WO2015131053A1 - Polypeptides derived from phl p and methods and uses thereof for immune response modulation - Google Patents

Polypeptides derived from phl p and methods and uses thereof for immune response modulation Download PDF

Info

Publication number
WO2015131053A1
WO2015131053A1 PCT/US2015/018026 US2015018026W WO2015131053A1 WO 2015131053 A1 WO2015131053 A1 WO 2015131053A1 US 2015018026 W US2015018026 W US 2015018026W WO 2015131053 A1 WO2015131053 A1 WO 2015131053A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
nos
pollen
amino acid
molecule
Prior art date
Application number
PCT/US2015/018026
Other languages
French (fr)
Inventor
Ilka HOOF
Lars Harder CHRISTENSEN
Peters BJOERN
Jason GREENBAUM
Original Assignee
Alk-Abelló A/S
La Jolla Institute For Allergy And Immunology
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 Alk-Abelló A/S, La Jolla Institute For Allergy And Immunology filed Critical Alk-Abelló A/S
Publication of WO2015131053A1 publication Critical patent/WO2015131053A1/en

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
    • A61K39/35Allergens
    • A61K39/36Allergens from pollen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/577Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 tolerising response

Definitions

  • the invention relates to pan pollen immunogenic molecules such as polypeptides, proteins and peptides, and methods and uses of such immunogenic molecules for modulating or relieving an immune response in a subject, such as treating a subject for an allergic immune response or inducing or promoting immunological tolerance to the immunogenic molecule or a pollen allergen in a subject.
  • allergen-specific T-cells play an important role in allergic inflammation and that induction of antigen specific T regulatory cells (Tregs) or elimination of allergen-specific T helper type 2 cells (Th2) might be a prerequisite for the induction of specific tolerance. Yet, cross-reactivity among multiple pollen families at the T-cell level is less explored.
  • Allergen-specific immunotherapy SIT is a hyposensitizing immunotherapy introduced in clinical medicine almost a century ago for the treatment of an allergic immune response using the allergens that the subject is sensitized to.
  • An allergic immune response may be mediated by activated allergen-specific Th2 cells, which produce cytokines such as IL-4, IL-5, and IL-13.
  • the allergen-specific T-cell response is mediated predominantly by Th1 cells.
  • SIT may reduce the ratio of Th2:Th1 cells and may alter the cytokine profile, reducing the production of IL-4, IL-5, and IL-13 and increasing the production of IFN-gamma in response to major allergens or allergen extracts.
  • SIT has several limitations, including safety concerns about giving patients allergenic substances. Because most SIT regimens involve the administration of whole, unfractionated, allergen extracts, adverse IgE-mediated events are a considerable risk. Significant efforts have been devoted to developing approaches to modulate allergen-specific T-cell responses without inducing IgE-meditated, immediate-type reactions. These approaches include developing hypoallergens that do not contain IgE- binding epitopes, allergens that are coupled to adjuvants and carriers of bacterial or viral origin or peptides that contain dominant T-cell epitopes and do not react with IgE in allergic individuals.
  • NTGA novel Timothy Grass antigens
  • NTGA's are unrelated to the known allergens of Timothy grass, which mainly are identified based on their high IgE reactivity.
  • International patent application, WO2013/119863 A1 relates to novel antigens (NTGA's) derived from Timothy grass pollen.
  • an immunogenic molecule derived from an allergenic pollen source is able to reduce an allergic immune response caused by an unrelated allergen of the same pollen source via bystander suppression.
  • immunogenic molecules with high sequence conservation/homology to polypeptides identified in several different pollen families.
  • Such immunogenic molecules have potential therapeutical utilization against immune responses triggered by pollen allergens of a broad array of pollen families for example via induction of bystander tolerance towards the offending pollen allergens.
  • immunogenic molecules in short immunogens
  • pan- pollen immunogens which have been detected in Timothy grass pollen (Phleum pratense (in short Phi p) and in at least one non-grass pollen species, which consist of or contain as part of their sequence an amino acid sequence conserved across the Phi p grass pollen species and at least one of the non-grass pollen species investigated.
  • a molecule e.g. an immunogenic molecule
  • a molecule of the invention may consist of or contain as part of its sequence a conserved amino acid sequence detected in several different pollen families, e.g. in a grass pollen species (e.g.
  • Phleum Pratense (Phi p) and at least one of the following non-grass pollen species: Ambrosia psilostachya (Amb p), Ambrosia artemisiifolia, (Amb a), Quercus alba (Que a) and Betula verrucosa, (Bet v).
  • Table 1 shows examples of amino acid sequences of "pan pollen” polypeptides detected and identified in Phi p grass pollen and Table 2 shows amino acid sequences of polypeptides homologous to the polypeptides of Table 1 , which is found in pollen of the species Cyn d, Amb a, Amb p, Que a, and Bet v. As those polypeptides can be detected in pollen, they are referred to herein as wild type polypeptides.
  • GWT Garnier-Weed-Tree
  • Table 3 shows GWT conserved regions identified in the Phi p sequences of Table 1.
  • Table 4 shows the corresponding GWT conserved regions identified in the other pollen species investigated.
  • a conserved region as used herein is defined as the region identified from merging overlapping conserved 15mer peptides in a Phi p sequence, wherein a conserved 15mer peptide is defined as a 15 amino acid peptide containing 0, 1 , or 2 mismatches to a 15mer peptide sequence of a homologous polypeptide in a weed pollen species (Amb a and/or Amb) and in a tree pollen species (Que a and/or Bet v).
  • conserved amino acid sequences of the polypeptides of Table 1 and 2 that are conserved across at least a set of homologous polypeptides found in Phi p grass pollen and in at least one weed pollen species (Amb a or Amp), but not in a tree pollen species (Que a or Bet v).
  • Such sequences are herein named GW (Grass-Weed) conserved stretches of amino acid residues (or alternatively GW conserved regions).
  • Table 5 shows GW conserved regions identified in the Phi p sequences of Table 1.
  • Table 6 shows the corresponding GW conserved regions identified inthe other pollen species.
  • conserved amino acid sequences of the polypeptides of Table 1 and 2 that are conserved across at least a set of homologous polypeptides found in Phi p grass pollen and in at least one tree pollen species (Que a or Bet v), but not in a weed pollen species (Amb a or Amp).
  • Such sequences are herein named GT (Grass- Tree) conserved stretches of amino acid residues (or alternatively GT conserved regions).
  • Table 7 shows GW conserved regions identified in the Phi p sequences of Table 1.
  • Table 8 shows the corresponding GT conserved regions identifiedin the other pollen species.
  • WT Wood-Tree
  • Table 9 shows WT conserved regions identified in polypeptides of Table 2.
  • the GWT, GW, GT or WT conserved sequences may be an immunogenic molecule in itself, or may give rise to additional immunogens comprising the conserved sequences or subsequences thereof.
  • a molecule e.g. an immunogenic molecule
  • an immunogenic molecule of the invention which is identified in Phi p pollen may be able to modify a T cell response in a grass allergic donor as well as in donors allergic to a weed pollen species and/or tree pollen species. This is evidenced using Phi p allergens and a selection of recently identified Phi p antigens as model immunogens (Example 8). It was found that a T cell response of grass allergic donors to a first immunogenic molecule derived from Phi p may also be activated in detectable levels by a second immunogenic molecule containing few mismatches to the first immunogenic molecule, e.g. preferably no more than 2 or 3 mismatches.
  • the presently provided conserved sequences shown in Tables 3 to 8 may be able to induce a T cell response in donors not only sensitized to Phi p pollen allergens, but to allergens of any of the pollen species Cyn d, Amb a, Amb p, Que a and Bet v as well as other pollen species containing polypeptides with similar conserved sequences comprising less than 2 or 3 mismatches to the Phi p sequence in question.
  • the invention relates in a first aspect to a molecule, such as an immunogenic molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293 set out in Table 3; SEQ ID NOS: 617-676, 294-616 and 677-767 set out in Table 4; SEQ ID NOS: 768-808 set out in Table 5; SEQ ID NOS: 809-951 set out in Table 6; SEQ ID NOS: 952-1023 set out in Table 7; SEQ ID NOS: 1024-1231 set out in Table 8 and SEQ ID NOS: 1232-1473 set out in Table 9; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NO
  • the invention also relates to the use of said molecule (e.g. said immunogenic molecule) as a medicament, in particularly for use in modulating an immune response, e.g. relieving an immune response, triggered by pollen (i.e. a pollen allergen and/or an immunogenic molecule disclosed herein) in a subject.
  • pollen i.e. a pollen allergen and/or an immunogenic molecule disclosed herein
  • the invention relates to a method for modulating an immune response, e.g. for relieving an immune response (e.g. triggered by pollen or an immunogen present in pollen), in a subject in need thereof, comprising administering an effective amount of a molecule (e.g. an immunogenic molecule) disclosed herein.
  • the invention relates to an immunogenic molecule as disclosed herein for use in modulating an immune response, e.g. for relieving an immune response, in a subject in need thereof, comprising administering an effective amount of a molecule (e.g. an immunogenic molecule) disclosed herein.
  • a molecule e.g. an immunogenic molecule
  • the invention also relates to the use of a molecule (e.g. an immunogenic molecule) as disclosed herein as a medicament, e.g. use of an immunogenic molecule as disclosed herein for the preparation of a medicament for modulating an immune response, e.g. relieving an immune response, in a subject in need thereof.
  • a molecule e.g. an immunogenic molecule
  • an immunogenic molecule as disclosed herein for the preparation of a medicament for modulating an immune response, e.g. relieving an immune response, in a subject in need thereof.
  • the immune response in question may be triggered by pollen, e.g. by pollen of one or more pollen species, pollen genera or pollen families. It follows that the immune response in question may be triggered by one or more pollen allergens from one or more pollen species, pollen genera or pollen families including one or more molecules (e.g.
  • immunogenic molecules disclosed herein, for example the immunogenic molecule itself or a subsequence thereof.
  • a polypeptide of option d) comprises an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 27-30 set out in Table 1 and SEQ ID NOS: 161-172 set out in Table 2;
  • a polypeptide of option a) comprises an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4;
  • a polypeptide of option b) comprises an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS
  • the molecule may comprise a combination of two or more polypeptides of option a), option b), option c) or option d), so as to construct molecules with desirable properties e.g. immunogenic molecules with high conservation throughout the entire amino acid sequence.
  • a polypeptide of option a) may contain as part of its sequence a combination of two or more amino acid sequences having at least 65% similarity or identity to a sequence selected from any one of SEQ ID NOS: 212-293 set out in Table 3 and SEQ ID NOS: 294-767 set out in Table 4.
  • a polypeptide of option b) and c) may comprise as part of its sequence a combination of two or more of said subsequences.
  • a cell expresses said molecule.
  • a cell is a eukaryotic or prokaryotic cell and may be a mammalian, insect, fungal or bacterium cell.
  • a molecule (e.g. an immunogenic molecule) of the present invention is suitable as a reagent, for example in immunotherapy against various pollen allergies in a subject.
  • nucleic acid molecules encoding a polypeptide of option a), b), c) or d) or a molecule comprising a polypeptide of option a), b), c) or d).
  • compositions for example pharmaceutical compositions comprising an immunogenic molecule of the invention.
  • a pharmaceutical composition is suitable for immunotherapy (e.g. treatment,
  • a pharmaceutical composition is a vaccine, e.g.
  • FIG. 1 Sensitization pattern of an immunogen of the invention (A0349): It is shown that the in vitro T-cell response towards A0349 is much weaker compared to the response to allergen Phi p 5.
  • FIG. 3 Tolerance induction investigated in mice shows that prophylactic sublingual immunotherapy treatment (SLIT) with A0349 in mice is capable of inducing tolerance towards Phi p extract as shown by the ability of A0349 to reduce the
  • an immunogenic molecule that is weaker than Phi p 5, the major allergen in Phi p extract, is able to reduce an immune response triggered by Phi p extract.
  • FIG 4 Bystander tolerance induction investigated in mice.
  • prophylactic SLIT treatment with A0349 could induce tolerance toward the allergen Phi p 5, which amino acid sequence is quite different from A0349.
  • Phi p 5 an unrelated antigen
  • FIG. 4A Treatment with A0349 resulted in the suppression of an immune response caused by an unrelated antigen, i.e. Phi p 5, possibly via bystander mechanisms, where the tolerance induction towards one immunogen (e.g. A0349) result in suppression of an immune response caused by another unrelated immunogen (e.g. Phi p 5).
  • a or “an” refers to an indefinite number and shall not only be interpreted as “one” but also may be interpreted to mean “some", “several” or one or more.
  • conserved sequence is in the present context meant to include that a given amino acid sequence of one pollen species contains at least 15 contiguous amino acids within the sequence that has less than 3 mismatches compared to an amino acid sequence of 15 amino acid residues from another pollen species. Longer stretches of conserved sequences may contain several stretches of at least 15 contiguous amino acids having less than 3 mismatches compared to another sequence of 15 amino acids from another pollen species, genera or family.
  • the conserved region is usually determined by multiple sequence alignments of three or more homologous pollen polypeptides.
  • polypeptide polypeptide
  • protein protein
  • peptide may be used interchangeably and mean any peptide-linked chain of amino acids regardless of post-translational modification. While polypeptides can be any length, the use of the term “protein” generally means longer polypeptides of at least 100 amino acid residues, whereas the term “peptide” generally means polypeptides shorter than 30 amino acid residues.
  • mismatch is meant to include any substitution of an amino acid residue within the 15mer peptide. Optionally, a mismatch may be a deletion or an addition of an amino acid residue within the 15mer peptide.
  • homologous polypeptides refer to a pollen polypeptide derived from a common ancestor and are typically, although not necessarily, polypeptides having one or more similar functions.
  • homologous pollen polypeptides will have a degree of sequence identity to each other across the entire length of each protein that is at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or greater.
  • homologous pollen polypeptides are found in pollens of different species, genera and families.
  • immunogenic molecule e.g. an allergen or an antigen
  • an immunogenic molecule e.g. an allergen or an antigen
  • the individual's adaptive immune system displays memory to the immunogenic molecule, for example that the immunogenic molecule has induced detectable IgE antibodies against the immunogenic molecule and thus qualifies as an IgE-reactive antigen
  • T-cells stimulated in vitro are able to proliferate under the presence of the immunogenic molecule or fragments of the immunogenic molecule (e.g. linear peptides derived from the immunogenic molecule).
  • allergic immune response is meant to encompass a hypersensitivity immune response, e.g. a type 1 hypersensitivity immune response, such as typically an immune response that is associated with the production of IgE antibodies (i.e. IgE-mediated immune response) and/or production of cytokines usually produced by Th2 cells.
  • An allergic immune response may be associated with an allergic disease, for example atopic dermatitis, urticaria, contact dermatitis, allergic conjunctivitis, allergic rhinitis, allergic asthma, anaphylaxis, food allergy and hay fever.
  • grass pollen is meant to designate pollen of the plant family Poaceae, for example pollen of the plant genus Anthoxanthum, Cynodon, Dactylis, Festuca, Holcus, Hordeum, Lolium, Oryza, Paspalum, Phalaris, Phleum, Poa, Secale, Sorghum, Triticum or Zea.
  • an "immunogenic molecule” refers to a substance, including but not limited to a molecule comprising as part of its structure or exclusively a protein, a polypeptide or a peptide, which optionally modifies, e.g. elicits, induces, stimulates, promotes enhances or decreases, reduces, inhibits, suppresses, relieves an immune response when administered to a subject, for example laboratory mice or modifies, e.g. elicits, induces, stimulates, promotes enhances or decreases, reduces, inhibits, suppresses or relieves a T cell response in vitro in response to the immunogenic molecule.
  • an immunogenic molecule may induce tolerance to itself in a subject.
  • An immune response elicited by an immunogenic molecule may include, but is not limited to, a B cell or a T cell response.
  • An immune response can include a cellular response with a particular pattern of lymphokine/cytokine production (e.g., Th1 , Th2), a humoral response (e.g., antibody production, like IgE, IgG or IgA), or a combination thereof, to a particular immunogenic molecule.
  • an antigen refers to a particular substance to which an immunoglobulin (Ig) isotype may be produced in response to the substance.
  • an immunoglobulin (Ig) refers to an antigen that induces an IgG antibody response.
  • an "IgE antigen” refers to an antigen that induces an IgE antibody response (and thus qualifies as an allergen);
  • an "IgA antigen” refers to a substance that induces an IgA antibody response, and so forth.
  • such an immunoglobulin (Ig) isotype produced in response to an antigen may also elicit production of other isotypes.
  • an IgG antigen may induce an IgG antibody response in combination with one more of an IgE, IgA, IgM or IgD antibody response.
  • an IgG antigen may induce an IgG antibody response without inducing a response to one more of an IgE, IgA, IgM or IgD antibody response.
  • an IgG antigen may induce an IgG antibody response without inducing an IgE, IgA, IgM or IgD antibody response.
  • allergen refers to a particular type of a substance that can elicit production of IgE antibodies, such as in predisposed subjects. For example, if a subject previously exposed to an allergen (i.e.
  • allergic asthma may develop due to a Th2 response characterized by an increased production of type 2 cytokines (e.g., IL-4, IL-5, IL-9, and/or IL-13) secreted by CD4+ T lymphocytes.
  • type 2 cytokines e.g., IL-4, IL-5, IL-9, and/or IL-13
  • the term "immunotherapy” is meant to encompass treatment of a disease by inducing, enhancing, or suppressing an immune response.
  • the therapeutically active agent is an immunogenic molecule, particularly an antigen, more particularly an allergen.
  • An immunogenic molecule may be a protein or a fragment thereof (e.g. immunogenic peptide including a linear peptide).
  • Immunotherapy in connection with allergy usually encompasses repeated administration of a sufficient dose of the immunogenic molecule/antigen/allergen, usually in microgram quantities, over a prolonged period of time, usually for more than 3 months, 6 months, 1 year, such as 2 or 3 years, during which period the immunogenic molecule may be administered daily or less frequently, such as several times a week, weekly, bi-weekly, or monthly, every second month or quarterly.
  • Immunotherapy can be effected by specific immunotherapy or may be effected by bystander tolerance induction.
  • specific immunotherapy in connection with allergy is meant to designate that immunotherapy is conducted with the administration of an immunogenic molecule to which the subject is sensitized to, particularly an immunogenic molecule to which the subject has raised specific IgE antibodies to, e.g. major allergens.
  • immunological tolerance refers to a) a decreased or reduced level of a specific immunological response (thought to be mediated at least in part by antigen-specific effector T lymphocytes, B lymphocytes, antibodies or a combination thereof); b) a delay in the onset or progression of a specific immunological response; or c) a reduced risk of the onset or progression of a specific immunological response to an immunogenic molecule, such as an antigen or an allergen.
  • Specific immunological tolerance occurs when tolerance is preferentially invoked against certain immunogens in comparison with other immunogens. Tolerance is an active immunogenic molecule dependent process and differs from non-specific immunosuppression and immunodeficiency.
  • an immunogenic molecule that elicits, induces, stimulates, promotes enhances or decreases, reduces, inhibits, suppresses, relieves an immune response against another unrelated immunogenic molecule, for example an allergen, e.g. major allergens of pollen.
  • an immunogenic molecule may induce immunological tolerance to itself, and may be able to reactivate T regulatory cells specific to the immunogenic molecule to down-regulate an immune response caused by another unrelated immunogenic molecule, e.g. an allergen.
  • an immunogenic molecule may induce immunological tolerance to another unrelated antigen, e.g. an allergen including a pollen allergen described herein.
  • one immunogenic molecule may provide bystander tolerance induction to another unrelated antigen, e.g. the immunogenic molecule may provide suppression of an immune response triggered by an unrelated antigen (e.g. allergen) via bystander mechanisms.
  • treatment refers to any type of treatment that conveys a benefit to a subject afflicted with allergy or at least sensitized to an allergen , including improvement in the condition of the subject (e.g., in one or more symptoms), delay in the onset of symptoms, slowing the progression of symptoms, or induce disease modification etc.
  • Typical symptoms of an allergic reaction are nasal symptoms, such as itchy nose, sneezing, runny nose, blocked nose; conjunctival symptoms, such asitchy eyes, red eyes, watery eyes; and respiratory symptoms, such as decreased lung function.
  • the treatment may also give the benefit that the patient needs less concomitant treatment with
  • treatment is not necessarily meant to imply cure or complete abolition of symptoms, but refers to any type of treatment that imparts a benefit to a patient.
  • Treatment may be initiated before the subject becomes sensitized to a protein. This may be realized by initiating immunotherapy before the subject has raised detectable serum IgE antibodies capable of binding specifically to the sensitizing protein or before any other biochemical marker indicative of an allergic immune response can be detected in biological samples isolated from the individual.
  • treatment may be initiated before the subject has evolved clinical symptoms of the allergic disease, such as symptoms of allergic rhinitis, allergic asthma or atopic dermatitis.
  • a therapeutically sufficient amount is meant to designate an amount effective to reduce, suppress, relieve or eliminate an allergic immune response, e.g. an amount sufficient to achieve the desirable reduction in clinical relevant symptoms or manifestations of the allergic immune response.
  • a therapeutically sufficient amount may be the accumulated dose of a polypeptide, a set of polypeptides administered during a course of immunotherapy in order to achieve the intended effect or it may be the maximal dose tolerated within a given period.
  • the total dose or accumulated dose may be divided into single doses administered daily, twice a week or more, weekly, every second or fourth week or monthly depending on the route of administration and the pharmaceutical formulation used.
  • the total dose or accumulated dose may vary. It is expected that a single dose is in the microgram range, such as in the range of 5 to 500 microgram dependent on the nature of the polypeptide.
  • Symptoms may be the clinically symptoms of allergic rhinitis, allergic asthma allergic conjunctivitis, atopic dermatitis, food allergy and/or hay fever.
  • the symptoms are the same as experienced with a flu/cold such as sneezing, itching, congestion, coughing, feeling of fatigue, sleepiness and body aches.
  • nasal symptoms may be itchy nose, sneezing, runny nose, blocked nose; conjunctival symptoms may be itchy eyes, red eyes, watery eyes; and respiratory symptoms may be decreased lung function.
  • a responder may also be evaluated by monitoring the patient's reduced need for concomitant treatment with corticosteroids or H1 antihistamines to suppress the clinical symptoms. Symptoms may be subjectively scored or in accordance with official guidelines used in clinical trials of SIT.
  • adjuvant refers to a substance that enhances the immune response to an immunogenic molecule. Depending on the nature of the adjuvant, it can promote either a cell-mediated immune response, humoral immune response or a mixture of the two.
  • an epitope refers to a region or part of an immunogenic molecule that elicits an immune response when administered to a subject.
  • an epitope is a T cell epitope, i.e., an epitope that elicits, stimulates, induces, promotes, increases or enhances a T cell activity, function or response.
  • An immunogenic molecule can be analyzed to determine whether it include at least one T cell epitope using any number of assays (e.g. T cell proliferation assays, lymphokine secretion assays, T cell non-responsiveness studies, etc.).
  • a T-cell epitope refers to an epitope that are MHC Class II binders (i.e. HLA-II binders), for example an epitope able to connect to/ associate with or bind to a HLA-II molecule shown in Tables 11 b or 11c.
  • MHC Class II binders i.e. HLA-II binders
  • immune response includes T cell (cellular) mediated and/or B cell (humoral) mediated immune responses, or both cellular and humoral responses.
  • exemplary immune responses include T cell responses, e.g., lymphokine production, cytokine production and cellular cytotoxicity.
  • T-cell responses include Th1 and/or Th2 responses.
  • immune response includes responses that are indirectly affected by T cell activation, e.g., antibody production (humoral responses) and activation of cytokine responsive cells, e.g., eosinophils, macrophages.
  • Immune cells involved in the immune response include lymphocytes, such as T cells (CD4+, CD8+, Th1 and Th2 cells, memory T cells) and B cells; antigen presenting cells (e.g., professional antigen presenting cells such as dendritic cells, macrophages, B lymphocytes, Langerhans cells, and non-professional antigen presenting cells such as keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes); natural killer (NK) cells; myeloid cells, such as macrophages, eosinophils, mast cells, basophils, and granulocytes.
  • lymphocytes such as T cells (CD4+, CD8+, Th1 and Th2 cells, memory T cells) and B cells
  • antigen presenting cells e.g., professional antigen presenting cells such as dendritic cells, macrophages, B lymphocytes, Langerhans cells, and non-professional antigen presenting cells such as
  • sequence means a fragment or part of a longer molecule, e.g. of a full length molecule (e.g. the wild type polypeptides shown in Tables 1 and 2) or of a conserved region thereof (e.g. GWT sequences shown in Tables 3 and 4).
  • subsequence therefore consists of one or more amino acids less than the wild type polypeptide or a conserved region thereof.
  • a molecule e.g. an immunogenic molecule of the invention comprises conserved amino acid sequences detected in a grass pollen species as well as in a weed pollen species and/or a tree pollen species.
  • such molecules can be used to broadly treat a subject with or at risk of developing an immune response to pollen of a variety of pollen species, genera or families, or to broadly induce or promote tolerance of a subject to pollen of a variety of pollen species, genera or families and may include promoting or inducing tolerance to the immunogenic molecule itself or another immunogenic molecule (e.g. an allergen) unrelated to the immunogenic molecule being administered, for example via induction of bystander tolerance towards an offending allergen.
  • an immunogenic molecule e.g. an allergen
  • a multisensitized subject e.g. a subject sensitized to both grass, weed and tree pollen
  • a molecule e.g. an immunogenic molecule
  • the molecule (e.g. the immunogenic molecule) comprises or consists of a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches compared to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 212-293 (e.g. SEQ ID NOS: 274, 270-273, 212-269 and 275-293) set out in Table 3 or SEQ ID NOS: 294-767 (e.g. SEQ ID NOS: 665-676, 617-664 and 677-767) set out in Table 4.
  • SEQ ID NOS: 212-293 e.g. SEQ ID NOS: 274, 270-273, 212-269 and 275-293
  • SEQ ID NOS: 294-767 e.g. SEQ ID NOS: 665-676, 617-664 and 677-767 set out in Table 4.
  • the molecule may comprise one or more T cell epitope(s) optionally a Th-2 cell epitope (i.e. the molecule may comprise one or more T cell epitope-containing amino acid sequence(s)).
  • the polypeptide of option a), option b), option c) and option d) comprises a T cell epitope, optionally a Th-2 cell epitope.
  • the GWT sequences (SEQ ID NOS:212-293 set out in Table 3 and SEQ ID NOS: 294-767 set out in Table 4), the GW sequences (SEQ ID NOS: 768-808 set out in Table 5 and SEQ ID NOS: 809-951 set out in Table 6), GT sequences (SEQ ID NOS: 952-1023 set out in Table 7 and SEQ ID NOS: 1024-1231 set out in Table 8) and WT sequences (SEQ ID NOS: 1232-1473 set out in Table 9) or subsequences thereof may comprise a T cell epitope, optionally a Th-2 cell epitope.
  • a subsequence of a polypeptide of option b) or option c) contains a T cell epitope, optionally a Th-2 cell epitope.
  • sequences selected from any of SEQ ID NOS: 1-37 e.g. SEQ ID NOS: 30, 1-29, 31-37 set out in Table 1
  • SEQ ID NOS: 38- 211 e.g SEQ ID NOS: 161-172, 38-160 and 173-211 set out in Table 2 contains a T cell epitope, optionally a Th-2 cell epitope.
  • immunogens of the present invention may be used in relieving an immune response against phi p grass pollen, non-phi p grass pollen as well as non-grass pollen to the extent that the pollen species contain polypeptides comprising essentially the same GWT, GW, GT or WT conserved region.
  • an immunogenic molecule of the present invention may at least be used in modulating, e.g. relieving, an immune response triggered by pollen of the grass pollen species Phi p and/or Cyn d as well as pollen of other species closely related to Phi p or Cyn d, e.g. pollen species also belonging to the plant family Poales, such as species of the plant genera selected from any of Anthoxanthum, Cynodon, Dactylis, Lolium, Phleum or Poa.
  • immunogenic molecule may also be used in modulating, e.g. relieving, an immune response triggered by pollen of the weed pollen species Amb a and/or Amb p and pollen of species closely related to Amb a and Amp p, e.g. pollen species also belonging to the plant family Asteraceae, such as species of the plant genera selected from any of Ambrosia, Artemisia, Helianthus.
  • the same immunogenic molecule may also be used in modulating, e.g. relieving, an immune response triggered by pollen of the tree pollen species Que a and/or Bet v and pollen of species closely related to Que a and/or Bet v, e.g.
  • pollen species also belonging to the plant families Betulaceae, Fagaceae and Oleaceae, such as species of the plant genera selected from any of Alnus, Betula, Carpinus, Castanea, Corylus, Fagus, Quercus, Fraxinus and Ligustrum.
  • Betulaceae Fagaceae and Oleaceae
  • species of the plant genera selected from any of Alnus, Betula, Carpinus, Castanea, Corylus, Fagus, Quercus, Fraxinus and Ligustrum.
  • a molecule e.g.
  • an immunogenic molecule of the present invention may be used in modulating, e.g., relieving, an immune response triggered by pollen of a plant genus selected from any of Phleum, Cynodon Ambrosia, Quercus and Betula.
  • conserved amino acid sequences disclosed herein have been detected in about five different pollen species and may be found in additional pollen species selected from any of the plant families Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, e.g. the plant genera Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea or Plantago using the methodology disclosed herein (see exemplary pollen species below):
  • conserved regions may be found in pollen of other weeds, grasses or tree species, e.g. found in one or more plant species selected from further plant families, like the plant family Chenopidiaceae including the plants Lambs quarters, Russian thistles and Kochias; plant family Amaranthaceae including Pigweeds, plant family Polygonaceae including for example Sheep sorrel, plant family Ulmacea including for example American Elm and Ralphberry, plant family Plantanaceae including for example Sycamore, plant family Salicaceae including for example White poplar and Cottonwood, plant family Aceraceae including for example Box elder and Red maple, plant family Cupressaceae including for example Common juniper and Cedar.
  • Non-limiting examples of species of the genus Ambrosia is Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida; a typical species of the genus Artemisia is Artemisia vulgaris; non-limiting examples of species of the genus Betula is Betula verrucosa; non-limiting examples of species of the genus Fagus is Fagus grandifolia or Fagus sylvatica; non-limiting examples of species of the genus Quercus is Quercus alba, non-limiting examples of species of the genus Fraxinus is Fraxinus excelsior; a typical species of the genus Olea is Olea Europaea, a typical species of the genus plantago is Plantago lanceolata, a typical species of the genus plantago is Plantago lanceolata, non- limiting examples of species of
  • a molecule of the invention may modulate an immune response at least triggered by a grass pollen and/or a weed pollen by administering to a subject in need thereof a molecule comprising a GWT conserved sequence set out in Tables 3 or 4 or a subsequence thereof or by administering to a subject in need thereof a molecule comprising a GW conserved sequence set out in Tables 5 or 6 or a
  • the molecule may modulate an immune response triggered by at least a grass pollen and/or a tree pollen by
  • the molecule modulate an immune response triggered by at least a grass pollen, a weed pollen and a tree pollen by administering to a subject in need thereof a molecule comprising a GWT conserved sequence set out in Tables 3 or 4 or a subsequence thereof.
  • a molecule of the invention may modulate, e.g. relieve, an immune response triggered by pollen.
  • Pollen may comprise allergens considered as major and minor allergens according to official guidelines in the art and may in addition comprise additional immunogens, for example a molecule as disclosed herein.
  • a molecule of the invention may be able to relieve an immune response triggered by a major and/or a minor pollen allergen and/or a molecule disclosed herein including the molecule itself.
  • Molecules (e.g. immunogens) eligible for relieving an immune response triggered by an allergen (e.g. a major allergen or minor allergen) unrelated to the molecule is thought, at least in part, to be mediated via induction of bystander tolerance, which mechanism requires, at least in part, co-existence of the triggering allergen and the unrelated molecule at the target organ for the immune response.
  • the co-existence may inherently be obtained by using molecules present in the same pollen source as the allergen.
  • the triggering allergen and the unrelated molecule may be released from the same pollen source within an overlapping period of time (co-release of triggering allergen and unrelated molecule (e,g. immunogenic molecule)) to ensure co-existence at the target organ.
  • Co-release can be examined in vitro using hydrated pollen as described in Example 4 herein.
  • the polypeptide of option a), b), c) or d) may be derived, be a part of or comprise a wild type polypeptide that co-releases/co-elutes from the same pollen source with the major or minor allergens the subject is sensitized to and to which allergens the immune response is sought to be relieved.
  • a molecule with the potential to relieve an immune response triggered by pollen of several different species, genera and families may be identified by investigating the presence of the molecule or a fragment thereof in pollen diffusates of pollen of different species, genera or families including the grass pollen, weed pollen or tree pollen species used for the present investigation within a time period overlapping with the relevant major and/or minor allergen.
  • co-release or “co-elute” may refer to when a molecule starts to release from pollen, e.g. hydrated pollen, within a period overlapping with a major allergen to which the allergic immune response is sought to be relieved.
  • Major allergens start to release from pollen within few minutes after hydration of pollen and continue to be released within the next 30 or 60 minutes.
  • co-release or “co-elute” may refer to when a molecule of the invention starts to release from pollen together with a major allergen, usually within 30 minutes after hydration of the pollen.
  • Co- release/co-elution may be determined by a method comprising extracting pollen in an aqueous solution having pH in the range of 6-8 for a period ranging from 1 to 200 minutes, optionally the aqueous solution comprises at least 60% of water, a buffering agent; a tonicity providing agent.
  • the aqueous solution has an ionic strength corresponding to that of isotonic saline (0.9 g of NaCI in 1 liter of water), e.g. the aqueous solution has an ionic strength in the range of 10 mM to 1000 mM and optionally the aqueous solution has a pH of about 7.
  • the period of extraction is in the range of 1-180, 1-120 minutes, 5-120 minutes, 5 to 90 minutes, for example 5-60 minutes, for example 5-45 minutes, for example 5-30 minutes.
  • some immunogenic molecules may be released from various distinct pollen species, such as from grass pollen species as well as weed and/or tree pollen species.
  • a polypeptide of option a), option b), option c) or option d), as described herein may be derived from a wild type polypeptide that co-releases with a major allergen from grass pollen (e.g. pollen of the genera Phleum and or Cynodon) and from a weed pollen (e.g. pollen of the genera Ambrosia) and from tree pollen (e.g. pollen of the genera Quercus or Betula).
  • grass pollen e.g. pollen of the genera Phleum and or Cynodon
  • a weed pollen e.g. pollen of the genera Ambrosia
  • tree pollen e.g. pollen of the genera Quercus or Betula
  • polypeptides are herein named "GWT polypeptides", such as the set of polypeptides referred to with polypeptide ID NOs: A0349, A0246, A0209, A0211 , A0325, A0357 and A0203.
  • GWT polypeptides such as the set of polypeptides referred to with polypeptide ID NOs: A0349, A0246, A0209, A0211 , A0325, A0357 and A0203.
  • these polypeptides have predicted HLA Class II allele binding sites in their conserved part of their sequence, thereby indicating the polypeptides potentially comprises a T cell epitope in their conserved region.
  • polypeptide A0262 (herein named GT polypeptide) was released from both grass and tree pollen, but not from weed pollen. It was also found that the set of polypeptides having the polypeptide ID NOs A0362,
  • A0316, A0356, A0376, A0336, A0377 and A0366 could be released from grass as well as weed pollen, but not the tree pollen investigated (herein named GW polypeptides).
  • the molecule comprises a conserved region or a subsequence thereof of the "GWT polypeptide" A0349, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least
  • the molecule comprises a conserved region or a
  • GWT polypeptide such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 233 set out in Table 3 and SEQ ID NOS: 414-418 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 233 set out in Table 3 and SEQ ID NOS: 414-418 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 233 set out in Table 3 and SEQ ID NO NOS: 414-418 set out in Table 4
  • the molecule comprises a conserved region or a
  • GWT polypeptide A0209
  • a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 217-220 set out in Table 3 and SEQ ID NOS: 321-344 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any of SEQ ID NOS: SEQ ID NOS: 217-220 set out in Table 3 and SEQ ID NOS: 321-344 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 217-220 set
  • the molecule comprises a conserved region or a
  • GWT polypeptide A0211
  • a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 221-225 set out in Table 3 and SEQ ID NOS: 345-386 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 221-225 set out in Table 3 and SEQ ID NOS: 345-386 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 221-225 set out in Table 3 and
  • the molecule comprises a conserved region or a
  • GWT polypeptide A0325
  • a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS:256 set out in Table 3 and SEQ ID NOS: 541-547 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS:256 set out in Table 3 and SEQ ID NOS: 541-547 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS:256 set out in Table 3 and SEQ ID NOS: 5
  • GWT polypeptide A0357
  • a molecule comprising or consisting of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 280-281 set out in Table 3 and SEQ ID NOS: 701-709 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 280-281 set out in Table 3 and SEQ ID NOS: 701-709 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 280-281 set out in Table 3 and SEQ ID NO NOS: 701-709 set out
  • the molecule comprises a conserved region or a
  • GWT polypeptide A0203, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 215, 216 set out in Table 3 and SEQ ID NOS: 309-320 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 215, 216 set out in Table 3 and SEQ ID NOS: 309-320 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 215, 216 set out in Table
  • a molecule of the present invention is an IgE reactive molecule, e.g. able to bind to IgE antibodies specific for the immunogenic molecule.
  • IgE reactivity towards a molecule of the invention may only be conferred by a low fraction of an allergic population.
  • a molecule of the invention may not fall under the usual definitions of a major allergen, which is an allergen with high prevalence in a population of donors allergic to a pollen species, for example the prevalence may be higher than at least 50%.
  • the molecule is able to react with, bind to or induce IgG antibodies in a subject, at least in detectable levels.
  • the molecule does not react with, bind to or induce IgG antibodies, at least in detectable levels. As demonstrated herein, a molecule of the invention seems to be less
  • polypeptides containing GWT, GT, GW or WT conserved regions can be detected in various pollen species and families and share high identity and similarity across the various pollen species. Therefore, a polypeptide of option d) comprises an amino acid sequence having at least 70%, such as at least 75%, 80%,
  • SEQ ID NOS: 1-37 e.g. SEQ ID NOS: 30, 1-29, 31- 37
  • SEQ ID NOS: 38-211 eg. SEQ ID NOS: 161-172, 38-160 and 173-211 set out in Table 2.
  • a polypeptide of option a) comprises an amino acid sequence having at least 70% such as at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity to a sequence selected from any one of SEQ ID NOS: 212- 293 (e.g. SEQ ID NOS: 270-274; 212-269; 275-293) set out in Table 3; SEQ ID NOS: 294-767 (e.g.
  • a polypeptide of option b) comprises an amino acid sequence having at least 70%, such as at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 212-293 (e.g. SEQ ID NOS: 270-274; 212-269; 275-293 set out in Table 3; SEQ ID NOS: 294-767 (e.g.
  • the at least 70% such as at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity is of a subsequence consisting of at least 16 contiguous amino acid residues, for example at least 17, 18, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 90, 100 or more contiguous amino acids.
  • a subsequence may comprise any number of amino acids, but typically the subsequence has a length of 15 to 50 amino acid residues, for example 15 to 45, 15 to 40, 15 to 35, 15 to 30; 15 to 25; 15 to 20; 15 to 19; 15 to 18; 15 to 17, or 15 to 16 amino acid residues.
  • 15 to 45, 15 to 40, 15 to 35, 15 to 30 15 to 25; 15 to 20; 15 to 19; 15 to 18; 15 to 17, or 15 to 16 amino acid residues.
  • the number of amino acid mismatches may be 0, 1 , 2, or 3, preferably 0, 1 or 2, more preferably 0 or 1.
  • the polypeptide of option b) or option c) has a length of 15-800 or more amino acid residues, for example 15-750, 15-700, 15-650 or 15-600 or more amino acid residues, for example 15-20, 20-25, 25-30, 30-35, 35-40, 45-50, 50-60, 60-70, 70-80, 90-100, 100-125, 125-150, 150-175, 175-200, 200-250, 250-300, 300-350, 350-400, 400- 450, 450-500, 500-550, 550-600, 600-650, 650-700, 700-800 or more amino acid residues.
  • the polypeptide of option a) or of option d) has a length of no more than 800 amino acid residues, for example no more than 750, 700, 650, 600, 550,
  • a polypeptide disclosed herein including a subsequence thereof may contain a T cell epitope, such as a Th2 cell epitope.
  • a subsequence or a polypeptide described herein may have HLA Class II binding properties. HLA Class II binding can be predicted using NetMHCIIpan-3.0 tool (Karosiene, Edita, Michael Rasmussen, Thomas Bö, Ole Lund, S0ren Buus, and Morten Nielsen.
  • a polypeptide of option b) or c) may have different lengths according to the desirable use, for example of about 15-800 or more amino acid residues in length, for example 15-750, 15-700, 15-650, 15-600, 15-500 or more amino acid residues, for example 15-20, 15-25, 15-30, 20-25, 25-30, 30-35, 35-40, 45-50, 50-60, 60-70, 70-80, 90-100, 100-125, 125- 150, 150-175, 175-200, 200-250, 250-300, 300-350, 350-400, 400-450, 450-500, 500- 550, 550-600, 600-650, 650-700, 700-800 or more amino acid residues.
