the peptide or nucleic acid molecules of the present invention against rheumatoid arthritis may be determined in an experimental animal model system. The experimental model system is adjuvant induced arthritis in rats, and the protocol is described by J. Holoshitz, et at., 1983, Science , 219:56, or by B. Waksman et al., 1963, Int. Arch. Allergy Appl. Immunol., 23:129. Induction of the disease can be caused by a single injection, generally intradermally, of a suspension of killed Mycobacterium tuberculosis in complete Freund's adjuvant (CFA). The route of injection can vary, but rats may be injected at the base of the tail with an adjuvant mixture. The polypeptide is administered in phosphate buffered solution (PBS) at a dose of about 1-5 mg/kg. The control consists of administering PBS only.

    [0036] The procedure for testing the effects of the test compound would consist of intradermally injecting killed Mycobacterium tuberculosis in CFA followed by immediately administering the test compound and subsequent treatment every other day until day 24. At 14, 15, 18, 20, 22, and 24 days after injection of Mycobacterium CFA, an overall arthritis score may be obtained as described by J. Holoskitz above. An analysis of the data would reveal that the test compound would have a dramatic affect on the swelling of the joints as measured by a decrease of the arthritis score.

    [0037] In an alternative embodiment, the present invention provides the use of a peptide or nucleic acid molecule as defined herein for the prevention and/or treatment of allergic conditions. Examples of allergic conditions and disorders which may be prevented and/or treated with the peptide or nucleic acid molecules of the present invention include eczema, dermatitis, allergic rhinitis (hay fever), allergic airways diseases, hyper-eosinophilic syndrome, contact dermatitis; respiratory diseases characterized by eosinophilic airway inflammation and airway hyper-responsiveness, such as asthma, including allergic asthma and intrinsic asthma, allergic bronchopulmonary aspergillosis, eosinophilic pneumonia, allergic bronchitis bronchiectasis, occupational asthma, reactive airway disease syndrome, interstitial lung disease, hyper-eosinophilic syndrome, parasitic lung disease; anaphylaxis, serum sickness, drug reactions, food allergies, insect venom allergies, mastocytosis, hypersensitivity pneumonitis, urticaria, angioedema, eczema, atopic dermatitis, allergic contact dermatitis, erythema multiforme, Stevens-Johnson syndrome, allergic conjunctivitis, atopic keratoconjunctivitis, venereal keratoconjunctivitis and giant papillary conjunctivitis.

    [0038] Other conditions, in which immune suppression is desired (including, for example, organ transplantation), may also be treatable using a peptide or nucleic acid molecule of the present invention. Preferred allergic disorders and conditions include asthma, allergic rhinitis, eczema and anaphylaxis.

    [0039] Within the terms "allergic disorders" and "allergic conditions" as used herein are included conditions associated with a T helper lymphocyte-2 (Th-2) type immune response. In allergic reaction, high IgE levels occur and Th-2 immune responses predominate over Th-1 responses, resulting in inflammatory response.

    [0040] The therapeutic effects of the polypeptides or antagonists thereof on allergic reactions can be evaluated by in vivo animals models such as the cumulative contact enhancement test (Lastborn et al., Toxicology 125: 59-66, 1998), skin prick test (Hoffmann et al., Allergy 54: 446-54, 1999), guinea pig skin sensitization test (Vohr et al., Arch. Toxocol. 73: 501-9), and murine local lymph node assay (Kimber et al., J. Toxicol. Environ. Health 53: 563-79).

    [0041] Suitable assays for thymocyte or splenocyte cytotoxicity include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan et al., Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Handa et al., J. Immunol. 135:1564-1572, 1985; Takai et al., I. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; Bowman et al., J. Virology 61:1992-1998; Bertagnolli et al., Cellular Immunology 133:327-341, 1991; Brown et al., J. Immunol. 153:3079-3092, 1994.

    [0042] Assays for T-cell-dependent immunoglobulin responses and isotype switching (which will identify, among others, proteins that modulate T-cell dependent antibody responses and that affect Th1/Th2 profiles) include, without limitation, those described in: Maliszewski, J. Immunol. 144:3028-3033, 1990; and Assays for B cell function: In vitro antibody production, Mond, J. J. and Brunswick, M. In Current Protocols in Immunology. J. E. Coligan et al., eds. Vol 1 pp. 3.8.1-3.8.16, John Wiley and Sons, Toronto. 1994.

    [0043] Mixed lymphocyte reaction (MLR) assays (which will identify, among others, proteins that generate predominantly Th1 and CTL responses) include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan et al., Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; Bertagnolli et al., J. Immunol. 149:3778-3783, 1992.