  • a polypeptide of option b) or a polypeptide of option c) has a length in the range of 15 to 30 amino acid residues, for example 15 to 25 amino acid residues.
  • a polypeptide of option a), b), c) or d) is a longer polypeptide which comprises a secondary or tertiary structure, e.g. folded.
  • a polypeptide of option a), b), c) or d) has a length in the range of 30 to 500 amino acid residues or more. It is considered that the length of the amino acid sequence of a polypeptide of option a), b), c) or d) is no more than 800 amino acid residues, for example no more than 750, 700, 650, 600, 550, 500 or 450 amino acid residues.
  • identity and “identical” and grammatical variations thereof, as used herein, mean that two or more referenced entities are the same (e.g. two or more amino acid sequences). Thus, where two polypeptides are identical, they have the same amino acid sequence.
  • the identity can be over a defined area (region or domain) of the sequence, e.g. over the sequence length of a sequence disclosed in Tables 1 to 9 or over a portion thereof e.g. at least 15 contiguous amino acid residues.
  • identity can be over the length of the sequence overlapping the two polypeptides, when aligned with best fit with gaps permitted.
  • the polypeptide may be aligned with a sequence of Tables 1 to 9 and the percent identity be calculated with reference to said sequence. Identity can be determined by comparing each position in aligned sequences. A degree of identity between amino acid sequences is a function of the number of identical or matching amino acids at positions shared by the sequences, i.e. over a specified region.
  • Optimal alignment of sequences for comparisons of identity may be conducted using a variety of algorithms, as are known in the art, including the Clustal Omega program available at http://www.ebi.ac.uk/Tools/msa/clustalo/, the local homology algorithm of Smith and Waterman, 1981 , Adv. Appl. Math 2: 482, the homology alignment algorithm of Needleman and Wunsch, 1970, J. Mol. Biol. 48:443, the search for similarity method of Pearson and Lipman, 1988, Proc. Natl. Acad. Sci.
  • Sequence identity may also be determined using the BLAST algorithm, described in Altschul et al., 1990, J. Mol. Biol. 215:403-10 (using the published default settings).
  • BLAST e.g., BLAST 2.0
  • search algorithm see, e.g., Altschul et al., J. Mol. Biol. 215:403 (1990), publicly available through NCBI
  • mismatch -2 e.g., Altschul et al., J. Mol. Biol. 215:403 (1990), publicly available through NCBI
  • mismatch -2 e.g., Altschul et al., J. Mol. Biol. 215:403 (1990), publicly available through NCBI
  • a BLASTP algorithm is typically used in combination with a scoring matrix, such as
  • FASTA e.g., FASTA2 and FASTA3
  • SSEARCH sequence comparison programs are also used to quantitate the extent of identity (Pearson et al., Proc. Natl. Acad. Sci. USA 85:2444 (1988); Pearson, Methods Mol Biol. 132: 185 (2000); and Smith et al., J. Mol. Biol. 147:195 (1981)).
  • Programs for quantitating protein structural similarity using Delaunay-based topological mapping have also been developed (Bostick et al., Biochem Biophys Res Commun. 304:320 (2003)).
  • a polypeptide sequence is a "homolog” of, or is “homologous” to, another sequence if the two sequences have substantial identity over a specified region and a functional activity of the sequences is preserved or conserved, at least in part. (As used herein, the term 'homologous' does not infer nor exclude evolutionary relatedness).
  • homologous polypeptides include polypeptides with high similarity or identity to Phi P polypeptides and detected in pollen species other than Phi p.
  • a homologous polypeptide to a polypeptide having an amino acid sequence set out in Table 1 or Table 2 may be found in pollen of plant families selected among Asteraceae, Betulaceae, Fagaceae, Oleaceae, or Plantaginaceae, e.g.
  • Two polypeptide sequences are considered to be substantially identical if, when optimally aligned (with gaps permitted), they share at least about 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, etc. identify over a specific region), for example, over all or a part of any amino acid sequence in Tables 1 to 9, or if the sequences share defined functional motifs (e.g., epitopes).
  • the length of the sequence sharing the percent identity is at least 15, 16, 17, 18, 19, 20, etc. contiguous amino acids, e.g. more than 25, 30, 35, 40, 45 or 50 or more contiguous amino acids, including the entire length of a reference sequence of Tables 1 to 9.
  • an "unrelated" or “non-homologous” sequence is considered to share less than 30% identity. More particularly, it may share less than about 25 % identity, with a polypeptide of the invention over a specified region of homology.
  • An amino acid sequence set out in any of Tables 1 to 9 may contain one or more modifications, which optionally may result in greater or less activity or function, for example in the ability to elicit, stimulate, induce, reduce, inhibit, suppress an in vitro immune response (e.g. T cell proliferation or T cell cytokine production); in the ability to bind HLA Class II alleles; in the ability to induce or enhance immunological
  • a modification includes deletions, including truncations and fragments; insertions and additions, substitutions, for example conservative substitutions, site-directed mutants and allelic variants.
  • Non-limiting examples of modifications include one or more amino acid substitutions (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20-25, 25-30, 30-50, 50-100 or more residues), additions and insertions (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15 or more residues) and deletions (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20-25, 25-30, 30-50, 50-100 or more) of a sequence set out in Tables 1 , 2, 3 and 4.
  • amino acid substitutions e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20-25, 25-30, 30-50, 50-100 or more
  • similarity and “similar” and grammatical variations thereof, as used herein, mean that two or more referenced amino acid sequences contains a limited number of conservative amino acid substitutions of the amino acid sequence.
  • a variety of criteria can be used to indicate whether amino acids at a particular position in a polypeptide are similar.
  • substitutions of like amino acid residues can be made on the basis of relative similarity of side-chain substituents, for example, their size, charge, hydrophobicity, hydrophilicity, and the like, and such substitutions may be assayed for their effect on the function of the peptide by routine testing.
  • a "conservative substitution” is the replacement of one amino acid by a biologically, chemically or structurally similar residue.
  • Biologically similar means that the substitution does not destroy a biological activity.
  • Structurally similar means that the amino acids have side chains with similar length, such as alanine, glycine and serine, or a similar size.
  • Chemical similarity means that the residues have the same charge, or are either hydrophilic or hydrophobic.
  • a conservative amino acid substitution is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain, which include amino acids with basic side chains (e.g., lysine, arginine, histidine); acidic side chains (e.g., aspartic acid, glutamic acid); uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, histidine); nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); beta-branched side chains (e.g., threonine, valine, isoleucine), and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan).
  • basic side chains e.g., lysine, arginine, histidine
  • Particular examples include the substitution of one hydrophobic residue, such as isoleucine, valine, leucine or methionine for another or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acids, or glutamine for asparagine, serine for threonine, and the like.
  • Proline which is considered more difficult to classify, shares properties with amino acids that have aliphatic side chains (e.g., Leu, Val, lie, and Ala).
  • substitution of glutamine for glutamic acid or asparagine for aspartic acid may be considered a similar substitution in that glutamine and asparagine are amide derivatives of glutamic acid and aspartic acid, respectively.
  • Conservative changes can also include the substitution of a chemically derivatized moiety for a non- derivatized residue, for example, by reaction of a functional side group of an amino acid.
  • Variants and derivatives of polypeptides include forms having a limited number of one or more substituted residues.
  • a polypeptide of option a), b), c) and d) may be longer than the reference sequence set out in Tables 1 to 9.
  • an addition can be one or more additional amino acid residues.
  • a polypeptide of option b) or c) may contain amino acid residues in addition to the subsequence of at least 15 amino acid residues.
  • the additional amino acid residues may be identical to those present in the wild type polypeptide from which the subsequence derives from.
  • the polypeptide of option b) comprises one or more amino acid residues in addition to the subsequence of at least 15 contiguous amino acids, wherein the additional amino acid residue(s) is/are selected from an amino acid residue or an amino acid sequence within the wild type polypeptide that the subsequence is a part of (e.g.
  • wild type polypeptide sequences of Tables 1 or 2 or a GWT sequence of Tables 3 or 4 may be adjacent to, subtended, comprised within, overlapping with or is a part of the subsequence, when present in its natural biological context within the wild type polypeptide.
  • a polypeptide of option a) may contain additional amino acid residues in addition to the GWT sequence set out in Tables 3 and 4.
  • a polypeptide of option a) may comprise one or more amino acid residues in addition to the GWT sequence set out in Tables 3 or 4, wherein the additional amino acid residue(s) is/are selected from an amino acid residue or an amino acid sequence within the wild type polypeptide of which the GWT sequence is a part of (e.g. a wild type polypeptide of Tables 1 or 2).
  • An illustrative example is a GWT sequence of A0349 set out in Table 3 that may be extended with amino acid residues from polypeptide A0349 set out in Tables 1 or 2, such as amino acid residues adjacent to the GWT sequence when aligned with the corresponding wild type polypeptide.
  • the additional amino acid residues may be added to the N- and/or C- terminal end of a sequence set out in Tables 3 to 9, such as additional amino acids selected from amino acids flanking the N- and/or C- terminal ends when sequence is aligned with the source protein it is present in, based upon or derived from.
  • the additional amino acids may be the amino acids flanking the N- and/or C- terminal ends of the sequence when aligned to polypeptide A0349 of Table 1 or 2.
  • a polypeptide of option a), b), c) or d) may include a number of
  • an amino acid sequence (parent sequence) disclosed in any of the Tables or a subsequence thereof may be modified to comprise: a) one or more (e.g. 1 , 2,3, 4, 5, 6, 7, 8, 9 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) amino acid substitutions in a sequence selected from any of the sequences disclosed in Tables 1 to 9, for example a glutamate residue at the N-terminus of an amino acid sequence disclosed in Tables 1 to 9 may be replaced with pyroglutamate and/or one or more cysteine residues in the an amino acid sequence disclosed in Tables 1 to 9 may be replaced with serine or 2-aminobutyric acid; and/or b) one or more amino acid additions (e.g.
  • a polypeptide of option a), b), c) or d) including
  • polypeptide is derivatized.
  • derivatization are covalent or non-covalent attachment of another molecule.
  • Specific examples include glycosylation, acetylation, phosphorylation, amidation, formylation, ubiquitination, and derivatization by protecting/blocking groups and any of numerous chemical modifications.
  • a derivative is formed by reacting a functional side group of an amino acid (e.g. amino, sulfhydryl or carboxy-group) with another molecule to form a covalent or non- covalent attachment of any type of molecule (naturally occurring or designed), such as a sugar moiety.
  • Specific examples of derivatives of a peptide include glycosylation, acylation (e.g.
  • acetylation phosphorylation
  • amidation formylation
  • ubiquitination phosphorylation
  • derivatization by protecting/blocking groups and any of numerous chemical modifications.
  • Additional specific non-limiting examples are tagged peptides, fusion peptides, chimeric peptides including peptides having one or more non-amino acyl groups (q.v., sugar, lipid, etc.) covalently linked to the peptide.
  • a derivative comprises one or more modifications, for example selected from any of: (a) N-terminal acylation (e.g. acetylation or formylation); (b) C-terminal amidation (e.g.
  • a derivative comprises a fusion (chimeric) sequence of peptides, which optionally may contain an amino acid sequence having one or more molecules not normally present in a reference (wild type) sequence covalently attached to the peptide amino acid sequence.
  • chimeric and grammatical variations thereof, when used in reference to a sequence, means that the sequence contains one or more portions that are derived from, obtained or isolated from, or based upon other physical or chemical entities.
  • a derivative is one in which a second heterologous sequence, i.e. a heterologous functional domain, is attached to a peptide disclosed herein, (covalent or non-covalent binding) that may confer a distinct or complementary function to a peptide disclosed herein.
  • Heterologous functional domains are not restricted to amino acid residues.
  • a heterologous functional domain can consist of any of a variety of different types of small or large functional moieties.
  • moieties include nucleic acid, peptide, carbohydrate, lipid or small organic compounds, such as a drug (e.g., an antiviral), a metal (gold, silver), or a radioisotope.
  • Linkers such as amino acid or peptidomimetic sequences, may be inserted between the peptide sequence and the addition (e.g., heterologous functional domain) so that the two entities maintain, at least in part, a distinct function or activity.
  • Linkers may have one or more properties that may include a flexible conformation, an inability to form an ordered secondary structure or a hydrophobic or charged character, which could promote or interact with either domain.
  • Amino acids typically found in flexible protein regions include Gly, Asn and Ser. Other near neutral amino acids, such as Thr and Ala, may also be used in the linker sequence.
  • polypeptides of option a), b), c) or d) or a combination of such polypeptides are not provided as individual polypeptides, but may be fused together or to a carrier molecule to form an isolated molecule (e.g. immunogenic molecule).
  • the polypeptides may be fused to the N- and C-terminus of a surface polypeptide of a virus, e.g. a virus of the hepadnaviridae family as disclosed in international patent application W012168487 A1.
  • a molecule of the invention including a polypeptide of option a), b), c) or d), a subsequence thereof, a modified polypeptide thereof, or a derivatized polypeptide thereof may bind to at least 70% of the group of Class HLA II alleles that the relevant parent polypeptide disclosed in Tables 1 to 9 binds to.
  • a molecule of the invention including a polypeptide of option a), b), c) or d), a modified polypeptide thereof, or a derivatized polypeptide binds to the same, substantially the same or at least 75%, 80%, such as at least 82%, 85%, 88%, 90%, 92%, 95%, 98% or more, of the group of HLA Class II alleles that binds to the relevant parent polypeptide disclosed in Tables 1 to 9, optionally wherein this is determined under same test conditions, either using prediction tools or in-vitro binding assay.
  • a subsequence disclosed herein binds to the same, substantially the same or at least 75%, 80%, such as at least 82%, 85%, 88%, 90%, 92%, 95%, 98% or more, of the group of HLA Class II alleles that binds to the relevant subsequent from which the modified sequence derives, optionally wherein this is determined under same test conditions, either using prediction tools or in-vitro binding assay.
  • the Class HLA II binding is determined with respect to a particular group of Class HLA II alleles, for example one or more or all of the following alleles: DPA1*02:01-DPB1 * 01 :01 ,
  • a polypeptide of option a), b), c) or d), including a modified or derivatized polypeptide thereof may have one or more of the same T cell epitopes or T cell activity (e.g. percent responders) as the amino acid sequence from which it derives (e.g. an amino acid sequence set out in Tables 1 to 9.
  • This may be determined by the ability to induce or stimulate in vitro T cell proliferation using cultured PBMCs (peripheral blood monocytes) in response to the polypeptide of option a), b), c) or d), including a modified or derivatized polypeptide compared to the amino acid sequence set out in Tables 1 to 9, optionally using same test conditions, or by the ability to induce or stimulate production of cytokines, (e.g.
  • T cell assays examples include cytokines, IL-5, IL-13 and/or IL-10) from T cells (obtained from cultured PBMC's) in response to the polypeptide of option a), b), c) or d), including a modified or derivatized polypeptide compared to the amino acid sequence set out in Tables 1 to 9. Examples of T cell assays are described herein in Example 6.
  • a derivative is a fusion (chimeric) sequence, an amino acid sequence having one or more molecules not normally present in the wild type sequence covalently attached to the sequence.
  • chimeric and grammatical variations thereof, when used in reference to a sequence, means that the sequence contains one or more portions that are derived from, obtained or isolated from, or based upon other physical or chemical entities. For example, a chimera of two or more different
  • polypeptides may have one part of a polypeptide, and a second part of the chimera may be from a different sequence, or unrelated protein sequence.
  • heterologous functional domain is attached (covalent or non- covalent binding) that confers a distinct or complementary function.
  • heterologous functional domains are not restricted to amino acid residues.
  • a heterologous functional domain can consist of any of a variety of different types of small or large functional moieties.
  • moieties include nucleic acid, peptide, carbohydrate, lipid or small organic compounds, such as a drug (e.g., an antiviral), a metal (gold, silver), and radioisotope.
  • a tag such as T7 or polyhistidine can be attached in order to facilitate purification or detection of a protein, peptide, etc.
  • a 6-HIS tag may be added to the C- or N-terminal end of a polypeptide of option a), b), c) or d), e.g. the 6- HIS sequence GHHHHHHGSGMLDI, which optionally may remain in the molecule when administered to a subject.
  • a polypeptide linked to a Tag containing histidines may easily be purified by use of a HIS tag affinity column).
  • polypeptides linked to a heterologous domain wherein the heterologous functional domain confers a distinct function on the polypeptide.
  • the polypeptide is derivatized for example to improve solubility, stability, bioavailability or biological activity.
  • tagged polypeptides and fusion proteins and modifications, including peptides having one or more non-amino acyl groups (q.v., sugar, lipid, etc.) covalently linked to the polypeptide and post-translational modifications.
  • Linkers such as amino acid or peptidomimetic sequences may be inserted between the sequence and the addition (e.g., heterologous functional domain) so that the two entities maintain, at least in part, a distinct function or activity.
  • Linkers may have one or more properties that include a flexible conformation, an inability to form an ordered secondary structure or a hydrophobic or charged character, which could promote or interact with either domain.
  • Amino acids typically found in flexible protein regions include Gly, Asn and Ser. Other near neutral amino acids, such as Thr and Ala, may also be used in the linker sequence.
  • the length of the linker sequence may vary without significantly affecting a function or activity of the fusion protein (see, e.g., U.S. Patent No. 6,087,329).
  • Linkers further include chemical moieties and conjugating agents, such as sulfo-succinimidyl derivatives (sulfo-SMCC, sulfo-SMPB), disuccinimidyl suberate (DSS), disuccinimidyl glutarate (DSG) and disuccinimidyl tartrate (DST).
  • sulfo-succinimidyl derivatives sulfo-SMCC, sulfo-SMPB
  • DSS disuccinimidyl suberate
  • DSG disuccinimidyl glutarate
  • DST disuccinimidyl tartrate
  • derivatives are detectable labels.
  • the invention provides polypeptides that are detectably labeled.
  • detectable labels include fluorophores, chromophores, radioactive isotopes (e.g., S 35 , P 32 , I 125 ), electron-dense reagents, enzymes, ligands and receptors.
  • Enzymes are typically detected by their activity.
  • horseradish peroxidase is usually detected by its ability to convert a substrate such as 3,3-',5,5-'-tetramethylbenzidine (TMB) to a blue pigment, which can be quantified.
  • TMB 3,3-',5,5-'-tetramethylbenzidine
  • Modified polypeptides also include one or more D-amino acids substituted for L-amino acids (and mixtures thereof), structural and functional analogues, for example, peptidomimetics having synthetic or non-natural amino acids or amino acid analogues and derivatized forms. Modifications include cyclic structures such as an end-to-end amide bond between the amino and carboxy-terminus of the molecule or intra- or inter- molecular disulfide bond.
  • Polypeptides may be provided in the form of a salt, for example as a pharmaceutically acceptable and/or a physiologically acceptable salt.
  • the salt may be an acid addition salt with an inorganic acid, an acid addition salt with an organic acid, a salt with a basic inorganic acid, a salt with a basic organic acid, a salt with an acidic or basic amino acid or a mixture thereof.
  • a salt, such as a pharmaceutically acceptable salt is an acetate salt.
  • the invention provides polypeptides and molecules in isolated and/or purified form.
  • isolated when used as a modifier of a composition, means that the compositions are made by the hand of man or are separated, completely or at least in part, from their naturally occurring in vivo environment. Generally, isolated compositions are substantially free of one or more materials with which they normally associate with in nature, for example, one or more protein, nucleic acid, lipid, carbohydrate, cell membrane.
  • isolated does not exclude alternative physical forms of the composition, such as fusions/chimeras, multimers/oligomers, modifications (e.g., phosphorylation, glycosylation, lipidation) or derivatized forms, or forms expressed in host cells produced by the hand of man.
  • an “isolated” composition e.g. polypeptides or molecules as defined herein
  • an isolated polypeptide that also is substantially pure or purified does not include polypeptides or polynucleotides present among millions of other sequences, such as polypeptide of a peptide library or nucleic acids in a genomic or cDNA library, for example.
  • compositions can be combined with one or more other molecules.
  • substantially pure or purified does not exclude combinations of compositions, such as combinations of polypeptides other antigens, agents, drugs or therapies.
  • Polypeptides can be prepared recombinant, chemically synthesized, isolated from a biological material or source, and optionally modified, or any combination thereof.
  • a biological material or source would include an organism that produced or possessed any polypeptide or molecule set forth herein.
  • a biological material or source may further refer to a preparation in which the morphological integrity or physical state has been altered, modified or disrupted, for example, by dissection, dissociation, solubilization,
  • polypeptides such as a molecule (e.g. immunogenic molecules) disclosed herein, may be modified by substituting, deleting or adding one or more amino acid residues in the amino acid sequence and screening for biological activity, for example eliciting an immune response.
  • a skilled person will understand how to make such derivatives or variants, using standard molecular biology techniques and methods, described for example in Sambrook et al. (2001) Molecular Cloning: a Laboratory Manual, 3 rd ed., Cold Spring Harbour Laboratory Press).
  • Molecules and polypeptides disclosed herein, including a modified polypeptide thereof, a derivatized polypeptide thereof, or salts thereof may be manufactured synthetically or recombinantly.
  • the molecule, including a polypeptide of option a), b), c), d) or a subsequence thereof is synthetic.
  • the molecule, including a polypeptide of option a), b), c), d) or a subsequence thereof may be isolated and/or purified, e.g. made by the hand of man, such as by synthesis.
  • the polypeptides may be combined after synthesis and freeze-dried or dissolved in aqueous solutions, DMSO, glycerol or the like or mixtures thereof.
  • Molecules e.g. immunogenic molecules
  • Such methods and uses include, for example, administration in vitro and in vivo of one or more molecules disclosed herein.
  • the methods and uses may include modulating an immune response (e.g. modulating an allergic immune response), including, among others, methods and uses for relieving an immune response (e.g. allergic immune response), protecting and treating subjects against a disorder, disease (e.g. allergic disease); and methods and uses of providing immunotherapy, such as specific immunotherapy against an immune response, e.g. an immune response triggered by an allergen (e.g. a pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • an allergen e.g. a pollen, such as a pollen allergen
  • methods and uses include administration or delivery of a molecule disclosed herein to modulate an immune response in a subject, including, for example, modulating an immune response triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • an allergen e.g. triggered by pollen, such as a pollen allergen
  • a molecule disclosed herein e.g. triggered by pollen, such as a pollen allergen
  • modulate means an alteration or effect on the term modified.
  • modulating involves decreasing, reducing, inhibiting, suppressing, relieving an immune response in a subject triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • modulating involves eliciting, stimulating, inducing, promoting, increasing or enhancing an immune response in a subject triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • modulate is used to modify the term "immune response triggered by an allergen (e.g.
  • an allergen e.g. pollen, such as a pollen allergen
  • a molecule disclosed herein in a subject this means that the immune response in the subject to the allergen, pollen, pollen allergen or molecule is altered or affected (e.g., decreased, reduced, inhibited, suppressed, limited, controlled, prevented, elicited, promoted, stimulated, increased, induced, enhanced, etc.
  • Methods and uses of modulating an immune response triggered by an allergen e.g. pollen, such as a pollen allergen
  • a molecule disclosed herein may be used to provide a subject with protection against an immune response or immune reaction to said allergen, pollen, pollen allergen and/or molecule (e.g.
  • methods and uses include administering a molecule (e.g.
  • immunogenic molecule of the invention to protect or treat a subject against an immune response, or one or more symptoms caused by, triggered by or associated with exposure to an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
  • an allergen e.g. pollen, such as a pollen allergen
  • a molecule disclosed herein e.g. a pollen allergen
  • the subject's administration of a therapeutically effective amount of a composition described herein may relieve one or more symptoms of the immune response.
  • the method may comprise relieving one or more symptoms associated with allergic rhinitis, allergic conjunctivitis, allergic asthma and/or allergic eczema (e.g. atopic dermatitis).
  • the one or more symptoms may be associated with allergic rhinitis.
  • the method may comprise reducing one or more of the following symptoms: intensity of itchy nose; number of sneezes within a given period (e.g. daily, weekly, monthly); intensity of blocked nose (e.g. congestion); amount of nasal secretions; eosinophilic count in nasal secretions; specific IgE antibody level (titer) in nasal secretions or in serum; and basophil histamine release of blood.
  • the one or more symptoms may be associated with allergic conjunctivitis.
  • the method may comprise reducing one or more of the following: intensity of itchy eyes, redness in the white of the eyes and/or watery eyes; eosinophilic count in conjunctival tissue scrapings; specific IgE antibody level (titer) in conjunctival tissue scrapings or in serum; and basophil histamine release in blood.
  • the one or more symptoms may be associated with allergic asthma.
  • the method may comprise reducing one or more of the following: number of or frequency of asthma exacerbations (optionally that require hospitalization), intensity and/or number of coughs within a given period (e.g. daily, weekly, monthly); intensity of wheezes; intensity of shortness of breath or congestion (e.g. improvement of being short of breath); reducing Forced Expiratory Volume (FEV1); reducing specific IgE antibody level (titer) in lung fluid or in serum and basophil histamine release in blood; or the method may comprise improving lung function.
  • the one or more symptoms may be symptoms associated with atopic dermatitis.
  • the method may comprise reducing one or more of the following: itch intensity of the skin; eczema score, and number of (peripheral) blood eosinophils.
  • a therapeutic or beneficial effect also includes reducing or eliminating the need, dosage frequency or amount of a second therapeutic method or therapeutically active drug (e.g. anti-inflammatory, decongestants or anti-allergic agent) used for treating a subject having an immune response or one or more symptoms caused by or associated with an allergen.
  • a second therapeutic method or therapeutically active drug e.g. anti-inflammatory, decongestants or anti-allergic agent
  • administration of a peptide combination described herein may reduce the amount of an adjunct therapy administered to a subject, such as reducing the subject's need for concomitant treatment with fast or long-acting p2-agonists, leukotriene modifiers, theophylline corticosteroids or H1 antihistamines (e.g. inhaled or oral) to reduce, relieve, or suppress one or more symptoms of the immune response.
  • an adjunct therapy administered to a subject, such as reducing the subject's need for concomitant treatment with fast or long-acting p2-agonists, leukotriene modifiers, theophylline corticosteroids or H1 antihistamines (e.g. inhaled or oral) to reduce, relieve, or suppress one or more symptoms of the immune response.
  • methods and uses include administering or delivering a molecule (e.g. an immunogenic molecule) of the invention to elicit, stimulate, induce, promote, increase or enhance immunological tolerance of a subject an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
  • a molecule e.g. an immunogenic molecule
  • an allergen e.g. pollen, such as a pollen allergen
  • a molecule disclosed herein include administering or delivering a molecule (e.g. an immunogenic molecule) of the invention to elicit, stimulate, induce, promote, increase or enhance immunological tolerance of a subject an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
  • a method or use includes administering to the subject an amount of an immunogenic molecule of the invention sufficient to provide the subject with protection against an immune response, or symptoms caused by or associated with exposure to an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
  • an allergen e.g. pollen, such as a pollen allergen
  • an immunogenic molecule disclosed herein includes administering to the subject an amount of an immunogenic molecule of the invention sufficient to provide the subject with protection against an immune response, or symptoms caused by or associated with exposure to an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
  • Methods and uses of the invention include providing a subject with protection against an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein, or symptoms caused by or associated with the subject's exposure to said allergen (e.g. pollen, such as pollen allergen) and/or a molecule disclosed herein, for example, vaccinating the subject to protect against an immune response to said allergen (e.g.
  • an allergen e.g. pollen, such as a pollen allergen
  • a molecule disclosed herein e.g. pollen, such as pollen allergen
  • methods and uses include protecting the subject against an immune response triggered by pollen, e.g. a pollen allergen or a molecule disclosed herein by inducing tolerance of the subject (desensitizing) to said allergen, pollen, pollen allergen and/or immunogenic molecule.
  • the terms "protection,” “protect” and grammatical variations thereof, when used in reference to an immune response or symptoms caused by or associated with the exposure to an allergen (e.g. pollen, such as a pollen allergen) and/or an immunogenic molecule disclosed herein, means preventing an immune response or symptoms caused by or associated with the exposure to the allergen, pollen, pollen allergen and/or a molecule disclosed herein, or reducing or decreasing susceptibility to an immune response or one or more symptoms caused by or associated with the exposure to the allergen, pollen, pollen allergen and/or a molecule disclosed herein.
  • an immune response includes but is not limited to an allergic immune response, such as an allergic reaction, hypersensitivity, an inflammatory response or inflammation.
  • an immune response may involve one or more of cell infiltration, production of antibodies, production of cytokines, lymphokines, chemokines, interferons and interleukins, cell growth and maturation factors (e.g., differentiation factors), cell proliferation, cell differentiation, cell accumulation or migration (chemotaxis) and cell, tissue or organ damage or remodeling.
  • an immune response may include an allergic immune response, such as allergic rhinitis; atopic dermatitis; allergic conjunctivitis and asthma. Allergic responses can occur systemically or locally in any region, organ, tissue, or cell.
  • an allergic immune response occurs in the skin, the upper respiratory tract, the lower respiratory tract, pancreas, thymus, kidney, liver, spleen, muscle, nervous system, skeletal joints, eye, mucosal tissue, gut or bowel.
  • Methods and uses herein include relieving a subject of, including treating a subject for, an immune response, or one or more symptoms caused by or associated with an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • an allergen e.g. pollen, such as a pollen allergen
  • Such methods and uses include administering to a subject an amount of an immunogenic molecule sufficient to relieve the subject of, such as treat the subject for, the allergic immune response, or one or more symptoms caused by or associated with the allergen, (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • Methods and uses of the invention include treating or administering to a subject previously exposed to an allergen (e.g.
  • methods and uses are for treating or protecting a subject from an allergic immune response, or one or more symptoms caused by or associated with secondary or subsequent exposure to an allergen, (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • an allergen e.g. pollen, such as a pollen allergen
  • a molecule disclosed herein e.g. a pollen allergen
  • Immunogens described herein may elicit, stimulate, induce, promote, increase or enhance immunological tolerance to an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • Methods and uses of the invention therefore further include inducing immunological tolerance of a subject to an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • an allergen e.g. pollen, such as a pollen allergen
  • molecules described herein can be effective in relieving, such as treating an immune response, including but not limited to an immune response following a secondary or subsequent exposure of a subject to an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
  • a method or use includes administering to the subject an amount of a molecule disclosed herein sufficient to induce tolerance in the subject to the allergen (e.g. pollen, such as a pollen allergen) or the molecule itself.
  • the immunological tolerance elicited, stimulated, induced, promoted, increased or enhanced may involve modulation of T cell activity, including but not limited to CD4+ T cells, CD8+ T cells, Th1 cells, Th2 cells and regulatory T cells.
  • immunological tolerance elicited, stimulated, induced, promoted, increased or enhanced from administration of the immunogenic molecule may involve modulation of the production or activity of pro-inflammatory or anti-inflammatory cytokines produced by T cells.
  • a method or use of inducing immunological tolerance in a subject to an allergen (e.g. pollen, such as a pollen allergen) and/or an immunogenic molecule disclosed herein includes a reduction in occurrence, frequency, severity, progression, or duration of physiological conditions, disorders, illnesses, diseases, symptoms or complications caused by or associated an allergic response to the allergen (e.g. pollen, such as a pollen allergen) and/or immunogenic molecule in the subject.
  • inducing immunological tolerance can protect a subject against or treat a subject for an immune response, or one or more symptoms caused by or associated with an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
  • Methods and uses of the invention include treating a subject via immunotherapy, including specific immunotherapy.
  • a method or use includes administering to the subject an amount of a molecule described herein.
  • a molecule administered to a subject during specific immunotherapy to treat the subject is the same immunogenic molecule to which the subject has been sensitized or is hypersensitive (e.g., allergic).
  • a molecule is administered to a subject to treat the subject to a different immunogenic molecule, e.g. an allergen, e.g. a pollen allergen, to which the subject has been sensitized or is hypersensitive (e.g., allergic).
  • the immunotherapeutic mechanism may involve bystander suppression of an immune response caused by an allergen, e.g. a pollen allergen, by administering an unrelated immunogenic molecule, e.g. a molecule disclosed herein.
  • a molecules of the invention may include T cell epitopes, such as Th2 cell epitopes.
  • the subject to be treated has a specific T- cell response to the a molecule before administering the first dose of the a molecule .
  • methods and uses of the invention include administering an amount of an a molecule (e.g., a T cell epitope-containing a molecule ) to a subject sufficient to provide the subject with protection against an immune response disclosed herein, or one or more symptoms disclosed herein.
  • a method includes administering an amount of an a molecule (e.g., a T cell epitope-containing a molecule ) to a subject sufficient to relieve, e.g. treat, vaccinate or immunize the subject against an immune response disclosed herein, or one or more symptoms caused by or associated with an allergen (e.g. pollen, such as a pollen allergen) and/or an a molecule disclosed herein.
  • an allergen e.g. pollen, such
  • the specific T-cell response may be monitored by determining by way of contacting a sample of PBMCs obtained from the subject with a molecule of the invention and measuring the IL-5 secretion or IL-5 mRNA gene expression in response to the molecule.
  • a method or use includes administering to a subject an amount of a polypeptide described herein or derivative thereof including an a molecule described herein, such as a T cell epitope, sufficient to modulate Th2 cell activity in the subject.
  • two or more a molecules may be administered to a subject, e.g. may be administered as a combination composition, or administered separately, such as concurrently or in series or sequentially.
  • methods and uses described herein comprise administration separately or as a combination: at least 2-25 polypeptides defined herein, or separately or as a combination of 3-25, 4-25, 5-25, 6-25, 7-25 polypeptides defined herein, or separately or as a combination of 2-20, 3-20, 4-20, 5-20, 6-20 defined herein, or separately or as a combination of 2-12, 3-12, 4-12, 5-12, 6-12, 7- 12 polypeptides defined herein, or separately or as a combination of 2-10, 3-10, 4-10, 5- 10, 6-10, 7-10 polypeptides defined herein.
  • a further aspect of the invention relates to a composition
  • a composition comprising a polypeptide of option a), b), c) or d) or a combination of two or more polypeptides of option a), b), c) or d).
  • Methods and uses of the invention therefore include any therapeutic or beneficial effect.
  • an immune response, or one or more symptoms caused by or associated with a pollen allergen or an a molecule disclosed herein is reduced, decreased, inhibited, limited, delayed or prevented.
  • Methods and uses of the invention moreover include reducing, decreasing, inhibiting, delaying or preventing onset, progression, frequency, duration, severity, probability or susceptibility of one or more adverse symptoms, disorders, illnesses, diseases or complications caused by or associated with an antigen/allergen.
  • methods and uses include improving, accelerating, facilitating, enhancing, augmenting, or hastening recovery of a subject from an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with a pollen allergen or an a molecule disclosed herein.
  • methods and uses include stabilizing an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with a pollen allergen or an a molecule disclosed herein.
  • a therapeutic or beneficial effect is therefore any objective or subjective measurable or detectable improvement or benefit provided to a particular subject.
  • a therapeutic or beneficial effect can but need not be complete ablation of all or any immune response, or one or more symptoms caused by or associated with a pollen allergen or an a molecule disclosed herein.
  • a satisfactory clinical endpoint is achieved when there is an incremental improvement or a partial reduction in an allergic immune response, or one or more symptoms caused by or associated with an allergen, or an inhibition, decrease, reduction, suppression, prevention, limit or control of worsening or progression of an immune response, or one or more symptoms caused by or associated with a pollen allergen or an a molecule disclosed herein, over a short or long duration (hours, days, weeks, months, etc.).
  • a therapeutic or beneficial effect also includes reducing or eliminating the need, dosage frequency or amount of a second therapeutic protocol or active such as another drug or other agent (e.g., anti-inflammatory) used for treating a subject having or at risk of having an allergic immune response, or one or more symptoms caused by or associated with an allergen.
  • a second therapeutic protocol or active such as another drug or other agent (e.g., anti-inflammatory) used for treating a subject having or at risk of having an allergic immune response, or one or more symptoms caused by or associated with an allergen.
  • reducing an amount of an adjunct therapy such as a reduction or decrease of a treatment for an allergic immune response, or one or more symptoms caused by or associated with an allergen, or a specific immunotherapy, vaccination or immunization protocol is considered a beneficial effect.
  • reducing or decreasing an amount of the a molecule used for specific immunotherapy, vaccination or immunization of a subject to provide protection to the subject is considered a beneficial effect.
  • Methods and uses described herein may relieve one or more symptoms of an allergic immune response or delays the onset of symptoms, slow the progression of symptoms, or induce disease modification.
  • the following symptoms may be decreased or eliminated; nasal symptoms in the form of itchy nose, sneezing, runny nose, blocked nose; conjunctival symptoms in the form of itchy eyes, red eyes, watery eyes; and respiratory symptoms in the form of decreased lung function.
  • the beneficial effect of methods and uses described herein may be observed by the patient's need for less concomitant treatment with corticosteroids or H1 antihistamines to suppress the symptoms.
  • an amount or dose of the immunogenic molecule to be administered, and the period of time required to achieve a desired outcome or result can be determined by one skilled in the art.
  • the immunogenic molecule may be administered to the patient through any route known in the art, including, but not limited to oral, inhalation, sublingual, epicutaneous, intranasal, and/or parenteral routes (intravenous, intramuscular, subcutaneously, intradermal, and intraperitoneal).
  • Methods and uses of the invention include administration of a molecule (e.g. an immunogenic molecule) to a subject prior to contact by or exposure to a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein; administration prior to, substantially contemporaneously with or after a subject has been contacted by or exposed to an allergen; and administration prior to, substantially contemporaneously with or after an allergic immune response, or one or more symptoms caused by or associated with a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein.
  • a molecule e.g. an immunogenic molecule
  • a "sufficient amount” or “effective amount” or an “amount sufficient” or an “amount effective” refers to an amount that provides, in single (e.g., primary) or multiple (e.g., booster) doses, a long term or a short term detectable or measurable improvement in a given subject or any objective or subjective benefit to a given subject of any degree or for any time period or duration (e.g., for minutes, hours, days, months, years, or cured).
  • prophylactically effective in each and every subject treated, nor a majority of subjects treated in a given group or population An amount sufficient or an amount effective means sufficiency or effectiveness in a particular subject, not a group of subjects or the general population. As is typical for such methods, different subjects will exhibit varied responses to a method of the invention, such as immunization, vaccination, specific immunotherapy and therapeutic treatments.
  • subject includes but is not limited to a subject at risk of allergen contact or exposure as well as a subject that has been contacted by or exposed to an allergen.
  • a subject also includes those having or at risk of having or developing an immune response to an antigen or an allergen.
  • Such subjects include mammalian animals (mammals), such domestic animal (dogs and cats), a farm animal (poultry such as chickens and ducks, horses, cows, goats, sheep, pigs), experimental animal (mouse, rat, rabbit, guinea pig) and humans.
  • Target subjects and subjects in need of treatment also include those at risk of allergen exposure or contact or at risk of having exposure or contact to an allergen. Accordingly, subjects include those at increased or elevated (high) risk of an allergic reaction; that have, or have previously had or are at risk of developing hypersensitivity to an allergen; and those that have or have previously had or are at risk of developing asthma.
  • the immune response to be treated in a subject in need thereof is against one or more allergens, e.g. one or more pollen, such as one or more pollen allergens.
  • immunological tolerance is desirable to be induced or promoted in a subject in need thereof to one or more allergens, e.g. one or more pollen, such as one or more pollen allergens.
  • the subject may be sensitized to one or more of the pollen allergens.
  • Non-limiting examples of the one or more pollen allergens to which the subject optionally is sensitized or the immune response is triggered by include proteins, polypeptides and peptides including allergens from a plant species selected from any of the plant families /Asferaceae, Betulaceae, Fagaceae, Oleaceae, Poaceae and/or Plantaginaceae, for example plant species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum, Poa, Ambrosia, Artemisia, Helianthus, AInus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and Plantago; for example plant species selected from any of the plant genera
  • Anthoxanthum Conydon, Dactylis, Lollium, Phleum, Poa, Ambrosia, Artemisia, AInus, Betula, Corylus, Fagus, Quercus, Olea and Plantago; for example plant species selected from the plant genera Ambrosia, Artemisia, Helianthus, AInus, Betula, Carpinus,
  • Quercus such as a plant species selected from any of the plant genera Phleum,
  • the one or more pollen allergens to which the subject optionally is sensitized to include proteins, polypeptides and peptides including allergens from species selected from further plant families, like the plant family Chenopidiaceae including the plants Lambs quarters, Russian thistles and Kochias; plant family Amaranthaceae including Pigweeds, plant family Polygonaceae including for example Sheep sorrel, plant family Ulmacea including for example American Elm and hackberry, plant family
  • Plantanaceae including for example Sycamore, plant family Salicaceae including for example White poplar and Cottonwood, plant family Aceraceae including for example Box elder and Red maple, plant family Cupressaceae including for example Common juniper and Cedar.
  • the subject may be sensitized to one or more pollen species from one or more plant families, for example selected from any of the plant families Poaceae,
  • the subject may be sensitized to a pollen species of the plant family Poaceae and a pollen species of a plant family selected from any one of Asteraceae, Fagaceae and Betulaceae, such as a subject that may be sensitized to a pollen species of the plant genus Phleum and a pollen species of a plant genus selected from any one of the genera Conydon, Ambrosia, Betula and Quercus and combinations thereof.
  • the subject may be sensitized to one or more pollen allergens from one or more plant families, for example selected from the any of the plant families Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, preferably Poaceae, Asteraceae, Fagaceae and Betulaceae.
  • the subject may be sensitized to a pollen allergen of the plant family Poaceae and a pollen allergen of a plant family selected from any one of Asteraceae, Fagaceae and Betulaceae and combinations thereof, such as a subject that may be sensitized to a pollen allergen of the plant genus Phleum and a pollen allergen of a plant genus selected from the group consisting of Conydon, Ambrosia, Betula and Quercus and combinations thereof.
  • the immune response may be triggered by pollen from one or more plant families, such as from plant families selected from any of Poaceae, Asteraceae,
  • the immune response is triggered by pollen of the plant family Poaceae and pollen of a plant family selected from any one of Asteraceae, Fagaceae and Betulaceae and combinations thereof, for example the immune response may be triggered by pollen of the plant genus Phleum and pollen of a plant genus selected from the any one of Conydon, Ambrosia, Betula and Quercus or combinations thereof.
  • the immune response may be triggered by one or more pollen allergens, such as one or more pollen allergens from a plant family selected from any of Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, preferably Poaceae, Asteraceae, Fagaceae and Betulaceae. Therefore, in some embodiments, the immune response is triggered by one or more pollen allergens from pollen of the plant family Poaceae and one or more pollen allergens from pollen of a plant family selected from any of Asteraceae, Fagaceae and Betulaceae.
  • pollen allergens such as one or more pollen allergens from a plant family selected from any of Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, preferably Poaceae, Asteraceae, Fagaceae and Betulaceae. Therefore, in
  • the one or more pollen allergens may be from pollen of the plant genus Phleum and from pollen of a plant genus selected from the group consisting of Conydon, Ambrosia, Betula and Quercus including combinations thereof.
  • the immune responses to be modulated or treated are against pollen allergens of grass species of various plant genera, for example of the genera
  • Phleum and Cynodon thus allowing the treatment or modulation of an immune response caused by a broad range of different grass pollen species. Therefore, in some
  • the one or more pollen allergens are from a grass species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum and/or Poa, for example from grass species selected from any of the genera Conydon and Phleum.
  • the immune responses to be modulated or treated are against grass pollen allergens as well as weed and/or tree pollen allergens.
  • the immune responses to be modulated or treated are against grass pollen allergens of the genera Phleum and against non-grass pollen of the genera Ambrosia and/or Quercus, thus allowing the treatment or modulation of an immune response caused by a broad range of different grass pollen species and non-grass species.
  • the one or more pollen allergens are from a grass species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum and Poa and from a non-grass species selected from any of the plant families Asteraceae, Betulaceae, Fagaceae, Oleaceae and Plantaginaceae, e.g.
  • non-grass species are selected from any of the plant genera Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and Plantago, for example against a non-grass species selected from any of the plant genera Ambrosia, Artemisia, Alnus, Betula, Quercus, Olea and Plantago, such as a non-grass species selected from any of the plant genera Ambrosia, Betula and Quercus.
  • the immune responses to be modulated or treated are against non-grass pollen allergens, such as against weed and/or tree pollen allergens.
  • the immune responses to be modulated or treated are against non-grass pollen allergens of the genera Ambrosia and against non-grass pollen allergens of the genera Quercus, thus allowing the treatment or modulation of an immune response caused by a broad range of different non-grass pollen species.
  • the one or more non-grass pollen allergens are from a plant genus selected from any of
  • Ambrosia Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and/or Plantago.
  • the one or more pollen allergens are from same grass species or alternatively from different grass species, optionally wherein the different grass species are from different plant genera selected from any of Anthoxanthum, Conydon, Dactylis, Lollium, Phleum and Poa, for example Conydon and Phleum.
  • the one or more non-grass pollen allergens are from same non-grass species or alternatively from different non-grass species, optionally wherein the different non-grass species are from different plant genera selected from any of
  • the immune response to be modulated or treated may be at least against one or more pollen allergens of various grass pollen species, e.g. against one or more pollen allergens of the plant genera Phleum. In some embodiments, the immune response to be modulated or treated may be at least against one or more pollen allergens of various weed pollen species, e.g. against one or more pollen allergens of the plant genera Ambrosia.
  • the immune response to be modulated or treated may be at least against one or more pollen allergens of various tree pollen species, e.g. against one or more pollen allergens of the plant genera Quercus. In some embodiments, the immune response to be modulated or treated may be at least against one or more pollen allergens of various tree pollen species, e.g. against one or more pollen allergens of the plant genera Betula.
  • the immune response to be modulated or treated may be against one or more pollen allergens of various grass pollen species and non-grass pollen species, such as two or more, three or more, four or more, five or more pollen allergens of different pollen species, like various grass pollen species and/or non-grass pollen species.
  • Non-limiting examples of the genus Ambrosia may be Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida; a typical species of the genus Artemisia may be Artemisia vulgaris; a typical species of the genus Betula may be Betula verrucosa; a typical species of the genus Fagus may be Fagus grandifolia or Fagus sylvatica; a typical species of the genus Quercus may be Quercus alba, a typical species of the genus Fraxinus may be Fraxinus excelsior; a typical species of the genus Olea may be Olea Europaea, a typical species of the genus plantago may be Plantago lanceolata, a typical species of the genus plantago may be Plantago lanceolata, a typical species of the genus
  • Non-limiting examples of non-grass pollen allergens to which a subject optionally may be sensitized to are Aln g 1 , Aln g 4, Amb a 1 , Amb a 2, Amb a 3, Amb a 4, Amb a 5, Amb a 6, Amb a 7, Amb a 8, Amb a 9, Amb a 10, Amb p 5, Amb t 5, Art v 1 , Art v 2, Art v 3, Art v 4, Art v 5, Art v 6, Bet v 1 , Bet v 2, Bet v 3, Bet v 4, Bet v 6, Bet v 7, Car b 1 , Cas s 1 , Cor a 6, Cor a 10, Fag s 1 , Fra e 1 , Hel a 1 , Hel a, Lig v 1 , Ole e 1 , Ole e 2, Ole e 3, Ole e 4, Oie e 5, Ole e 6, Ole e 7, Ole e 8, Ole e 9,
  • Non-limiting examples of grass pollen allergens to which a subject optionally may be sensitized to are Ant o 1 , Cyn d 1 , Cyn d 7, Cyn d 12, Cyn d 15, Cyn d 22w, Cyn d 23, Cyn d 24, Dac g 1 , Dac g 2, Dac g 3, Dac g 4, Dac g 5, Fes p 4, Hoi 1 1 , Hoi I 5, Hor v 1 , Hor v 5, Lol p 1 , Lol p 2, Lol p 3, Lol p 4, Lol p 5, Lol p 11 , Ory s 1 , Pas n 1 , Pha a 1 , Pha a 5, Phi p 1 , Phi p 2, Phi p 4, Phi p 5, Phi p, Phi p 7, Phi p 1 1 , Phi p 12, Phi p 13, Poa p 1 , Poa p 5, Sec c 1 , Sec c 5, Sec
  • the group 1 allergens e.g. Ant o 1 , Cyn d 1 , Dac g 1 , Hoi 1 , Lol p 1 , Pha a 1 , Phi p 1 and Poa p 1
  • group 5 allergens Dac g 5, Lol p 5, Pha a 5, Phi p 5, Poa p 5
  • “Prophylaxis” and grammatical variations thereof mean a method or use in which contact, administration or in vivo delivery to a subject is prior to contact with or exposure to a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein.
  • a pollen allergen or a molecule e.g. an immunogenic molecule
  • administration or in vivo delivery to a subject can be performed prior to manifestation of an allergic immune response, or one or more symptoms caused by or associated with an allergen.
  • a subject can be provided protection against an allergic immune response, or one or more symptoms caused by or associated with a pollen allergen or a molecule disclosed herein or provided immunotherapy with a molecule (e.g.
  • an immunogenic molecule of the present invention.
  • a method or use can eliminate, prevent, inhibit, suppress, limit, decrease or reduce the probability of or susceptibility towards an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with an a pollen allergen or a molecule disclosed herein.
  • “Prophylaxis” can also refer to a method or use in which contact, administration or in vivo delivery to a subject is prior to a secondary or subsequent exposure to an allergen (e.g. pollen such as a pollen allergen) and/or a molecule disclosed herein.
  • an allergen e.g. pollen such as a pollen allergen
  • a subject may have had a prior contact or exposure to a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein.
  • an acute allergic reaction may but need not be resolved.
  • Such a subject typically may have developed anti- allergen antibodies due to the prior exposure. Immunization or vaccination, by
  • Such a method or use can eliminate, prevent, inhibit, suppress, limit, decrease or reduce the probability of or susceptibility towards a secondary or subsequent allergic immune response, or one or more symptoms caused by or associated with a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein.
  • a method or use includes providing specific immunotherapy to the subject to eliminate, prevent, inhibit, suppress, limit, decrease or reduce the probability of or susceptibility towards a secondary or subsequent allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with an a pollen allergen or a molecule disclosed herein.
  • Treatment of an allergic reaction or response can be at any time during the reaction or response.
  • a molecule e.g. an immunogenic molecule
  • a molecule can be administered as a single or multiple dose e.g., one or more times hourly, daily, weekly, monthly or annually or between about 1 to 10 weeks, or for as long as appropriate (e.g. 3 months, 6 months or more, for example, to achieve a reduction in the onset, progression, severity, frequency, duration of one or more symptoms or complications associated with or caused by an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with an antigen/allergen.
  • methods and uses of the invention can be practiced one or more times (e.g., 1-10, 1-5 or 1-3 times) an hour, day, week, month, or year.
  • Doses can be based upon current existing protocols, empirically determined, using animal disease models or optionally in human clinical trials.
  • Initial study doses can be based upon animal studies, e.g. a mouse, and the sufficient amount of immunogenic molecule to be administered for being effective can be determined.
  • Exemplary non-limiting amounts (doses) are in a range of about 0.1 mg/kg to about 100 mg/kg, and any numerical value or range or value within such ranges.
  • doses can be administered, for example, 0.01-500 mg/kg, and any numerical value or range or value within such ranges.
  • the dose can be adjusted according to the mass of a subject, and will generally be in a range from about 1-10 ug/kg, 10-25 ug/kg, 25-50 ug/kg, 50-100 ug/kg, 100-500 ug/kg, 500-1 ,000 ug/kg, 1-5 mg/kg, 5-10 mg/kg, 10-20 mg/kg, 20-50 mg/kg, 50-100 mg/kg, 100-250 mg/kg, 250-500 mg/kg, or more, two, three, four, or more times per hour, day, week, month or annually.
  • a typical range will be from about 0.3 mg/kg to about 50 mg/kg, 0-25 mg/kg, or 1.0-10 mg/kg, or any numerical value or range or value within such ranges.
  • Doses can vary and depend upon whether the treatment is prophylactic or therapeutic, whether a subject has been previously exposed to the antigen/allergen, the onset, progression, severity, frequency, duration, probability of or susceptibility of the symptom, condition, pathology or complication, or vaccination or specific immunotherapy to which treatment is directed, the clinical endpoint desired, previous or simultaneous treatments, the general health, age, gender, race or immunological competency of the subject and other factors that will be appreciated by the skilled artisan. The skilled artisan will appreciate the factors that may influence the dosage and timing required to provide an amount sufficient for providing a therapeutic or prophylactic benefit.
  • Immunogens of the invention can be provided in compositions, and in turn such compositions can be used in accordance with the invention methods and uses.
  • Such compositions, methods and uses include pharmaceutical compositions and formulations.
  • a pharmaceutical composition includes one or more
  • compositions and formulations may be a vaccine, including but not limited to a vaccine to protect against (e.g. modify, relieve or suppress) an immune response disclosed herein, or one or more symptoms caused by or associated with an allergen and/or a molecule (e.g. an immunogenic molecule) disclosed herein.
  • a vaccine to protect against (e.g. modify, relieve or suppress) an immune response disclosed herein, or one or more symptoms caused by or associated with an allergen and/or a molecule (e.g. an immunogenic molecule) disclosed herein.
  • a pharmaceutical composition comprises a molecule (e.g. an immunogenic molecule) of the invention and a pharmaceutically acceptable ingredient or carrier.
  • a pharmaceutically acceptable and “physiologically acceptable” mean a biologically acceptable formulation, gaseous, liquid or solid, or mixture thereof, which is suitable for one or more routes of administration, in vivo delivery or contact.
  • Such formulations include solvents (aqueous or non-aqueous), solutions (aqueous or non-aqueous), emulsions (e.g., oil-in-water or water-in-oil), suspensions, syrups, elixirs, dispersion and suspension media, coatings, isotonic and absorption promoting or delaying agents, compatible with pharmaceutical administration or in vivo contact or delivery.
  • Aqueous and non-aqueous solvents, solutions and suspensions may include suspending agents and thickening agents.
  • Such pharmaceutically acceptable carriers include tablets (coated or uncoated), capsules (hard or soft), microbeads, powder, granules and crystals.
  • Supplementary active compounds e.g., preservatives,
  • antibacterial, antiviral and antifungal agents can also be incorporated into the composition.
  • a pharmaceutically acceptable or physiologically acceptable excipient, carrier and/or adjuvants are well-known to the person skilled in the art and may include, but are not limited to, solvents, emulsifiers, wetting agents, plasticizers, solubilizers (e.g. solubility enhancing agents) coloring substances, fillers, preservatives, anti-oxidants, anti-microbial agents, viscosity adjusting agents, buffering agents, pH adjusting agents, isotonicity adjusting agents, mucoadhesive substances, and the like. Examples of formulation strategies are well-known to the person skilled in the art.
  • the pharmaceutical composition may be formulated for parenteral administration, such as formulated for injection, e.g.
  • the pharmaceutical composition may be a liquid (i.e. formulated as a liquid), including a solution, a suspension, a dispersion, and a gelled liquid.
  • a liquid pharmaceutical composition may be formed by dissolving a powder, granulate or lyophilizate of a molecule (e.g. an immunogenic molecule) combination described herein in a suitable solvent and then administering to a subject.
  • Suitable solvents may be any solvent having physiologically acceptable properties and able to dissolve the immunogenic molecule combination in desired concentrations. A desired concentration may depend on the aliquot to be administered (i.e. to be injected) and the desired single dose.
  • a liquid composition comprises each of the immunogenic molecules of the combination in a concentration of 10 to 800 ⁇ , for example 20 to 500 ⁇ or20 to 300 ⁇ .
  • concentration of each immunogenic molecule is the same, such as in an equimolar concentration, but each immunogenic molecule of the composition may be present in different concentrations.
  • the solvent is an aqueous solution, optionally mixed with other solvents.
  • a solvent may comprise at least 60% w/w of water, e.g. at least 65% w/w, 70% w/w, 75% w/w, 80% w/w , 85% w/w, 90% w/w or 95% w/w , 99% w/w of water, such as distilled water, such as sterile water.
  • the solvent is sterile distilled water, e.g. water for injection.
  • An aqueous solution may comprise other solvents than water, for example
  • the aqueous phase of the solvent may be in a physiological acceptable range, typically in the range of 3 to 9, such as in the range of pH 3 to 8, such as in the range of pH 4 to 8, such as in the range of 5 to 8, such as in the range of 6 to 8.
  • the liquid formulation may comprise a pH controlling agent or buffering agent (e.g. citrate buffer, phosphate buffer, acetate buffer), optionally the pH may be adjusted with dilutions of strong base (e.g. sodium hydroxide or the like) and/or dilutions of strong acids (e.g.
  • the liquid formulation is isotonic, and optionally sterile. Therefore, in some embodiments, the formulation comprises saline, such as isotonic saline.
  • the liquid may contain additional excipients, such as another solvent, a solubilizing enhancing agent (e.g. polyoxyethylene (20) sorbitan monolaurate (Tween® 20), ionic and non-ionic emulsifiers (e.g. poloxamers (Kolliphor®)), a dispersant, a thickener, a preservative, an anti-microbial agent, and/or an antioxidant.
  • a solubilizing enhancing agent e.g. polyoxyethylene (20) sorbitan monolaurate (Tween® 20
  • ionic and non-ionic emulsifiers e.g. poloxamers (Kolliphor®)
  • a dispersant e.g. poloxamers (Kolliphor®)
  • a thickener e.g.
  • Non-limiting illustrative examples of solvents include water, saline, DMSO, glycerol, ethanol, acetonitrile, vegetable or synthetic oils.
  • solvents include water, saline, DMSO, glycerol, ethanol, acetonitrile, vegetable or synthetic oils.
  • a pharmaceutical composition may be formulated to contain only a limited amount of water or aqueous solution, e.g. containing less than 10% w/w of water or aqueous solution, such as less than 9, 8, 7, 6, 5, 4, 3, 2, 1 , 0.5% w/w of water or aqueous solution.
  • aqueous solution e.g. containing less than 10% w/w of water or aqueous solution, such as less than 9, 8, 7, 6, 5, 4, 3, 2, 1 , 0.5% w/w of water or aqueous solution.
  • Examples of pharmaceutical compositions with limited levels of water may include granulates, powders, for example lyophilizates, i.e. freeze-dried powders.
  • the freeze-dried composition may be dissolved before use, for example dissolved in an aqueous, optionally sterile, solution, for example a solution having a pH in the range of 3-9, such as pH in the range of 3 to 8, such as pH in the range of 4 to 8.
  • a lyophilizate may contain additional ingredients, e.g. bulking agents and lyoprotectants (e.g. sucrose, lactose, trehalose, mannose, mannitol, sorbitol, glucose, raffinose, glycine, histidine or mixtures thereof), buffering agents (e.g.
  • solubilizers e.g. polyoxyethylene (20) sorbitan monolaurate (Tween® 20)
  • a freeze-dried composition may also be formulated into a solid dosage form that is administered for example by the oral route such as by oral mucosa.
  • the pharmaceutical composition may be formulated for oral administration, for example for sublingual administration. Therefore, the pharmaceutical composition may be a solid dosage form, such as a freeze-dried solid dosage form, typically a tablet, a capsule or sachet, which optionally may be formulated for fast disintegration.
  • compositions, methods and uses of the invention are known in the art (see, e.g., Remington: The Science and Practice of Pharmacy (2003) 20th ed., Mack Publishing Co., Easton, PA; Remington's Pharmaceutical Sciences (1990) 18th ed., Mack Publishing Co., Easton, PA; The Merck Index (1996) 12th ed., Merck Publishing Group, Whitehouse, NJ;
  • compositions can be formulated to be compatible with a particular route of administration, such as by intradermal or by sublingual administration.
  • pharmaceutical compositions may include carriers, diluents, or excipients suitable for administration by various routes.
  • routes of administration for contact or in vivo delivery for which a composition can optionally be formulated include inhalation, intranasal, oral, buccal, sublingual, subcutaneous, intradermal, epicutaneous, rectal, transdermal, or intralymphatic.
  • a composition may take the form of, for example, tablets or capsules, optionally formulated as fast-integrating tablets/capsules or slow-release tablets/capsules.
  • the tablet is freeze-dried, optionally a fast-disintegrating tablet or capsule suitable for being administered under the tongue.
  • the pharmaceutical composition may also be formulated into a "unit dosage form", which used herein refers to physically discrete units, wherein each unit contains a
  • Unit dosage forms also include, for example, ampules and vials, which may include a composition in a freeze-dried or lyophilized state (a lyophilizate) or a sterile liquid carrier, for example that can be added prior to administration or delivery in vivo.
  • Unit dosage forms also include a composition in a freeze-dried or lyophilized state; a sterile liquid carrier, for example, can be added prior to administration or delivery in vivo.
  • Pharmaceutical formulations can be packaged in single or multiple unit dosage form for ease of administration and uniformity of dosage.
  • Immunogenic molecules may be prone to degradation when exposed to oxygen, for example when exposed to air or solvents containing air. Therefore, in some
  • the pharmaceutical composition comprises an inert gas, e.g. argon or nitrogen.
  • Another aspect of the invention relates to a kit comprising a compartment and
  • kits wherein the compartment comprises a pharmaceutical composition as described herein and wherein the instructions are for use in reducing an immune response triggered by pollen, such as a pollen allergen, e.g. of a grass, weed or tree pollen species disclosed herein, such as instructions for use in treating allergy to pollen (e.g. grass, weed and/or tree pollen allergy to a plant family or species disclosed herein.
  • a kit may further comprise packaging material comprising corrugated fiber, glass, plastic, foil, ampules, vials, blister pack, preloaded syringes or tubes, optionally that maintain sterility of the components.
  • a kit may further comprise labels or inserts comprising printed matter or computer readable medium optionally including identifying components, dose amounts, clinical pharmacology and instructions for the clinician or for a subject using one or more of the kit components, prophylactic or therapeutic benefits, adverse side effects or manufacturer information.
  • the kit additionally comprises a container comprising a solvent for dissolving the composition before use. Examples of suitable solvents are described supra.
  • the kit may also comprise a device for use in parenteral injection, e.g. for injecting the composition (e.g. dissolved composition) to a subcutaneous or intradermal tissue.
  • a device may be any suitable device for that purpose, such as a needle or microneedle adapted for intradermal or subcutaneous delivery of the composition.
  • the device may be a microneedle or a device comprising a plurality of microneedles designed for intradermal delivery of liquids, e.g. as described in international patent applications W014064543 A1 , WO05049107 A2, WO06054280 A2, WO07066341 A3 and W014188429 A1.
  • a composition may be lyophilized so as to enhance stability and ease of transportation.
  • the composition may be sterile.
  • compositions can be formulated to be compatible with a particular route of administration.
  • pharmaceutical compositions include carriers, diluents, or excipients suitable for administration by various routes. Exemplary routes of
  • the pharmaceutical composition is aqueous and, in other embodiments, the composition is non-aqueous solutions, suspensions or emulsions of the immunogenic molecule/protein, which compositions are typically sterile and can be isotonic with the biological fluid or organ of the intended recipient.
  • Non-limiting illustrative examples include water, saline, dextrose, fructose, ethanol, vegetable or synthetic oils.
  • a composition can take the form of for example a solid dosage form, e.g. tablets or capsules, optionally formulated as fast- integrating tablets/capsules or slow-release tablets/capsules.
  • the tablet is a freeze-dried, optionally fast-disintegrating tablet suitable for being administered under the tongue.
  • a solid dosage form optionally is sterile, optionally anhydrous.
  • immunogenic molecules can be mixed with adjuvants.
  • Adjuvants include, for example: oil (mineral or organic) emulsion adjuvants such as Freund's complete (CFA) and incomplete adjuvant (I FA) (WO 95/17210; WO 98/56414; WO 99/12565; WO 99/11241 ; and U.S. Patent No.
  • oil mineral or organic
  • I FA incomplete adjuvant
  • metal and metallic salts such as aluminum and aluminum salts, such as aluminum phosphate or aluminum hydroxide, alum (hydrated potassium aluminum sulfate); bacterially derived compounds, such as Monophosphoryl lipid A and derivatives thereof (e.g., 3 De-O-acylated monophosphoryl lipid A, aka 3D-MPL or d3-MPL, to indicate that position 3 of the reducing end glucosamine is de-O-acylated, 3D-MPL consisting of the tri and tetra acyl congeners), and enterobacterial lipopolysaccharides (LPS); plant derived saponins and derivatives thereof, for example Quil A (isolated from the Quilaja Saponaria Molina tree, see, e.g., "Saponin adjuvants", Archiv.
  • Quil A isolated from the Quilaja Saponaria Molina tree, see, e.g., "Saponin adjuvants", Archiv.
  • Cosolvents may be added to the composition.
  • cosolvents contain hydroxyl groups or other polar groups, for example, alcohols, such as isopropyl alcohol; glycols, such as propylene glycol, polyethyleneglycol, polypropylene glycol, glycol ether; glycerol; polyoxyethylene alcohols and polyoxyethylene fatty acid esters.
  • cosolvents contain hydroxyl groups or other polar groups, for example, alcohols, such as isopropyl alcohol; glycols, such as propylene glycol, polyethyleneglycol, polypropylene glycol, glycol ether; glycerol; polyoxyethylene alcohols and polyoxyethylene fatty acid esters.
  • Supplementary compounds can also be incorporated into the compositions.
  • Pharmaceutical compositions may therefore include preservatives, anti-oxidants and antimicrobial agents.
  • Preservatives can be used to inhibit microbial growth or increase stability of ingredients thereby prolonging the shelf life of the pharmaceutical formulation.
  • Suitable preservatives are known in the art and include, for example, EDTA, EGTA, benzalkonium chloride or benzoic acid or benzoates, such as sodium benzoate.
  • Antioxidants include, for example, ascorbic acid, vitamin A, vitamin E, tocopherols, and similar vitamins or provitamins.
  • An antimicrobial agent or compound directly or indirectly inhibits, reduces, delays, halts, eliminates, arrests, suppresses or prevents contamination by or growth, infectivity, replication, proliferation, reproduction, of a pathogenic or non- pathogenic microbial organism.
  • Classes of antimicrobials include antibacterial, antiviral, antifungal and antiparasitics.
  • Antimicrobials include agents and compounds that kill or destroy (-cidal) or inhibit (-static) contamination by or growth, infectivity, replication, proliferation, reproduction of the microbial organism.
  • compositions, methods and uses of the invention are known in the art (see, e.g. Remington: The Science and Practice of Pharmacy (David B. Troy, Paul Beringer Lippincott Williams & Wilkins) 2006).
  • compositions peptides, proteins, antigens, allergens
  • substituents described herein are disclosed by the application to the same extent as if each composition or group of compositions was set forth individually. Thus, selection of particular peptides, proteins, antigens, allergens, etc. is clearly within the scope of the invention.
  • any concentration range, percentage range, ratio range or other integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • numerical values are often presented in a range format throughout this document, a range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention.
  • range expressly includes all possible sub ranges, all individual numerical values within that range, and all numerical values or numerical ranges including integers within such ranges and fractions of the values or the integers within ranges unless the context clearly indicates otherwise.
  • This construction applies regardless of the breadth of the range and in all contexts throughout this patent document.
  • reference to a range of 90-100% includes 91-99%, 92-98%, 93-95%, 91-98%, 91-97%, 91-96%, 91-95%, 91- 94%, 91-93%, and so forth.
  • Reference to a range of 90-100% includes 91%, 92%, 93%, 94%, 95%, 95%, 97%, etc., as well as 91.1%, 91.2%, 91.3%, 91.4%, 91.5%, etc., 92.1%, 92.2%, 92.3%, 92.4%, 92.5%, etc., and so forth.
  • Reference to a range of 5-10, 10-20, 20- 30, 30-40, 40-50, 50-75, 75-100, 100-150, and 150-175, includes ranges such as 5-20, 5- 30, 5-40, 5-50, 5-75, 5-100, 5-150, 5-171 , and 10-30, 10-40, 10-50, 10-75, 10-100, 10- 150, 10-175, and 20-40, 20-50, 20-75, 20-100, 20-150, 20-175, and so forth.
  • reference to a series of ranges of 2-72 hours, 2-48 hours, 4-24 hours, 4-18 hours and 6-12 hours includes ranges of 2-6 hours, 2, 12 hours, 2-18 hours, 2-24 hours, etc., and 4-27 hours, 4-48 hours, 4-6 hours, etc.
  • Bostick D Vaisman II. A new topological method to measure protein structure similarity. Biochem Biophys Res Commun. 304, 320-325, 2003.
  • HLA human leukocyte antigen
  • Needleman SB Wunsch CD. A general method applicable to the search for similarities in the amino acid sequence of two proteins, J. Mol. Biol. 48:443, 1970.
  • This example includes a description of transcriptomic analysis of various pollen species.
  • RNA-sequencing was performed on pollen samples of the following species: Timothy grass (Phleum pratense (Phi p)), Bermuda grass (Cynodon dactylon (Cyn d)), Western ragweed (Ambrosia psilostachya (Amb p)), Short ragweed (Ambrosia artemisiifolia (Amb a)), White oak (Quercus alba (Que a)), and European white birch (Betula verrucosa (Bet v)) (See Table below).
  • Timothy grass Phleum pratense
  • Bermuda grass Cronodon dactylon
  • Western ragweed Ambrosia psilostachya
  • Short ragweed Ambrosia artemisiifolia (Amb a)
  • White oak Quercus alba (Que a)
  • European white birch Betula verrucosa (Bet v)
  • the table below shows the number of reads assembled for each of the different pollens (top), with over 500 million reads over two replicate runs per allergen. Sequences were assembled into transcripts using Trinity (bottom), resulting in over 50 thousand transcripts per allergen with minimum lengths of 200 nucleotides.
  • the transcripts include related variants, such as isoforms, and homologs.
  • RNA sequences of additional species may also be performed on RNA sequences of additional species as shown in the Table below showing pollen RNA-seq reads for the pollen species Kentucky blue grass, Sweet vernal grass, Rye grass and Ash tree, Olive tree and English plantain:
  • This example includes a description of the identification of polypeptides in extracts of Phi p grass pollen and homologous polypeptides thereof in other pollen species.
  • the Phi p grass transcriptome was translated into amino acid sequences in all six reading frames, and they were used to search 17 mass spectrometry samples of Phi p grass pollen extract using the Mascot software. This identified 354 polypeptide sequences. Polypeptide sequences shorter than 50 aa were discarded, resulting in 275 sequences.
  • the closest homologous sequence in the rice (Oryza sativa japonica) proteome via Blast. Rice was chosen since it is a species closely related to Phi p grass with a completely sequenced and annotated genome. Homologous rice sequences were identified for 180 Timothy grass sequences.
  • homologous sequences were identified (via Blast) in the translated transcriptomes of Cyn d, Amb a, Amb p, Que a, and Bet v of all identified sequences, the one(s) sharing the largest number of conserved peptides with the Phi p sequence was selected as homolog. This resulted in 66 sets of homologous polypeptides. In total, there was found evidence of release of the polypeptides from Phi p grass pollen for 52 of these polypeptides upon extracting pollen in a buffered aqueous solution (see Example 4).
  • Table 1 shows amino acid sequences of the polypeptides detected and identified in Phi p grass pollen and Table 2 shows amino acid sequences of polypeptides homologous to the Phi p sequence of Table 1 found in pollen of the species Cyn d, Amb a, Amb p, Que a, and Bet v.

Abstract

The invention relates to polypeptides identified in pollen of several different plant families. The polypeptides comprises sequences of amino acid residues conserved across the different plant families, such as across grass, weed and tree pollen families. The invention further relates to the use of such polypeptides or their conserved amino acid residues as a molecule (e.g. an immunogenic molecule) in modifying an immune response against itself or another unrelated antigen (e.g. major allergen of a pollen source) possible via induction of bystander tolerance.

Description

POLYPEPTIDES DERIVED FROM PHL P AND METHODS AND USES THEREOF FOR IMMUNE RESPONSE MODULATION
Government support
This invention received government support from the National Institutes Health contract NIH-NIAIDHHSN272200700048C. The government has certain rights in the invention.
Field of the invention
The invention relates to pan pollen immunogenic molecules such as polypeptides, proteins and peptides, and methods and uses of such immunogenic molecules for modulating or relieving an immune response in a subject, such as treating a subject for an allergic immune response or inducing or promoting immunological tolerance to the immunogenic molecule or a pollen allergen in a subject. Introduction
Patients with pollen allergies are typically poly-sensitized as evidenced by positive RAST- and/or skin prick tests to multiple pollen allergens, like grass, weed and tree pollen allergens. However, today it is not possible to treat multisensitized patients with one immunotherapeutic product. Although several investigators have suggested that immunotherapy with a single grass species such as Timothy grass (Phleum Pratense (Phi p)) is sufficient to also treat allergies to other grass pollens due to observed cross- reactivity at the IgE level, it has not been suggested to treat multiple pollen allergies with one single immunogenic molecule.
It is firmly established that allergen-specific T-cells play an important role in allergic inflammation and that induction of antigen specific T regulatory cells (Tregs) or elimination of allergen-specific T helper type 2 cells (Th2) might be a prerequisite for the induction of specific tolerance. Yet, cross-reactivity among multiple pollen families at the T-cell level is less explored. Allergen-specific immunotherapy (SIT) is a hyposensitizing immunotherapy introduced in clinical medicine almost a century ago for the treatment of an allergic immune response using the allergens that the subject is sensitized to. An allergic immune response may be mediated by activated allergen-specific Th2 cells, which produce cytokines such as IL-4, IL-5, and IL-13. In healthy individuals, the allergen-specific T-cell response is mediated predominantly by Th1 cells. SIT may reduce the ratio of Th2:Th1 cells and may alter the cytokine profile, reducing the production of IL-4, IL-5, and IL-13 and increasing the production of IFN-gamma in response to major allergens or allergen extracts.
Despite its efficacy, SIT has several limitations, including safety concerns about giving patients allergenic substances. Because most SIT regimens involve the administration of whole, unfractionated, allergen extracts, adverse IgE-mediated events are a considerable risk. Significant efforts have been devoted to developing approaches to modulate allergen-specific T-cell responses without inducing IgE-meditated, immediate-type reactions. These approaches include developing hypoallergens that do not contain IgE- binding epitopes, allergens that are coupled to adjuvants and carriers of bacterial or viral origin or peptides that contain dominant T-cell epitopes and do not react with IgE in allergic individuals.
It was recently shown that a large fraction of Timothy Grass-specific T cells target epitopes contained in novel Timothy Grass antigens (NTGA). NTGA's are unrelated to the known allergens of Timothy grass, which mainly are identified based on their high IgE reactivity. International patent application, WO2013/119863 A1 , relates to novel antigens (NTGA's) derived from Timothy grass pollen.
It has also recently been shown and described in International patent application
WO2012/049310 that an immunogenic molecule derived from an allergenic pollen source is able to reduce an allergic immune response caused by an unrelated allergen of the same pollen source via bystander suppression.
As disclosed herein, it is possible to provide immunogenic molecules with high sequence conservation/homology to polypeptides identified in several different pollen families. Such immunogenic molecules have potential therapeutical utilization against immune responses triggered by pollen allergens of a broad array of pollen families for example via induction of bystander tolerance towards the offending pollen allergens. Summary
Disclosed herein are immunogenic molecules (in short immunogens), also named pan- pollen immunogens, which have been detected in Timothy grass pollen (Phleum pratense (in short Phi p) and in at least one non-grass pollen species, which consist of or contain as part of their sequence an amino acid sequence conserved across the Phi p grass pollen species and at least one of the non-grass pollen species investigated. For example, a molecule (e.g. an immunogenic molecule) of the invention may consist of or contain as part of its sequence a conserved amino acid sequence detected in several different pollen families, e.g. in a grass pollen species (e.g. Phleum Pratense (Phi p) and at least one of the following non-grass pollen species: Ambrosia psilostachya (Amb p), Ambrosia artemisiifolia, (Amb a), Quercus alba (Que a) and Betula verrucosa, (Bet v). Table 1 shows examples of amino acid sequences of "pan pollen" polypeptides detected and identified in Phi p grass pollen and Table 2 shows amino acid sequences of polypeptides homologous to the polypeptides of Table 1 , which is found in pollen of the species Cyn d, Amb a, Amb p, Que a, and Bet v. As those polypeptides can be detected in pollen, they are referred to herein as wild type polypeptides.
Disclosed herein are also conserved regions, i.e. stretches of amino acid residues of the polypeptides of Table 1 and 2 that are conserved across at least one set of homologous polypeptides found in Phi p grass pollen and in at least one weed pollen species (Amb a or Amp) and in at least one tree pollen species (Que a or Bet v). Such stretches are herein named GWT (Grass-Weed-Tree) conserved stretches of amino acid residues (or alternatively GWT conserved regions). Table 3 shows GWT conserved regions identified in the Phi p sequences of Table 1. Table 4 shows the corresponding GWT conserved regions identified in the other pollen species investigated. A conserved region as used herein is defined as the region identified from merging overlapping conserved 15mer peptides in a Phi p sequence, wherein a conserved 15mer peptide is defined as a 15 amino acid peptide containing 0, 1 , or 2 mismatches to a 15mer peptide sequence of a homologous polypeptide in a weed pollen species (Amb a and/or Amb) and in a tree pollen species (Que a and/or Bet v). In addition, there is provided herein conserved amino acid sequences of the polypeptides of Table 1 and 2 that are conserved across at least a set of homologous polypeptides found in Phi p grass pollen and in at least one weed pollen species (Amb a or Amp), but not in a tree pollen species (Que a or Bet v). Such sequences are herein named GW (Grass-Weed) conserved stretches of amino acid residues (or alternatively GW conserved regions). Table 5 shows GW conserved regions identified in the Phi p sequences of Table 1. Table 6 shows the corresponding GW conserved regions identified inthe other pollen species.
Also there is provided herein conserved amino acid sequences of the polypeptides of Table 1 and 2 that are conserved across at least a set of homologous polypeptides found in Phi p grass pollen and in at least one tree pollen species (Que a or Bet v), but not in a weed pollen species (Amb a or Amp). Such sequences are herein named GT (Grass- Tree) conserved stretches of amino acid residues (or alternatively GT conserved regions). Table 7 shows GW conserved regions identified in the Phi p sequences of Table 1. Table 8 shows the corresponding GT conserved regions identifiedin the other pollen species.
Also provided herein are conserved amino acid sequences of the polypeptides of Table 1 and 2 that are conserved across at least a set of homologous polypeptides found in Amb a pollen and in at least one tree pollen species (Que a or Bet v), but not found in Phi p pollen. Such sequences are herein named WT (Weed-Tree) conserved stretches of amino acid residues (or alternatively WT conserved regions). Table 9 shows WT conserved regions identified in polypeptides of Table 2.
The GWT, GW, GT or WT conserved sequences may be an immunogenic molecule in itself, or may give rise to additional immunogens comprising the conserved sequences or subsequences thereof. In certain embodiments, a molecule (e.g. an immunogenic molecule) may contain at least one T cell epitope, as may be determined by the T cell response observed against immunogens disclosed in Tables 1 to 9 when cultured together with PBMC's obtained from grass pollen allergic donors or alternatively from ragweed, oak and/or birch pollen allergic donors. In some embodiments, an immunogenic molecule of the invention which is identified in Phi p pollen may be able to modify a T cell response in a grass allergic donor as well as in donors allergic to a weed pollen species and/or tree pollen species. This is evidenced using Phi p allergens and a selection of recently identified Phi p antigens as model immunogens (Example 8). It was found that a T cell response of grass allergic donors to a first immunogenic molecule derived from Phi p may also be activated in detectable levels by a second immunogenic molecule containing few mismatches to the first immunogenic molecule, e.g. preferably no more than 2 or 3 mismatches.
Therefore, the presently provided conserved sequences shown in Tables 3 to 8 may be able to induce a T cell response in donors not only sensitized to Phi p pollen allergens, but to allergens of any of the pollen species Cyn d, Amb a, Amb p, Que a and Bet v as well as other pollen species containing polypeptides with similar conserved sequences comprising less than 2 or 3 mismatches to the Phi p sequence in question.
Therefore, the invention relates in a first aspect to a molecule, such as an immunogenic molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293 set out in Table 3; SEQ ID NOS: 617-676, 294-616 and 677-767 set out in Table 4; SEQ ID NOS: 768-808 set out in Table 5; SEQ ID NOS: 809-951 set out in Table 6; SEQ ID NOS: 952-1023 set out in Table 7; SEQ ID NOS: 1024-1231 set out in Table 8 and SEQ ID NOS: 1232-1473 set out in Table 9; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293 set out in Table 3; SEQ ID NOS: 617-676, 294-616 and 677-767 set out in Table 4; SEQ ID NOS: 768-808 set out in Table 5; SEQ ID NOS: 809-951 set out in Table 6; SEQ ID NOS: 952- 1023 set out in Table 7; SEQ ID NOS: 1024-1231 set out in Table 8 and SEQ ID NOS: 1232-1473 set out in Table 9; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293 set out in Table 3; SEQ ID NOS: 617-676, 294-616 and 677-767 set out in Table 4; SEQ ID NOS: 768-808 set out in Table 5; SEQ ID NOS: 809-951 set out in Table 6; SEQ ID NOS: 952-1023 set out in Table 7; SEQ ID NOS: 1024-1231 set out in Table 8 and SEQ ID NOS: 1232-1473 set out in Table 9; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 30, 1-29, 31- 37 set out in Table 1 or SEQ ID NOS: 161-172, 38-160 and 173-211 set out in Table 2.
The invention also relates to the use of said molecule (e.g. said immunogenic molecule) as a medicament, in particularly for use in modulating an immune response, e.g. relieving an immune response, triggered by pollen (i.e. a pollen allergen and/or an immunogenic molecule disclosed herein) in a subject. Thus, in further aspects, the invention relates to a method for modulating an immune response, e.g. for relieving an immune response (e.g. triggered by pollen or an immunogen present in pollen), in a subject in need thereof, comprising administering an effective amount of a molecule (e.g. an immunogenic molecule) disclosed herein. In still further aspects, the invention relates to an immunogenic molecule as disclosed herein for use in modulating an immune response, e.g. for relieving an immune response, in a subject in need thereof, comprising administering an effective amount of a molecule (e.g. an immunogenic molecule) disclosed herein.