    [0044] Dendritic cell-dependent assays (which will identify, among others, proteins expressed by dendritic cells that activate naive T-cells) include, without limitation, those described in: Guery et al., J. Immunol. 134:536-544, 1995; Inaba et al., J. Experimental Medicine 173:549-559, 1991; Macatonia et al., J. Immunol. 154:5071-5079, 1995; Porgador et al., J. Experimental Medicine 182:255-260, 1995; Nair et al., J. Virology 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., J. Experimental Medicine 169:1255-1264, 1989; Bhardwaj et al., J. Clinical Investigation 94:797-807, 1994; and Inaba et al., J. Experimental Medicine 172:631-640, 1990.

    [0045] Assays for lymphocyte survival/apoptosis (which will identify, among others, proteins that prevent apoptosis after superantigen induction and proteins that regulate lymphocyte homeostasis) include, without limitation, those described in: Darzynkiewicz et al., Cytometry 13:795-808, 1992; Gorczyca et al., Leukemia 7:659-670, 1993; Gorczyca et al., Cancer Research 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, J. Immunol. 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., Int. J. Oncol. 1:639-648, 1992.

    [0046] Assays for proteins that influence early steps of T-cell commitment and development include, without limitation, those described in: Antica et al., Blood 84:111-117, 1994; Fine et al., Cellular Immunology 155:111-122, 1994; Galy et al., Blood 85:2770-2778, 1995; Toki et al., Proc. Natl. Acad Sci. USA 88:7548-7551, 1991.

    [0047] As used herein "treatment" means reducing alleviating or eliminating one or more symptoms of the condition which is being treated, relative to the symptoms prior to treatment. For example, symptoms which may be affected include eosinophilia, decreased secretion of particular cytokines, a Th2-baised immune response, allergic response and the presence of autoantibodies.

    [0048] As used herein "prevention" means delaying or preventing the onset of a condition or reducing its severity, as assessed by the appearance or extent of one or more symptoms of said condition.

    [0049] In a further aspect, the invention provides a pharmaceutical composition comprising or consisting of a peptide or nucleic acid molecule of the present invention and a pharmaceutically-acceptable excipient.

    [0050] The molecules, medicaments and pharmaceutical compositions of the present invention may be delivered using an injectable sustained-release drug delivery system. These are designed specifically to reduce the frequency of injections. An example of such a system is Nutropin Depot which encapsulates recombinant human growth hormone (rhGH) in biodegradable microspheres that, once injected, release rhGH slowly over a sustained period. Preferably, delivery is performed intra-muscularly (i.m.) and/or sub-cutaneously (s.c.) and/or intravenously (i.v.).

    [0051] The molecules, medicaments and pharmaceutical compositions of the present invention can be administered by a surgically implanted device that releases the drug directly to the required site. For example, Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis. The direct application of this toxic agent to the site of disease achieves effective therapy without the drug's significant systemic side-effects.

    [0052] Electroporation therapy (EPT) systems can also be employed for the administration of the agents, medicaments and pharmaceutical compositions of the invention. A device which delivers a pulsed electric field to cells increases the permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery.

    [0053] The molecules, medicaments and pharmaceutical compositions of the invention can also be delivered by electroincorporation (EI). EI occurs when small particles of up to 30 microns in diameter on the surface of the skin experience electrical pulses identical or similar to those used in electroporation. In EI, these particles are driven through the stratum corneum and into deeper layers of the skin. The particles can be loaded or coated with drugs or genes or can simply act as "bullets" that generate pores in the skin through which the drugs can enter.

    [0054] An alternative method of delivery of the molecules, medicaments and pharmaceutical compositions of the invention is the ReGel injectable system that is thermo-sensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The active substance is delivered over time as the biopolymers dissolve.

    [0055] The molecules, medicaments and pharmaceutical compositions of the invention can also be delivered orally. The process may employ a natural process for oral uptake of vitamin B12 and/or vitamin D in the body to co-deliver proteins and peptides. By riding the vitamin B12 and/or vitamin D uptake system, the nucleic acids, molecules and pharmaceutical formulations of the invention can move through the intestinal wall. Complexes are synthesised between vitamin B12 analogues and/or vitamin D analogues and the drug that retain both significant affinity for intrinsic factor (IF) in the vitamin B12 portion/vitamin D portion of the complex and significant bioactivity of the active substance of the complex.

    [0056] The molecules, medicaments and pharmaceutical compositions of the invention can be introduced to cells by "Trojan peptides". These are a class of polypeptides called penetratins which have translocating properties and are capable of carrying hydrophilic compounds across the plasma membrane. This system allows direct targeting of oligopeptides to the cytoplasm and nucleus, and may be non-cell type specific and highly efficient. See Derossi et al. (1998), Trends Cell Biol 8, 84-87.

    [0057] Preferably, the medicament and/or pharmaceutical composition of the present invention is a unit dosage containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of the active ingredient.

    [0058] The molecules, medicaments and pharmaceutical compositions of the invention will normally be administered orally or by any parenteral route, in the form of a pharmaceutical composition comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions may be administered at varying doses.

    [0059] In human therapy, the molecules, medicaments and pharmaceutical compositions of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient, diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.

    [0060] For example, the molecules, medicaments and pharmaceutical compositions of the invention can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed- or controlled-release applications. The molecules, medicaments and pharmaceutical compositions of the invention may also be administered via intracavernosal injection.

    [0061] Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.

    [0062] Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the agents of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.

    [0063] The molecules, medicaments and pharmaceutical compositions of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.

    [0064] Medicaments and pharmaceutical compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The medicaments and compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.

    [0065] For oral and parenteral administration to human patients, the daily dosage level of the molecules, medicaments and pharmaceutical compositions of the invention will usually be from 0.1 to 100 mg per adult per day administered in single or divided doses.

    [0066] Thus, for example, the tablets or capsules of the molecules of the invention may contain from 0.1 mg to 100mg of active agent for administration singly or two or more at a time, as appropriate. The physician in any event will determine the actual dosage which will be most suitable for any individual patient and it will vary with the age, weight and response of the particular patient. The above dosages are exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited and such are within the scope of this invention.

    [0067] The molecules, medicaments and pharmaceutical compositions of the invention can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EA3), carbon dioxide or other suitable gas. In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active agent, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g. sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a agent of the invention and a suitable powder base such as lactose or starch.

    [0068] Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff" contains at least 0.1 mg of a molecule of the invention for delivery to the patient. It will be appreciated that he overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided doses throughout the day.

    [0069] Alternatively, the molecules, medicaments and pharmaceutical compositions of the invention can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, gel, ointment or dusting powder. The molecules, medicaments and pharmaceutical compositions of the invention may also be transdermally administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.

    [0070] For ophthalmic use, the molecules, medicaments and pharmaceutical compositions of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.

    [0071] For application topically to the skin, the molecules, medicaments and pharmaceutical compositions of the invention can be formulated as a suitable ointment containing the active agent suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene agent, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.

    [0072] Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouth-washes comprising the active ingredient in a suitable liquid carrier.

    [0073] Generally, in humans, oral or parenteral administration of the molecules, medicaments and pharmaceutical compositions of the invention agents of the invention is the preferred route, being the most convenient.

    [0074] For veterinary use, the molecules, medicaments and pharmaceutical compositions of the invention is administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.

    [0075] Conveniently, the formulation is a pharmaceutical formulation. Advantageously, the formulation is a veterinary formulation.

    [0076] Advantageously, in the use according to the invention, the daily dosage level will be from 0.0001 to 100,000 mg, administered in single or divided doses; preferably, the daily dosage level is 0.0001 to 1000 mg.

    [0077] Preferred pharmaceutical formulations include those in which the active ingredient is present in at least 1% (such as at least 10%, preferably in at least 30% and most preferably in at least 50%) by weight. That is, the ratio of active ingredient to the other components (i.e. the addition of adjuvant, diluent and carrier) of the pharmaceutical composition is at least 1:99 (e.g. at least 10:90, preferably at least 30:70 and most preferably at least 50:50) by weight.

    [0078] Typically, the time between dose administration to the patient is between six and twelve hours; in a preferred embodiment, the time between dose administration to the patient is between nine and twelve hours after the previous dose; more preferably, the time between dose administration to the patient is between twelve hours and twelve days; even more preferably, the time between dose administration to the patient is between twelve days and six months.

    [0079] In a preferred embodiment, the invention provides a use wherein the medicament of the invention is used to relieve pain in a human or animal patient.

    [0080] Preferably, the pharmaceutical composition or the medicament of the invention is formulated to permit administration by at least one route selected from the group comprising or consisting of: intranasal; oral; parenteral; topical; ophthalmic; suppository; pessary; or inhalation routes. Formulations suitable for such administration routes are well known to those in the art of pharmacy and medicine and exemplary formulations are described above and in the accompanying examples.