The invention also relates to the use of a molecule (e.g. an immunogenic molecule) as disclosed herein as a medicament, e.g. use of an immunogenic molecule as disclosed herein for the preparation of a medicament for modulating an immune response, e.g. relieving an immune response, in a subject in need thereof.
The immune response in question may be triggered by pollen, e.g. by pollen of one or more pollen species, pollen genera or pollen families. It follows that the immune response in question may be triggered by one or more pollen allergens from one or more pollen species, pollen genera or pollen families including one or more molecules (e.g.
immunogenic molecules) disclosed herein, for example the immunogenic molecule itself or a subsequence thereof.
Below is provided example embodiments specifically related to each set of pan-pollen immunogens presently identified. For example, in embodiment X, the set of polypeptides identified in Phi p grass pollen and the other pollen species investigated are commonly numbered A0349. In such embodiments, a polypeptide of option d) comprises an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 27-30 set out in Table 1 and SEQ ID NOS: 161-172 set out in Table 2; a polypeptide of option a) comprises an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4; a polypeptide of option b) comprises an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4, and a polypeptide of option c) includes at least one amino acid sequence with 0, 1 , 2, 3 or 4 mismatches compared to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4. Other embodiments (A to AE) may be constructed the same way using the list below:
Figure imgf000009_0001
Figure imgf000010_0001
In some embodiments, the molecule (e.g. immunogenic molecule) may comprise a combination of two or more polypeptides of option a), option b), option c) or option d), so as to construct molecules with desirable properties e.g. immunogenic molecules with high conservation throughout the entire amino acid sequence. For example, a polypeptide of option a) may contain as part of its sequence a combination of two or more amino acid sequences having at least 65% similarity or identity to a sequence selected from any one of SEQ ID NOS: 212-293 set out in Table 3 and SEQ ID NOS: 294-767 set out in Table 4. Likewise, a polypeptide of option b) and c) may comprise as part of its sequence a combination of two or more of said subsequences.
Also provided are cells expressing a molecule (e.g. immunogenic molecule) described herein. In various embodiments, a cell expresses said molecule. In certain aspects, a cell is a eukaryotic or prokaryotic cell and may be a mammalian, insect, fungal or bacterium cell. A molecule (e.g. an immunogenic molecule) of the present invention is suitable as a reagent, for example in immunotherapy against various pollen allergies in a subject.
In other embodiments, there are provided nucleic acid molecules encoding a polypeptide of option a), b), c) or d) or a molecule comprising a polypeptide of option a), b), c) or d).
In additional aspects, there are provided compositions, for example pharmaceutical compositions comprising an immunogenic molecule of the invention. In one embodiment, a pharmaceutical composition is suitable for immunotherapy (e.g. treatment,
desensitization, tolerance induction, bystander suppression (bystander tolerance induction). In certain embodiments, a pharmaceutical composition is a vaccine, e.g.
suitable formulated for the purpose of vaccination.
Brief Description of the Drawings
Figure 1 : Conservation in transcriptome predicts peptide cross-reactivity. For each peptide, Phi p allergic donors were selected that reacted to the peptide after expanding PBMCs in vitro with Phi p extract. PBMCs were stimulated with individual peptides for 14 days and IL-5 responses were measured by ELISPOT to i) the peptide itself, ii) Phi p extract, iii) non-Phi p extracts (e.g. Amb a, Que, Ole e, Bet v, Cyn d), iv) pools of predefined peptide pools that did or did not contain the peptide as relevant and irrelevant controls. T cell cultures that did not induce a robust response (>=200 SFC) to the peptide itself were excluded. Reponses to extracts and peptide pools are expressed as the relative fraction of the response to the peptide itself, and capped at 100%.
Figure 2: Sensitization pattern of an immunogen of the invention (A0349): It is shown that the in vitro T-cell response towards A0349 is much weaker compared to the response to allergen Phi p 5.
Figures 3 Tolerance induction investigated in mice: shows that prophylactic sublingual immunotherapy treatment (SLIT) with A0349 in mice is capable of inducing tolerance towards Phi p extract as shown by the ability of A0349 to reduce the
proliferation of cells of splenocytes from treated mice compared to buffer (sham) treated mice. Thus, an immunogenic molecule that is weaker than Phi p 5, the major allergen in Phi p extract, is able to reduce an immune response triggered by Phi p extract.
Figure 4: Bystander tolerance induction investigated in mice. As shown in Figure 4A, prophylactic SLIT treatment with A0349 could induce tolerance toward the allergen Phi p 5, which amino acid sequence is quite different from A0349. Thus, treatment with A0349 resulted in the suppression of an immune response caused by an unrelated antigen, i.e. Phi p 5, possibly via bystander mechanisms, where the tolerance induction towards one immunogen (e.g. A0349) result in suppression of an immune response caused by another unrelated immunogen (e.g. Phi p 5).
Detailed description
The following terms and phrases shall have the following meaning:
The term "a" or "an" refers to an indefinite number and shall not only be interpreted as "one" but also may be interpreted to mean "some", "several" or one or more. The term "conserved sequence" is in the present context meant to include that a given amino acid sequence of one pollen species contains at least 15 contiguous amino acids within the sequence that has less than 3 mismatches compared to an amino acid sequence of 15 amino acid residues from another pollen species. Longer stretches of conserved sequences may contain several stretches of at least 15 contiguous amino acids having less than 3 mismatches compared to another sequence of 15 amino acids from another pollen species, genera or family. The conserved region is usually determined by multiple sequence alignments of three or more homologous pollen polypeptides. The terms "polypeptide", "protein" and "peptide" may be used interchangeably and mean any peptide-linked chain of amino acids regardless of post-translational modification. While polypeptides can be any length, the use of the term "protein" generally means longer polypeptides of at least 100 amino acid residues, whereas the term "peptide" generally means polypeptides shorter than 30 amino acid residues. In the present context, e.g. for the purpose of detecting a conserved sequence of a polypeptide herein, the term "mismatch" is meant to include any substitution of an amino acid residue within the 15mer peptide. Optionally, a mismatch may be a deletion or an addition of an amino acid residue within the 15mer peptide.
As used herein, the term "homologous polypeptides" refer to a pollen polypeptide derived from a common ancestor and are typically, although not necessarily, polypeptides having one or more similar functions. Typically, homologous pollen polypeptides will have a degree of sequence identity to each other across the entire length of each protein that is at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or greater. Typically, homologous pollen polypeptides are found in pollens of different species, genera and families.
The term "sensitized to" is generally meant to encompass that the subject has been exposed to an immunogenic molecule, e.g. an allergen or an antigen, in a manner that the individual's adaptive immune system displays memory to the immunogenic molecule, for example that the immunogenic molecule has induced detectable IgE antibodies against the immunogenic molecule and thus qualifies as an IgE-reactive antigen
(allergen) and/or that T-cells stimulated in vitro are able to proliferate under the presence of the immunogenic molecule or fragments of the immunogenic molecule (e.g. linear peptides derived from the immunogenic molecule).
The term "allergic immune response" is meant to encompass a hypersensitivity immune response, e.g. a type 1 hypersensitivity immune response, such as typically an immune response that is associated with the production of IgE antibodies (i.e. IgE-mediated immune response) and/or production of cytokines usually produced by Th2 cells. An allergic immune response may be associated with an allergic disease, for example atopic dermatitis, urticaria, contact dermatitis, allergic conjunctivitis, allergic rhinitis, allergic asthma, anaphylaxis, food allergy and hay fever. The term "grass pollen" is meant to designate pollen of the plant family Poaceae, for example pollen of the plant genus Anthoxanthum, Cynodon, Dactylis, Festuca, Holcus, Hordeum, Lolium, Oryza, Paspalum, Phalaris, Phleum, Poa, Secale, Sorghum, Triticum or Zea.
As used herein, an "immunogenic molecule" refers to a substance, including but not limited to a molecule comprising as part of its structure or exclusively a protein, a polypeptide or a peptide, which optionally modifies, e.g. elicits, induces, stimulates, promotes enhances or decreases, reduces, inhibits, suppresses, relieves an immune response when administered to a subject, for example laboratory mice or modifies, e.g. elicits, induces, stimulates, promotes enhances or decreases, reduces, inhibits, suppresses or relieves a T cell response in vitro in response to the immunogenic molecule. For example, an immunogenic molecule may induce tolerance to itself in a subject. An immune response elicited by an immunogenic molecule may include, but is not limited to, a B cell or a T cell response. An immune response can include a cellular response with a particular pattern of lymphokine/cytokine production (e.g., Th1 , Th2), a humoral response (e.g., antibody production, like IgE, IgG or IgA), or a combination thereof, to a particular immunogenic molecule.
The term "an antigen" refers to a particular substance to which an immunoglobulin (Ig) isotype may be produced in response to the substance. For example, an "IgG antigen" refers to an antigen that induces an IgG antibody response. Likewise, an "IgE antigen" refers to an antigen that induces an IgE antibody response (and thus qualifies as an allergen); an "IgA antigen" refers to a substance that induces an IgA antibody response, and so forth. In certain embodiments, such an immunoglobulin (Ig) isotype produced in response to an antigen may also elicit production of other isotypes. For example, an IgG antigen may induce an IgG antibody response in combination with one more of an IgE, IgA, IgM or IgD antibody response. In other embodiments, an IgG antigen may induce an IgG antibody response without inducing a response to one more of an IgE, IgA, IgM or IgD antibody response. Accordingly, in certain embodiments, an IgG antigen may induce an IgG antibody response without inducing an IgE, IgA, IgM or IgD antibody response. The term "allergen" refers to a particular type of a substance that can elicit production of IgE antibodies, such as in predisposed subjects. For example, if a subject previously exposed to an allergen (i.e. is sensitized or is hypersensitive) comes into contact with the allergen again, allergic asthma may develop due to a Th2 response characterized by an increased production of type 2 cytokines (e.g., IL-4, IL-5, IL-9, and/or IL-13) secreted by CD4+ T lymphocytes.
The term "immunotherapy" is meant to encompass treatment of a disease by inducing, enhancing, or suppressing an immune response. Typically, the therapeutically active agent is an immunogenic molecule, particularly an antigen, more particularly an allergen. An immunogenic molecule may be a protein or a fragment thereof (e.g. immunogenic peptide including a linear peptide). Immunotherapy in connection with allergy usually encompasses repeated administration of a sufficient dose of the immunogenic molecule/antigen/allergen, usually in microgram quantities, over a prolonged period of time, usually for more than 3 months, 6 months, 1 year, such as 2 or 3 years, during which period the immunogenic molecule may be administered daily or less frequently, such as several times a week, weekly, bi-weekly, or monthly, every second month or quarterly. Immunotherapy can be effected by specific immunotherapy or may be effected by bystander tolerance induction.
The term "specific immunotherapy" in connection with allergy is meant to designate that immunotherapy is conducted with the administration of an immunogenic molecule to which the subject is sensitized to, particularly an immunogenic molecule to which the subject has raised specific IgE antibodies to, e.g. major allergens.
As used herein, the term "immunological tolerance" refers to a) a decreased or reduced level of a specific immunological response (thought to be mediated at least in part by antigen-specific effector T lymphocytes, B lymphocytes, antibodies or a combination thereof); b) a delay in the onset or progression of a specific immunological response; or c) a reduced risk of the onset or progression of a specific immunological response to an immunogenic molecule, such as an antigen or an allergen. "Specific" immunological tolerance occurs when tolerance is preferentially invoked against certain immunogens in comparison with other immunogens. Tolerance is an active immunogenic molecule dependent process and differs from non-specific immunosuppression and immunodeficiency.
The term "bystander tolerance induction" in connection with treatment of allergy is meant to encompass that immunotherapy is conducted with the administration of an
immunogenic molecule that elicits, induces, stimulates, promotes enhances or decreases, reduces, inhibits, suppresses, relieves an immune response against another unrelated immunogenic molecule, for example an allergen, e.g. major allergens of pollen. For example, an immunogenic molecule may induce immunological tolerance to itself, and may be able to reactivate T regulatory cells specific to the immunogenic molecule to down-regulate an immune response caused by another unrelated immunogenic molecule, e.g. an allergen. Thus, an immunogenic molecule may induce immunological tolerance to another unrelated antigen, e.g. an allergen including a pollen allergen described herein. Thus, one immunogenic molecule may provide bystander tolerance induction to another unrelated antigen, e.g. the immunogenic molecule may provide suppression of an immune response triggered by an unrelated antigen (e.g. allergen) via bystander mechanisms.
The term "treatment" refers to any type of treatment that conveys a benefit to a subject afflicted with allergy or at least sensitized to an allergen , including improvement in the condition of the subject (e.g., in one or more symptoms), delay in the onset of symptoms, slowing the progression of symptoms, or induce disease modification etc. Typical symptoms of an allergic reaction are nasal symptoms, such as itchy nose, sneezing, runny nose, blocked nose; conjunctival symptoms, such asitchy eyes, red eyes, watery eyes; and respiratory symptoms, such as decreased lung function. The treatment may also give the benefit that the patient needs less concomitant treatment with
corticosteroids or H1 antihistamines to suppress the clinical symptoms. As used herein, "treatment" is not necessarily meant to imply cure or complete abolition of symptoms, but refers to any type of treatment that imparts a benefit to a patient. Treatment may be initiated before the subject becomes sensitized to a protein. This may be realized by initiating immunotherapy before the subject has raised detectable serum IgE antibodies capable of binding specifically to the sensitizing protein or before any other biochemical marker indicative of an allergic immune response can be detected in biological samples isolated from the individual. Furthermore, treatment may be initiated before the subject has evolved clinical symptoms of the allergic disease, such as symptoms of allergic rhinitis, allergic asthma or atopic dermatitis. The phrase "therapeutically sufficient amount" or "sufficient amount" is meant to designate an amount effective to reduce, suppress, relieve or eliminate an allergic immune response, e.g. an amount sufficient to achieve the desirable reduction in clinical relevant symptoms or manifestations of the allergic immune response. For example, a therapeutically sufficient amount may be the accumulated dose of a polypeptide, a set of polypeptides administered during a course of immunotherapy in order to achieve the intended effect or it may be the maximal dose tolerated within a given period. The total dose or accumulated dose may be divided into single doses administered daily, twice a week or more, weekly, every second or fourth week or monthly depending on the route of administration and the pharmaceutical formulation used. The total dose or accumulated dose may vary. It is expected that a single dose is in the microgram range, such as in the range of 5 to 500 microgram dependent on the nature of the polypeptide.
The term "patient responding to therapy," such as "immunotherapy" is meant to designate that the patient has improvement in the symptoms of the allergic immune response caused by a pollen allergen. Symptoms may be the clinically symptoms of allergic rhinitis, allergic asthma allergic conjunctivitis, atopic dermatitis, food allergy and/or hay fever. Typically, the symptoms are the same as experienced with a flu/cold such as sneezing, itching, congestion, coughing, feeling of fatigue, sleepiness and body aches. For example nasal symptoms may be itchy nose, sneezing, runny nose, blocked nose; conjunctival symptoms may be itchy eyes, red eyes, watery eyes; and respiratory symptoms may be decreased lung function. A responder may also be evaluated by monitoring the patient's reduced need for concomitant treatment with corticosteroids or H1 antihistamines to suppress the clinical symptoms. Symptoms may be subjectively scored or in accordance with official guidelines used in clinical trials of SIT. The term "adjuvant" refers to a substance that enhances the immune response to an immunogenic molecule. Depending on the nature of the adjuvant, it can promote either a cell-mediated immune response, humoral immune response or a mixture of the two.
As used herein an "epitope" refers to a region or part of an immunogenic molecule that elicits an immune response when administered to a subject. In particular embodiments, an epitope is a T cell epitope, i.e., an epitope that elicits, stimulates, induces, promotes, increases or enhances a T cell activity, function or response. An immunogenic molecule can be analyzed to determine whether it include at least one T cell epitope using any number of assays (e.g. T cell proliferation assays, lymphokine secretion assays, T cell non-responsiveness studies, etc.). In the context of the present invention, a T-cell epitope refers to an epitope that are MHC Class II binders (i.e. HLA-II binders), for example an epitope able to connect to/ associate with or bind to a HLA-II molecule shown in Tables 11 b or 11c.
As used herein, the term "immune response" includes T cell (cellular) mediated and/or B cell (humoral) mediated immune responses, or both cellular and humoral responses. Exemplary immune responses include T cell responses, e.g., lymphokine production, cytokine production and cellular cytotoxicity. T-cell responses include Th1 and/or Th2 responses. In addition, the term immune response includes responses that are indirectly affected by T cell activation, e.g., antibody production (humoral responses) and activation of cytokine responsive cells, e.g., eosinophils, macrophages. Immune cells involved in the immune response include lymphocytes, such as T cells (CD4+, CD8+, Th1 and Th2 cells, memory T cells) and B cells; antigen presenting cells (e.g., professional antigen presenting cells such as dendritic cells, macrophages, B lymphocytes, Langerhans cells, and non-professional antigen presenting cells such as keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes); natural killer (NK) cells; myeloid cells, such as macrophages, eosinophils, mast cells, basophils, and granulocytes.
The term "subsequence" or "stretch" means a fragment or part of a longer molecule, e.g. of a full length molecule (e.g. the wild type polypeptides shown in Tables 1 and 2) or of a conserved region thereof (e.g. GWT sequences shown in Tables 3 and 4). A
subsequence therefore consists of one or more amino acids less than the wild type polypeptide or a conserved region thereof.
As disclosed herein, a molecule (e.g. an immunogenic molecule) of the invention comprises conserved amino acid sequences detected in a grass pollen species as well as in a weed pollen species and/or a tree pollen species. Thus, such molecules can be used to broadly treat a subject with or at risk of developing an immune response to pollen of a variety of pollen species, genera or families, or to broadly induce or promote tolerance of a subject to pollen of a variety of pollen species, genera or families and may include promoting or inducing tolerance to the immunogenic molecule itself or another immunogenic molecule (e.g. an allergen) unrelated to the immunogenic molecule being administered, for example via induction of bystander tolerance towards an offending allergen.
Thus, by the present invention it is now possible to modify, such as relieving, an immune response in a multisensitized subject, e.g. a subject sensitized to both grass, weed and tree pollen, by administering a molecule (e.g. an immunogenic molecule) described herein. Likewise, it is also now possible to treat subjects with different pollen allergies using the same molecule or set of molecules.
In certain embodiments, the molecule (e.g. the immunogenic molecule) comprises or consists of a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches compared to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 212-293 (e.g. SEQ ID NOS: 274, 270-273, 212-269 and 275-293) set out in Table 3 or SEQ ID NOS: 294-767 (e.g. SEQ ID NOS: 665-676, 617-664 and 677-767) set out in Table 4.
The molecule (e.g. the immunogenic molecule) may comprise one or more T cell epitope(s) optionally a Th-2 cell epitope (i.e. the molecule may comprise one or more T cell epitope-containing amino acid sequence(s)). Thus, in some embodiments, the polypeptide of option a), option b), option c) and option d) comprises a T cell epitope, optionally a Th-2 cell epitope. Accordingly, the GWT sequences (SEQ ID NOS:212-293 set out in Table 3 and SEQ ID NOS: 294-767 set out in Table 4), the GW sequences (SEQ ID NOS: 768-808 set out in Table 5 and SEQ ID NOS: 809-951 set out in Table 6), GT sequences (SEQ ID NOS: 952-1023 set out in Table 7 and SEQ ID NOS: 1024-1231 set out in Table 8) and WT sequences (SEQ ID NOS: 1232-1473 set out in Table 9) or subsequences thereof may comprise a T cell epitope, optionally a Th-2 cell epitope.
Thus, a subsequence of a polypeptide of option b) or option c) contains a T cell epitope, optionally a Th-2 cell epitope. It follows that the sequences selected from any of SEQ ID NOS: 1-37 (e.g. SEQ ID NOS: 30, 1-29, 31-37 set out in Table 1 and SEQ ID NOS: 38- 211 (e.g SEQ ID NOS: 161-172, 38-160 and 173-211) set out in Table 2 contains a T cell epitope, optionally a Th-2 cell epitope.
Advantageously, immunogens of the present invention may be used in relieving an immune response against phi p grass pollen, non-phi p grass pollen as well as non-grass pollen to the extent that the pollen species contain polypeptides comprising essentially the same GWT, GW, GT or WT conserved region. For example, an immunogenic molecule of the present invention may at least be used in modulating, e.g. relieving, an immune response triggered by pollen of the grass pollen species Phi p and/or Cyn d as well as pollen of other species closely related to Phi p or Cyn d, e.g. pollen species also belonging to the plant family Poales, such as species of the plant genera selected from any of Anthoxanthum, Cynodon, Dactylis, Lolium, Phleum or Poa. The same
immunogenic molecule may also be used in modulating, e.g. relieving, an immune response triggered by pollen of the weed pollen species Amb a and/or Amb p and pollen of species closely related to Amb a and Amp p, e.g. pollen species also belonging to the plant family Asteraceae, such as species of the plant genera selected from any of Ambrosia, Artemisia, Helianthus. In addition, the same immunogenic molecule may also be used in modulating, e.g. relieving, an immune response triggered by pollen of the tree pollen species Que a and/or Bet v and pollen of species closely related to Que a and/or Bet v, e.g. pollen species also belonging to the plant families Betulaceae, Fagaceae and Oleaceae, such as species of the plant genera selected from any of Alnus, Betula, Carpinus, Castanea, Corylus, Fagus, Quercus, Fraxinus and Ligustrum. As disclosed herein, there can be found several GWT conserved sequences in pollen species of the plant genera Phleum, Cynodon, Ambrosia and Quercus and Betula. Thus, in certain embodiments, a molecule (e.g. an immunogenic molecule) of the present invention may be used in modulating, e.g., relieving, an immune response triggered by pollen of a plant genus selected from any of Phleum, Cynodon Ambrosia, Quercus and Betula.
The following Table lists pollen species of the plant families Asteraceae, Betulaceae, Fagaceae, Oleaceae, Plantaginaceae and Poaceae. Pollen species used for the conservation analysis described herein are highlighted in grey color.
As mentioned, conserved amino acid sequences disclosed herein have been detected in about five different pollen species and may be found in additional pollen species selected from any of the plant families Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, e.g. the plant genera Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea or Plantago using the methodology disclosed herein (see exemplary pollen species below):
CateID Common Latin name of Genus Family Order gory Name species
Weed Giant ragweed Ambrosia trifida Ambrosia Asteraceae Asterales Weed Amb a Short Ambrosia Ambrosia Asteraceae Asterales
Ragweed artemisiifolia
Weed Amb p Western Ambrosia Ambrosia Asteraceae Asterales ragweed psilostachya
Weed Mugwort Artemisia Artemisia Asteraceae Asterales vulgaris
Weed Sunflower Helianthus Helianthus Asteraceae Asterales annuus
Tree Common Alnus glutinosa Alnus Betulaceae Fagales
Alder
Tree Bet European Betula Betula Betulaceae Fagales white birch Verrucosa Cate- ID Common Latin name of Genus Family Order gory Name species
Tree Common Carpinus betulus Carpinus Betulaceae Fagales
Hornbeam
Tree European Castanea sativa Castanea Betulaceae Fagales
Chestnuts
Tree Common Corylus avellana Corylus Betulaceae Fagales
Hazel
Tree European Ostrya Ostrya Betulaceae Fagales
Hop- carpinifolia
hornbeam
Tree Hazel- Ostryopsis Ostryopsis Betulaceae Fagales hornbeam
Tree American Fagus Fagus Fagaceae Fagales
Beech grandifolia
Tree European Fagus sylvatica Fagus Fagaceae Fagales beech
Tree Que a White Oak Quercus alba Quercus Fagaceae Fagales Tree Fra e European Ash Fraxinus Fraxinus Oleaceae Lamiales
Excelsior (Oleales)
Tree Common Ligustrum Ligustrum Oleaceae Lamiales
Privet vulgare (Oleales)
Tree Lilac Syringa vulgaris Lilac Oleaceae Lamiales
(Oleales)
Tree Ole e European Olea Europaea Olea Oleaceae Lamiales
Olive (Oleales)
Herb Pla I English Plantago Plantago Plantagina- Lamiales plantain lanceolata ceae (Oleales)
Grass Ant o Sweet vernal Anthoxanthum Anthoxanthum Poaceae Poales grass odoratum
Grass Cyn d Bermuda Cynodon Cynodon Poaceae Poales grass dactylon
Grass Orchard Grass Dactylis Dactylis Poaceae Poales glomerata L.
Grass Meadow Festuca Festuca Poaceae Poales fescue pratensis
Grass Velvet Grass Holcus lanatus Holcus Poaceae Poales Grass Barley Hordeum Hordeum Poaceae Poales vulgare
Grass Lol p Rye grass Lolium Perenne Lolium Poaceae Poales
Grass Rice Oryza sativa Oryza Poaceae Poales
Grass Bahia grass Paspalum Paspalum Poaceae Poales notatum
Grass Canary Grass Phalaris Phalaris Poaceae Poales aquatica CateID Common Latin name of Genus Family Order gory Name species
Grass Phl p Timothy grass Phleum Phleum Poaceae Poales
Pratense
Grass Poa p Kentucky blue Poa pratensis Poa Poaceae Poales grass
Grass Rye Secale Cereale Secale Poaceae Poales
Grass Johnson grass Sorghum Sorghum Poaceae Poales halepense
Grass Wheat Triticum Triticum Poaceae Poales aestivum
Grass Maize Zea mays Zea Poaceae Poales
For example, conserved regions may be found in pollen of other weeds, grasses or tree species, e.g. found in one or more plant species selected from further plant families, like the plant family Chenopidiaceae including the plants Lambs quarters, Russian thistles and Kochias; plant family Amaranthaceae including Pigweeds, plant family Polygonaceae including for example Sheep sorrel, plant family Ulmacea including for example American Elm and Hackberry, plant family Plantanaceae including for example Sycamore, plant family Salicaceae including for example White poplar and Cottonwood, plant family Aceraceae including for example Box elder and Red maple, plant family Cupressaceae including for example Common juniper and Cedar.
Non-limiting examples of species of the genus Ambrosia is Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida; a typical species of the genus Artemisia is Artemisia vulgaris; non-limiting examples of species of the genus Betula is Betula verrucosa; non-limiting examples of species of the genus Fagus is Fagus grandifolia or Fagus sylvatica; non-limiting examples of species of the genus Quercus is Quercus alba, non-limiting examples of species of the genus Fraxinus is Fraxinus excelsior; a typical species of the genus Olea is Olea Europaea, a typical species of the genus plantago is Plantago lanceolata, a typical species of the genus plantago is Plantago lanceolata, non- limiting examples of species of the genus Anthoxanthum is Anthoxanthum odoratum, non-limiting examples of species of the genus Conydon is Conydon dactylon, non-limiting examples of species of the genus Lollium is Lollium perenne, non-limiting examples of species of the genus Phleum is Phleum pratense, non-limiting examples of species of the genus Poa is Poa pratensis.
In particular embodiments, a molecule of the invention may modulate an immune response at least triggered by a grass pollen and/or a weed pollen by administering to a subject in need thereof a molecule comprising a GWT conserved sequence set out in Tables 3 or 4 or a subsequence thereof or by administering to a subject in need thereof a molecule comprising a GW conserved sequence set out in Tables 5 or 6 or a
subsequence thereof. In still other particular embodiments the molecule may modulate an immune response triggered by at least a grass pollen and/or a tree pollen by
administering to a subject in need thereof a molecule comprising a GWT conserved sequence set out in Tables 3 or 4 or a subsequence thereof or by administering to a subject in need thereof a molecule comprising a GT conserved sequence set out in Tables 7 or 8 or a subsequence thereof. In more interesting embodiments thereof, the molecule modulate an immune response triggered by at least a grass pollen, a weed pollen and a tree pollen by administering to a subject in need thereof a molecule comprising a GWT conserved sequence set out in Tables 3 or 4 or a subsequence thereof.
As mentioned, a molecule of the invention may modulate, e.g. relieve, an immune response triggered by pollen. Pollen may comprise allergens considered as major and minor allergens according to official guidelines in the art and may in addition comprise additional immunogens, for example a molecule as disclosed herein. A molecule of the invention may be able to relieve an immune response triggered by a major and/or a minor pollen allergen and/or a molecule disclosed herein including the molecule itself.
Molecules (e.g. immunogens) eligible for relieving an immune response triggered by an allergen (e.g. a major allergen or minor allergen) unrelated to the molecule is thought, at least in part, to be mediated via induction of bystander tolerance, which mechanism requires, at least in part, co-existence of the triggering allergen and the unrelated molecule at the target organ for the immune response. The co-existence may inherently be obtained by using molecules present in the same pollen source as the allergen. It is also considered that, in an optimal setting, the triggering allergen and the unrelated molecule may be released from the same pollen source within an overlapping period of time (co-release of triggering allergen and unrelated molecule (e,g. immunogenic molecule)) to ensure co-existence at the target organ. Co-release can be examined in vitro using hydrated pollen as described in Example 4 herein.
Therefore, in some embodiments, the polypeptide of option a), b), c) or d) may be derived, be a part of or comprise a wild type polypeptide that co-releases/co-elutes from the same pollen source with the major or minor allergens the subject is sensitized to and to which allergens the immune response is sought to be relieved. Thus, a molecule with the potential to relieve an immune response triggered by pollen of several different species, genera and families may be identified by investigating the presence of the molecule or a fragment thereof in pollen diffusates of pollen of different species, genera or families including the grass pollen, weed pollen or tree pollen species used for the present investigation within a time period overlapping with the relevant major and/or minor allergen.
In the present context, the term "co-release" or "co-elute" may refer to when a molecule starts to release from pollen, e.g. hydrated pollen, within a period overlapping with a major allergen to which the allergic immune response is sought to be relieved. Major allergens start to release from pollen within few minutes after hydration of pollen and continue to be released within the next 30 or 60 minutes. Thus, the term "co-release" or "co-elute" may refer to when a molecule of the invention starts to release from pollen together with a major allergen, usually within 30 minutes after hydration of the pollen. Co- release/co-elution may be determined by a method comprising extracting pollen in an aqueous solution having pH in the range of 6-8 for a period ranging from 1 to 200 minutes, optionally the aqueous solution comprises at least 60% of water, a buffering agent; a tonicity providing agent. For example, the aqueous solution has an ionic strength corresponding to that of isotonic saline (0.9 g of NaCI in 1 liter of water), e.g. the aqueous solution has an ionic strength in the range of 10 mM to 1000 mM and optionally the aqueous solution has a pH of about 7. Typically, the period of extraction is in the range of 1-180, 1-120 minutes, 5-120 minutes, 5 to 90 minutes, for example 5-60 minutes, for example 5-45 minutes, for example 5-30 minutes. For example some immunogenic molecules may be released from various distinct pollen species, such as from grass pollen species as well as weed and/or tree pollen species.
For example, a polypeptide of option a), option b), option c) or option d), as described herein, may be derived from a wild type polypeptide that co-releases with a major allergen from grass pollen (e.g. pollen of the genera Phleum and or Cynodon) and from a weed pollen (e.g. pollen of the genera Ambrosia) and from tree pollen (e.g. pollen of the genera Quercus or Betula).
For example, it was found that some of the immunogens disclosed herein can be released from grass pollen and weed pollen as well as tree pollen, which polypeptides are herein named "GWT polypeptides", such as the set of polypeptides referred to with polypeptide ID NOs: A0349, A0246, A0209, A0211 , A0325, A0357 and A0203. As shown in Table 11a, these polypeptides have predicted HLA Class II allele binding sites in their conserved part of their sequence, thereby indicating the polypeptides potentially comprises a T cell epitope in their conserved region.
By the present analysis, it was also found that polypeptide A0262 (herein named GT polypeptide) was released from both grass and tree pollen, but not from weed pollen. It was also found that the set of polypeptides having the polypeptide ID NOs A0362,
A0316, A0356, A0376, A0336, A0377 and A0366 could be released from grass as well as weed pollen, but not the tree pollen investigated (herein named GW polypeptides).
Accordingly, in specific embodiments, the molecule (e.g. the immunogenic molecule) comprises a conserved region or a subsequence thereof of the "GWT polypeptide" A0349, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and SEQ ID NOS: 617-676 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 set out in Table 3 and
SEQ ID NOS: 617-676 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 27-30 set out in Table 1 and SEQ ID NOS: 161-172 set out in Table 2. In still specific embodiments, the molecule comprises a conserved region or a
subsequence thereof of the "GWT polypeptide" A0246, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 233 set out in Table 3 and SEQ ID NOS: 414-418 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 233 set out in Table 3 and SEQ ID NOS: 414-418 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 233 set out in Table 3 and SEQ ID NOS: 414-418 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 12 set out in Table 1 and SEQ ID NOS: 77-81 set out in Table 2.
In still specific embodiments, the molecule comprises a conserved region or a
subsequence thereof of the "GWT polypeptide" A0209, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 217-220 set out in Table 3 and SEQ ID NOS: 321-344 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any of SEQ ID NOS: SEQ ID NOS: 217-220 set out in Table 3 and SEQ ID NOS: 321-344 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 217-220 set out in Table 3 and SEQ ID NOS: 321-344 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 4,5 set out in Table 1 and SEQ ID NOS: 49-54 set out in Table 2.
In still specific embodiments, the molecule comprises a conserved region or a
subsequence thereof of the "GWT polypeptide" A0211 , such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 221-225 set out in Table 3 and SEQ ID NOS: 345-386 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 221-225 set out in Table 3 and SEQ ID NOS: 345-386 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 221-225 set out in Table 3 and
SEQ ID NOS: 345-386 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS:6-8 set out in Table 1 and SEQ ID NOS: 55-63 set out in Table 2. In still specific embodiments, the molecule comprises a conserved region or a
subsequence thereof of the "GWT polypeptide" A0325, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS:256 set out in Table 3 and SEQ ID NOS: 541-547 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS:256 set out in Table 3 and SEQ ID NOS: 541-547 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS:256 set out in Table 3 and SEQ ID NOS: 541-547 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 22 set out in Table 1 and SEQ ID NOS: 132-138 set out in Table 2. In still specific embodiments, the molecule comprises a conserved region or a
subsequence thereof of the "GWT polypeptide" A0357, such as a molecule comprising or consisting of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 280-281 set out in Table 3 and SEQ ID NOS: 701-709 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 280-281 set out in Table 3 and SEQ ID NOS: 701-709 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 280-281 set out in Table 3 and SEQ ID NOS: 701-709 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 32 set out in Table 1 and SEQ ID NOS: 179-183 set out in Table 2.
In still specific embodiments, the molecule comprises a conserved region or a
subsequence thereof of the "GWT polypeptide" A0203, such as a molecule comprising, consisting of or consisting essentially of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 215, 216 set out in Table 3 and SEQ ID NOS: 309-320 set out in Table 4; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 215, 216 set out in Table 3 and SEQ ID NOS: 309-320 set out in Table 4; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 215, 216 set out in Table 3 and SEQ ID NOS: 309-320 set out in Table 4; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 2, 3 set out in Table 1 and SEQ ID NOS: 43-48 set out in Table 2.
In some embodiments, a molecule of the present invention is an IgE reactive molecule, e.g. able to bind to IgE antibodies specific for the immunogenic molecule. However, IgE reactivity towards a molecule of the invention may only be conferred by a low fraction of an allergic population. Thus, a molecule of the invention may not fall under the usual definitions of a major allergen, which is an allergen with high prevalence in a population of donors allergic to a pollen species, for example the prevalence may be higher than at least 50%. In some embodiments, the molecule is able to react with, bind to or induce IgG antibodies in a subject, at least in detectable levels. In still other embodiments, the molecule does not react with, bind to or induce IgG antibodies, at least in detectable levels. As demonstrated herein, a molecule of the invention seems to be less
immunogenic than the major allergen Phi p 5 (Figure 2), but still able to induce bystander tolerance (i.e. reducing an immune response) towards an unrelated molecule, such as Phi p 5 allergen (Figure 4).
As disclosed herein, polypeptides containing GWT, GT, GW or WT conserved regions can be detected in various pollen species and families and share high identity and similarity across the various pollen species. Therefore, a polypeptide of option d) comprises an amino acid sequence having at least 70%, such as at least 75%, 80%,
85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity to a sequence selected from any one of SEQ ID NOS: 1-37 (e.g. SEQ ID NOS: 30, 1-29, 31- 37) set out in Table 1 and SEQ ID NOS: 38-211 (eg. SEQ ID NOS: 161-172, 38-160 and 173-211) set out in Table 2. Accordingly, a polypeptide of option a) comprises an amino acid sequence having at least 70% such as at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity to a sequence selected from any one of SEQ ID NOS: 212- 293 (e.g. SEQ ID NOS: 270-274; 212-269; 275-293) set out in Table 3; SEQ ID NOS: 294-767 (e.g. SEQ ID NOS: 617-676, 294-616 and 677-767) set out in Table 4; SEQ ID NOS: 768-808 set out in Table 5; SEQ ID NOS: 809-951 set out in Table 6; SEQ ID NOS: 952-1023 set out in Table 7; SEQ ID NOS: 1024-1231 set out in Table 8 and SEQ ID NOS: 1232-1473 set out in Table 9.
Accordingly, a polypeptide of option b) comprises an amino acid sequence having at least 70%, such as at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 212-293 (e.g. SEQ ID NOS: 270-274; 212-269; 275-293 set out in Table 3; SEQ ID NOS: 294-767 (e.g. SEQ ID NOS: 617-676, 294-616 and 665-676, 617-664, 294-616, 677-767) set out in Table 4; SEQ ID NOS: 768-808 set out in Table 5; SEQ ID NOS: 809-951 set out in Table 6; SEQ ID NOS: 952-1023 set out in Table 7; SEQ ID NOS: 1024-1231 set out in Table 8 and SEQ ID NOS: 1232-1473 set out in Table 9. Optionally, the at least 70%, such as at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% similarity or identity is of a subsequence consisting of at least 16 contiguous amino acid residues, for example at least 17, 18, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 90, 100 or more contiguous amino acids. A subsequence may comprise any number of amino acids, but typically the subsequence has a length of 15 to 50 amino acid residues, for example 15 to 45, 15 to 40, 15 to 35, 15 to 30; 15 to 25; 15 to 20; 15 to 19; 15 to 18; 15 to 17, or 15 to 16 amino acid residues. As shown herein, there is established evidence that an amino acid sequences having less than 3 mismatches to a T cell epitope containing amino acid sequence may be
recognized by the same T cell population. Therefore, in some embodiments, the number of amino acid mismatches may be 0, 1 , 2, or 3, preferably 0, 1 or 2, more preferably 0 or 1. In some embodiments, the polypeptide of option b) or option c) has a length of 15-800 or more amino acid residues, for example 15-750, 15-700, 15-650 or 15-600 or more amino acid residues, for example 15-20, 20-25, 25-30, 30-35, 35-40, 45-50, 50-60, 60-70, 70-80, 90-100, 100-125, 125-150, 150-175, 175-200, 200-250, 250-300, 300-350, 350-400, 400- 450, 450-500, 500-550, 550-600, 600-650, 650-700, 700-800 or more amino acid residues. The polypeptide of option a) or of option d) has a length of no more than 800 amino acid residues, for example no more than 750, 700, 650, 600, 550, 500, 450, 400, 350 or 300 amino acid residues.
As mentioned, a polypeptide disclosed herein including a subsequence thereof may contain a T cell epitope, such as a Th2 cell epitope. A subsequence or a polypeptide described herein may have HLA Class II binding properties. HLA Class II binding can be predicted using NetMHCIIpan-3.0 tool (Karosiene, Edita, Michael Rasmussen, Thomas Blicher, Ole Lund, S0ren Buus, and Morten Nielsen. "NetMHCIIpan-3.0, a Common Pan- specific MHC Class II Prediction Method Including All Three Human MHC Class II Isotypes, HLA-DR, HLA-DP and HLA-DQ." Immunogenetics) available at the internet site <URL: http://www.cbs. dtu.dk/services/NetMHCIIpan-3.0>. A polypeptide of option b) or c) may have different lengths according to the desirable use, for example of about 15-800 or more amino acid residues in length, for example 15-750, 15-700, 15-650, 15-600, 15-500 or more amino acid residues, for example 15-20, 15-25, 15-30, 20-25, 25-30, 30-35, 35-40, 45-50, 50-60, 60-70, 70-80, 90-100, 100-125, 125- 150, 150-175, 175-200, 200-250, 250-300, 300-350, 350-400, 400-450, 450-500, 500- 550, 550-600, 600-650, 650-700, 700-800 or more amino acid residues. One may consider utilizing short linear peptides, which when administered to a subject need not to be processed by an antigen presenting cells to interact with a relevant T cell receptor, but rather be freely loaded onto a MHC class II molecule to interact with the relevant T cell receptor. Thus, in some embodiments, a polypeptide of option b) or a polypeptide of option c) has a length in the range of 15 to 30 amino acid residues, for example 15 to 25 amino acid residues. In other embodiments, a polypeptide of option a), b), c) or d) is a longer polypeptide which comprises a secondary or tertiary structure, e.g. folded. Thus, in some embodiments, a polypeptide of option a), b), c) or d) has a length in the range of 30 to 500 amino acid residues or more. It is considered that the length of the amino acid sequence of a polypeptide of option a), b), c) or d) is no more than 800 amino acid residues, for example no more than 750, 700, 650, 600, 550, 500 or 450 amino acid residues.