    [0081] In a further aspect, the invention provides the use of a peptide molecule according to the invention and/or a nucleic acid molecule according to the invention as an adjuvant.

    [0082] The term "adjuvant" means any substance which, when incorporated into or administered simultaneously with antigen, potentiates the immune response.

    [0083] In a further aspect, the invention provides an adjuvant system comprising (i) a peptide molecule according to the invention and/or a nucleic acid molecule according to the invention and (ii) an antigen.

    [0084] Preferably, the antigen is selected from the group comprising or consisting of: anthrax antigen; cholera antigen; diphtheria antigen; haemophilus influenza b (Hib) antigen; hepatitis A antigen; hepatitis B antigen; influenza antigen; Japanese encephalitis antigen; measles, mumps and rubella (MMR) antigen; meningococcal antigen; pertussis antigen; pneumococcal antigen; poliomyelitis antigen; rabies antigen; rubella antigen; smallpox and/or vaccinia antigen; tetanus antigen; tick-borne encephalitis antigen; tuberculosis antigen; typhoid antigen; varicella/herpes zoster antigen; yellow fever antigen; and veterinary vaccine antigen.

    Description of Figures



    [0085] 

    Figure 1
    The nucleotide and amino acid sequence of chaperonin 60.1 from M. tuberculosis.

    Figure 2
    Peptides covering the equatorial domain of chaperonin 60.1 from M. tuberculosis.

    Figure 3
    Effect of peptide fragments of chaperonin 60.1 from M. tuberculosis in a lipopolysaccharide (LPS) model of non-allergic inflammation


    Examples



    [0086] The following abbreviations are used in the examples to refer to peptides of the present invention:

    F1 - DGSVVVNKVSELPAGH

    F2 - GLNVNTLSYGDLAAD

    F3 - SELPAGHGLNVNTLS


    Experimental section



    [0087] The peptides F1-F3 were synthesized and isolated according to the following procedures:

    F1



    [0088] The peptide was synthesised on Fmoc-His(trt)-Wang LL resin (200mg) from Merck Chemicals. A Protein Technologies Symphony Automated synthesiser was used to add the remaining amino acid residues. All coupling reactions were 10 minute double couplings with HBTU coupling agent. The peptide was cleaved in 100% Trifluoroacetic Acid in the presence of a scavenger mix containing 1:1 v/v Triisopropylsilane:Water. The peptide was purified upon a LC-ABZ+ (Supelcosil) column, 5 micron particle size, 110 Angstrom pore size, 250 mm x 10 mm, from Supelco. Analysis was performed with the same buffers, but using a C-18, 3.5 micron, 90 angstrom pore size, 4.6mm X 150 mm column from agilent. The run conditions were:

    absorption 216 nm, flow rate 1 mL/ min

    t=0, 0% B

    t=2, 0% B

    t=22, 80% B

    % Yield = 31%


    F2



    [0089] The peptide was synthesised on Fmoc-Asp(OtBu)-Wang LL resin (150mg) from Merck Chemicals. A Protein Technologies Symphony Automated synthesiser was used to add the remaining amino acid residues. All coupling reactions were 10 minute double couplings with HBTU coupling agent. The peptide was cleaved in 100% Trifluoroacetic Acid in the presence of a scavenger mix containing 1:1 v/v Triisopropylsilane:Water. The peptide was purified upon a LC-ABZ+ (Supelcosil) column, 5 micron particle size, 110 Angstrom pore size, 250 mm x 10 mm, from Supelco. Analysis was performed with the same buffers, but using a C-18, 3.5 micron, 90 angstrom pore size, 4.6mm X 150 mm column from agilent. The run conditions were:

    absorption 216 nm, flow rate 1 mL/ min

    t=0, 0% B

    t=2, 0% B

    t=22, 80% B

    % Yield = 23%


    F3



    [0090] The peptide was synthesised on Fmoc-Ser(tBu)-Wang LL resin (150mg) from Merck Chemicals. A Protein Technologies Symphony Automated synthesiser was used to add the remaining amino acid residues. All coupling reactions were 10 minute double couplings with HBTU coupling agent. The peptide was cleaved in 100% Trifluoroacetic Acid in the presence of a scavenger mix containing 1:1 v/v Triisopropylsilane:Water. The peptide was purified upon a LC-ABZ+ (Supelcosil) column, 5 micron particle size, 110 Angstrom pore size, 250 mm x 10 mm, from Supelco. Analysis was performed with the same buffers, but using a C-18, 3.5 micron, 90 angstrom pore size, 4.6mm X 150 mm column from agilent. The run conditions were:

    absorption 216 nm, flow rate 1 mL/ min

    t=0, 0% B

    t=2, 0% B

    t=22, 80% B

    % Yield = 34%


    Example 1


    Effect of peptide fragments F1, F2 and F3 in a lipopolysaccharide (LPS) model of non-allergic inflammation


    Aim



    [0091] The purpose of these experiments was to determine whether the peptides F1, F2 and F3 are capable of inhibiting neutrophil recruitment in an in vivo system.