The term "identity" and "identical" and grammatical variations thereof, as used herein, mean that two or more referenced entities are the same (e.g. two or more amino acid sequences). Thus, where two polypeptides are identical, they have the same amino acid sequence. The identity can be over a defined area (region or domain) of the sequence, e.g. over the sequence length of a sequence disclosed in Tables 1 to 9 or over a portion thereof e.g. at least 15 contiguous amino acid residues. Moreover, the identity can be over the length of the sequence overlapping the two polypeptides, when aligned with best fit with gaps permitted.
For example, to determine whether a polypeptide has at least 65% similarity or identity to a sequence set out in Tables 1 to 9, the polypeptide may be aligned with a sequence of Tables 1 to 9 and the percent identity be calculated with reference to said sequence. Identity can be determined by comparing each position in aligned sequences. A degree of identity between amino acid sequences is a function of the number of identical or matching amino acids at positions shared by the sequences, i.e. over a specified region. Optimal alignment of sequences for comparisons of identity may be conducted using a variety of algorithms, as are known in the art, including the Clustal Omega program available at http://www.ebi.ac.uk/Tools/msa/clustalo/, the local homology algorithm of Smith and Waterman, 1981 , Adv. Appl. Math 2: 482, the homology alignment algorithm of Needleman and Wunsch, 1970, J. Mol. Biol. 48:443, the search for similarity method of Pearson and Lipman, 1988, Proc. Natl. Acad. Sci. USA 85: 2444, and the computerized implementations of these algorithms (such as GAP, BESTFIT, FASTA and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, Madison, Wl, U.S.A.). Sequence identity may also be determined using the BLAST algorithm, described in Altschul et al., 1990, J. Mol. Biol. 215:403-10 (using the published default settings).
Software for performing BLAST analysis may be available through the National Center for Biotechnology Information (through the internet at http://www.ncbi.nlm.nih.gov/). Such algorithms that calculate percent sequence identity (homology) generally account for sequence gaps and mismatches over the comparison region or area. For example, a BLAST (e.g., BLAST 2.0) search algorithm (see, e.g., Altschul et al., J. Mol. Biol. 215:403 (1990), publicly available through NCBI) has exemplary search parameters as follows: Mismatch -2; gap open 5; gap extension 2. For polypeptide sequence comparisons, a BLASTP algorithm is typically used in combination with a scoring matrix, such as
PAM100, PAM 250, BLOSUM 62 or BLOSUM 50. FASTA (e.g., FASTA2 and FASTA3) and SSEARCH sequence comparison programs are also used to quantitate the extent of identity (Pearson et al., Proc. Natl. Acad. Sci. USA 85:2444 (1988); Pearson, Methods Mol Biol. 132: 185 (2000); and Smith et al., J. Mol. Biol. 147:195 (1981)). Programs for quantitating protein structural similarity using Delaunay-based topological mapping have also been developed (Bostick et al., Biochem Biophys Res Commun. 304:320 (2003)).
A polypeptide sequence is a "homolog" of, or is "homologous" to, another sequence if the two sequences have substantial identity over a specified region and a functional activity of the sequences is preserved or conserved, at least in part. (As used herein, the term 'homologous' does not infer nor exclude evolutionary relatedness).
Examples of "homologous polypeptides" of the invention include polypeptides with high similarity or identity to Phi P polypeptides and detected in pollen species other than Phi p. For example, a homologous polypeptide to a polypeptide having an amino acid sequence set out in Table 1 or Table 2 may be found in pollen of plant families selected among Asteraceae, Betulaceae, Fagaceae, Oleaceae, or Plantaginaceae, e.g. the plant genera Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea or Plantago.
Two polypeptide sequences are considered to be substantially identical if, when optimally aligned (with gaps permitted), they share at least about 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, etc. identify over a specific region), for example, over all or a part of any amino acid sequence in Tables 1 to 9, or if the sequences share defined functional motifs (e.g., epitopes). In particular aspects, the length of the sequence sharing the percent identity is at least 15, 16, 17, 18, 19, 20, etc. contiguous amino acids, e.g. more than 25, 30, 35, 40, 45 or 50 or more contiguous amino acids, including the entire length of a reference sequence of Tables 1 to 9.
An "unrelated" or "non-homologous" sequence is considered to share less than 30% identity. More particularly, it may share less than about 25 % identity, with a polypeptide of the invention over a specified region of homology.
An amino acid sequence set out in any of Tables 1 to 9 may contain one or more modifications, which optionally may result in greater or less activity or function, for example in the ability to elicit, stimulate, induce, reduce, inhibit, suppress an in vitro immune response (e.g. T cell proliferation or T cell cytokine production); in the ability to bind HLA Class II alleles; in the ability to induce or enhance immunological
tolerance(desensitize) to a relevant pollen allergen or a molecule disclosed herein; or in the ability to dissolve in solvents e.g. in an aqueous solution.
A modification includes deletions, including truncations and fragments; insertions and additions, substitutions, for example conservative substitutions, site-directed mutants and allelic variants.
Non-limiting examples of modifications include one or more amino acid substitutions (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20-25, 25-30, 30-50, 50-100 or more residues), additions and insertions (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15 or more residues) and deletions (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20-25, 25-30, 30-50, 50-100 or more) of a sequence set out in Tables 1 , 2, 3 and 4. The term "similarity" and "similar" and grammatical variations thereof, as used herein, mean that two or more referenced amino acid sequences contains a limited number of conservative amino acid substitutions of the amino acid sequence. A variety of criteria can be used to indicate whether amino acids at a particular position in a polypeptide are similar. In making such changes, substitutions of like amino acid residues can be made on the basis of relative similarity of side-chain substituents, for example, their size, charge, hydrophobicity, hydrophilicity, and the like, and such substitutions may be assayed for their effect on the function of the peptide by routine testing. A "conservative substitution" is the replacement of one amino acid by a biologically, chemically or structurally similar residue. Biologically similar means that the substitution does not destroy a biological activity. Structurally similar means that the amino acids have side chains with similar length, such as alanine, glycine and serine, or a similar size. Chemical similarity means that the residues have the same charge, or are either hydrophilic or hydrophobic. For example, a conservative amino acid substitution is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain, which include amino acids with basic side chains (e.g., lysine, arginine, histidine); acidic side chains (e.g., aspartic acid, glutamic acid); uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, histidine); nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); beta-branched side chains (e.g., threonine, valine, isoleucine), and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan). Particular examples include the substitution of one hydrophobic residue, such as isoleucine, valine, leucine or methionine for another or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acids, or glutamine for asparagine, serine for threonine, and the like. Proline, which is considered more difficult to classify, shares properties with amino acids that have aliphatic side chains (e.g., Leu, Val, lie, and Ala). In certain circumstances, substitution of glutamine for glutamic acid or asparagine for aspartic acid may be considered a similar substitution in that glutamine and asparagine are amide derivatives of glutamic acid and aspartic acid, respectively. Conservative changes can also include the substitution of a chemically derivatized moiety for a non- derivatized residue, for example, by reaction of a functional side group of an amino acid. Variants and derivatives of polypeptides include forms having a limited number of one or more substituted residues.
As mentioned, a polypeptide of option a), b), c) and d) may be longer than the reference sequence set out in Tables 1 to 9.
An addition can be one or more additional amino acid residues. For example, a polypeptide of option b) or c) may contain amino acid residues in addition to the subsequence of at least 15 amino acid residues. Optionally, the additional amino acid residues may be identical to those present in the wild type polypeptide from which the subsequence derives from. Thus, in some embodiments, the polypeptide of option b) comprises one or more amino acid residues in addition to the subsequence of at least 15 contiguous amino acids, wherein the additional amino acid residue(s) is/are selected from an amino acid residue or an amino acid sequence within the wild type polypeptide that the subsequence is a part of (e.g. wild type polypeptide sequences of Tables 1 or 2 or a GWT sequence of Tables 3 or 4). For example, the wild type amino acid residue or wild type amino acid sequence to be added may be adjacent to, subtended, comprised within, overlapping with or is a part of the subsequence, when present in its natural biological context within the wild type polypeptide.
Likewise, a polypeptide of option a) may contain additional amino acid residues in addition to the GWT sequence set out in Tables 3 and 4. Thus, a polypeptide of option a) may comprise one or more amino acid residues in addition to the GWT sequence set out in Tables 3 or 4, wherein the additional amino acid residue(s) is/are selected from an amino acid residue or an amino acid sequence within the wild type polypeptide of which the GWT sequence is a part of (e.g. a wild type polypeptide of Tables 1 or 2). An illustrative example is a GWT sequence of A0349 set out in Table 3 that may be extended with amino acid residues from polypeptide A0349 set out in Tables 1 or 2, such as amino acid residues adjacent to the GWT sequence when aligned with the corresponding wild type polypeptide.
The additional amino acid residues may be added to the N- and/or C- terminal end of a sequence set out in Tables 3 to 9, such as additional amino acids selected from amino acids flanking the N- and/or C- terminal ends when sequence is aligned with the source protein it is present in, based upon or derived from. Thus, where a sequence derives from A0349, the additional amino acids may be the amino acids flanking the N- and/or C- terminal ends of the sequence when aligned to polypeptide A0349 of Table 1 or 2.
As mentioned, a polypeptide of option a), b), c) or d) may include a number of
modifications compared to the parent amino acid sequences set out in any of Table 1 to 9, for example to increase or decrease physical or chemical properties of the parent sequence, for example to decrease its ability to resist oxidation, to improve or increase solubility in aqueous solution, to decrease aggregation, to decrease manufacturing problems, e.g. synthesis problems, etc.
Accordingly, in some embodiments of the invention, an amino acid sequence (parent sequence) disclosed in any of the Tables or a subsequence thereof may be modified to comprise: a) one or more (e.g. 1 , 2,3, 4, 5, 6, 7, 8, 9 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) amino acid substitutions in a sequence selected from any of the sequences disclosed in Tables 1 to 9, for example a glutamate residue at the N-terminus of an amino acid sequence disclosed in Tables 1 to 9 may be replaced with pyroglutamate and/or one or more cysteine residues in the an amino acid sequence disclosed in Tables 1 to 9 may be replaced with serine or 2-aminobutyric acid; and/or b) one or more amino acid additions (e.g. 1 , 2,3, 4, 5, 6, 7, 8, 9 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) to an amino acid sequence disclosed in Tables 1 to 9 or a subsequence thereof, for example the addition of one or more (e.g. 1 , 2, 3, or 4) lysine residue(s) and/or arginine residue(s) and/or positively charged residues at the N- and/or C-terminus of amino acid sequence disclosed in Tables 1 to 9 or a subsequence thereof); and/or c) one or more amino acid deletions from the amino acid sequence disclosed in Tables 1 to 9 or a subsequence thereof, for example the deletion of a hydrophobic residue in the up to three amino acids from the N- or C- terminus of said sequence; and/or of any two consecutive amino acids comprising the sequence Asp-Gly up to four amino acids from the N- or C- terminus of said sequence. In one embodiment, a polypeptide of option a), b), c) or d) including a modified
polypeptide is derivatized. Specific non-limiting examples of derivatization are covalent or non-covalent attachment of another molecule. Specific examples include glycosylation, acetylation, phosphorylation, amidation, formylation, ubiquitination, and derivatization by protecting/blocking groups and any of numerous chemical modifications. Typically, a derivative is formed by reacting a functional side group of an amino acid (e.g. amino, sulfhydryl or carboxy-group) with another molecule to form a covalent or non- covalent attachment of any type of molecule (naturally occurring or designed), such as a sugar moiety. Specific examples of derivatives of a peptide include glycosylation, acylation (e.g. acetylation), phosphorylation, amidation, formylation, ubiquitination and derivatization by protecting/blocking groups and any of numerous chemical modifications. Additional specific non-limiting examples are tagged peptides, fusion peptides, chimeric peptides including peptides having one or more non-amino acyl groups (q.v., sugar, lipid, etc.) covalently linked to the peptide. Typically, a derivative comprises one or more modifications, for example selected from any of: (a) N-terminal acylation (e.g. acetylation or formylation); (b) C-terminal amidation (e.g. reaction with ammonia or an amine); (c) one or more hydrogens on the side chain amines of arginine and/or lysine replaced with a methylene group; (d) glycosylation and/or (e) phosphorylation. In particular embodiments, a derivative comprises a fusion (chimeric) sequence of peptides, which optionally may contain an amino acid sequence having one or more molecules not normally present in a reference (wild type) sequence covalently attached to the peptide amino acid sequence. The term "chimeric" and grammatical variations thereof, when used in reference to a sequence, means that the sequence contains one or more portions that are derived from, obtained or isolated from, or based upon other physical or chemical entities.
Another particular example of a derivative is one in which a second heterologous sequence, i.e. a heterologous functional domain, is attached to a peptide disclosed herein, (covalent or non-covalent binding) that may confer a distinct or complementary function to a peptide disclosed herein. Heterologous functional domains are not restricted to amino acid residues. Thus, a heterologous functional domain can consist of any of a variety of different types of small or large functional moieties. Such moieties include nucleic acid, peptide, carbohydrate, lipid or small organic compounds, such as a drug (e.g., an antiviral), a metal (gold, silver), or a radioisotope.
Linkers, such as amino acid or peptidomimetic sequences, may be inserted between the peptide sequence and the addition (e.g., heterologous functional domain) so that the two entities maintain, at least in part, a distinct function or activity. Linkers may have one or more properties that may include a flexible conformation, an inability to form an ordered secondary structure or a hydrophobic or charged character, which could promote or interact with either domain. Amino acids typically found in flexible protein regions include Gly, Asn and Ser. Other near neutral amino acids, such as Thr and Ala, may also be used in the linker sequence.
In a particular aspect of the invention, the polypeptides of option a), b), c) or d) or a combination of such polypeptides are not provided as individual polypeptides, but may be fused together or to a carrier molecule to form an isolated molecule (e.g. immunogenic molecule). For example, the polypeptides may be fused to the N- and C-terminus of a surface polypeptide of a virus, e.g. a virus of the hepadnaviridae family as disclosed in international patent application W012168487 A1. A molecule of the invention, including a polypeptide of option a), b), c) or d), a
subsequence thereof, a modified polypeptide thereof, or a derivatized polypeptide thereof may share the same functionality or activity as the relevant parent polypeptide disclosed in Tables 1 to 9 or may have improved functionality or activity. For example, a molecule of the invention, including a polypeptide of option a), b), c) or d), a subsequence thereof, a modified polypeptide thereof, or a derivatized polypeptide thereof may bind to at least 70% of the group of Class HLA II alleles that the relevant parent polypeptide disclosed in Tables 1 to 9 binds to. Thus, in some embodiments a molecule of the invention, including a polypeptide of option a), b), c) or d), a modified polypeptide thereof, or a derivatized polypeptide binds to the same, substantially the same or at least 75%, 80%, such as at least 82%, 85%, 88%, 90%, 92%, 95%, 98% or more, of the group of HLA Class II alleles that binds to the relevant parent polypeptide disclosed in Tables 1 to 9, optionally wherein this is determined under same test conditions, either using prediction tools or in-vitro binding assay. Moreover, in some embodiments a subsequence disclosed herein, including a modified subsequence thereof, binds to the same, substantially the same or at least 75%, 80%, such as at least 82%, 85%, 88%, 90%, 92%, 95%, 98% or more, of the group of HLA Class II alleles that binds to the relevant subsequent from which the modified sequence derives, optionally wherein this is determined under same test conditions, either using prediction tools or in-vitro binding assay. Optionally, the Class HLA II binding is determined with respect to a particular group of Class HLA II alleles, for example one or more or all of the following alleles: DPA1*02:01-DPB1*01 :01 ,
DPA1*01 :03-DPB1*02:01 , DPA1*01 :03-DPB1*03:01 , DPA1*01 :03-DPB1*04:01 ,
DPA1*01 :03-DPB1*04:02, DPA1*02:02-DPB1*05:01 , DPA1*02:01-DPB1*14:01 ,
DQA1*05:01-DQB1*02:01 , DQA1*05:01-DQB1*03:01 , DQA1*03:01-DQB1*03:02, DQA1*04:01-DQB1*04:02, DQA1*01 :01-DQB1*05:01 , DQA1*01 :02-DQB1*06:02, DRB1*01 :01 , DRB1*03:01 , DRB1*04:01 , DRB1*04:05, DRB1*07:01 , DRB1*09:01 , DRB1*11 :01 , DRB1*12:01 , DRB1*13:02, DRB1*15:01 , DRB3*01 :01 , DRB3*02:02, DRB4*01 :01 and DRB5*01 :01 , or the alleles disclosed in Tables 11 b or 11 c. Assays for measuring Class HLA II binding in vitro is well known in the art, e.g. as described herein in Example 5.
Furthermore, a polypeptide of option a), b), c) or d), including a modified or derivatized polypeptide thereof may have one or more of the same T cell epitopes or T cell activity (e.g. percent responders) as the amino acid sequence from which it derives (e.g. an amino acid sequence set out in Tables 1 to 9. This may be determined by the ability to induce or stimulate in vitro T cell proliferation using cultured PBMCs (peripheral blood monocytes) in response to the polypeptide of option a), b), c) or d), including a modified or derivatized polypeptide compared to the amino acid sequence set out in Tables 1 to 9, optionally using same test conditions, or by the ability to induce or stimulate production of cytokines, (e.g. cytokines, IL-5, IL-13 and/or IL-10) from T cells (obtained from cultured PBMC's) in response to the polypeptide of option a), b), c) or d), including a modified or derivatized polypeptide compared to the amino acid sequence set out in Tables 1 to 9. Examples of T cell assays are described herein in Example 6.
In particular embodiments, a derivative is a fusion (chimeric) sequence, an amino acid sequence having one or more molecules not normally present in the wild type sequence covalently attached to the sequence. The term "chimeric" and grammatical variations thereof, when used in reference to a sequence, means that the sequence contains one or more portions that are derived from, obtained or isolated from, or based upon other physical or chemical entities. For example, a chimera of two or more different
polypeptides may have one part of a polypeptide, and a second part of the chimera may be from a different sequence, or unrelated protein sequence.
Another particular example of a derivatized polypeptide is one in which a second heterologous sequence, i.e., heterologous functional domain is attached (covalent or non- covalent binding) that confers a distinct or complementary function. Heterologous functional domains are not restricted to amino acid residues. Thus, a heterologous functional domain can consist of any of a variety of different types of small or large functional moieties. Such moieties include nucleic acid, peptide, carbohydrate, lipid or small organic compounds, such as a drug (e.g., an antiviral), a metal (gold, silver), and radioisotope. For example, a tag such as T7 or polyhistidine can be attached in order to facilitate purification or detection of a protein, peptide, etc. For example, a 6-HIS tag may be added to the C- or N-terminal end of a polypeptide of option a), b), c) or d), e.g. the 6- HIS sequence GHHHHHHGSGMLDI, which optionally may remain in the molecule when administered to a subject. Thus, a polypeptide linked to a Tag containing histidines may easily be purified by use of a HIS tag affinity column).
Accordingly, there are provided polypeptides linked to a heterologous domain, wherein the heterologous functional domain confers a distinct function on the polypeptide.
In some embodiments, the polypeptide is derivatized for example to improve solubility, stability, bioavailability or biological activity. For example, tagged polypeptides and fusion proteins; and modifications, including peptides having one or more non-amino acyl groups (q.v., sugar, lipid, etc.) covalently linked to the polypeptide and post-translational modifications.
Linkers, such as amino acid or peptidomimetic sequences may be inserted between the sequence and the addition (e.g., heterologous functional domain) so that the two entities maintain, at least in part, a distinct function or activity. Linkers may have one or more properties that include a flexible conformation, an inability to form an ordered secondary structure or a hydrophobic or charged character, which could promote or interact with either domain. Amino acids typically found in flexible protein regions include Gly, Asn and Ser. Other near neutral amino acids, such as Thr and Ala, may also be used in the linker sequence. The length of the linker sequence may vary without significantly affecting a function or activity of the fusion protein (see, e.g., U.S. Patent No. 6,087,329). Linkers further include chemical moieties and conjugating agents, such as sulfo-succinimidyl derivatives (sulfo-SMCC, sulfo-SMPB), disuccinimidyl suberate (DSS), disuccinimidyl glutarate (DSG) and disuccinimidyl tartrate (DST). Further non-limiting examples of derivatives are detectable labels. Thus, in another embodiment, the invention provides polypeptides that are detectably labeled. Specific examples of detectable labels include fluorophores, chromophores, radioactive isotopes (e.g., S35, P32, I125), electron-dense reagents, enzymes, ligands and receptors. Enzymes are typically detected by their activity. For example, horseradish peroxidase is usually detected by its ability to convert a substrate such as 3,3-',5,5-'-tetramethylbenzidine (TMB) to a blue pigment, which can be quantified.
Modified polypeptides also include one or more D-amino acids substituted for L-amino acids (and mixtures thereof), structural and functional analogues, for example, peptidomimetics having synthetic or non-natural amino acids or amino acid analogues and derivatized forms. Modifications include cyclic structures such as an end-to-end amide bond between the amino and carboxy-terminus of the molecule or intra- or inter- molecular disulfide bond.
Polypeptides may be provided in the form of a salt, for example as a pharmaceutically acceptable and/or a physiologically acceptable salt. For example, the salt may be an acid addition salt with an inorganic acid, an acid addition salt with an organic acid, a salt with a basic inorganic acid, a salt with a basic organic acid, a salt with an acidic or basic amino acid or a mixture thereof. In particular embodiments of the invention a salt, such as a pharmaceutically acceptable salt, is an acetate salt.
The invention provides polypeptides and molecules in isolated and/or purified form. The term "isolated," when used as a modifier of a composition, means that the compositions are made by the hand of man or are separated, completely or at least in part, from their naturally occurring in vivo environment. Generally, isolated compositions are substantially free of one or more materials with which they normally associate with in nature, for example, one or more protein, nucleic acid, lipid, carbohydrate, cell membrane. The term "isolated" does not exclude alternative physical forms of the composition, such as fusions/chimeras, multimers/oligomers, modifications (e.g., phosphorylation, glycosylation, lipidation) or derivatized forms, or forms expressed in host cells produced by the hand of man.
An "isolated" composition (e.g. polypeptides or molecules as defined herein) can also be "substantially pure" or "purified" when free of most or all of the materials with which it typically associates with in nature. Thus, an isolated polypeptide that also is substantially pure or purified does not include polypeptides or polynucleotides present among millions of other sequences, such as polypeptide of a peptide library or nucleic acids in a genomic or cDNA library, for example.
A "substantially pure" or "purified" composition can be combined with one or more other molecules. Thus, "substantially pure" or "purified" does not exclude combinations of compositions, such as combinations of polypeptides other antigens, agents, drugs or therapies.
Polypeptides can be prepared recombinant, chemically synthesized, isolated from a biological material or source, and optionally modified, or any combination thereof. A biological material or source would include an organism that produced or possessed any polypeptide or molecule set forth herein. A biological material or source may further refer to a preparation in which the morphological integrity or physical state has been altered, modified or disrupted, for example, by dissection, dissociation, solubilization,
fractionation, homogenization, biochemical or chemical extraction, pulverization, lyophilization, sonication or any other means of manipulating or processing a biological source or material. As mentioned, polypeptides, such as a molecule (e.g. immunogenic molecules) disclosed herein, may be modified by substituting, deleting or adding one or more amino acid residues in the amino acid sequence and screening for biological activity, for example eliciting an immune response. A skilled person will understand how to make such derivatives or variants, using standard molecular biology techniques and methods, described for example in Sambrook et al. (2001) Molecular Cloning: a Laboratory Manual, 3rd ed., Cold Spring Harbour Laboratory Press).
Molecules and polypeptides disclosed herein, including a modified polypeptide thereof, a derivatized polypeptide thereof, or salts thereof may be manufactured synthetically or recombinantly. Thus, in certain embodiments, the molecule, including a polypeptide of option a), b), c), d) or a subsequence thereof is synthetic. The molecule, including a polypeptide of option a), b), c), d) or a subsequence thereof may be isolated and/or purified, e.g. made by the hand of man, such as by synthesis. Typically, the polypeptides may be combined after synthesis and freeze-dried or dissolved in aqueous solutions, DMSO, glycerol or the like or mixtures thereof.
Molecules (e.g. immunogenic molecules) disclosed herein can be employed in various therapeutic or diagnostic methods and uses. Such methods and uses include, for example, administration in vitro and in vivo of one or more molecules disclosed herein. The methods and uses may include modulating an immune response (e.g. modulating an allergic immune response), including, among others, methods and uses for relieving an immune response (e.g. allergic immune response), protecting and treating subjects against a disorder, disease (e.g. allergic disease); and methods and uses of providing immunotherapy, such as specific immunotherapy against an immune response, e.g. an immune response triggered by an allergen (e.g. a pollen, such as a pollen allergen) and/or a molecule disclosed herein.
In particular embodiments, methods and uses include administration or delivery of a molecule disclosed herein to modulate an immune response in a subject, including, for example, modulating an immune response triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein.
As used herein, the term "modulate," means an alteration or effect on the term modified. In certain embodiments, modulating involves decreasing, reducing, inhibiting, suppressing, relieving an immune response in a subject triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein. In other embodiments, modulating involves eliciting, stimulating, inducing, promoting, increasing or enhancing an immune response in a subject triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein. Thus, where the term "modulate" is used to modify the term "immune response triggered by an allergen (e.g. triggered by pollen, such as a pollen allergen) and/or a molecule disclosed herein in a subject" this means that the immune response in the subject to the allergen, pollen, pollen allergen or molecule is altered or affected (e.g., decreased, reduced, inhibited, suppressed, limited, controlled, prevented, elicited, promoted, stimulated, increased, induced, enhanced, etc. Methods and uses of modulating an immune response triggered by an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein may be used to provide a subject with protection against an immune response or immune reaction to said allergen, pollen, pollen allergen and/or molecule (e.g. immunogenic molecule) disclosed herein, or symptoms or complications caused by or associated with the allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein. Accordingly, in other embodiments, methods and uses include administering a molecule (e.g.
immunogenic molecule) of the invention to protect or treat a subject against an immune response, or one or more symptoms caused by, triggered by or associated with exposure to an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
Therefore, in still other aspects, the subject's administration of a therapeutically effective amount of a composition described herein may relieve one or more symptoms of the immune response. For example, the method may comprise relieving one or more symptoms associated with allergic rhinitis, allergic conjunctivitis, allergic asthma and/or allergic eczema (e.g. atopic dermatitis).
In some embodiments, the one or more symptoms may be associated with allergic rhinitis. For example, the method may comprise reducing one or more of the following symptoms: intensity of itchy nose; number of sneezes within a given period (e.g. daily, weekly, monthly); intensity of blocked nose (e.g. congestion); amount of nasal secretions; eosinophilic count in nasal secretions; specific IgE antibody level (titer) in nasal secretions or in serum; and basophil histamine release of blood.
In other embodiments, the one or more symptoms may be associated with allergic conjunctivitis. For example, the method may comprise reducing one or more of the following: intensity of itchy eyes, redness in the white of the eyes and/or watery eyes; eosinophilic count in conjunctival tissue scrapings; specific IgE antibody level (titer) in conjunctival tissue scrapings or in serum; and basophil histamine release in blood.
In some embodiments, the one or more symptoms may be associated with allergic asthma. For example, the method may comprise reducing one or more of the following: number of or frequency of asthma exacerbations (optionally that require hospitalization), intensity and/or number of coughs within a given period (e.g. daily, weekly, monthly); intensity of wheezes; intensity of shortness of breath or congestion (e.g. improvement of being short of breath); reducing Forced Expiratory Volume (FEV1); reducing specific IgE antibody level (titer) in lung fluid or in serum and basophil histamine release in blood; or the method may comprise improving lung function. In some embodiments, the one or more symptoms may be symptoms associated with atopic dermatitis. For example, the method may comprise reducing one or more of the following: itch intensity of the skin; eczema score, and number of (peripheral) blood eosinophils. A therapeutic or beneficial effect also includes reducing or eliminating the need, dosage frequency or amount of a second therapeutic method or therapeutically active drug (e.g. anti-inflammatory, decongestants or anti-allergic agent) used for treating a subject having an immune response or one or more symptoms caused by or associated with an allergen. For example, administration of a peptide combination described herein may reduce the amount of an adjunct therapy administered to a subject, such as reducing the subject's need for concomitant treatment with fast or long-acting p2-agonists, leukotriene modifiers, theophylline corticosteroids or H1 antihistamines (e.g. inhaled or oral) to reduce, relieve, or suppress one or more symptoms of the immune response.
In still other embodiments, methods and uses include administering or delivering a molecule (e.g. an immunogenic molecule) of the invention to elicit, stimulate, induce, promote, increase or enhance immunological tolerance of a subject an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
In various embodiments, there are provided methods and uses of providing a subject with protection against an immune response, or one or more symptoms caused by or associated with exposure to an allergen (e.g. pollen, such as a pollen allergen) or an immunogenic molecule disclosed herein. In various aspects, a method or use includes administering to the subject an amount of an immunogenic molecule of the invention sufficient to provide the subject with protection against an immune response, or symptoms caused by or associated with exposure to an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
Methods and uses of the invention include providing a subject with protection against an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein, or symptoms caused by or associated with the subject's exposure to said allergen (e.g. pollen, such as pollen allergen) and/or a molecule disclosed herein, for example, vaccinating the subject to protect against an immune response to said allergen (e.g.
pollen, such as pollen allergen) and/or a molecule, for example with an immunogenic molecule provided herein. In certain embodiments, methods and uses include protecting the subject against an immune response triggered by pollen, e.g. a pollen allergen or a molecule disclosed herein by inducing tolerance of the subject (desensitizing) to said allergen, pollen, pollen allergen and/or immunogenic molecule.
As used herein, the terms "protection," "protect" and grammatical variations thereof, when used in reference to an immune response or symptoms caused by or associated with the exposure to an allergen (e.g. pollen, such as a pollen allergen) and/or an immunogenic molecule disclosed herein, means preventing an immune response or symptoms caused by or associated with the exposure to the allergen, pollen, pollen allergen and/or a molecule disclosed herein, or reducing or decreasing susceptibility to an immune response or one or more symptoms caused by or associated with the exposure to the allergen, pollen, pollen allergen and/or a molecule disclosed herein.
An immune response includes but is not limited to an allergic immune response, such as an allergic reaction, hypersensitivity, an inflammatory response or inflammation. In certain embodiments an immune response may involve one or more of cell infiltration, production of antibodies, production of cytokines, lymphokines, chemokines, interferons and interleukins, cell growth and maturation factors (e.g., differentiation factors), cell proliferation, cell differentiation, cell accumulation or migration (chemotaxis) and cell, tissue or organ damage or remodeling. In particular aspects, an immune response may include an allergic immune response, such as allergic rhinitis; atopic dermatitis; allergic conjunctivitis and asthma. Allergic responses can occur systemically or locally in any region, organ, tissue, or cell. In particular aspects, an allergic immune response occurs in the skin, the upper respiratory tract, the lower respiratory tract, pancreas, thymus, kidney, liver, spleen, muscle, nervous system, skeletal joints, eye, mucosal tissue, gut or bowel.
Methods and uses herein include relieving a subject of, including treating a subject for, an immune response, or one or more symptoms caused by or associated with an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein. Such methods and uses include administering to a subject an amount of an immunogenic molecule sufficient to relieve the subject of, such as treat the subject for, the allergic immune response, or one or more symptoms caused by or associated with the allergen, (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein. Methods and uses of the invention include treating or administering to a subject previously exposed to an allergen (e.g. pollen, such as a pollen allergen) and/or an immunogenic molecule disclosed herein. Thus, in certain embodiments, methods and uses are for treating or protecting a subject from an allergic immune response, or one or more symptoms caused by or associated with secondary or subsequent exposure to an allergen, (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein.
Immunogens described herein may elicit, stimulate, induce, promote, increase or enhance immunological tolerance to an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein. Methods and uses of the invention therefore further include inducing immunological tolerance of a subject to an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein. Thus, for example, molecules described herein can be effective in relieving, such as treating an immune response, including but not limited to an immune response following a secondary or subsequent exposure of a subject to an allergen (e.g. pollen, such as a pollen allergen) and/or a molecule disclosed herein. In one embodiment, a method or use includes administering to the subject an amount of a molecule disclosed herein sufficient to induce tolerance in the subject to the allergen (e.g. pollen, such as a pollen allergen) or the molecule itself. In particular aspects, the immunological tolerance elicited, stimulated, induced, promoted, increased or enhanced may involve modulation of T cell activity, including but not limited to CD4+ T cells, CD8+ T cells, Th1 cells, Th2 cells and regulatory T cells. For example, immunological tolerance elicited, stimulated, induced, promoted, increased or enhanced from administration of the immunogenic molecule, may involve modulation of the production or activity of pro-inflammatory or anti-inflammatory cytokines produced by T cells.
In additional embodiments, a method or use of inducing immunological tolerance in a subject to an allergen (e.g. pollen, such as a pollen allergen) and/or an immunogenic molecule disclosed herein includes a reduction in occurrence, frequency, severity, progression, or duration of physiological conditions, disorders, illnesses, diseases, symptoms or complications caused by or associated an allergic response to the allergen (e.g. pollen, such as a pollen allergen) and/or immunogenic molecule in the subject. Thus, in certain embodiments, inducing immunological tolerance can protect a subject against or treat a subject for an immune response, or one or more symptoms caused by or associated with an allergen (e.g. pollen, such as a pollen allergen) or a molecule disclosed herein.
Methods and uses of the invention include treating a subject via immunotherapy, including specific immunotherapy. In one embodiment, a method or use includes administering to the subject an amount of a molecule described herein. In one aspect, a molecule administered to a subject during specific immunotherapy to treat the subject is the same immunogenic molecule to which the subject has been sensitized or is hypersensitive (e.g., allergic). In another non-limiting aspect, a molecule is administered to a subject to treat the subject to a different immunogenic molecule, e.g. an allergen, e.g. a pollen allergen, to which the subject has been sensitized or is hypersensitive (e.g., allergic). Thus, the immunotherapeutic mechanism may involve bystander suppression of an immune response caused by an allergen, e.g. a pollen allergen, by administering an unrelated immunogenic molecule, e.g. a molecule disclosed herein.
As described herein, a molecules of the invention may include T cell epitopes, such as Th2 cell epitopes. In methods and uses herein, the subject to be treated has a specific T- cell response to the a molecule before administering the first dose of the a molecule . Accordingly, methods and uses of the invention include administering an amount of an a molecule (e.g., a T cell epitope-containing a molecule ) to a subject sufficient to provide the subject with protection against an immune response disclosed herein, or one or more symptoms disclosed herein. In another embodiment, a method includes administering an amount of an a molecule (e.g., a T cell epitope-containing a molecule ) to a subject sufficient to relieve, e.g. treat, vaccinate or immunize the subject against an immune response disclosed herein, or one or more symptoms caused by or associated with an allergen (e.g. pollen, such as a pollen allergen) and/or an a molecule disclosed herein.
The specific T-cell response may be monitored by determining by way of contacting a sample of PBMCs obtained from the subject with a molecule of the invention and measuring the IL-5 secretion or IL-5 mRNA gene expression in response to the molecule.
In accordance with the invention, methods and uses of modulating anti-allergen activity of T cells, including but not limited to CD8+ T cells, CD4+ T cells, Th1 cells or Th2 cells, in a subject are provided. In one embodiment, a method or use includes administering to a subject an amount of a polypeptide described herein or derivative thereof including an a molecule described herein, such as a T cell epitope, sufficient to modulate Th2 cell activity in the subject.
In certain embodiments, two or more a molecules may be administered to a subject, e.g. may be administered as a combination composition, or administered separately, such as concurrently or in series or sequentially. For example, methods and uses described herein comprise administration separately or as a combination: at least 2-25 polypeptides defined herein, or separately or as a combination of 3-25, 4-25, 5-25, 6-25, 7-25 polypeptides defined herein, or separately or as a combination of 2-20, 3-20, 4-20, 5-20, 6-20 defined herein, or separately or as a combination of 2-12, 3-12, 4-12, 5-12, 6-12, 7- 12 polypeptides defined herein, or separately or as a combination of 2-10, 3-10, 4-10, 5- 10, 6-10, 7-10 polypeptides defined herein.
Therefore, a further aspect of the invention relates to a composition comprising a polypeptide of option a), b), c) or d) or a combination of two or more polypeptides of option a), b), c) or d).
Methods and uses of the invention therefore include any therapeutic or beneficial effect. In various embodiments, an immune response, or one or more symptoms caused by or associated with a pollen allergen or an a molecule disclosed herein is reduced, decreased, inhibited, limited, delayed or prevented. Methods and uses of the invention moreover include reducing, decreasing, inhibiting, delaying or preventing onset, progression, frequency, duration, severity, probability or susceptibility of one or more adverse symptoms, disorders, illnesses, diseases or complications caused by or associated with an antigen/allergen. In further various particular embodiments, methods and uses include improving, accelerating, facilitating, enhancing, augmenting, or hastening recovery of a subject from an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with a pollen allergen or an a molecule disclosed herein. In yet additional various embodiments, methods and uses include stabilizing an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with a pollen allergen or an a molecule disclosed herein.
A therapeutic or beneficial effect is therefore any objective or subjective measurable or detectable improvement or benefit provided to a particular subject. A therapeutic or beneficial effect can but need not be complete ablation of all or any immune response, or one or more symptoms caused by or associated with a pollen allergen or an a molecule disclosed herein. Thus, a satisfactory clinical endpoint is achieved when there is an incremental improvement or a partial reduction in an allergic immune response, or one or more symptoms caused by or associated with an allergen, or an inhibition, decrease, reduction, suppression, prevention, limit or control of worsening or progression of an immune response, or one or more symptoms caused by or associated with a pollen allergen or an a molecule disclosed herein, over a short or long duration (hours, days, weeks, months, etc.).
A therapeutic or beneficial effect also includes reducing or eliminating the need, dosage frequency or amount of a second therapeutic protocol or active such as another drug or other agent (e.g., anti-inflammatory) used for treating a subject having or at risk of having an allergic immune response, or one or more symptoms caused by or associated with an allergen. For example, reducing an amount of an adjunct therapy, such as a reduction or decrease of a treatment for an allergic immune response, or one or more symptoms caused by or associated with an allergen, or a specific immunotherapy, vaccination or immunization protocol is considered a beneficial effect. In addition, reducing or decreasing an amount of the a molecule used for specific immunotherapy, vaccination or immunization of a subject to provide protection to the subject is considered a beneficial effect.
Methods and uses described herein may relieve one or more symptoms of an allergic immune response or delays the onset of symptoms, slow the progression of symptoms, or induce disease modification. For example, the following symptoms may be decreased or eliminated; nasal symptoms in the form of itchy nose, sneezing, runny nose, blocked nose; conjunctival symptoms in the form of itchy eyes, red eyes, watery eyes; and respiratory symptoms in the form of decreased lung function. Furthermore, the beneficial effect of methods and uses described herein may be observed by the patient's need for less concomitant treatment with corticosteroids or H1 antihistamines to suppress the symptoms.
When an immunogenic molecule is administered to induce tolerance, an amount or dose of the immunogenic molecule to be administered, and the period of time required to achieve a desired outcome or result (e.g., to desensitize or develop tolerance to the allergen or immunogenic molecule) can be determined by one skilled in the art. The immunogenic molecule may be administered to the patient through any route known in the art, including, but not limited to oral, inhalation, sublingual, epicutaneous, intranasal, and/or parenteral routes (intravenous, intramuscular, subcutaneously, intradermal, and intraperitoneal).
Methods and uses of the invention include administration of a molecule (e.g. an immunogenic molecule) to a subject prior to contact by or exposure to a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein; administration prior to, substantially contemporaneously with or after a subject has been contacted by or exposed to an allergen; and administration prior to, substantially contemporaneously with or after an allergic immune response, or one or more symptoms caused by or associated with a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein.
As used herein, a "sufficient amount" or "effective amount" or an "amount sufficient" or an "amount effective" refers to an amount that provides, in single (e.g., primary) or multiple (e.g., booster) doses, a long term or a short term detectable or measurable improvement in a given subject or any objective or subjective benefit to a given subject of any degree or for any time period or duration (e.g., for minutes, hours, days, months, years, or cured).
An amount sufficient or an amount effective need not be therapeutically or
prophylactically effective in each and every subject treated, nor a majority of subjects treated in a given group or population. An amount sufficient or an amount effective means sufficiency or effectiveness in a particular subject, not a group of subjects or the general population. As is typical for such methods, different subjects will exhibit varied responses to a method of the invention, such as immunization, vaccination, specific immunotherapy and therapeutic treatments.
The term "subject" includes but is not limited to a subject at risk of allergen contact or exposure as well as a subject that has been contacted by or exposed to an allergen. A subject also includes those having or at risk of having or developing an immune response to an antigen or an allergen. Such subjects include mammalian animals (mammals), such domestic animal (dogs and cats), a farm animal (poultry such as chickens and ducks, horses, cows, goats, sheep, pigs), experimental animal (mouse, rat, rabbit, guinea pig) and humans.
Target subjects and subjects in need of treatment also include those at risk of allergen exposure or contact or at risk of having exposure or contact to an allergen. Accordingly, subjects include those at increased or elevated (high) risk of an allergic reaction; that have, or have previously had or are at risk of developing hypersensitivity to an allergen; and those that have or have previously had or are at risk of developing asthma.
It should be understood that in some embodiments, the immune response to be treated in a subject in need thereof is against one or more allergens, e.g. one or more pollen, such as one or more pollen allergens. Likewise, immunological tolerance is desirable to be induced or promoted in a subject in need thereof to one or more allergens, e.g. one or more pollen, such as one or more pollen allergens. For example, the subject may be sensitized to one or more of the pollen allergens.
Non-limiting examples of the one or more pollen allergens to which the subject optionally is sensitized or the immune response is triggered by include proteins, polypeptides and peptides including allergens from a plant species selected from any of the plant families /Asferaceae, Betulaceae, Fagaceae, Oleaceae, Poaceae and/or Plantaginaceae, for example plant species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum, Poa, Ambrosia, Artemisia, Helianthus, AInus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and Plantago; for example plant species selected from any of the plant genera
Anthoxanthum, Conydon, Dactylis, Lollium, Phleum, Poa, Ambrosia, Artemisia, AInus, Betula, Corylus, Fagus, Quercus, Olea and Plantago; for example plant species selected from the plant genera Ambrosia, Artemisia, Helianthus, AInus, Betula, Carpinus,
Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and Plantago; for example plant species selected from the plant genera Ambrosia, Artemisia, AInus, Betula, Corylus, and Quercus; for example plant species selected from the plant genera Ambrosia and Quercus, for example plant species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum, Poa, Ambrosia, Artemisia, Plantago, Fraxinus, Olea and Quercus; for example a plant species selected from any of the plant genera Phleum, Cynodon, Ambrosia, Artemisia, Betula and
Quercus, such as a plant species selected from any of the plant genera Phleum,
Cynodon, Ambrosia, Betula and Quercus.
Further examples of the one or more pollen allergens to which the subject optionally is sensitized to include proteins, polypeptides and peptides including allergens from species selected from further plant families, like the plant family Chenopidiaceae including the plants Lambs quarters, Russian thistles and Kochias; plant family Amaranthaceae including Pigweeds, plant family Polygonaceae including for example Sheep sorrel, plant family Ulmacea including for example American Elm and Hackberry, plant family
Plantanaceae including for example Sycamore, plant family Salicaceae including for example White poplar and Cottonwood, plant family Aceraceae including for example Box elder and Red maple, plant family Cupressaceae including for example Common juniper and Cedar.
As mentioned, the subject may be sensitized to one or more pollen species from one or more plant families, for example selected from any of the plant families Poaceae,
Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, preferably Poaceae, Asteraceae, Fagaceae and Betulaceae. Therefore, in some embodiments, the subject may be sensitized to a pollen species of the plant family Poaceae and a pollen species of a plant family selected from any one of Asteraceae, Fagaceae and Betulaceae, such as a subject that may be sensitized to a pollen species of the plant genus Phleum and a pollen species of a plant genus selected from any one of the genera Conydon, Ambrosia, Betula and Quercus and combinations thereof. The subject may be sensitized to one or more pollen allergens from one or more plant families, for example selected from the any of the plant families Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, preferably Poaceae, Asteraceae, Fagaceae and Betulaceae. Therefore, in some embodiments, the subject may be sensitized to a pollen allergen of the plant family Poaceae and a pollen allergen of a plant family selected from any one of Asteraceae, Fagaceae and Betulaceae and combinations thereof, such as a subject that may be sensitized to a pollen allergen of the plant genus Phleum and a pollen allergen of a plant genus selected from the group consisting of Conydon, Ambrosia, Betula and Quercus and combinations thereof. As mentioned, the immune response may be triggered by pollen from one or more plant families, such as from plant families selected from any of Poaceae, Asteraceae,
Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, e.g. of Poaceae, Asteraceae, Fagaceae, Betulaceae. Therefore, in some embodiments, the immune response is triggered by pollen of the plant family Poaceae and pollen of a plant family selected from any one of Asteraceae, Fagaceae and Betulaceae and combinations thereof, for example the immune response may be triggered by pollen of the plant genus Phleum and pollen of a plant genus selected from the any one of Conydon, Ambrosia, Betula and Quercus or combinations thereof.
As mentioned, the immune response may be triggered by one or more pollen allergens, such as one or more pollen allergens from a plant family selected from any of Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae, preferably Poaceae, Asteraceae, Fagaceae and Betulaceae. Therefore, in some embodiments, the immune response is triggered by one or more pollen allergens from pollen of the plant family Poaceae and one or more pollen allergens from pollen of a plant family selected from any of Asteraceae, Fagaceae and Betulaceae. For example, the one or more pollen allergens may be from pollen of the plant genus Phleum and from pollen of a plant genus selected from the group consisting of Conydon, Ambrosia, Betula and Quercus including combinations thereof.
In some embodiments the immune responses to be modulated or treated are against pollen allergens of grass species of various plant genera, for example of the genera
Phleum and Cynodon, thus allowing the treatment or modulation of an immune response caused by a broad range of different grass pollen species. Therefore, in some
embodiments, the one or more pollen allergens are from a grass species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum and/or Poa, for example from grass species selected from any of the genera Conydon and Phleum. In some embodiments, the immune responses to be modulated or treated are against grass pollen allergens as well as weed and/or tree pollen allergens. For example the immune responses to be modulated or treated are against grass pollen allergens of the genera Phleum and against non-grass pollen of the genera Ambrosia and/or Quercus, thus allowing the treatment or modulation of an immune response caused by a broad range of different grass pollen species and non-grass species. Thus, in some
embodiments, the one or more pollen allergens are from a grass species selected from any of the plant genera Anthoxanthum, Conydon, Dactylis, Lollium, Phleum and Poa and from a non-grass species selected from any of the plant families Asteraceae, Betulaceae, Fagaceae, Oleaceae and Plantaginaceae, e.g. wherein the non-grass species are selected from any of the plant genera Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and Plantago, for example against a non-grass species selected from any of the plant genera Ambrosia, Artemisia, Alnus, Betula, Quercus, Olea and Plantago, such as a non-grass species selected from any of the plant genera Ambrosia, Betula and Quercus.
In some embodiments, the immune responses to be modulated or treated are against non-grass pollen allergens, such as against weed and/or tree pollen allergens. For example the immune responses to be modulated or treated are against non-grass pollen allergens of the genera Ambrosia and against non-grass pollen allergens of the genera Quercus, thus allowing the treatment or modulation of an immune response caused by a broad range of different non-grass pollen species. Thus, in some embodiments, the one or more non-grass pollen allergens are from a plant genus selected from any of
Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and/or Plantago.
It should be understood that in some embodiments of the invention, the one or more pollen allergens are from same grass species or alternatively from different grass species, optionally wherein the different grass species are from different plant genera selected from any of Anthoxanthum, Conydon, Dactylis, Lollium, Phleum and Poa, for example Conydon and Phleum. Likewise, it should be understood that in some embodiments of the invention the one or more non-grass pollen allergens are from same non-grass species or alternatively from different non-grass species, optionally wherein the different non-grass species are from different plant genera selected from any of
Ambrosia, Artemisia, Helianthus, Alnus, Betula, Carpinus, Castanea, Corylus, Ostrya, Ostryopsis, Fagus, Quercus, Fraxinus, Ligustrum, Lilac, Olea and Plantago. In some embodiments, the immune response to be modulated or treated may be at least against one or more pollen allergens of various grass pollen species, e.g. against one or more pollen allergens of the plant genera Phleum. In some embodiments, the immune response to be modulated or treated may be at least against one or more pollen allergens of various weed pollen species, e.g. against one or more pollen allergens of the plant genera Ambrosia. In some embodiments, the immune response to be modulated or treated may be at least against one or more pollen allergens of various tree pollen species, e.g. against one or more pollen allergens of the plant genera Quercus. In some embodiments, the immune response to be modulated or treated may be at least against one or more pollen allergens of various tree pollen species, e.g. against one or more pollen allergens of the plant genera Betula.
As mentioned, the immune response to be modulated or treated may be against one or more pollen allergens of various grass pollen species and non-grass pollen species, such as two or more, three or more, four or more, five or more pollen allergens of different pollen species, like various grass pollen species and/or non-grass pollen species.
Non-limiting examples of the genus Ambrosia may be Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida; a typical species of the genus Artemisia may be Artemisia vulgaris; a typical species of the genus Betula may be Betula verrucosa; a typical species of the genus Fagus may be Fagus grandifolia or Fagus sylvatica; a typical species of the genus Quercus may be Quercus alba, a typical species of the genus Fraxinus may be Fraxinus excelsior; a typical species of the genus Olea may be Olea Europaea, a typical species of the genus plantago may be Plantago lanceolata, a typical species of the genus plantago may be Plantago lanceolata, a typical species of the genus Anthoxanthum may be Anthoxanthum Odoratum, a typical species of the genus Conydon may be Conydon Dactylon, a typical species of the genus Lollium may be Lollium Perenne, a typical species of the genus Phleum may be Phleum Pratense, a typical species of the genus Poa may be Poa Pratensis.
Non-limiting examples of non-grass pollen allergens to which a subject optionally may be sensitized to are Aln g 1 , Aln g 4, Amb a 1 , Amb a 2, Amb a 3, Amb a 4, Amb a 5, Amb a 6, Amb a 7, Amb a 8, Amb a 9, Amb a 10, Amb p 5, Amb t 5, Art v 1 , Art v 2, Art v 3, Art v 4, Art v 5, Art v 6, Bet v 1 , Bet v 2, Bet v 3, Bet v 4, Bet v 6, Bet v 7, Car b 1 , Cas s 1 , Cor a 6, Cor a 10, Fag s 1 , Fra e 1 , Hel a 1 , Hel a, Lig v 1 , Ole e 1 , Ole e 2, Ole e 3, Ole e 4, Oie e 5, Ole e 6, Ole e 7, Ole e 8, Ole e 9, Ole e 10, Ole e 11 , Ost c 1 , Pla I, Que a 1 , Syr v 1 , Syr v 3, and any combination thereof, for example the major allergens Amb a 1 , Que a 1 , Bet v 1 , Bet v 2, Ole e 1 and any combination thereof. Non-limiting examples of grass pollen allergens to which a subject optionally may be sensitized to are Ant o 1 , Cyn d 1 , Cyn d 7, Cyn d 12, Cyn d 15, Cyn d 22w, Cyn d 23, Cyn d 24, Dac g 1 , Dac g 2, Dac g 3, Dac g 4, Dac g 5, Fes p 4, Hoi 1 1 , Hoi I 5, Hor v 1 , Hor v 5, Lol p 1 , Lol p 2, Lol p 3, Lol p 4, Lol p 5, Lol p 11 , Ory s 1 , Pas n 1 , Pha a 1 , Pha a 5, Phi p 1 , Phi p 2, Phi p 4, Phi p 5, Phi p, Phi p 7, Phi p 1 1 , Phi p 12, Phi p 13, Poa p 1 , Poa p 5, Sec c 1 , Sec c 5, Sec c 38 and/or Sor h 1 , and any combination thereof. The group 1 allergens (e.g. Ant o 1 , Cyn d 1 , Dac g 1 , Hoi 1 , Lol p 1 , Pha a 1 , Phi p 1 and Poa p 1 ) or group 5 allergens (Dac g 5, Lol p 5, Pha a 5, Phi p 5, Poa p 5) are considered major allergens of grass pollen
"Prophylaxis" and grammatical variations thereof mean a method or use in which contact, administration or in vivo delivery to a subject is prior to contact with or exposure to a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein. In certain situations it may not be known that a subject has been contacted with or exposed to an allergen, but administration or in vivo delivery to a subject can be performed prior to manifestation of an allergic immune response, or one or more symptoms caused by or associated with an allergen. For example, a subject can be provided protection against an allergic immune response, or one or more symptoms caused by or associated with a pollen allergen or a molecule disclosed herein or provided immunotherapy with a molecule (e.g. an immunogenic molecule) of the present invention. In such case, a method or use can eliminate, prevent, inhibit, suppress, limit, decrease or reduce the probability of or susceptibility towards an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with an a pollen allergen or a molecule disclosed herein.
"Prophylaxis" can also refer to a method or use in which contact, administration or in vivo delivery to a subject is prior to a secondary or subsequent exposure to an allergen (e.g. pollen such as a pollen allergen) and/or a molecule disclosed herein. In such a situation, a subject may have had a prior contact or exposure to a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein. In such subjects, an acute allergic reaction may but need not be resolved. Such a subject typically may have developed anti- allergen antibodies due to the prior exposure. Immunization or vaccination, by
administration or in vivo delivery to such a subject, can be performed prior to a secondary or subsequent allergen exposure. Such a method or use can eliminate, prevent, inhibit, suppress, limit, decrease or reduce the probability of or susceptibility towards a secondary or subsequent allergic immune response, or one or more symptoms caused by or associated with a pollen allergen or a molecule (e.g. an immunogenic molecule) disclosed herein. In certain embodiments, such a method or use includes providing specific immunotherapy to the subject to eliminate, prevent, inhibit, suppress, limit, decrease or reduce the probability of or susceptibility towards a secondary or subsequent allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with an a pollen allergen or a molecule disclosed herein.
Treatment of an allergic reaction or response can be at any time during the reaction or response. A molecule (e.g. an immunogenic molecule) can be administered as a single or multiple dose e.g., one or more times hourly, daily, weekly, monthly or annually or between about 1 to 10 weeks, or for as long as appropriate (e.g. 3 months, 6 months or more, for example, to achieve a reduction in the onset, progression, severity, frequency, duration of one or more symptoms or complications associated with or caused by an allergic immune response, or one or more physiological conditions, symptoms or complications caused by or associated with an antigen/allergen.
Accordingly, methods and uses of the invention can be practiced one or more times (e.g., 1-10, 1-5 or 1-3 times) an hour, day, week, month, or year. The skilled artisan will know when it is appropriate to delay or discontinue administration. Doses can be based upon current existing protocols, empirically determined, using animal disease models or optionally in human clinical trials. Initial study doses can be based upon animal studies, e.g. a mouse, and the sufficient amount of immunogenic molecule to be administered for being effective can be determined. Exemplary non-limiting amounts (doses) are in a range of about 0.1 mg/kg to about 100 mg/kg, and any numerical value or range or value within such ranges. Greater or lesser amounts (doses) can be administered, for example, 0.01-500 mg/kg, and any numerical value or range or value within such ranges. The dose can be adjusted according to the mass of a subject, and will generally be in a range from about 1-10 ug/kg, 10-25 ug/kg, 25-50 ug/kg, 50-100 ug/kg, 100-500 ug/kg, 500-1 ,000 ug/kg, 1-5 mg/kg, 5-10 mg/kg, 10-20 mg/kg, 20-50 mg/kg, 50-100 mg/kg, 100-250 mg/kg, 250-500 mg/kg, or more, two, three, four, or more times per hour, day, week, month or annually. A typical range will be from about 0.3 mg/kg to about 50 mg/kg, 0-25 mg/kg, or 1.0-10 mg/kg, or any numerical value or range or value within such ranges. Doses can vary and depend upon whether the treatment is prophylactic or therapeutic, whether a subject has been previously exposed to the antigen/allergen, the onset, progression, severity, frequency, duration, probability of or susceptibility of the symptom, condition, pathology or complication, or vaccination or specific immunotherapy to which treatment is directed, the clinical endpoint desired, previous or simultaneous treatments, the general health, age, gender, race or immunological competency of the subject and other factors that will be appreciated by the skilled artisan. The skilled artisan will appreciate the factors that may influence the dosage and timing required to provide an amount sufficient for providing a therapeutic or prophylactic benefit.
Immunogens of the invention can be provided in compositions, and in turn such compositions can be used in accordance with the invention methods and uses. Such compositions, methods and uses include pharmaceutical compositions and formulations. In certain embodiments, a pharmaceutical composition includes one or more
immunogenic molecules (e.g. polypeptides of option a), b), c) or d). In particular, aspects, such compositions and formulations may be a vaccine, including but not limited to a vaccine to protect against (e.g. modify, relieve or suppress) an immune response disclosed herein, or one or more symptoms caused by or associated with an allergen and/or a molecule (e.g. an immunogenic molecule) disclosed herein.
A pharmaceutical composition comprises a molecule (e.g. an immunogenic molecule) of the invention and a pharmaceutically acceptable ingredient or carrier. As used herein the term "pharmaceutically acceptable" and "physiologically acceptable" mean a biologically acceptable formulation, gaseous, liquid or solid, or mixture thereof, which is suitable for one or more routes of administration, in vivo delivery or contact. Such formulations include solvents (aqueous or non-aqueous), solutions (aqueous or non-aqueous), emulsions (e.g., oil-in-water or water-in-oil), suspensions, syrups, elixirs, dispersion and suspension media, coatings, isotonic and absorption promoting or delaying agents, compatible with pharmaceutical administration or in vivo contact or delivery. Aqueous and non-aqueous solvents, solutions and suspensions may include suspending agents and thickening agents. Such pharmaceutically acceptable carriers include tablets (coated or uncoated), capsules (hard or soft), microbeads, powder, granules and crystals. Supplementary active compounds (e.g., preservatives,
antibacterial, antiviral and antifungal agents) can also be incorporated into the
compositions.
A pharmaceutically acceptable or physiologically acceptable excipient, carrier and/or adjuvants are well-known to the person skilled in the art and may include, but are not limited to, solvents, emulsifiers, wetting agents, plasticizers, solubilizers (e.g. solubility enhancing agents) coloring substances, fillers, preservatives, anti-oxidants, anti-microbial agents, viscosity adjusting agents, buffering agents, pH adjusting agents, isotonicity adjusting agents, mucoadhesive substances, and the like. Examples of formulation strategies are well-known to the person skilled in the art. In some embodiments, the pharmaceutical composition may be formulated for parenteral administration, such as formulated for injection, e.g. subcutaneous and/or intradermal injection. Therefore, in some embodiments, the pharmaceutical composition may be a liquid (i.e. formulated as a liquid), including a solution, a suspension, a dispersion, and a gelled liquid. For example, a liquid pharmaceutical composition may be formed by dissolving a powder, granulate or lyophilizate of a molecule (e.g. an immunogenic molecule) combination described herein in a suitable solvent and then administering to a subject. Suitable solvents may be any solvent having physiologically acceptable properties and able to dissolve the immunogenic molecule combination in desired concentrations. A desired concentration may depend on the aliquot to be administered (i.e. to be injected) and the desired single dose. It is emphasized that for the purposes of injection the aliquot is in the range of about 10 to 500 microliters, e.g. 50 to 300 microliters or less and a desired single dose is within range of 1 to 1000 nanomoles. Therefore, a suitable solvent should be able to dissolve any immunogenic molecule of the combination to achieve a final concentration of about 1 to 1000 μΜ for each of the immunogenic molecules. Thus, in one embodiment, a liquid composition comprises each of the immunogenic molecules of the combination in a concentration of 10 to 800 μΜ, for example 20 to 500 μΜ or20 to 300 μΜ. Typically, the concentration of each immunogenic molecule is the same, such as in an equimolar concentration, but each immunogenic molecule of the composition may be present in different concentrations. Typically, the solvent is an aqueous solution, optionally mixed with other solvents. Thus, a solvent may comprise at least 60% w/w of water, e.g. at least 65% w/w, 70% w/w, 75% w/w, 80% w/w , 85% w/w, 90% w/w or 95% w/w , 99% w/w of water, such as distilled water, such as sterile water. In some embodiments, the solvent is sterile distilled water, e.g. water for injection. An aqueous solution may comprise other solvents than water, for example
DMSO (dimethylsulfoxide), glycerol, ethanol, acetonitrile, vegetable or synthetic oils. The pH of the aqueous phase of the solvent may be in a physiological acceptable range, typically in the range of 3 to 9, such as in the range of pH 3 to 8, such as in the range of pH 4 to 8, such as in the range of 5 to 8, such as in the range of 6 to 8. Thus, the liquid formulation may comprise a pH controlling agent or buffering agent (e.g. citrate buffer, phosphate buffer, acetate buffer), optionally the pH may be adjusted with dilutions of strong base (e.g. sodium hydroxide or the like) and/or dilutions of strong acids (e.g.
hydrochloric acid).
Typically, the liquid formulation is isotonic, and optionally sterile. Therefore, in some embodiments, the formulation comprises saline, such as isotonic saline. The liquid may contain additional excipients, such as another solvent, a solubilizing enhancing agent (e.g. polyoxyethylene (20) sorbitan monolaurate (Tween® 20), ionic and non-ionic emulsifiers (e.g. poloxamers (Kolliphor®)), a dispersant, a thickener, a preservative, an anti-microbial agent, and/or an antioxidant. Non-limiting illustrative examples of solvents include water, saline, DMSO, glycerol, ethanol, acetonitrile, vegetable or synthetic oils. Some immunogenic molecules are known to be prone to oxidation or being unstable when exposed to water for a long period. Therefore, to achieve storage stable
compositions, a pharmaceutical composition may be formulated to contain only a limited amount of water or aqueous solution, e.g. containing less than 10% w/w of water or aqueous solution, such as less than 9, 8, 7, 6, 5, 4, 3, 2, 1 , 0.5% w/w of water or aqueous solution. Examples of pharmaceutical compositions with limited levels of water may include granulates, powders, for example lyophilizates, i.e. freeze-dried powders.
Typically, the freeze-dried composition may be dissolved before use, for example dissolved in an aqueous, optionally sterile, solution, for example a solution having a pH in the range of 3-9, such as pH in the range of 3 to 8, such as pH in the range of 4 to 8. A lyophilizate may contain additional ingredients, e.g. bulking agents and lyoprotectants (e.g. sucrose, lactose, trehalose, mannose, mannitol, sorbitol, glucose, raffinose, glycine, histidine or mixtures thereof), buffering agents (e.g. sodium citrate, sodium phosphate, disodium phosphate, sodium hydroxide, Tris base, Tris acetate, Tris HCI or mixtures thereof), antioxidants, antimicrobial agents, solubilizers (e.g. polyoxyethylene (20) sorbitan monolaurate (Tween® 20)).
A freeze-dried composition may also be formulated into a solid dosage form that is administered for example by the oral route such as by oral mucosa. Thus, in some embodiments, the pharmaceutical composition may be formulated for oral administration, for example for sublingual administration. Therefore, the pharmaceutical composition may be a solid dosage form, such as a freeze-dried solid dosage form, typically a tablet, a capsule or sachet, which optionally may be formulated for fast disintegration.
Pharmaceutical formulations and delivery systems appropriate for the compositions, methods and uses of the invention are known in the art (see, e.g., Remington: The Science and Practice of Pharmacy (2003) 20th ed., Mack Publishing Co., Easton, PA; Remington's Pharmaceutical Sciences (1990) 18th ed., Mack Publishing Co., Easton, PA; The Merck Index (1996) 12th ed., Merck Publishing Group, Whitehouse, NJ;
Pharmaceutical Principles of Solid Dosage Forms (1993), Technonic Publishing Co., Inc., Lancaster, Pa.; Ansel ad Soklosa, Pharmaceutical Calculations (2001) 11th ed.,
Lippincott Williams & Wilkins, Baltimore, MD; and Poznansky et al., Drug Delivery Systems (1980), R. L. Juliano, ed., Oxford, N.Y., pp. 253-315). As mentioned, pharmaceutical compositions can be formulated to be compatible with a particular route of administration, such as by intradermal or by sublingual administration. Thus, pharmaceutical compositions may include carriers, diluents, or excipients suitable for administration by various routes. Exemplary routes of administration for contact or in vivo delivery for which a composition can optionally be formulated include inhalation, intranasal, oral, buccal, sublingual, subcutaneous, intradermal, epicutaneous, rectal, transdermal, or intralymphatic.
For oral, buccal or sublingual administration, a composition may take the form of, for example, tablets or capsules, optionally formulated as fast-integrating tablets/capsules or slow-release tablets/capsules. In some embodiments, the tablet is freeze-dried, optionally a fast-disintegrating tablet or capsule suitable for being administered under the tongue.
The pharmaceutical composition may also be formulated into a "unit dosage form", which used herein refers to physically discrete units, wherein each unit contains a
predetermined quantity of a immunogenic molecule or immunogenic molecule
combination, optionally in association with a pharmaceutical carrier (excipient, diluent, vehicle or filling agent) which, when administered in one or more doses, may produce a desired effect. Unit dosage forms also include, for example, ampules and vials, which may include a composition in a freeze-dried or lyophilized state (a lyophilizate) or a sterile liquid carrier, for example that can be added prior to administration or delivery in vivo. Unit dosage forms also include a composition in a freeze-dried or lyophilized state; a sterile liquid carrier, for example, can be added prior to administration or delivery in vivo. Pharmaceutical formulations can be packaged in single or multiple unit dosage form for ease of administration and uniformity of dosage.
Immunogenic molecules may be prone to degradation when exposed to oxygen, for example when exposed to air or solvents containing air. Therefore, in some
embodiments, the pharmaceutical composition comprises an inert gas, e.g. argon or nitrogen.
Another aspect of the invention relates to a kit comprising a compartment and
instructions, wherein the compartment comprises a pharmaceutical composition as described herein and wherein the instructions are for use in reducing an immune response triggered by pollen, such as a pollen allergen, e.g. of a grass, weed or tree pollen species disclosed herein, such as instructions for use in treating allergy to pollen (e.g. grass, weed and/or tree pollen allergy to a plant family or species disclosed herein. A kit may further comprise packaging material comprising corrugated fiber, glass, plastic, foil, ampules, vials, blister pack, preloaded syringes or tubes, optionally that maintain sterility of the components. A kit may further comprise labels or inserts comprising printed matter or computer readable medium optionally including identifying components, dose amounts, clinical pharmacology and instructions for the clinician or for a subject using one or more of the kit components, prophylactic or therapeutic benefits, adverse side effects or manufacturer information.
In one embodiment, the kit additionally comprises a container comprising a solvent for dissolving the composition before use. Examples of suitable solvents are described supra. Optionally, the kit may also comprise a device for use in parenteral injection, e.g. for injecting the composition (e.g. dissolved composition) to a subcutaneous or intradermal tissue. A device may be any suitable device for that purpose, such as a needle or microneedle adapted for intradermal or subcutaneous delivery of the composition. For example, the device may be a microneedle or a device comprising a plurality of microneedles designed for intradermal delivery of liquids, e.g. as described in international patent applications W014064543 A1 , WO05049107 A2, WO06054280 A2, WO07066341 A3 and W014188429 A1.
A composition may be lyophilized so as to enhance stability and ease of transportation. For the purpose of being used as a vaccine, the composition may be sterile.
Pharmaceutical compositions can be formulated to be compatible with a particular route of administration. Thus, pharmaceutical compositions include carriers, diluents, or excipients suitable for administration by various routes. Exemplary routes of
administration for contact or in vivo delivery for which a composition can optionally be formulated include inhalation, intranasal, oral, buccal, sublingual, subcutaneous, intradermal, epicutaneous, rectal, transdermal, or intralymphatic. In some embodiments, the pharmaceutical composition is aqueous and, in other embodiments, the composition is non-aqueous solutions, suspensions or emulsions of the immunogenic molecule/protein, which compositions are typically sterile and can be isotonic with the biological fluid or organ of the intended recipient. Non-limiting illustrative examples include water, saline, dextrose, fructose, ethanol, vegetable or synthetic oils. For oral, buccal or sublingual administration, a composition can take the form of for example a solid dosage form, e.g. tablets or capsules, optionally formulated as fast- integrating tablets/capsules or slow-release tablets/capsules. In some embodiments, the tablet is a freeze-dried, optionally fast-disintegrating tablet suitable for being administered under the tongue. A solid dosage form optionally is sterile, optionally anhydrous.
To increase an immune response, immunological tolerance or protection against an allergic immune response, or one or more symptoms caused by or associated with an allergen, immunogenic molecules can be mixed with adjuvants.
Adjuvants include, for example: oil (mineral or organic) emulsion adjuvants such as Freund's complete (CFA) and incomplete adjuvant (I FA) (WO 95/17210; WO 98/56414; WO 99/12565; WO 99/11241 ; and U.S. Patent No. 5,422,109); metal and metallic salts, such as aluminum and aluminum salts, such as aluminum phosphate or aluminum hydroxide, alum (hydrated potassium aluminum sulfate); bacterially derived compounds, such as Monophosphoryl lipid A and derivatives thereof (e.g., 3 De-O-acylated monophosphoryl lipid A, aka 3D-MPL or d3-MPL, to indicate that position 3 of the reducing end glucosamine is de-O-acylated, 3D-MPL consisting of the tri and tetra acyl congeners), and enterobacterial lipopolysaccharides (LPS); plant derived saponins and derivatives thereof, for example Quil A (isolated from the Quilaja Saponaria Molina tree, see, e.g., "Saponin adjuvants", Archiv. fur die gesamte Virusforschung, Vol. 44, Springer Verlag, Berlin, p243-254; U.S. Patent No. 5,057,540), and fragments of Quil A which retain adjuvant activity without associated toxicity, for example QS7 and QS21 (also known as QA7 and QA21), as described in W096/33739, for example; surfactants such as, soya lecithin and oleic acid; sorbitan esters such as sorbitan trioleate; and
polyvinylpyrrolidone; oligonucleotides such as CpG (WO 96/02555, and WO 98/16247), polyriboA and polyriboU; block copolymers; and immunostimulatory cytokines such as GM-CSF and IL-1 , and Muramyl tripeptide (MTP). Additional examples of adjuvants are described, for example, in "Vaccine Design-the subunit and adjuvant approach" (Edited by Powell, M. F. and Newman, M. J.; 1995, Pharmaceutical Biotechnology (Plenum Press, New York and London, ISBN 0-306-44867-X) entitled "Compendium of vaccine adjuvants and excipients" by Powell, M. F. and Newman M.
Cosolvents may be added to the composition. Non-limiting examples of cosolvents contain hydroxyl groups or other polar groups, for example, alcohols, such as isopropyl alcohol; glycols, such as propylene glycol, polyethyleneglycol, polypropylene glycol, glycol ether; glycerol; polyoxyethylene alcohols and polyoxyethylene fatty acid esters. Non-limiting examples of cosolvents contain hydroxyl groups or other polar groups, for example, alcohols, such as isopropyl alcohol; glycols, such as propylene glycol, polyethyleneglycol, polypropylene glycol, glycol ether; glycerol; polyoxyethylene alcohols and polyoxyethylene fatty acid esters. Supplementary compounds (e.g., preservatives, antioxidants, antimicrobial agents including biocides and biostats such as antibacterial, antiviral and antifungal agents) can also be incorporated into the compositions. Pharmaceutical compositions may therefore include preservatives, anti-oxidants and antimicrobial agents. Preservatives can be used to inhibit microbial growth or increase stability of ingredients thereby prolonging the shelf life of the pharmaceutical formulation. Suitable preservatives are known in the art and include, for example, EDTA, EGTA, benzalkonium chloride or benzoic acid or benzoates, such as sodium benzoate. Antioxidants include, for example, ascorbic acid, vitamin A, vitamin E, tocopherols, and similar vitamins or provitamins. An antimicrobial agent or compound directly or indirectly inhibits, reduces, delays, halts, eliminates, arrests, suppresses or prevents contamination by or growth, infectivity, replication, proliferation, reproduction, of a pathogenic or non- pathogenic microbial organism. Classes of antimicrobials include antibacterial, antiviral, antifungal and antiparasitics. Antimicrobials include agents and compounds that kill or destroy (-cidal) or inhibit (-static) contamination by or growth, infectivity, replication, proliferation, reproduction of the microbial organism.
Pharmaceutical formulations and delivery systems appropriate for the compositions, methods and uses of the invention are known in the art (see, e.g. Remington: The Science and Practice of Pharmacy (David B. Troy, Paul Beringer Lippincott Williams & Wilkins) 2006).
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described herein.
All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the invention is not entitled to antedate such publication by virtue of prior invention.
As used in this specification and the appended claims, the use of an indefinite article or the singular forms "a," "an" and "the" include plural reference unless the context clearly dictates otherwise. In addition, it should be understood that the individual peptides, proteins, antigens, allergens (referred to collectively as compositions), or groups of compositions, modeled or derived from the various components or combinations of the compositions, and substituents described herein, are disclosed by the application to the same extent as if each composition or group of compositions was set forth individually. Thus, selection of particular peptides, proteins, antigens, allergens, etc. is clearly within the scope of the invention.
As used in this specification and the appended claims, the terms "comprise",
"comprising", "comprises" and other forms of these terms are intended in the non-limiting inclusive sense, that is, to include particular recited elements or components without excluding any other element or component. Unless defined otherwise all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. As used herein, "about" means + or - 5%. The use of the wild type (e.g., "or") should be understood to mean one, both, or any combination thereof of the wild types, i.e., "or" can also refer to "and." As used in this specification and the appended claims, any concentration range, percentage range, ratio range or other integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated. For example, although numerical values are often presented in a range format throughout this document, a range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the use of a range expressly includes all possible sub ranges, all individual numerical values within that range, and all numerical values or numerical ranges including integers within such ranges and fractions of the values or the integers within ranges unless the context clearly indicates otherwise. This construction applies regardless of the breadth of the range and in all contexts throughout this patent document. Thus, to illustrate, reference to a range of 90-100% includes 91-99%, 92-98%, 93-95%, 91-98%, 91-97%, 91-96%, 91-95%, 91- 94%, 91-93%, and so forth. Reference to a range of 90-100%, includes 91%, 92%, 93%, 94%, 95%, 95%, 97%, etc., as well as 91.1%, 91.2%, 91.3%, 91.4%, 91.5%, etc., 92.1%, 92.2%, 92.3%, 92.4%, 92.5%, etc., and so forth. Reference to a range of 5-10, 10-20, 20- 30, 30-40, 40-50, 50-75, 75-100, 100-150, and 150-175, includes ranges such as 5-20, 5- 30, 5-40, 5-50, 5-75, 5-100, 5-150, 5-171 , and 10-30, 10-40, 10-50, 10-75, 10-100, 10- 150, 10-175, and 20-40, 20-50, 20-75, 20-100, 20-150, 20-175, and so forth. Further, for example, reference to a series of ranges of 2-72 hours, 2-48 hours, 4-24 hours, 4-18 hours and 6-12 hours, includes ranges of 2-6 hours, 2, 12 hours, 2-18 hours, 2-24 hours, etc., and 4-27 hours, 4-48 hours, 4-6 hours, etc.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims. The invention is further exemplified by way of the following non-limited examples.
References
Altschul, S.F., Gish, W., Miller, W., Myers, E.W. & Lipman D.J. Basic local alignment search tool, J. Mol. Biol. 215, 403-410, 1990
Bostick D, Vaisman II. A new topological method to measure protein structure similarity. Biochem Biophys Res Commun. 304, 320-325, 2003.
Greenbaum J, Sidney J, Chung J, Brander C, Peters B, Sette A. Functional classification of class II human leukocyte antigen (HLA) molecules reveals seven different supertypes and a surprising degree of repertoire sharing across supertypes. Immunogenetics, 63, 325-335, 2011.
Karosiene, Edita, Michael Rasmussen, Thomas Blicher, Ole Lund, Soren Buus, and Morten Nielsen. "NetMHCIIpan-3.0, a Common Pan-specific MHC Class II Prediction Method Including All Three Human MHC Class II Isotypes, HLA-DR, H LA-DP and HLA- DQ." Immunogenetics, Vol. 65, No. 10, 2013, p. 711-724.
McKinney DM, Southwood S, Hinz D, Oseroff C, Arlehamn CS, Schulten V, et al. A strategy to determine HLA class II restriction broadly covering the DR, DP, and DQ allelic variants most commonly expressed in the general population. Immunogenetics; 65:357- 70, 2013.
Needleman SB, Wunsch CD. A general method applicable to the search for similarities in the amino acid sequence of two proteins, J. Mol. Biol. 48:443, 1970.
Oseroff C, Sidney J, Kotturi MF, Kolla R, Alam R, Broide DH, et al. Molecular
determinants of T cell epitope recognition to the common Timothy grass allergen. Journal of immunology 2010; 185:943-55. Pearson and Lipman, 1988: Pearson WR, Lipman DJ. Improved tools for biological sequence comparison, Proc. Natl. Acad. Sci. USA 85(8): 2444-2448,1988.
Pearson WR1 , Flexible sequence similarity searching with the FASTA3 program package, Methods Mol Biol. 132, 185-219, 2000. Sidney J, Southwood S, Oseroff C, del Guercio MF, Sette A, Grey HM. Measurement of MHC/peptide interactions by gel filtration. Curr Protoc Immunol. Chapter 18(Unit 18): 13. doi: 10.1002/0471142735.im1803s31 , 2001
Sidney J, Assarsson E, Moore C, Ngo S, Pinilla C, Sette A, Peters B. Quantitative peptide binding motifs for 19 human and mouse MHC class I molecules derived using positional scanning combinatorial peptide libraries. Immunome Res. 4:2, doi: 10.1186/1745-7580-4- 2, 2008
Sidney J, Steen A, Moore C, Ngo S, Chung J, Peters B, Sette A. Divergent motifs but overlapping binding repertoires of six HLA-DQ molecules frequently expressed in the worldwide human population. J Immunol, 185(7), 4189-4198, 2010a. Sidney J, Steen A, Moore C, Ngo S, Chung J, Peters B, Sette A. Five HL.A-DP molecules frequently expressed in the worldwide human population share a common HLA supertypic binding specificity. J Immunol, 184(5), 2492-2503, 2010b.
Sidney J, Southwood S, Grey HM, Moore C, Oseroff C, Pinilla C, Sette A. Measurement of MHC/peptide interactions by gel filtration or monoclonal antibody capture. Current protocols in immunology / edited by John E. Coligan et al, Chapter 18():Unit 18.3, 2013.
Smith and Waterman 1981 : Smith and Waterman. Comparison of biosequences, Adv. Appl. Math 2: 482, 1981.
Examples
Example 1 - Transcriptomic analysis
This example includes a description of transcriptomic analysis of various pollen species.
RNA-sequencing was performed on pollen samples of the following species: Timothy grass (Phleum pratense (Phi p)), Bermuda grass (Cynodon dactylon (Cyn d)), Western ragweed (Ambrosia psilostachya (Amb p)), Short ragweed (Ambrosia artemisiifolia (Amb a)), White oak (Quercus alba (Que a)), and European white birch (Betula verrucosa (Bet v)) (See Table below). RNA-seq was run at UCSD, using an lllumina HiSeq 2000. The table below shows the number of reads assembled for each of the different pollens (top), with over 500 million reads over two replicate runs per allergen. Sequences were assembled into transcripts using Trinity (bottom), resulting in over 50 thousand transcripts per allergen with minimum lengths of 200 nucleotides. The transcripts include related variants, such as isoforms, and homologs.
Transcriptomic analysis may also be performed on RNA sequences of additional species as shown in the Table below showing pollen RNA-seq reads for the pollen species Kentucky blue grass, Sweet vernal grass, Rye grass and Ash tree, Olive tree and English plantain:
Figure imgf000066_0001
Raw read counts (millions)
Additional species
Sweet Ash Rye grass Olive English Kentucky vernal plantain blue grass
Ant O FE Lol P Ole E PL Poa P
1st run 394 410 332 385 303 363
2nd run 360 346 319 350 287 309
Total 754 756 651 735 590 672
Transcripts after Trinity assembly
Count 317,874 81 ,401 122,266 74,333 57,102 128,174 min 201 201 201 201 201 201 median 544 722 631 710 696 635
max 11 ,515 9,838 9,631 8,133 8,090 10,100 Example 2 - Identification of polypeptides in Phi p grass pollen and homologous polypeptides thereof
This example includes a description of the identification of polypeptides in extracts of Phi p grass pollen and homologous polypeptides thereof in other pollen species. In order to identify polypeptides in Phi p grass pollen, the Phi p grass transcriptome was translated into amino acid sequences in all six reading frames, and they were used to search 17 mass spectrometry samples of Phi p grass pollen extract using the Mascot software. This identified 354 polypeptide sequences. Polypeptide sequences shorter than 50 aa were discarded, resulting in 275 sequences. In order to identify the correct coding region of each transcript, there was identified the closest homologous sequence in the rice (Oryza sativa japonica) proteome (via Blast). Rice was chosen since it is a species closely related to Phi p grass with a completely sequenced and annotated genome. Homologous rice sequences were identified for 180 Timothy grass sequences.