    Method



    [0092] Female Balb/c mice (Charles River, UK) were pre-treated intranasally with F1, F2 or F3 (5pg or 0.5pg). After 15 minutes, LPS (25 µg) was administered intranasally. Neutrophil influx in the lungs was determined by bronchoalveolar lavage 4 hours later.

    Results



    [0093] The results obtained are summarised in Figure 3. LPS induced a dose dependent increase in neutrophilia compared to control. A significant decrease in neutrophil migration to the lung was observed for all peptides F1 - F3 at both dosage concentrations.

    Conclusions



    [0094] The peptides of the present invention (F1 - F3) have been shown to inhibit neutrophil cell migration in vivo. The observed biological activity is indicative of a direct anti-inflammatory effect in non-allergic inflammation.

    SEQUENCE LISTING



    [0095] 

    <110> Peptinnovate Limited

    <120> Novel Peptides

    <130> P045177PCT

    <150> GB 1118201.1
    <151> 2011-10-21

    <160> 19

    <170> PatentIn version 3.5

    <210> 1
    <211> 31
    <212> PRT
    <213> Mycobacterium tuberculosis

    <400> 1

    <210> 2
    <211> 16
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of Peptide 4)

    <400> 2

    <210> 3
    <211> 15
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of Peptide 4)

    <400> 3

    <210> 4
    <211> 15
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of Peptide 4)

    <400> 4

    <210> 5
    <211> 10
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of Peptide 4)

    <400> 5

    <210> 6
    <211> 10
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of Peptide 4)

    <400> 6

    <210> 7
    <211> 11
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of Peptide 4)

    <400> 7

    <210> 8
    <211> 29
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 8



    <210> 9
    <211> 21
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 9

    <210> 10
    <211> 33
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 10

    <210> 11
    <211> 28
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 11

    <210> 12
    <211> 25
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 12

    <210> 13
    <211> 27
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 13

    <210> 14
    <211> 28
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 14

    <210> 15
    <211> 31
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 15

    <210> 16
    <211> 25
    <212> PRT
    <213> Artificial Sequence

    <220>
    <223> Peptide (fragment of MtCpn60.1)

    <400> 16

    <210> 17
    <211> 1620
    <212> DNA
    <213> Mycobacterium tuberculosis

    <220>
    <221> CDS
    <222> (1)..(1620)

    <400> 17





    <210> 18
    <211> 539
    <212> PRT
    <213> Mycobacterium tuberculosis

    <400> 18







    <210> 19
    <211> 540
    <212> PRT
    <213> Mycobacterium tuberculosis

    <400> 19








    Claims

    1. An isolated or recombinant peptide molecule consisting of an amino acid sequence selected from the group:

    (i) DGSVVVNKVSELPAGH;

    (ii) GLNVNTLSYGDLAAD; and

    (iii) SELPAGHGLNVNTLS.


     
    2. An isolated or recombinant peptide molecule according to claim 1, consisting of an amino acid sequence DGSVVVNKVSELPAGH.
     
    3. An isolated or recombinant peptide molecule according to claim 1, consisting of an amino acid sequence GLNVNTLSYGDLAAD.
     
    4. An isolated or recombinant peptide molecule according to claim 1, consisting of an amino acid sequence SELPAGHGLNVNTLS.
     
    5. An isolated or recombinant nucleic acid molecule consisting of a polynucleotide sequence which encodes a peptide consisting of an amino acid sequence selected from the group:

    1. DGSVVVNKVSELPAGH;

    2. GLNVNTLSYGDLAAD; and

    3. SELPAGHGLNVNTLS.


     
    6. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine.
     
    7. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 6 wherein the use in medicine is the modulation of diapedesis in a human subject.
     
    8. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 6 wherein the use in medicine is the prevention and/or treatment of a condition, disease and/or disorder in a human subject which is associated with an increase in the flow of white blood cells across the endothelium.
     
    9. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 6 wherein the use in medicine is the treatment and/or prevention of acute and/or chronic inflammatory conditions, preferably for the treatment and/or prevention of chronic obstructive pulmonary disease.
     
    10. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 9 wherein the inflammatory condition is associated with eosinophilia and/or neutrophilia.
     
    11. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 9 wherein the inflammatory condition is selected from the group consisting of inflammation associated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine-induced lung injury, non-allergic asthma, inflammatory bowel disease, and Crohn's disease.
     
    12. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 6 wherein the use in medicine is the treatment and/or prevention of autoimmune disorders, preferably wherein the autoimmune disorder is selected from the group consisting of haemolytic anaemia, thrombocytopenia, pernicious anaemia, Addison's disease, autoimmune diabetes, insulin dependent diabetes mellitus, myasthenia gravis, rheumatoid arthritis, systemic lupus erythematosus, atherosclerosis, autoimmune encephalitis, connective tissue disease, multiple sclerosis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, graft-versus-host disease and autoimmune inflammatory eye disease.
     
    13. A peptide according to any of claims 1 to 4 or a nucleic acid molecule according to claim 5 for use in medicine according to claim 6 wherein the use in medicine is the treatment and/or prevention of allergic disorders, preferably wherein the allergic disorder is selected from the group consisting of eczema, dermatitis, allergic rhinitis (hay fever), allergic airways diseases, hyper-eosinophilic syndrome, contact dermatitis; respiratory diseases characterized by eosinophilic airway inflammation and airway hyper-responsiveness, such as asthma, including allergic asthma and intrinsic asthma, allergic bronchopulmonary aspergillosis, eosinophilic pneumonia, allergic bronchitis bronchiectasis, occupational asthma, reactive airway disease syndrome, interstitial lung disease, hyper-eosinophilic syndrome, parasitic lung disease; anaphylaxis, serum sickness, drug reactions, food allergies, insect venom allergies, mastocytosis, hypersensitivity pneumonitis, urticaria, angioedema, eczema, atopic dermatitis, allergic contact dermatitis, erythema multiforme, Stevens-Johnson syndrome, allergic conjunctivitis, atopic keratoconjunctivitis, vernal keratoconjunctivitis and giant papillary conjunctivitis.
     
    14. A pharmaceutical composition comprising a peptide molecule according to claim 1 or a nucleic acid molecule according to claim 5 and a pharmaceutically-acceptable excipient.
     
    15. An adjuvant system comprising (i) a peptide molecule according to claim 1 and/or a nucleic acid molecule according to claim 5 and (ii) an antigen.
     


    Ansprüche

    1. Isoliertes oder rekombinantes Peptidmolekül, bestehend aus einer Aminosäuresequenz ausgewählt aus der Gruppe:

    (i) DGSVVVNKVSELPAGH;

    (ii) GLNVNTLSYGDLAAD; und

    (iii) SELPAGHGLNVNLTS.


     
    2. Isoliertes oder rekombinantes Peptidmolekül nach Anspruch 1, bestehend aus einer Aminosäuresequenz DGSVVVNKVSELPAGH.
     
    3. Isoliertes oder rekombinantes Peptidmolekül nach Anspruch 1, bestehend aus einer Aminosäuresequenz GLNVNTLSYGDLAAD.
     
    4. Isoliertes oder rekombinantes Peptidmolekül nach Anspruch 1, bestehend aus einer Aminosäuresequenz SELPAGHGLNVNLTS.
     
    5. Isoliertes oder rekombinantes Nukleinsäuremolekül, bestehend aus einer Polynukleotidsequenz, die ein Peptid codiert, das aus einer Aminosäuresequenz ausgewählt aus der Gruppe:

    1. DGSVVVNKVSELPAGH;

    2. GLNVNTLSYGDLAAD; und

    3. SELPAGHGLNVNLTS

    besteht.
     
    6. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung.
     
    7. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 6, wobei es sich bei der medizinischen Verwendung um die Modulation von Diapedese bei einem menschlichen Individuum handelt.
     
    8. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 6, wobei es sich bei der medizinischen Verwendung um die Vorbeugung und/oder Behandlung eines Leidens, einer Krankheit und/oder einer Störung in Zusammenhang mit einem Anstieg des Flusses weißer Blutzellen durch das Endothel bei einem menschlichen Individuum handelt.
     
    9. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 6, wobei es sich bei der medizinischen Verwendung um die Behandlung und/oder Vorbeugung von akuten und/oder chronischen Entzündungsleiden handelt, vorzugsweise für die Behandlung und/oder Vorbeugung von chronisch obstruktiver Lungenerkrankung.
     