Subsequently, homologous sequences were identified (via Blast) in the translated transcriptomes of Cyn d, Amb a, Amb p, Que a, and Bet v of all identified sequences, the one(s) sharing the largest number of conserved peptides with the Phi p sequence was selected as homolog. This resulted in 66 sets of homologous polypeptides. In total, there was found evidence of release of the polypeptides from Phi p grass pollen for 52 of these polypeptides upon extracting pollen in a buffered aqueous solution (see Example 4). Table 1 shows amino acid sequences of the polypeptides detected and identified in Phi p grass pollen and Table 2 shows amino acid sequences of polypeptides homologous to the Phi p sequence of Table 1 found in pollen of the species Cyn d, Amb a, Amb p, Que a, and Bet v.
Table 1. Polypeptides of Timothy Grass Pollen (Phi p)
Table 1 - SEQ ID NO: 1-37
SEQ ID NO: SpeAmino Acid Sequence
ID cies
NO:
1 A0202 Phi p HGIQQRKNSWQDGTPGAMCPIMPGTNFTYKMQFKDQIGTFFYFPSIGMQRAAG
GYGLITIHSRLLIPVPFDPPADDFSVLVGDWYTKD
2 A0203 Phi p KTIPLEVESSATIDNVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQ
3 A0203 Phi p MQIFVKILTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLIFAGKQLEDGR
4 A0209 Phi p ASFGDAPAGNAASGE IFRTKCAQCHTVERGGAHKQGPNLSGLFGRQSGTT
AGYAFSAGN NAAWWGDDTLYDYLLNPKKYIPSTKMVFPGLKKPQERTDLIAY LKESTA
5 A0209 Phi p ASFSEAPPGNAAAGEKIF TKCAQCHTVEKGAGHKQGPNLNGLFGRQSGTTP
GYSYSTANKNMAVIWEEKTLYDYLLNPKKYIPGTKMVFPGLKKPQERADLIAYLK DATA
6 A0211 Phl p MANSASGLAVHDDCKIKFLDLKARRSFRFIVFKIDEKAMEIKVERLGEASHGYEE
FTNSLPADECRYAVYDLDFVTDENCQKSKIFFFSWSPDTARTRSK LYASSKDR
FRREMDGIQCEIQATDPSEMSLDIIKSRAH
7 A0211 Phi p MANSASGMAVSDECKLKFQDLKSKRSFRFITFKINEQTQQVWDKVGQPGDTY
SDFTATMPEGECRYAVFDFDFVTDENCQKSKIFFFSWSPDTARTRSKMLYASS Table 1 - SEQ ID NO: 1-37
SEQ ID NO: SpeAmino Acid Sequence
ID cies
NO:
KDRFRREMDGIQCEIQATDPSEMSLDIIKSRAH
8 A0211 Phi p MSNSASGMAVCDECKLKFQELKAKRSLRFIVFKINEKVQQVWDRLGQPNESY
DDFAACLPADECRYAVFDFDFVTDENCQKSKI
9 A0215 Phi p AITVDDACRQYTKHRSYCAHALSAKAGPSETTLPALAEQAVTLAAESGEEAVAF
VRNLEKMPGGMPLGCLERCVGKFQAAVAELTRSRAAIVEHRDVARVKAWVKAA
RADGETCMDGCHTEGGADPTIIHRIGDLGKLCSIALSLTDA
10 A0219 Phl p LLKVNQIGSVTESIEAVKMS RAGWGV TSHRSGETEDTFIADLAVGLSTGQIKT
GAPCRSERLAKYNQLLRIEEELGAAAVYAGLKFRAP
11 A0233 Phi p M GS LS YVLALAVLAALVTGG AC IT VP PG P N ITTKYDTKWLP AKATWYG KPTG A
GPKDNGGACGIKDVNMAPYLGMTACGNVPIFKDGKGCGSCYQVKCKKPEPCS
DKPITIFITDKNYEPIAPYHFDLSGKAFGL ALPGKDQALRSVGELELQFRRVQC
KYPPGTKITFHVEKGSNPNYLAVLVKFVADDGDIVQVDIQDTQSPAWKPMTESW
GANA/VRWDGVPPLKGPISLRLTSESGKKLTAKDVIPATWKADTVYPSKIQF
12 A0246 Phl p DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGGTMIKVKTLTG
KEIEIDIEPTDTIDRIKERVEEKEGIPPVQQRLIYAGKQLADDKTAKDYNIEGGSVL HLVLALRGGQ
13 A0260 Phi p MAGRGELWSFGE LIDFVPTVAGVSLAEAPAFVKAPGGAPANVAIAVARLGGK
AAFAGKLGDDEFGRMLAAILRDNGVDDSAWFDAGARTALAFVTLRADGDREF
MFYRNPSADMLLTADELNVGLLKRAAVFHYGSISLITEPCRSAHLRAMEIAKEAG
ALLSYDPNLREALWPSLEEARTKILSIWDQADIVKVSEVEVEFLTGVDSVEDDHV
MKLWRPTFKLMLVTLGDQGCKYYARDFRGMVSSYKVQQVDTTGAGDAFVGAL
LRKIVQDPSSLQDQKKLEGIIKFANACGAITTLTKGAIPSLPTETAVLRLMENA
14 A0262 Phi p VEYRCFVGGLAWATDDFNLKQAFSQFGEILDAKIINDRETGRSRGFGFVTFDSA
DAMNAAIEAMNGKDLDGRNITVNKANDRPRRRR
15 A0265 Phi p MGFRSDGSPATGGGISEKGRFSYGFASCTG RASMEDFYETRVDEVDGDTVG
LFGVFDGHGGARAAEYVKKHLFSNLIKHPKFITDTKSAIAETFNHTDSEFLKADS
SQSRDAGSTASTAIIVRGRLWANVGDSRAWSKGGKAIAVSRDHKPDQTDERQ
RIEEAGGFVMWAGTWRVGGVLAVSRAFGDKLLKQYWADPEIKEEWDGSLEF
LILASDGLWDWTNDEAVAMVKPIEDPEQAAKRLLEEASIRGSADNITWIVRFLD
GTTTVG
16 A0270 Phi p PWEAPTPILGEPMDLMTALQLVMKKSSAHDGLVKGLREAAKAIEKHVAQLCVL
AEDCDQPDYVKLVKALCAEHNVHLVTVPSAKTLGEWSGLCKIDSEGKARKWG CSCVWKDYGEESEGLNIVQEYVKSH
17 A0300 Phi p INDAVSRILRVKFTMGLFEDPLPDRRLAEHLGCKSHRELARDAVRKSLVLLKNGK
KNATAVLPLSKNAKKILVAGSHAHDLGLQCGGWTKSWQGQSGNNITGQGTTIL
EAIKSAVNN TVIDYSEHPDKDSISKSEDKYEYAVWVGEQAYAETAGDNQNLTI
PSPGPDVIRDVCELVKCVWLVSGRPLVIEPYLDTMDAFVAAWLPGTEGHGVAD
LLFGDYGFTGKLPRTWFRSVDQLPMNYGDKLYDPLFSFGYGLT
18 A0304 Phi p WFLEINPEGKVPVFNSGDGKWIADSDVITQVIEEKYPTPSLVTPPEYASVGSKIF
STFIAFLKSKDATDGTEKALVDELQALNEHLKAHGPYINGESVSAADLSLGPKLF HLQVALEHFKGWKIPETLTSVHAYT
19 A0311 Phl MSFTGTQDKCKACDKTVHFIDLLTADGVSYHKTCFKCSHCKGTLSMCSYSSMD
GVLYCKTHFEQLFKETGSFSKNFTPGG STDKNDQSKAPNKMSSIFSGTC.DKC AACQKTVYPLEKLSLEGECYHKSCFKCSHGGCILTTSSYAALNGILYCKIHFSQL FKEKGSYNHLIKTAQTKKENEEAAAAAEAEADADAKDNEEEVPPQDAT
20 A0316 Phi p MADAEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPRHTGVMVGMGQK
DAYVGDEAQSKRGILTLKYPIEHGIVSNWDDMEKIWHHTFYNELRVAPEEHPVL
LTEAPLNPKANREKMTQI FETFNTPA YVAIQAVLSLYASGRTTGIVLDSGDGV
SHTVPIYEGYALPHAILRLDLAGRDLTDYLMKILTERGYSh I I I AEREIVRDVKEK
LSYIALDYDQELDTSKTSSSVEKSYELPDGQVITIGSERFRCPEVLFQPSFIGMEA
AGIHETTYNSIMKCDVDIRKDLYGNIVLSGGTTMFPGIADRMSKEITALAPSS KI
KWAPPERKYS\ WIGGSILASLSTFQQ WIAKAEYDESGPSIVHRKCF
21 A0317 Phi p GCITTKELGTV RSLGQNPTEAELQDMINEVDADGNGTIDFPEFLNL ARKMKD
TDSEEELKEAFRVFDKD
22 A0325 Phi p VKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGGMAKK
RKKKNYTTPKKIKHKRKPVKMAILKYFRVDSDGKIKRLRRECPAETCGPSVF A
QHADRQYCGKCGLTYKFDAE
23 A0326 Phl p TGDELLSDSFPYTELFEGVLWEVNGKWWNGSVDVDIGANPSAEGGCDDEGV
DDQAVKWDIVDTFRLQEQPPFD KQFLGFMKGYIKKLTAKLEGDELDCFKKNIE
GATKFLLGKIKDLQFFVGESMHDDCAWCAYYKEGAIDPTFLYFAHGLNEVKC
24 A0327 Phl p LLAVAVAFSAAAAINVEVAAVCKGTPYAELCSATAGKQASHYATVDALAVLGMQ Table 1 - SEQ ID NO: 1-37
SEQ ID NO: SpeAmino Acid Sequence
ID cies
NO:
VDAFSKRASAASIHATQLIARGKLSPGALIALRKCNEMYVNVEDNLGAARRAISF KDAVTIRAMMS AAQDMKNCDEEFRKNAAVNPMTRFDKSLLDISENCRALSNM
I
25 A0334 Phi p MALRNVAIRVFFLLLWSAAYAGKKEEKKDEKKESGDAASGADGTYDITKLGAK
PDGKTDCTKEVEEAWASACGGTGKNTIVIP GDFLTGPLNFTGPCKGDSVTIKL
DGNLLSSNDLAKYKANWIEIMRIKKLTITGKGTLDGQGKAVWGKNSCAKNYNCKI
LPNTLVLDFCDDALIEGITLLNA FFHMNIYECKGVTVKDVTITAPGDSPNTDGIHI
GDSSNINVTGTTIGTGDDCISIGGGSSRIIVTGVTCGPGQGISVGCLGRYKDEKD
VSDVTVKDCVLRSSTNGVRVKTYEDAVKSITASKLTFENIK DDVANPIIIDQNYC
PEKVCTAKSKSAVTVKDVIFRNITGTSSTPAAVSLLCSDKQPCSGVELIDVNVEY
SGKNNKTIGVCNNAKGTAKDTLQALACL
26 A0336 Phi p MEQTFIMIKPDGVQRGLIGDIISRFEKKGFYLKAMKFMNVERSFAQQHYADLSGK
PFFPKLVDYIISGPWAMVWEGKDWLTGRRIIGATRPWEAAPGTIRGDYAVEVG RNVIHGSDSVENGQKEIALWFPGGLAEWRSNLHPWIYEN
27 A0349 Phi p KELKDLQRDPPTSCSAGPVAEDMFHWQATIMGPAESPYAGGVFLVTIHFPPDY
PFKPPKVAFKTKVFHPNINSNGSICLDILKEQWSPALTVSKVLLSICSLLTDPNPD DPLVPEIAHMYKSDRVKYESTARSWTQKYAMG
28 A0349 Phi p ASKRILKELKDLQKDPPTSCSAGPVGEDMFHWQATIMGPSDSPFTGGLFLVNI
HFPPDYPFKPPKVSFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAH YKTDRAKYESTARSWTQKYAMG
29 A0349 Phi p DMFHWQATI GPSDSPYSGGVFLVTIHFPPDYPFKPPKVAFKTKVFHPNVNSN
GSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKADRAKYEST ARSWTQ
30 A0349 Phi p MASKRILKELKDLQKDPPTSCSAGPAGEDMFHWQATIMGPPDSPYAGGVFLVNI
HFPPDYPFKPPKVSFKTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMYKTDRSKYETTARSWTQKYAMG
31 A0356 Phi p MGKEKLHISIWIGHVDSGKSTTTGHLIYKLGGIDKRVIERFEKEAAEMNKRSFKY
AWVLDKLKAERERGITIDIALWKFETTKYSCTVIDAPGHRDFIKNMITGTSQADCA
VLIIDSTTGGFEAGISKDGQTREHALLAFTLGVKQMICCCNKMDATTPKYSKSRF
EEIVKEVSSYLKKVGYNPDKVPFVPISGFEGDN IERSTNLDWYKGPTLLEALDQ
VTEPKRPTDKPLRLPLQDVYKIGGIGTVPVGRVETGLIKPGMLVTFGPTGLTTEV
KSVEMHHESLLEAGPGDNVGFNV NVAVKDIKRGYVASNAKDDPAKESASFVA
QVIIMNHPGQIGNGYAPVLDCHTSHIAVKFAEIQTKVDRRSGKWEEFPKFLKNG
DAGFVKMVPTKPMWETFSQYPPLGRFAVRDMRQTVAVGVIKSVEKKDPTG
32 A0357 Phl p DMERIFKRFDTNGDGKISLSELTDALRTLGSTSADEVQRMMAEIDTDGDGFIDFN
EFISFCNANPGLMKDVAKVF
33 A0358 Phi p MSTPAAQVSEKPALRKPVFLKVDQLKPGTNGHNLIVKWSANPVPGRVRPGAP
ASSSARAPRIAECLVGDETGCIIFTARNDQVDLLKAGATAILRNAKIDMFKSSMRL
AVDKWGRVDSSEPASFTVNEENNLSQVEYELVNVAE
34 A0362 Phi p FSPEEISAMILGKMKETAEAYLGKKINDAWTVPAYFNDAQRQATKDAGVIAGLN
VARIINEPTAAAIAYGLDKKGSEKNILVFDLGGGTFDVSILTIDNGVFEVLATNGDT
HLGGEDFDHRIMDYFIKLIKKKYSKDISKDNRALGKLRREAERA RALSNQHQVR
VEIESLFDGTDFSEPLTRARFEELNNDLFRKTMGPVK AMDDAGLEKSQIHEIVL
VGGSTRIPKVQQLLRDYFDGKEPSKGVNPDEAVAFGAAVQGSILSGEGGDETK
DILLLDVAPLTLGIETVGGVMTKLIPRNTVIPTKKSQVFTTYQDQQTTVSIQVFEG
ERSMTKDCRLLGKFDLNGIPAAPRGTPQIEVTFEVDANGILNVKAEDKGTGKSE
KITITNEKGRLSQEEIDRMVKEAEEFAEEDKKVK
35 A0366 Phi p GTSAVSSLAATVPVEAVLFDIDGTLCDSDPLHHVAFQEMLLAIGYNNGVPIDEEF
FIKNIAGRSDVEAAQNLFPDWPLEKGLKFLEDKETKYRSLAMERLEPVNGLGKV VQWVKDHGYKRAAVTNAPRINAELML LLGLSDFFQAVIVGGECEKPKPAPFPY LKAL ELDVSAAHTFIFEDSASGTRAGVAAGMPWAVLTRNPEKSLEEAGAALIV KDYADPKLWSALEEIDGEEAKLKKGAA
36 A0376 Phi p MAQNFLLGGVGAILWAVWGWATVTHSGNKAGDNFTVPGEATLATSGKSVQ
SLCAPTLYKESCE TLTSASNGTENPKEVFSTMAMSAMESIKSAVERSKTIGEAK
SSDPLTQGAREDCKELLEDSVDDL GMVEMAGGDIKVLLSRSDDIEHWITGVMT
FIDTCADGFADEKLKADMQLILRNATELSSNALAITNSLGAIFKKLDLDVFKKDSR
RRLLSEKDEQGYPAWMKSPERKLLATGAMPAPNAWAKDGSGKFKTIQEAVNA
MPKGHPGRYVIYVKTGLYDEIVMIPKDKVNIFMYGDGPKQSRVTGRKSFKDGITT
MKTATFSIEMGFICKNMGFHNTAGAENHQAVALRVQGDLAAFYNCRFDAFQD
TLYVHARRQFFRNCVISGTIDFIFGNSAAIFQNCLIITRRP DNQQNSVTAHGRTD
PN KSGLVIQNCRLVPDQKLFPDRFKIPSYLGRPWKEYSRLVIMESTIADFIKPE Table 1 - SEQ ID NO: 1-37
SEQ ID NO: SpeAmino Acid Sequence
ID cies
NO:
GYMPWNGDFGIKTLYYAEYANRGPGAGTSKRVNWPGFRVIGRKEAEQFTTGP FVDGATWL FTGMPNYLGFKV
37 A0377 Phi p RLKTGFEKFKTDVYD KPDMFEPLKAGQAPKYMVFACADSRVCPSVTLGLEPG
EAFTVRNIANMVPSYCKNKYAGVGSAIEYAVCALKVEVIWIGHSRCGGIKALLSL KDGADDSFHFVEDWVRIGFPAKKKVQ ECASLSFDDQCGVLEKEAVNVSLENL LTYPFVKEGVSAGTLKLVGGHYDFVSGKFDTWE
Table 2. Polypeptides homologous to Phi p polypeptides shown in Table 1.
Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
38 A0202 Amb a NGMQHRKNSWMDGMPGTQCPILPNTNFTYKWQPKDQIGSF
YYFPSIGMQRAAGGYGGISVYSRLUPVPFDQPPPENDHWLI GDWYTKD
39 A0202 Amb p NGMQQRKNSWQDGLPGTNCPVAPGTNYTFKWQAKDQIGS
FFYFPSLGMQRAAGGYGMISWSRLLIPVPFDPPADDHWLI GDWYTKD
40 A0202 Bet v GIQHRKNSWQDGVLGTMCPIPPGTNYTYHFQVKDQIGSYIY
YPTTATHRAAGAFGGLRVNSRLLIPVPYADPEDDYTVLIGDW
YAK
41 A0202 Que a NGIQHRKNSWQDGLPGTNCPVAPGTNYTFKWQAKDQIGSF
FYFPSLGMQRAAGGYGMISWSRLLIPVPFDPPADDFQVLVG
DWYTKD
42 A0202 Cyn d NGMQHRKNSWMDGLPGTNCPIAPGTNFTYKWQPKDQIGSF
FYFPSIAMQRSAGGYGLISVHSRDLIPVPFDIPADDFAVLAGD WYTKD
43 A0203 Amb a MQIFVKTLTGKTITLEVESSDTIDNVKSKIQDKEGIPPDQQRLI
FAGKQLEDGRTLSDYNIQKESTLH
44 A0203 Amb a MQIFVKTLTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLI
FAGKQLEDGRTLADYNIQKESTLHLVLRLRGGF
45 A0203 Amb p MQIFVKTLTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLI
FAGKQLEDGRTLADYNIQKESTLHLVLRLRGGF
46 A0203 Bet MQIFVKTLTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLI
FAGKQLEDGRTLADYNIQKESTLHLVLRLRGGMQIFVKT
47 A0203 Que a MQIFVKTLTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLI
FAGKQLEDGRTLADYNIQKESTL
48 A0203 Cyn d MQIFVKTLTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLI
FAGKQLEDGRTLADYNIQKESTLHLVLRLRGGL
49 A0209 Amb a MAS FAE AP AG N PTVG E Kl F KTKCAQC HTVE KG AG H KQG P N L
NGLFGRQSGTTAGYSYSAGNKNKAVIWEENTLYDYLLNPKK YIPGTKMVFTGLKKPQERADLISYLKQSTA
50 A0209 Amb a MASFEEAPAGDAKVGEKIFKTKCAQCHTWKGAGHKQGPNL
NGLFGRQSGTTAGYSYSAANKNMAVIWEEKTLYEYLLNPKK
YIPGTKMVFPGLKKPKDRADLIAYLKESTA
51 A0209 Amb p M AS FE E APAG DAKVG E Kl F KTKCAQC HTWKG AG H KQG P N L
NGLFGRQSGTTAGYSYSAANKNMAVIWEEKTLYEYLLNPKK
YIPGTKMVFPGLKKPKDRADLIAYLKESTA Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
52 A0209 Bet v MASFDEAPPGNPKVGEKIFKTKCAQCHTVEKGAGHKQGPNL
NGLFGRQSGTTAGYSYSSANKNMAVNWEEKTLYDYLLNPK
KYIPGTKMVFPGLKKPQDRADLISYLKESTA
53 A0209 Que a SGEKIFKTKCAQCHTVEKGAGHKQGPNLNGLFGRQSGTTA
GYSYSTANKNMAVIWEEKTLYDYLLNPKKYIPGTKMVFPGLK
KPQERADLIAYLKSSTA
54 A0209 Cyn d PPGNPKAGEKIFKTKCAQCHTVDKGAGHKQGPNLNGLFGR
QSGTTPGYSYSAANKNRAVIWEENTLYDYLLNPKKYIPGTKM
VFPGLKKPQERADLIAYLKEATS
55 A0211 Amb a MANAASGMAVDDECKLKFQELKAKRSYRFITFKIEGQQVW
DKFGMPDETYEDFTNSLPADECRYAVFDFDFTTDENCQKSK
IFFIAWSPDTSRVRMKMVYASSKD
56 A0211 Amb p MANAASGMAVHDECKLKFLELKAKRTFRYIIFKIEEKQKEVIV
EKVGEPTQSHGDFAAALPDAECRYAVFDYDFVTEENCQKSR IFFIAWSPDTARVRNKMIYASSKDRFKRELDGIQVELQATDPT EMDLDVFKSRAN
57 A021 1 Amb p LKFQELKAKRSYRFITFKIEGQQVWDKFGMPDETYEDFTNS
LPADECRYAVFDFDFTTDENCQKSKIFFIAWSPDTSRVRMK
MVYASSK
58 A0211 Amb p MANAASGMAVNDECKLKFSELKSKRNYRFIVFKIEQQQVW
EKVGSPDESYEDFTNSLPANECRYAVFDFDFTTDENCQKSKI FFIAWAPDTSKVREKMVYASSKDRFKRELDGIQVEVQATDP SEMSLDIVKARAL
59 A0211 Amb p DECRYAVFDFDFVTDENCQKSKIFFISWAPDIARVRSKMLYA
SSKDRFKRELDGIQVELQATDPSEMSMDIVKARAL
60 A0211 Bet v MANSASGMAVHDECKLKFLELKAKRNHRFIVFKIDEKIQQVIV
EKVGSPDETYDDFNASMPPNECRYAVFDFDFTTDENVQKSK IFFIAWAPDTSRVRSKMLYASSKDRFRRELDGVQVELQATDP SEMSLDIIKGRAL
61 A0211 Bet v MANAASGMAVSDECKLKFLELKTKRNYRFIIFKIENQEVWEK
VGCPDETYEDFTASIPADECRYAVFDFDFITDENCQKSKIFFI AWS P DTS RVRS KMVYAS SKDRFKRELDGI Q VE LQ ATD PS E M SLDIVKGRAL
62 A0211 Que a MANSASGMAVHDDCKLKFLELKAKRNYRFIVFKIEEKIQQVIV
EKLGGPEETYDDFTASMPANECRYAVFDFDFITDENCQKSKI FFISWSPDTSRVRSKMLYASSKDRFRRELDGVQVELQATDP SEMSLDIIKGRAL
63 A0211 Cyn d MSNSASGMAVCDECKLKFQELKAKRSFRFIVFKINEKVQQW
VDRLGQTGESYDDFTACLPADECRYAVFDFDFVTDENCQKS KIFFISWSPDTSRVRSKMLYASSKDRFKRELDGIQVELQATD PSEMSMDIVKARAL
64 A0215 Amb p AITVDEACRQYTKHPSYCKRTLAPKVDPSAPTTLPALAEQAV
LLAAESGVAAVSYVKNLEKIPGGMPLGCLERCVGRFQAAVA ALMQSRVAIVEHRDVARVKAWVKTARVDGENCMAGCHTEG GVDPAVKRRISKLGKICSI
65 A0215 Que a AITVDEACRQYTKHPSYCTHALSAKAGPPETTLPALAVQAVT
LAAESGGSAVLFVKNLEKMPGGMPLGCLERCVGKFQAAVA
ELTLSRVAIVEHRDVARVKAWVKAARADGETCMDGCHTEG Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
GADPTIIHRIGDLGKLCSIALSLTD
66 A0215 Cyn d AITVDEACRKYTKHPSYCTHALSAKAGPPETTLPALAVQAVT
LAAESGGSAVLFVKNLEKMPGGMPLGCLERCVGKFQAAVA ELTLSRVAIVEHR D VAR VKAWVKAARADG ETC DGCHTEG GADPTIIHRIGDLGKLCSIALALTDA
67 A0219 Amb a LLKVNQIGSVTESIEAVKMSKRAGWGVMASHRSGETEDTFIA
DLSVGLATGQIKTGAPCRSERLAKYNQLLRIEEELGAAAVYA
GSKFRAP
68 A0219 Amb p LLKVNQIGSVTESIEAVKMSKRAGWGVMASHRSGETEDTFIA
DLSVGLATGQIKTGAPCRSERLAKYNQLLRIEEELGAAAVYA
GSKFRAP
69 A0219 Bet v LLKVNQIGSVTESIEAVRMSKRAGWGVMASHRSGETEDTFIA
DLSVGLATGQIKTGAPCRSERLAKYNQLLRIEEELGSAAVYA
GSKYRAP
70 A0219 Que a LLKVNQIGSVTESIEAVKMSKHAGWGVMASHRSGETEDTFIA
DLSVGLATGQIKTGAPCRSERLAKYNQLLRIEEELGPAAVYA
GSKFRAP
71 A0219 Cyn d LLKVNQIGSVTESIEAVKMSKHAGWGVMTSHRSGETEDTFIA
DLAVGLATGQIKTGAPCRSERLAKYNQLLRIEEELGAAAVYA
GAKFRAP
72 A0233 Amb a KFPPGPNITTNYNGQWLTARATWYGQPNGAGPDDNGGAC
GIKGVNLPPYNGMTACGNIPIFKDGKGCGSCYEVRCKEKPE CSGQPITVFITDMNYEPIAPYHFDFSGKAFGSLAKPGLNDKL RHCGIMDVEFRRVRCKLPGQKILFHVEKGCNPNYLAVLVKN VADDG
73 A0233 Amb p CITKVPPGPNITTKYDTKWLPAKATWYGKPTGAGPKDNGGA
CGIKDVNLAPYLGMTACGNVPIFKDGKGCGSCYELKCQKPE PCADKP
74 A0233 Que a MGSLSYVLAVAVLAALVTGGVCITKVPPGPNITTKYDTKWLP
AKATWYG KPTGAG PKDNGGACG I KDVN LAPYLG MTACG N V
PIFKDGKGCGSCYELKCQKPAPCADKPITIFITDKNYEPIAPY
HFDLSGKAFGLMALPGQDQKLRSVGELELQFRRVQCKYPA
GTKITFHVEKGSNPNYLAVLVKFVANDGDIVQMEIQDSQSAA
WKPMTESWGAVWRWDGAPPLKGPLSLRLTSESGKKLIAKD
VIPATWKADNVYPSNIQF
75 A0233 Cyn d GSLSYVLAVAVLAALVTGGACITKVPPGPNITTKYDTKWLP
AKATWYGKPTGAGPKDNGGACGIKDVNLAPYLGMTACGNV
PIFKDGKGCGSCYELKCQKPEPCADKPITVFITDKNYEPIAPY
HFDLSGHAFGLMALPGKDQALRSVGELELEFRRVRCKYPPG
TKITFHVEKGSN
76 A0233 Amb p EPCSDKPITVFITDKNYEPIAPYHFDLSGKAFGLMALPGKDQA
LRSVGELELQFRRVQCKYPAGTKITFHVEKGSNPNYLAVLVK FVANDGDIVQ EIQDSQSAAWKPMTESWGAVWRWDGAPP LKGPLSLRLTSESGKKLIAKDVIPATWKADNVYPSNIQF
77 A0246 Amb a DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLR
GGMQIFVKTLTGKTITLEVESSDTIDNVKAKIQDKEGIPPDQQ
RLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGG
78 A0246 Amb p NIQKESTLHLVLRLRGGTMIKVKTLTGKEIEIDIEPTDTIDRIKE Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
RVEEKEGIPPVQQRLIYAGKQLADDKTAKDYNIEGGSVLHLV LALRGG
79 A0246 Bet v DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLR
GGTMIKVKTLTGKEIEIDIEPTDTIDRIKERVEEKEGIPPVQQRL IYAGKQLGDDKTAKDYNIEGGSVLHLVLALRGGS
80 A0246 Que a QQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGGTMIKV
KTLTGKEIEIDIEPTDTIDRIKERVEEKEGIPPVQQRLIYAGKQL
GDDKTAKDYNIEGGSVLHLVLALRGGS
81 A0246 Cyn d DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLR
GGTMIKVKTLTGKEIEIDIEPTDTIDRIKERVEEKEGIPPVQQRL IYAGKQLADDKTAKDYNIEGGSVLHLVLALRGGQ
82 A0260 Amb a NGSLWCFGEMLIDFVPSVSGVSLAEAPAFHKAPGGAPANV
AVGISRLGGSSAFIGKVGDDEFGRMLADILKENKVDNSGMRF
DQKARTALAFVTLRSDGEREFMFFRNPSADMLLHESELDVN
LIKQASIFHYGSISLIEEPCKSTHLAAMAIAKKSGSILSYDVNLR
LPLWPSEDAARDGIMSIWDQAD
83 A0260 Amb p NGSLWCFGEMLIDFVPSVSGVSLAEAPAFHKAPGGAPANV
AVGISRLGGSSAFIGKVGDDEFGRMLADILKENKVDNSG RF
DQKARTALAFVTLRSDGEREFMFFRNPSADMLLHESELDVN
LIKQASIFHYGSISLIEEPCKSTHLAAMAIAKKSGSILSYDVNLR
LPLWPSEDAARDGIMSIWDQADVIKVSEDEITFLTRGDDPYD
DNWLNKLFHPNLKLLLVSEGPDGCRYYTQNFKGRVPGVKV
KPVDTTGAGDAFVGGILSVLASDTDLYKDEAKLREALLFANA
CGALTVTKKGAIPAMPTRDEVQKILK
84 A0260 Bet v LIVSFGEMLIDFVPTVSGVSLAEAPGFVKAPGGAPANVAIAVS
RMGGKASFVGKLGEDDFGRMLAGILDQNGVDCSGITFDQG
ARTALAFVTLRADGEREFMFYRNPSADMLLRPDELNLELIRS
AKAFHYGSISLIVEPCRSAHLKAMEVARDAGALLSYDPNLRL
PLWPSPEEAREQIMSIWDKADVIKVSDVELEFLTGSDKIDDA
SAFSLLHPNVKLLLVTLGEHGCRYYSKNFHGAVEAFHVTQV
DTTGAGDSFVGALLSKIVDDRSILEDEQRLRRVLTFANACGAI
I I I KKGAIPALPTEAEAVTLLQ
85 A0260 Que a LIVSFGEMLIDFVPTVSGVSLAEAPGFLKAPGGAPANVAIAVS
RLGGKAAFVGKLGDDEFGHMLAGILRENGWDEGINFDKGA
RTALAFVTLRADGEREFMFYRNPSADMLLTPDELNLDLIRSA
KVFHYGSISLIVEPCRTAHLKAMEVAKDAGLLLSYDPNLRLPL
WPSPEEARVQIKSIWDKADLIKVSDVELEFLTGSNKIDDESA
MSLWHPNLKLLLVTLGDLGCRYYTKNFHGEVEAFRVNTV'DT
TGAGDSFVGALLCKIVDDQSILEDEPRLRAVLKFANACGAITT
TKKGAIPALPTESDALSLIK
86 A0260 Cyn d MADRRDLWSFGEMLIDFVPTVAGVSLAEAPAFVKAPGGAP
ANVAIAVSRLGGAAAFVGKLGDDEFGRMLAGILRENGVEDG
GWFDSGARTALAFVTLRADGEREFMFYRNPSADMLLTADE
LNVELITRAAVFHYGSISLIAEPCRSAHLRAMEIAKEAGALLSY
DPNLREALWPSREEARTQILSIWDQADIVKVSEVELEFLTGID
SVEDDWMKLWRPTMKLLLVTLGDQGCKYYARDFRGAVPS
YKVQQVDTTGAGDAFVGALVRKIVQDPSSLQDQKKLEEAIKF
ANACGAITATKKGAIPSLPTETEVLQLIERA
87 A0262 Amb p VEYRCFVGGLAWATSDQSLGDAFSKYGEWDTKIINDRETG Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
RSRGFGFVTFRDEQSLRSAIEGMNGQSLDGRNITVNEAQ
88 A0262 Bet v VEYRCFVGGLAWATDDQALERAFSPYGDILESKIINDRETGR
SRGFGFVTFGNEKA RDAIEGMNGQNLDGRNITVNEA
89 A0262 Que a VEFRCFVGGLAWATDDQALERAFSPYGDILESKIINDRETGR
SRGFGFVTFSNEKSMRDAIEGMNGQNLDGRNITVNEA
90 A0262 Cyn d VEYRCFVGGLAWATSDDSLHAAFSPFGEVLESKIINDRETGR
SRGFGFVTFANEQAMRDAIEQMNGKQLDGRNITVNEA
91 A0262 Amb a KVFVGGLSWATDDKSLREAFSSYGEVTEAKVIRDRETGRSR
GFGFVTFADPDSCPNAIIALDQWELHGRTVSVRWADDRPR
92 A0265 Amb a SPDSGGGVSEDSKFSYGFASTTGKRSSMEDFYEARIDCVDG
EMIGLFGVFDGHGGARAAEYVKHHLFNTLLKHPKFITDTKAAI
SDAYSHTDTEFL SENNQNRDAGSTASTAILVGDRLLVANVG
DSRAVISRGGKAFAVSRDHKPDQSDERQRIEDAGGFVMWA
GTWRVGGVLAVSRAFGDKLLKQYWADPEIQEEKVDNSLEF
LILASDGLWDWTNDEAVAMVKPIESPEDAAKRLVQEASERG
SADNITWWRFLEN
93 A0265 Amb p SPDSGGGVSEDSKFSYGFASTTGKRSSMEDFYEARIDCVDG
EMIGLFGVFDGHGGARAAEYVKHHLFNTLLKHPKFITDTKAAI
SDAYSHTDTEFLKSENNQNRDAGSTASTAILVGDRLLVANVG
DSRAVISRGGKAFAVSRDHKPDQSDERQRIEDAGGFVMWA
GTWRVGGVLAVSRAFGDKLLKQYWADPEIQEEKVDNSLEF
LILASDGLWDWTNDEAVAMVKPIESPEDAAKRLVQEASERG
SADNITWWRFLEN
94 A0265 Bet v HAEDAPVSGGGLSQNGKFSYGYASSPGKRSSMEDFYETRI
DGVEGEIVGLFGVFDGHGGARAAEYVKQNLFSNLIRHPKFIS
DTKSAIADAYNHTDSEFLKSENSQNRDAGSTASTAILVGDRL
LVANVGDSRAVICRGGNAIAVSRDHKPDQTDERQRIEEAGG
FVMWAGTWRVGGVLAVSRAFGDRLLKQYWADPEIQEEKID
SSLEFLILASDGLWDWTNEEAVAMVKPIPEPEEAAKRLMQE
AYQRGSADNITIWVRFL
95 A0265 Cyn d MGFPGERSPTSGGGFSENGKFSFGYASSPGKRSSMEDFYE
TRVDGVDGETVGLFGVFDGHGGARAAEFVKQNLFTNLINHP
KLFSDTKSAIAETYTHTDSELLKGETSHNRDAGSTASTAILVG
DRLWANVGDSRAVICRGGDAIAVSRDHKPDQSDERQRIED
AGGFVMWAGTWRVGGVLAVSRAFGDKLLKQYWADPEIKE
EKVDSSLEFLILASDGLWDWTNEEAVAMVKPIIDSEQAAKKL
LQEASQRGSADNITCWVRFLD
96 A0265 Que a ARAAEFVKQNLFSNLIKHPKFFTDTKSAIAETFTRTDSELLKA
DTTHNRDAGSTASTAILVGDRLWANVGDSRAVICRGGDAIA
VSRDHKPDQTDERQRIEDAGGFVMWAGTWRVGGVLAVSR
AFGDKLLKQYWADPEIKEEWDSSLEFLILASDGLWDWTN
EEAVAMVKPIVDSQEAAKKLLVEATQRGSADNITCWVRFLD
97 A0270 Amb a VEAPAPALGEPMDIMTALQLVLRKSLAHGGLIRGLHEAAKVIE
KHAALLCVLAEDCNQPDYQKLVKALCADHNVSLVTVPSAKTL GEWAGLCKI
98 A0270 Amb p VEAPAPALGEPMDIMTALQLVLRKSLAHGGLIRGLHEAAKVIE
KHAALLCVLAEDCNQPDYQKLVKALCADHNVSLVTVPSAKTL GEWAGLCKIDSEGKARKWGCSCLWKDYGEESEGLHIVQE YVKSH Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID
NO:
99 A0270 Bet v VTETPVAPLGEAMDVMTALQHVLRKSLAHGGLVRGLHEAAK
VIEKHAAQLCVLAEDCNQADYLKLVKALCADHNVKLMTVPSA KTLGEWAGLCKIDSEGKARKWGCSCVWKDFGEQTEGYN VIQEYVKSH
100 A0270 Que a LGEAMDIMTALQWLRKSLAHGGLARGLHEGAKVIEKHAAQL
CVLAEDCNQPDYVKLVKGLCADHNVSLMTVPSAKTLGEWA GLCKIDSEGKARKWGCSCVWKDFGEVHEALHIVQEHVKS
H
101 A0270 Cyn d VEAPTPVLGEPMDLMTALQLVMKKSSAHDGLVKGLREAAKA
I E KH AAQ LCVLAE DC D Q P D YVKLVKALCAE H N VH LVTVPSAK TLGEWAGLCKIDSEGKARKWGCSCVWKDYGEESEGLNIV QEYVKSH
102 A0300 Amb a IDDAVSRILRVKFTMGLFENPLADYSMVNEVGSQAHREIARE
AVRKSLVLLKNGKTAVDPMLPLPKMSSKILVAGSHADNLGYQ
CG
103 A0300 Amb p IDDAVERILRVKFAAGLFEHPMTDRSLLNIVGSKPHRELAREA
VRKSLVLLKNGKDPNKPFLPLNRNVKKVLVAGKHADDLGYQ
CGGWTSTWEGTSGRITIGTTILDAVKETIGDNAEWYEENPS
AETLSEHDFSYAIVWGEAPYVESGGDNSELIIPFKGDELLKL
VAQQIPTLAILISGRPLVLEPSVLETLDALIAAWLPGTEGNGIT
DVIFGDHEFHGRLPVSWFKSVDQLPMDSNKSSYDPLFPLGY
GL
104 A0300 Bet v INDAVKRILRVKFSLGLFENPLADLSLADKLGCKEHRELAREA
VRKSLVLLKNGKSAPAGPLLPLAKKVPKILVAGSHADNLGFQ
CGGWTI EWQGRDG N N LTVGTTI LNG I KATVDTSTQWFN E N
PDPAFVKSSGFSYAIVWGEQPYAETNGDNLNLTLPEPGPST
INNVCGAVKCWIWSGRPLVIEPYLASVDALVAAWLPGSQG
EGVADVLFGDYGFTGKLARTWFKRVDQLPMNVGDAHYDPL
FPFGFGLT
105 A0300 Que a IDDAVKRILRVKFTMGLFENPLADDSQADLLGAQEHRELARE
AVRKSLVLLKNGKSAYAGPVLPLPRKSQRILVAGTHADNLGY
QCGGWTIEWQGLSGNNLTSGTTILKAIKDTVDSSTQWFKEN
PDPAFINSDDFAYAIVWGEQPYAETNGDNLNLTLPAPGPDTI
KNVCSHIKCVWWSGRPLVIEPYLESMDALVAAWLPGTEGQ
GVADVLFG D YG FTG KLARTWFKRVDQLPM N VG DAH YDPLF
PFEFGLT
106 A0300 Cyn d VNDAVSRILRVKFAMGLFEDPLPDPRLAKELGAQNHRAIARE
AVRKSLVLLKNGKKGKAVLPLAKKAKKILVAGSHAHNLGYQC GGWTGTWQGQSG N N FTG VGKTI LEAI KSTVDNSTTVEYS E E PDKETIEKAGDYEYAWAVGELPYAETAGDNQNLTIPAPGAK VIKEVCGLVKCWLIVSGRPLWEPYVDYMDALVAAWLPGTE GQGVTDVLFGDYGFTGKLPRTWFKSVDQLPMNYGDKRYDP LFPFGYGLT
107 A0300 Amb a LGCEEHRDLAREAVRKTLVLLKNGKSSMQPLLLLSKKTSKILV
AGSHADNLGYQCGGWTIEWQGLSGDVTSVEKTVDPNTQVI
YNENPNQDFIKSNNFDFAIVWGERPYAETFGDSSNLTILEPG
PSTIRNVCGSVKCVWLITGRPWMQPFVDTIDALVAAWLPG
TEGQGVTDVLFGDYGFTGKLARTWFKSVNQLPMNVGDPHY
DPLYRFGFGL Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
108 A0300 Amb a GTEGQGVTDLLFGDYGFTGKLARTWFKSVDQLPMNVGDKH
YDPLFPFGFGLT
109 A0304 Amb a WLFEVNPEGKVPLAKFDDNWVSDSDVIVGIIEDKYPHPSLVT
PPELASVGSNIFSKFIAFLKSKDANDGTEQALLDELNALNEHL KSKGPYVNGETITAVDLSLGPKLYHLQVALGHYKKWTVPESL THLHNY
110 A0304 Amb p WFLSISPEGKAPLAKLDDKWISDSDVITQTIEEKFPDPSLVTP
PEKASVGSKIFSTFIAFLKSKDANDGTEQALLNELSAFNDYIK ENG
111 A0304 Bet v WFLKINPDGKVPAIKFDEKWIPDSDVITQALEEKYPVPPLGTP
PEKASVGSKIFSTFIGFLKSKNPSDGTEQALLNELSSFNDYLK ENGPFINGKEVSAVDLSLGPKLHHLEIALGHYKNWSVPDSLP HVKSY
112 A0304 Que a WFLKLNPGGKVPVIKLNEKWISDSDVIAQALEEKYPDPPLGT