    10. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 9, wobei das Entzündungsleiden in Zusammenhang mit Eosinophilie und/oder Neutrophilie steht.
     
    11. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 9, wobei das Entzündungsleiden ausgewählt ist aus der Gruppe bestehend aus Entzündung in Zusammenhang mit Infektion (z. B. septischer Schock, Sepsis oder SIRS (Systemic Inflammatory Response Syndrome)), Ischämie-Reperfusionsschaden, Endotoxinletalität, Arthritis, komplementvermittelter hyperakuter Abstoßung, Nephritis, cytokin- oder chemokininduziertem Lungenschaden, nichtallergischem Asthma, entzündlicher Darmerkrankung und Morbus Crohn.
     
    12. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 6, wobei es sich bei der medizinischen Verwendung um die Behandlung und/oder Vorbeugung von Autoimmunkrankheiten handelt, vorzugsweise wobei die Autoimmunkrankheit ausgewählt ist aus der Gruppe bestehend aus hämolytischer Anämie, Thrombozytopenie, perniziöser Anämie, Morbus Addison, Autoimmundiabetes, insulinabhängigem Diabetes mellitus, Myasthenia gravis, rheumatoider Arthritis, systemischem Lupus erythematodes, Atherosklerose, Autoimmunenzephalitis, Bindegewebekrankheit, multipler Sklerose, Autoimmun-Lungenentzündung, Guillain-Barre-Syndrom, Autoimmunthyreopathie, Graft-versus-Host-Krankheit und Autoimmun-Augenentzündung.
     
    13. Peptid nach einem der Ansprüche 1 bis 4 oder Nukleinsäuremolekül nach Anspruch 5 zur medizinischen Verwendung nach Anspruch 6, wobei es sich bei der medizinischen Verwendung um die Behandlung und/oder Vorbeugung von allergischen Erkrankungen handelt, vorzugsweise wobei die allergische Erkrankung ausgewählt ist aus der Gruppe bestehend aus Ekzem, Dermatitis, allergischer Rhinitis (Heuschnupfen), allergischen Erkrankungen der Luftwege, Hypereosinophilie-Syndrom, Kontaktdermatitis; durch eosinophile Entzündung und Überreaktivität der Luftwege gekennzeichneten Atemwegskrankheiten wie Asthma, einschließlich allergischem Asthma und intrinsischem Asthma, allergischer bronchopulmonaler Aspergillose, eosinophiler Lungenentzündung, allergischer Bronchitis Bronchiektasie, berufsbedingtem Asthma, RADS (Reactive Airway Disease Syndrome), interstitieller Lungenerkrankung, Hypereosinophilie-Syndrom, parasitärer Lungenerkrankung; Anaphylaxie, Serumkrankheit, Arzneistoffreaktionen, Lebensmittelallergien, Insektengiftallergien, Mastozytose, Hypersensitivitätspneumonitis, Urtikaria, Angioödem, Ekzem, atopischer Dermatitis, allergischer Kontaktdermatitis, Erythema multiforme, Stevens-Johnson-Syndrom, allergischer Konjunktivitis, atopischer Keratokonjunktivitis, Frühlingskeratokonjunktivitis und Riesenpapillenkonjunktivitis.
     
    14. Pharmazeutische Zusammensetzung, umfassend ein Peptidmolekül nach Anspruch 1 oder ein Nukleinsäuremolekül nach Anspruch 5 und einen pharmazeutisch unbedenklichen Exzipienten.
     
    15. Adjuvanssystem, umfassend (i) ein Peptidmolekül nach Anspruch 1 und/oder ein Nukleinsäuremolekül nach Anspruch 5 und (ii) ein Antigen.
     


    Revendications

    1. Molécule de peptide isolé ou recombiné, constitué d'une séquence d'acides aminés choisie dans le groupe constitué par :

    (i) DGSVVVNKVSELPAGH ;

    (ii) GLNVNTLSYGDLAAD ; et

    (iii) SELPAGHGLNVNTLS.


     
    2. Molécule de peptide isolé ou recombiné selon la revendication 1, constitué d'une séquence d'acides aminés DGSVVVNKVSELPAGH.
     
    3. Molécule de peptide isolé ou recombiné selon la revendication 1, constitué d'une séquence d'acides aminés GLNVNTLSYGDLAAD.
     
    4. Molécule de peptide isolé ou recombiné selon la revendication 1, constitué d'une séquence d'acides aminés SELPAGHGLNVNTLS.
     
    5. Molécule d'acide nucléique isolé ou recombiné, constitué d'une séquence polynucléotidique codant pour un peptide constitué d'une séquence d'acides aminés choisie dans le groupe constitué par :

    1. DGSVVVNKVSELPAGH ;

    2. GLNVNTLSYGDLAAD ; et

    3. SELPAGHGLNVNTLS.


     
    6. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine.
     
    7. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 6, où l'utilisation en médecine consiste en la modulation de la diapédèse chez un sujet humain.
     
    8. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 6, où l'utilisation en médecine consiste en la prévention et/ou le traitement d'une condition, d'une maladie et/ou d'un trouble chez un sujet humain, qui est associé (e) à une augmentation du flux de leucocytes à travers l'endothélium.
     
    9. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 6, où l'utilisation en médecine consiste en le traitement et/ou la prévention de conditions inflammatoires aiguës et/ou chroniques, préférablement le traitement et/ou la prévention de la bronchopneumopathie chronique obstructive.
     
    10. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 9, où la condition inflammatoire est associée à une éosinophilie et/ou une neutrophilie.
     
    11. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 9, où la condition inflammatoire est choisie dans le groupe constitué par une inflammation associée à une infection (telle qu'un choc septique, une sepsie ou un syndrome de réponse inflammatoire systémique (SRIS)), les lésions d'ischémie-reperfusion, une létalité des endotoxines, l'arthrite, le rejet hyper-aigu médié par le complément, la néphrite, les lésions pulmonaires induites par des cytokines ou des chémokines, l'asthme non allergique, l'affection abdominale inflammatoire, et la maladie de Crohn.
     
    12. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 6, où l'utilisation en médecine consiste en le traitement et/ou la prévention de troubles auto-immuns, préférablement où le trouble auto-immun est choisi dans le groupe constitué par l'anémie hémolytique, la thrombocytopénie, l'anémie pernicieuse, la maladie d'Addison, le diabète auto-immun, le diabète sucré insulinodépendant, la myasthénie gravis, la polyarthrite rhumatoïde, le lupus érythémateux disséminé, l'athérosclérose, l'encéphalite auto-immune, la maladie du tissu conjonctif, la sclérose en plaques, une inflammation pulmonaire auto-immune, le syndrome de Guillain-Barré, la thyroïdite auto-immune, la réaction de greffe contre hôte, et une maladie oculaire inflammatoire auto-immune.
     
    13. Peptide selon l'une quelconque des revendications 1 à 4, ou molécule d'acide nucléique selon la revendication 5, pour une utilisation en médecine selon la revendication 6, où l'utilisation en médecine consiste en le traitement et/ou la prévention de troubles allergiques, préférablement où le trouble allergique est choisi dans le groupe constitué par l'eczéma, la dermatite, la rhinite allergique (rhume des foins), les maladies allergiques des voies respiratoires, le syndrome hyper-éosinophilique, la dermatite de contact ; les maladies respiratoires caractérisées par une inflammation des voies respiratoires éosinophilique et une hyperréactivité des voies respiratoires, telles que l'asthme, y compris l'asthme allergique et l'asthme intrinsèque, l'aspergillose bronchopulmonaire allergique, la pneumonie éosinophilique, la bronchite allergique, la bronchiectasie, l'asthme professionnel, le syndrome des voies respiratoires réactives, la maladie pulmonaire interstitielle, le syndrome hyper-éosinophilique, une maladie pulmonaire parasitaire ; l'anaphylaxie, la maladie du sérum, les réactions aux médicaments, les allergies alimentaires, les allergies au venin d'insectes, la mastocytose, le pneumopathie d'hypersensibilité, l'urticaire, l'angio-œdème, l'eczéma, la dermatite atopique, la dermatite allergique de contact, l'érythème polymorphe, le syndrome de Stevens-Johnson, la conjonctivite allergique, la kératoconjonctivite atopique, la kératoconjonctivite vernale, et la conjonctivite giganto-papillaire.
     
    14. Composition pharmaceutique comprenant une molécule de peptide selon la revendication 1 ou une molécule d'acide nucléique selon la revendication 5, et un excipient pharmaceutiquement acceptable.
     
    15. Système d'adjuvant, comprenant (i) une molécule de peptide selon la revendication 1 et/ou une molécule d'acide nucléique selon la revendication 5, et (ii) un antigène.
     




    Drawing




















    Cited references

    REFERENCES CITED IN THE DESCRIPTION



    This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

    Patent documents cited in the description




    Non-patent literature cited in the description