PPEKASVGSKIFSTFIGFLKSKDPSDGTEKALISELSSFNNHIK
DNGPYVNGKEVSAVDLSLADR
113 A0304 Cyn d WFLKINPEGKVPVFNSGDGKWIADSDVIVQIVEEKYPTPSLVT
PPEYASVGSKIFSTFVTFLKSKDANDGSEQALLNELQALEEH LKAHGPYINGQNVSAADLSLAPKLYHLHVALEHFKGWKIPES LTNVNAYS
114 A0311 Amb a MSFIGTQQKCKACGKTVYPVELLSADGIDYHKSCFKCSHCK
GTLKLSNYSSMEGVLYCKPHFEQLFKESGNFNKNFTSPA A
ADKLSPMLTKSPSKAAG FSGTQEKCATCGKTAYPLEKVTV
ENQAYHKSCFKCSHGGCSLSPSNYAALEGILYCKHHFSQLF
KEKGSYNHLIKSASIKR
1 15 A0311 Amb a TFSGTQDKCKACDKTVHFIDLLSADGVPYHKTCFKCSHCKG
TLSMSGYSSMEGVLYCKPHFEQLFKETGSFTKKFPSSGAEK REMTRTPSKLSAFFSGTQDKCSTCKKTV
116 A0311 Amb a MGFGGTIDKCNACDKTVHFVDLLTVDSVIYHKRCFKCSHCK
GTLVMSNYSSMDGVLYCMPHFEQLFKETGNFSKNFPSKPN
RDNSTPQLPNKFSFFFSGTQDKCRLCDKTVYFIDKMTMEGE
SYHKQCFRCIHGGCPLTHSSYAALNGVLYCRHHFAQLFLEK
GTLSHVLMAADRKKNTVPPDQETELADDDKLLKQEE
117 A0311 Amb a MSFTGTLDKCKACDKTVYFVDLLSVDGVTYHKACFRCSHCK
GTLAMSNYSSMDGVLYCKPHFEQLFKESGNFSKNFHTSKPD
RESAHSRAPNKLSSLFSGTLDKCRQCNKTVYPLEKVTMEGE
PYHKSCFRCAHGGCPLTHSSYAALDGILYCKHHFAQLFMEK
GNFSHVLEAANRKSNAKAEDIAPEQPP
118 A0311 Amb p MTFTGTQDKCKACDKTVHFIDLLTADSISYHKSCFKCSHCKG
TLSMCNYSSMDGVLYCKTHFEQLFKETGNFNKNFPTCAKAN
NEQSKVPNRLSSVFCGTQDKCAACKKTVYPLEKMTLEEEPY
H TCFKCAHGGCLLTTASYAALNGILYCQHHFWQLFKETGS
YDNLLKPAKKTEEPEATREEVPPEE
119 A0311 Bet MSFTGTQQKCKVCEKTVYPVEQLSADGVIYHKSCFKCSHCK
GTLKLSNYSSMEGVLYCKPHFEQLFKETGSFNKNFQSPAKS AEKLTPEMTRSPSKAASMFSGTQEKCATCGKTAYPLEKVTV ESQAYHKSCFKCSHGGCPITPSNYAALEGILYCKHHFAQLFK E KGS YN H L I KS AS I KRAAAAS VP EAAP A Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
120 A0311 Que a MTFTGTQDKCKACDKTVHFIDLLTADSISYHKSCFKCSHCKG
TLSMCNYSSMDGVLYCKTHFEQLFKETGNFNKNFPTCAKAN
NEQSKVPNRLSSVFCGTQDKCAACKKTVYPLEKMTLEEEPY
HKTCF CAHGGCLLTTASYAALNGILYCQHHFWQLFKETGS
YDNLLKPAKKTEEPEATREEVPPEE
121 A0311 Cyn d MSFTGTQDKCNACDKTVHFIDLLTADGVIYHKTCFKCSHCKG
ILSMCSYSSMDGVLYCKTHFEQLFKETGSFSKKFTPGGKSS
EKGELARAPSKLSSAFSGTQDKCAACQKTVYPLEKLTLEGE
PYHKSCFKCSHGGCILTTSSYAALNGVLFCKIHFSQLFMEKG
SYNHMKKKSESQEALPDWADEQPAASPQDEEV
122 A0316 Amb a MADGEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPR
HTGVMVGMGQKDAYVGDEAQSKRGILTLKYPIEHGIVNNWD
DMEKIWHHTFYNELRVAPEEHPVLLTEAPLNPKANREKMTQI
MFETFNSPAMYVAIQAVLSLYASGRTTGIVLDSGDGVSHTVP
IYEGYALPHAILRLDLAGRDLTDSLMKILTERGYSFTTSAEREI
VRDMKEKLAYIALDFEQELETSKTSSSVEKSFELPDGQVITIG
AERFRCPEVLFQPSMIGMESAGIHETTYNSIMKCDVDIRKDL
YGNIVLSGGTTMFPGIADRMSKEITALAPSSMKIKWAPPERK
YSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
123 A0316 Amb p MADGEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPR
HTGVMVGMGQKDAYVGDEAQSKRGILTLKYPIEHGIVNNWD
DMEKIWHHTFYNELRVAPEEHPVLLTEAPLNPKANREKMTQI
MFETFNSPAMYVAIQAVLSLYASGRTTGIVLDSGDGVSHTVP
lYEGYALPHAILRLDLAGRDLTDSLMKILTERGYSFTTSAEREI
VRDMKEKLAYIALDFEQELETSKTSSSVEKSFELPDGQVITIG
AERFRCPEVLFQPSMIGMESAGIHETTYNSIMKCDVDIRKDL
YGNIVLSGGTTMFPGIADRMSKEITALAPSSMKIKWAPPERK
YSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
124 A0316 Bet v MAEADDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPR
HTGVMVGMGQKDAYVGDEAQSKRGILTLKYPIEHGIVSNWD
DMEKIWHHTFYNELRVAPEEHPVLLTEAPLNPKANREKMTQI
M FETFNTPAM YVAI QAVLS LYASG RTTG I VLDSG DG VS HTVP I
YEGYALPHAILRLDLAGRDLTDALMKILTERGYSFTTTAEREI
VRDMKEKLAYIALDYEQELETAKTSSSVEKSYELPDGQVITIG
AERFRCPEVLFQPSMIGMEAAGIHETTYNSIMKCDVDIRKDL
YGNIVLSGGSTMFPGIADRMSKEITALAPNSMKIKWAPPERK
YSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
125 A0316 Que a MAETEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPR
HTGVMVGMGQKDAYVGDEAQSKRGILTLKYPIEHGIVSNWD
DMEKIWHHTFYNELRVAPEEHPVLLTEAPLNPKANREKMTQI
MFETFNTPAMYVAIQAVLSLYASGRTTGIVLDSGDGVSHTVPI
YEGYALPHAILRLDLAGRDLTDHLMKILTERGYSh I I I AEREI
VRDMKEKLAYIALDYEQEIETSKTSSSVEKSYELPDGQVITIG
AERFRCPEVLFQPSMIGMEAAGIHETTYNSIMKCDVDIRKDL
YGNIVLSGGSTMFPGIADRMSKEITALAPSSMKIKWAPPERK
YSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
126 A0316 Cyn d MADAEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPR
HTGVMVGMGQKDAYVGDEAQSKRGILTLKYPIEHGIVSNWD DMEKIWHHTFYNELRVAPEEHPVLLTEAPLNPKANREKMTQI Table 2 - SEQ ID OS:38-21 1
SE ID NO: SpeAmino Acid Sequence
Q cies
ID
NO:
MFETFNTPAMYVAIQAVLSLYASGRTTGIVLDSGDGVSHTVPI
YEGYALPHAILRLDLAGRDLTDYLMKILTERGYSh I I I AEREI
VRDMKEKLAYIALDYDQEMETAKTSSSVEKSYELPDGQVITI
GAERFRCPEVLFQPSFIGMEAAGIHETTYNSIMKCDVDIRKDL
YGNIVLSGGTTMFPGIADRMSKEITALAPSSMKIKWAPPERK
YSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
127 A0317 Amb a GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFP
EFLNLMARKMKDTDSEEELKEAFRVFDKD
128 A0317 Amb p GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFP
EFLNLMARKMKDTDSEEELKEAFRVFDKD
129 A0317 Bet v GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFP
EFLNLMARKMKDTDSEEELKEAFRVFDKD
130 A0317 Que a GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFP
EFLNLMARKMKDTDSEEELKEAFRVFDKD
131 A0317 Cyn d GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFP
EFLNLMARKMKDTDSEEELKEAFRVFDKD
132 A0325 Amb a AKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLV
LRLRGGAKKRKKKNYSTPKKIKHKKKKIKLAVLKYYKVDENG
KISRLRRECPSEQCGAGVFMAAMEDRHYCGKCSYTLVFN
133 A0325 Amb p VKSKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLH
LVLRLRGGMAKKRKKKVYTTPKKIKHKRKKTKLAVLKYYKVD GDGKIERLRRECPQPECGAGVFMAAMHNRQYCGKCHLTYV FD
134 A0325 Bet v AGKQLEDGRTLADYNIQKESTLHLVLRLRGGAKKRKKKQYTT
PKKIKHKRKKVKMAVLKYYKVDADGALKRLRRECPAPECGA
GTFMAWHENRQYCGKCHLTYLF
135 A0325 Bet v VKSKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLH
LVLRLRGGMAKKRKKKVYTTPKKIK
136 A0325 Bet v FAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG-
AKKRKKKTYTKPKKLKHKKKKVKLSVLQFYKVDDSGKVQRL
RKECPNTECGAGTFMANHFDRHYCGKCGLTYVY
137 A0325 Que a VKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLH
LVLRLRGGAKKRKKKTYTKPKKIKHKHKKVKLAILQFYKVDDS GKVQRLRKECPNSECGAGTFMANHFDRHYCGKCGLTYVY
138 A0325 Cyn d VKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLH
LVLRLRGGMAKKRKKKNYTTPKKIKHKRKPVKMAILKYYRVD SDGKIKRLRRECPSETCGPSVMMAQHADRQYCGKCGLTYK FDAE
139 A0326 Amb a DDEGVDDQAVKWDIVDTFRLQEQPPFDKKQFVAYIKKYIKSI
TPKLDEEKQDFFKKNIEAATKFLLSKLSDLQFFVGESMHDDS TIVFAYYKDGASDPTFLYFGVGLKEVKC
140 A0326 Amb p VWVQGAVDVDIGANPSAEGADEDEGVDDQAVKWDIVDTFR
LQEQPAFNKKQFVAYIKNYIKNVTPKLDEEKQEFFKKNIEAAT
KFLLGKLSDLQFFVGESMHDDS
141 A0326 Bet TGDELLSDSFPYKEIENGMLWEVEGKWWQGAVDVNIGANP
SAEGGDEDEGVDDQHVKWDIVDTFRLQEQPPFEKKQFVTF MKRYIKNLAAKLEPEQQALFKKHIEGAIKYLLPKISDLQFFVGE SMHDDGSLVFAYYKEGATDPTFIYLAYGLKEVKC Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
142 A0326 Que a TGDELLSDSFPYKEIENGMLWEVEGKWWQGAVDVDIGANP
SAEGGGEDEGVDDQAVKWDIVDTFRLQEQPSFDKKQFVTF MKRYIKLLTPKLEGEQQDIFKKNIEGATKFLLSKLSDLQFFVG ESMHDDGGLVFAYYKEGATDPTFLYFAHGLKEIKC
143 A0326 Cyn d TGDELLSDSFPYKEIENGILWEVEGKWWQGPVDVDIGANPS
AEGGDEEGVDDQAVKWDIVDTFRLQEQPAFDKKQFVTFIK RYIKNLTAKLEPEKAEEFKKGIEGATKFLLGKLKDLQFFVGES MHDDGTLVFAYYKDGATNPTFLYFGHGLKEVKC
144 A0327 Amb a LAEICKGTAFPDICTSTVGSDPASHGPLDPMAVLRMQVEQFL
KRTEAARAHVKEAALTASPKARTVLDLCNNLYLDVEDNLGAC RRAIGFHDAVTIRATMGMAAQDMQNCDEQFRQIGEKNPME QFDASLVEMSENCRSLSNMI
145 A0327 Amb p DVAFVCKGTPYAE LC I ATAG KQASH YPTVDALTVLG MQVDA
FSKRAAAARLHAVQISASGKVTPGALAALKTCDELYSNVEDN LGATRRAIGFKDPVTIRAMMSMSAQAMKSCDEEFKKNAAVN PLTRFDQSLLNISENCRALSNMI
146 A0327 Que a DVAFVCKGTPYAELCIATAGKQASHYPTVDALTVLGMQVDA
FSKRAAAARLHAVQISASGKVTPGALAALKTCDELYSNVEDN LGATRRAIGFKDPVTIRAMMS SAQAMKSCDEEFKKNAAVN PLTRFDQSLLNISENCRALSNMI
147 A0327 Cyn d ICKATSFPDVCIKTAGKHVKHYSSVDALSVLHMQVDAFSKRA
AAAR RVASKVRKASPAARNALTMCGKFYLDVEDNLGACRR AMRHRDGVTIRATMSMAAQDMQNCDEQFRQAGENKNPLE RFNKSLGQMAEVCRSLSNMI
148 A0334 Amb a DIAQLGAKGDGKSDSTPMLLKAWKNACDATGVQKIVIPPGN
YLTGGLELKGPCKSSIIIRLDGNLLGTGDLSQYKRNWIEIENV
DNLSINGHGTIDGQGALVWSKNECQKSYNCKILPNSLVLDFV
TNAQIRGITLANSKFFHLNIFSSKNWIDKVTVKAPGNSPNTD
GIHIGDSSNVTITGTTIGVGDDCISIGPGSKTIRIEGVKCGPGH
GISIGSLGRYKDEKDVEDVKVKGCTLVGTSNGLRIKSYEDSK
SSPKCTKFVYEDVTMDNVSYPIIIDQKYCPNNICVKSGASKVA
VTDVIFKNIHGTSNTPEAITLNCANNLPCEGLQLINVDIKYNGS
GNKTMAVCKNAVGKSSGLAKELACL
149 A0334 Amb a LVITGKGTLDGQGKEVWNNNKCAQKYDCKILPNTLVFDFCN
NGTVSGITLLNAKFFHLNVFQCKGMTLKDLTITAPGDSPNTD GIHIGDSSKVTISDTTIGTGDDCISIGPGSTGINITGVTCGPGH GISIGSLGRYADEKDVTDI
150 A0334 Amb p DIAKLGAICDGKTDSTKALQAAWASACGGAGQQTLLIPKGDY
MTGSLRFVGPCQGQVTIQLDGNLLGTSDLSQYKNDKWILIKD
VSNLVITGNGTLDGQGPGVWQKNSCDHKYDCKKLPTSMEL
EFVNSSIITGITFKDAKFFHLKVTDSTDITITGVTVTAPEDSPNT
DGIHIGRSSRVTISGATIGTGDDCVSIGAGTRDINVTGVTCGP
GHGISVGSLGRYKDETDVCNVNIRDCTLRNTANGIRIKTYDD
AACALTASKIDFENIRMEDVNNPIIIDMKYCPNKICPKLAGKKV
TVRDVTFKHISGTSLNPEWKFMCSDKMPCTGMQMIDVNVQ
YAGKRNKTMAICNNAWKAMGCLSALAC
151 A0334 Bet v KSTCQKDKDCDSLPMNIRFDFITNALVRDITSRDSKNFHVNV
LGCKNLTFQHFTVRAPGESVNTDGIHIGRSTGIYIIDSKIGTGD
DCISVGDGTEELHITGVTCGPGHGISVGSLG Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
152 A0334 Que a PPGNYLTGNLELVGPCTSSIIIRLDGNLLGTGDLNAYKKNWIEI
MHVKDFSINGHGTIDGQGPLVWKRNECAKNYNCKILPNSLVL
DYVTNVQIRGITLLNAKFFHMNIYESKNVLVERVNITAPGDSP
NTDGIHIGDSTNVTIAETNIGTGDDCISIGPGSKTIRVRGVRCG
PG HG I SVGS LG RYKDEKDVS DVTVRDCVLRSTTNG VRI KS Y
EDAESVITASRLTFENIRMEDVAKPIIIDQYYCPEKVCPGKKSS
TSHVDVKDWFRNITGTSSTPEAVSLLCSATQPCSGVELIDV
NVEYAGKNNKTMAVCTNAKGVAKGSIEALGCLV
153 A0334 Cyn d APSVPAGPLDITKLGARGDGKSDSTQAIMQAWKHACAATGT
QKIVIPPGNYLTGNLELVGPCTSSIIIRLDGNLLGTGDLNAYKK
NWIEIMHVKDFSINGHGTIDGQGPLVWKRNECAKNYNCKILP
NSLVLDYVTNVQIRGITLLNAKFFHMNIYESKNVLVERVNITAP
GDSPNTDGIHIGDSTNVTIAETNIGTGDDCISIGPGSKTIRVRG
VRCGPGHGISVGSLGRYKDEKDVSDVTVRDCVLKSTTNGVR
IKSYEDAESVITASRLTFENIRMEDVAKPIIIDQYYCPEKVCPG
KKSSTSHVAVKDWFRNITGTSSTPEAVSLLCSETQPCSGVE
LIDVKVEYAGKNNKTMAVCTNAKGVAKGSVEALGCL
154 A0334 Bet v ITGVTCGPGHGISVGSLGKYPNEKPVSGIFVKNCTISDTTNGV
R I KSWP AL YGG VAS NMHFEDIVMN N VQ N P VI I DQVYC PWNQ CSLKAPSKVKISDVSFKSIRGTSATPVWRIACSSGFPCQKVK LANIN
155 A0334 Amb p ASRLTFENIRMEDVAKPIIIDQYYCPEKVCPGKKSSTSHVAVK
DWFRNITGTSSTPEAVSLLCSETQPCS
156 A0336 Amb a MEQTFIMIKPDGVQRGLVGEIIGRFEKKGFTLKGLKLLTVDQA
FAEKHYADLSAKPFFNGLVEYIISGPWAMVWEGKNWTTGR KIIGATNPAESAPGTIRGDFAIDIGRNVIHGSDAVESARKEIGL WFPEGVANWSSSLHPWIYE
157 A0336 Amb p MEQTFIMIKPDGVQRGLVGEIIGRFEKKGFTLKGLKLLTVDQA
FAEKHYADLSSKPFFNGLVEYIISGPWAMVWEGKNWTTGR KIIGATNPAESAPGTIRGDFAIDIGRNVIHGSDAVESARKEIGL WFPEGVANWSSSLHPWIYE
158 A0336 Bet v MEQTFIMIKPDGVQRGLVGEIISRFEKRGFFMKGLKLITVDRS
FAEKHYADLSAKPFFSGLVDYIISGPWATVWEGKNWKTGR KIIGATNPSDSDPGTIRGDFAVEIGRNVIHGSDSVESARKEIAL WFPEAPLSWESSLHSWIYE
159 A0336 Que a MEQTFIMIKPDGVQRGLVGEIISRFEKKGFSLKGLKLITVDRP
FAEKHYADLSAKPFFSGLVDYIISGPWAMIWEGKNVWTGR KIIGATNPSESAPGTIRGDFAVEIGRNVIHGSDSVESARKE!AL WFPDGPVNWESSLHHWVYE
160 A0336 Cyn d MEQTFIMIKPDGVQRGLIGDIISRFEKKGFYLKGMKFMNVEKS
FAQQHYADLSDKPFFAGLVEYIISGPWAMVWEGKDWATG RRMIGATRPWEATPGTIRGDYAVEVGRNVIHGSDSVENGKK EIALWFPEGLAEWRSNLHPWVYES
161 A0349 Amb a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPP
DSPYSGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDRNKYETTARSWTQKYAMG
162 A0349 Amb p MASKRIIKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPS
DSPYSGGVFLVNIHFPPDYPFKPPKVAFRTKVFHPNINSNGSI Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDKS KYEATARSWTQ KYA
163 A0349 Amb p MASKRILKELKDLQKDPPTSCSAGPVAED FHWQATIMGPS
DSPYSGGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDKSKYEATARSWTQKYAMG
164 A0349 Amb p MASKRILKEL DLQKDPPTSCSAGPVAEDMFHWQATIMGPP
DSPYSGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDRNKYETTARSWTQKYAMG
165 A0349 Bet v EDMFHWQATIMGPADSPYAGGVFLVSIHFPPDYPFKPPKVA
FRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTD PNPDDPLVPEIAHMYKTDRAKYEATARSWTQKYAMG
166 A0349 Bet v RILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPY
AGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDIL KEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRG KYETTARSWTQKYAMG
167 A0349 Bet v MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPP
DSPYAGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDRNKYETTARSWTQKYAMG
168 A0349 Que a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPA
DSPYAGGVFLVSIHFPPDYPFKPPKVAFRTKVFHPN!NSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDRAKYEATARSWTQKYAMG
169 A0349 Que a RILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPTDSPY
AGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDIL KEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRA KYETTARSWTQKYAMG
170 A0349 Que a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPP
DSPYAGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDRSKYETTARSWTQKYAMG
171 A0349 Cyn d MASKRILKELKDLQKDPPTSCSAGPVGEDMFHWQATIMGPS
DSPFAGGVFLVNIHFPPDYPFKPPKVSFRTKVFHPNINSNGSI CLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYK TDRAKYESTARSWTQKYAMG
172 A0349 Cyn d MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPS
DSPYAGGVFLVTIHFPPDYPFKPPKVAFKTKVFHPNVNSNGS ICLDILKEQWSPALTVSKVLLSICSLLTDPNPDDPLVPEIAHMY KTDRAKYESTARSWTQKYAMG
173 A0356 Amb a MGKEKIHINIWIGHVDSGKSTTTGHLIYKCGGIDKRTIEKFEK
EAQEMGKGSFKYAWVLDKLKAERERGITIDIALWKFETSKYY VTIIDAPGHRDFIKNMITGTSQADCAVLIVAAGTGEFEAGISKN GQTREHALLAFTLGVKQ
174 A0356 Bet v MGKEKFHINIWIGHVDSGKSTTTGHLIYKLGGIDKRVIERFEK
EAAEMNKRSFKYAWVLDKLKAERERGITIDIALWKFETTKYY
CTVIDAPGHRDFIKNMITGTSQADCAILIIDSTTGGFEAGISKD Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
GQTREHALLAFTLGVRQMICCCNKMDATTPKYSKARYDEIVK
EVSSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGP
TLLEALDLISEPKRPSDKPLRLPLQDVYKIGGIGTVPVGRVET
GVI KPGMWTFG PTG LTTEVKSVEM H H EALQ EALPG DN VG F
NVKNVAVKDLKRGFVASNSKDDPAREAANFTAQVIIMNHPG
QIGNGYAPVLDCHTCHIAVKFAELVTKIDRRSGKEIEKEPKFL
KNGDAGIVKMIPTKPMWETFAEYPPLGRFAVRDMRQTVAV
GVIKGVEKKDPSG
175 A0356 Que a MGKEKVHISIWIGHVDSGKSTTTGHLIYKLGGIDKRVIERFEK
EAAEMNKRSFKYAWVLDKLKAERERGITIDIALWKFETTRYY
CTVIDAPGHRDFIKNMITGTSQADCAVLIIDSTTGGFEAGISKD
GQTREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIIK
EVSSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGP
TLLEALDQISEPKRPSDKPLRLPLQDVYKIGGIGTVPVGRVET
GIIKPGMWTFGPTGLTTEVKSVEMHHEALLEALPGDNVGFN
VKNVAVKDLKRGFVASNSKDDPAREAANFTSQVIIMNHPGQI
GNGYAPVLDCHTSHIAVKFAELVTKIDRRSGKEIEKEPKFLKN
GDAGMVKMIPTKPMWETFSEYPPLGRFAVRDMRQTVAVG
VIKSVEKKDPSG
176 A0356 Cyn d MGKEKSHINIWIGHVDSGKSTTTGHLIYKLGGIDKRVIERFEK
EAAEMNKRSFKYAWVLDKLKAERERGITIDIALWKFETTKYY
CTVIDAPGHRDFI NMITGTSQADCAVLIIDSTTGGFEAGISKD
GQTREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIVK
EVSSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGP
TLLEALDQINEPKRPSDKPLRLPLQDVYKIGGIGTVPVGRVET
G VLKPG MWTFGPSG LTTEVKSVEM H H EALQEALPG DNVG F
NVKNVAVKDIKRGYVASNSKDDPAKEAASFTSQVIIMNHPGQ
IGNGYAPVLDCHTSHIAVKFAELLTKIDRRSGKELEKEPKFLK
NGDAGMVKMIPTKPMWETFSQYPPLGRFAVRDMRQTVAV
GVI KSVEKKDPTG
177 A0356 Amb a QTREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIVKE
VSSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGPT
LLEALDQINEPKRPSDKPLRLPLQDVYKIGGIGTVPVGRVETG
VIKPGMWTFGPSGLTTEVKSVEMHHEALQEALPGDNVGFN
VKNVAVKDLKRGYVASNSKDDPAKGTASFTSQVIIMNHPGQI
GNGYAPVLDCHTSHIAVKFSELLTKIDRRSGKELEKEPKFLKN
GDAGMVKMLPTKPMWETFAEYPPLGRFAVRDMRQTVAVG
VIKSVDKKDPT
178 A0356 Amb p TREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIVKEV
SSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGPTL
LEALDQINEPKRPSDKPLRLPLQDVYKIGGIGTVPVGRVETG
VIKPGMWTFGPSGLTTEVKSVEMHHEALQEALPGDNVGFN
VKNVAVKDLKRGYVASNSKDDPAKGAASFTSQVIIMNHPGQI
GNGYAPVLDCHTSHIAVKFSELLTKIDRRSGKELEKEPKFLKN
GDAGMVKMLPTKPMWETFAEYPPLGRFAVRDMRQTVAVG
VIKSVDKKDPTG
179 A0357 Amb a DRERIFKRFDTNGDGKISSTELGDALKTLGSVTSEEIVRMMA
EIDTDGDGFISFEEFTDFAGANRGLIKDVAKIF
180 A0357 Bet v ELERIFKRFDLNGDGQISAAELGDCLKTLGSVTAEEIKRMMA Table 2 - SEQ ID OS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
EIDTDGDGFISFQEFLDFAKANSGLMKDVAKIF
181 A0357 Que a DMERIFKRFDTNGDGKISLAELTDALRTLGSTSADEVQRMMA
EIDTDGDGFIDFNEFISFCNANPGLMKDVAKVF
182 A0357 Cyn d DMERIFKRFDTNGDGKISLAELTDALRTLGSTSADEVQRMMA
EIDTDGDGFIDFDEFISFCNANPGLMKDVAKVF
183 A0357 Amb p TLGSTSADEVQRMMAEIDTDGDGFIDFNEFISFCNANPGLMK
DVAKVF
184 A0358 Amb a RKPVFVKVGDLKPGTNGHTLAVKVLSSTTVLDKKVRNTSSFS
GRPGGSNTRIAECLVGDETGTILFTARNDQVDLM AGSTVIIR NAKIDMFKGSMRLAVDKWGRIEVTDPATFTVKEDNNLSLVEY ELVNVTE
185 A0358 Amb p RKPVFVKVGDLKPGTNGHTLAVKVLSSNTVLDKKVRNTSSF
SGRPGGSNTRIAECLVGDETGTILFTARNDQVDLMKAGGTVI IRNAKIDMFKGSMRLAVDKWGRIEVTDPATFTVKEDNNLSLV EYELVNVTE
186 A0358 Bet v KPGLRKPVFTKVDQLRPGTTGHTLTVKWSTKMVLQKGRAD
GPQVRQMRIAECLVGDETGMIIFTARNDQVDLMKEGGTVILR NAKIDMFKGSMRLAVDKWGRVEVTEPADFTVKEDNNLSLIE YELVNWE
187 A0358 Que a QSNAKPALRKPEFKKVDQLKPGTTGHTLWKWSSNTVLAK
GRSVSQHLRHTRIAECLVGDETATIVFTARNEQVDLMQPGAT VILRNAKIDMFKGSMRLAVDKWGRVEVTEPAKFWKEDNNL SLVEYELVNWE
188 A0358 Cyn d RKPVFVKVDQLKPGTAGHTLIAKVMSSKTVLQKGRPGAAAG
PGARPTRIAECLIGDETGCILFTARNEQVDLMKPDSTVIIRNA KIDMFKGSMRLAVDKWGRIEVTEPAGFNVKEDNNLSLVEYE LVNVSE
189 A0362 Amb a FTN E E I SSM VLVKM KE I AEAYI G ETVKSAVITVPAYFN DAQRQ
ATKDAGVIAGLNVARIINEPTAAAIAYGLDKKGGEKNILVFDLG
GGTFDVSILTIDNGVFEVLATNGDTHLGGEDFDQRIMEYFIKLI
KKKHGKDISKDHRALGKLRREAERAKRALSSQHQVRVEIESL
FDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMEDAGLEK
RQIDEIVLVGGSTRIPKVQQLLKDYFDGKEPNKGVNPDEAVA
YGAAVQGGILSGEGGDETKDILLLDVAPLTLGIETVGGVMTKL
IPRNTVIPTKKSQVFTTYQDQQTWSIKVYEGERS
190 A0362 Amb p FS P E E I SAM I LTKM KETAE AF LG KKI KD A WTVPAYF N DAQ RQ
ATKDAGVIAGLNVARIINEPTAAAIAYGLDKKGGEKNILVFDLG
GGTFDVSILTIDNGVFEVLATNGDTHLGGEDFDQRIMEYFIKLI
KKKHGKDISKDHRALGKLRREAERAKRALSSQHQVRVEIESL
FDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMEDAGLEK
RQIDEIVLVGGSTRIPKVQQLLKDYFDGKEPNKGVNPDEAVA
YGAAVQGGILSGEGGDETKDILLLDVAPLTLGIETVGGVMTKL
I PRNTVI PTKKSQVh I I YQDQQTWSIKVYEGERSLTKDCRLL
GTFDLTGIPPAPRGTPQIEVTFEVDANGILNVKAEDKASGKSE
KITITNEKGRLSQEEIERMVREAEEFAEEDKKVK
191 A0362 Bet v FSPEEVSAMILTKMKETAEAFLGKKIKDAWTVPAYFNDAQR
Q ATKD AG 11 AG LN VAR 11 N E PTAAAI AYG LD KKGG EK I LVF D L GGGTFDVSILTIDNGVFEVLATNGDTHLGGEDFDHRIMEYFIK LIKKKHGKDISKDNRALGKLRREAERAKRALSSQHQVRVEIE Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
SLFDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMDDAGL EKNQIDEIVLVGGSTRIPKVQQLLKDYFDGKEPNKGVNPDEA VAYGAAVQGSILSGEGGEETKDILLLDVAPLTLGIETVGGVMT KLIPRNTVIPTKKSQVh I I YQDQQTTVSIQVYEGERSLTKDCR N LGKFDLSG I PPAPRGTPQI EVTFEVDANG I LNVKAEDKGTG KSEKITITNDKGRLSQEEIDRMVREAEEFAEEDKKVK
192 A0362 Que a FSPEEISA VLTKMKETAEAFLGKKIKDAWTVPAYFNDAQR
QATKDAGVIAGLNVARIINEPTAAAIAYGLDKKGGEKNILVFDL
GGGTFDVSILTIDNGVFEVLATNGDTHLGGEDFDHRVMDYFI
KLIKKKHSKDISKDNRALGKLRREAERAKRALSSQHQVRVEI
ESLFDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMDDAG
LEKRQIDEIVLVGGSTRIPKVQQLLKDFFDGKEPNKGVNPDE
AVAYGAAVQGGILSGEGGDETKDILLLDVAPLTLGIETVGGV
MTKLIPRNTVIPSKKSQVFTTYQDQQTTVTIQVFEGERSLTKD
CRLLGNFDLTGIAPAPRGTPQIEVTFEVDANGILNVKAEDKAS
GKSEKITITNDKGRLSQEEIDRMVQEAEEFAEEDKKVK
193 A0362 Cyn d FS P E E I SAM I LG KMKDTAEAYLG KKI N DAWTVPAYFN DAQR
QATKDAGVIAGLNVARIINEPTAAAIAYGLDKKGGEKNILVFDL
GGGTFDVSILTIDNGVFEVLATNGDTHLGGEDFDHRIMEYFIK
LIKKKYSKDISKDNRALGKLRREAERAKRALSNQHQVRVEIE
SLFDGTDFSEPLTRARFEELNNDLFRKTMGPVKKAMEDAGL
EKSQIHEIVLVGGSTRIPKVQQLLRDYFDGKEPNKGVNPDEA
VAYGAAVQGSILSGEGGEETKDILLLDVAPLTLGIETVGGVMT
KLIPRNTVIPTKKSQVFTTYQDQQTTVSIQVFEGERSMTKDC
RLLGKFDLSGIPPAPRGTPQIEVTFEVDANGILNVKAEDKGTG
KSEKITITNEKGRLSQEEIDRMVREAEEFAEEDKKVK
194 A0366 Amb a AAFQALAPLEAVLFDVDGTLCDSDPIHHIAFKEMLLEIGFNGG
VPIDEEFFIQNIAGKHNDDIAAVLFPDDIERGLKFCVDKEAYFR
KLVKEKVEPIKGLYKLTKWVEDNGLKRAAVTNAPRPNAELMI
STLGLTDFFHHVIIGDECERAKPAPDPYLKAIELLNVSKDHTFI
CEDSVSGIKAGVAAGMPWGLTTRNPEQMLMTANPTLLIKDY
EDPKLWAVLEEL
195 A0366 Amb p AAFQALAPLEAVLFDVDGTLCDSDPIHHIAFKEMLLEIGFNGG
VPIDEEFFIQNIAGKHNDDIAAVLFPDDIERGLKFCVDKEAYFR
KLVKEKVEPIKGLYKLTKWVEDNGLKRAAVTNAPRPNAELMI
STLGLTDFFHHVIIGDECERAKPAPDPYLKAIELLNVSKDHTFI
CEDSVSGIKAGVAAGMPWGLTTRNPEQLLMTANPTLLIKDY
EDPKLWAVLEEL
196 A0366 Bet v SSLAALAPLQAVLFDVDGTLCDSDPLHYYAFREMLQEIGFNG
GVPITEEFYIENIAGKHNDDVAVLLFPDDVQRGLKFTDDKEA
MFRRLAAEQLEPVNGLYKVTKWVEDRGLKRAAVTNAPRPN
AELMISRLGLSDFFEAVILGSECDHAKPHPEPYLKALEILNVS
KDHTFVFEDSVSGIKAGVAAGLPWGLTTRNPERFLMEAKPT
FLVKDYDDPKLWAALEELD
197 A0366 Que a TTLAELAPLQAVLFDIDGTICDSDPLHHYAFREMLQEIGFNGG
VPITEEFFVDHIAGKHNDDIAKFLFPDDIQRGLKFVDDKEAMF
RRLASEQLKPVNGLYKVKKWIEDRGIKRAAVTNAPKANAELM
ISSLGLSDFFEALIVGSECEHAKPHPDPYLKALEIINVSKDHTF
VFEDSVSGIKAGVAAGMPWGIATRNPEHLLMEAKPVFLIKD Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
YEDPKLWAALEELD
198 A0366 Cyn d TTVSSLASTVPVQAVLFDIDGTLCDSDPLHHVAFQEMLLAIGY
NNGVPIDDEFFIKNIAGRSDVEAAQNLFPDWELEKGLKFLED
KEEKYRSLAKERLVPVKGLEKWQWVKDHGYKRAAVTNAPR
INAELMISLLGLSDFFQAVIVGGECEKPKPAPYPYLKAIKELDV
SAEHTFIFEDSASGIRAGVAAGIPWGVATRNPEKSLLEAGAS
LLIKDYEDPKLWAALEEIDREEAKLKNQGSA
199 A0376 Amb a DIKVIMGRSDDLETWLTGVMTFMDTCIDGFVDEKLKADMHS
VLRNATELSSNALAITNSLGGILKKLDLDMFKKDSRRRLLSEQ
DEKGWPNA/VMKSPERKLLAAGNQPKPNAWAKDGSGQFKTI
QQAVDAVPKGQLGRYVIYVKTGLYDEIVMVPKDKPNIFMYGD
GPKQSRVTGKKSFADGITTMKTATFSIEAAGF!CKNMGFHNT
AGAEKHQAVALRVQGDFAAFYNCRFDAFQDTLYVHARRQF
FRNCVISGTIDFIFG
200 A0376 Bet v HNGSKSAGDSLSPQMKIVKDICSKTDYQEACQNSLSPAADK
GNTDPKEFIKSAIQATINEVAKSANFSDALVQNVSSIPRVKMA
AEDCKDLLSFATDELQASFSTVGDSDMHTINDRSADLKNWL
SAVISYQQTCLDGFDAPEYRTLMEQNLQDASLLTSNALAIVS
ELADILKAFGLEFNIKPSGRRRLLSQDGYPTWFSGSDRKLLA
LVDNGRIRPNAIVAKDGSGQFKTIAAALAAYPKNLRGRYVIYV
KAGIYNEYITVEKTQPNIFMFGDGPRKTIVTGRKSYRDGITTF
KTASFSTMGEGFIAKSMGFQNTAGPEGHQAVALRVQADRS
AFFNCRMDGYQDTLYVQTHRQFFRNCVISGTVDFIFGDAAA
VIQNSLIIVRKPMDNQQNTVTAQGRHDKRETTGLVIHNCRIVP
EQKLFAQRFTIPTYLGRPWKEYARTVIMETTLADFIQPVGYM
PWAGSFALSTCS YFE YAN RG PGARTVRRVRWKGARVI S RS
EALQFTAGPFLQGNLWLKATGFPFLLGLR
201 A0376 Cyn d LGGLSAILWAVWGWATVTRSGNKAGDNFTVPGEATLATS
GKSVKSLCAPTLYKESCEKTLTQASNGTENPKEVFQAVAKT
ALESVKAAYEQSQNIGRKLLDVDDSMTASAREDCKKLLDDSI
DDLRGMIDMAGGDIKVLLSRSDDLETWLTGVMTFLDTCSDG
FTDEKLKADMKLVLRNATELSSNALAITNSLGGIFKKLDLDML
KKDATSRRRLLSDQDEKGWP\AtyMKSPERKLLASGDANRPQ
PNAWAKDGSGQFKTVQDAVNAYPKGLQGRYVIYVKAGWD
EMV ITKEKVNIFMYGDGPKRSRVTGRKSFADGITTMKTATF
SIEASGFICKNMGFHNTAGAERHQAVALRVQGDLAAFFNCR
FDAFQDTLYVHARRQFFRNCVISGTIDFIFGNSAAVFQNCLIIT
RRPMDNQQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFP
DRFKIPSYLGRPWKEFSRLVIMESTIADFIKPEGYMPWDRDF
AL KTL Y YAE YN N KG PGAGTS KRVNWPG FRVI G KQ E AAQ FTA
GPFIDGATWLKFTGTPHILGFK
202 A0376 Amb a RYVIYVKAGIYEEIVLIPKDKTNIYMYGDGPKKSRVLGHKSNA
DGLTTQDTATFSVIGQGFICKNMGFSNTAGAIKHQAVALRLQ
SDFAAFYNCRMDGYQDTLYVQARRQFFRNCWSGTIDFIFG
NAAAVFQNCLIIVRRPMDNQQNTVTAHGRTDPKMKSGLVIQ
NCRIVPDQALFPDRFKIPSYLGRPWKEYSRTVIMESTIGDAIR
PEGWMAWNGDFALKTLYYAEYANRGPGANTRSRVRWPGF
HVINRNEAQQFTVGNF!SGNLWLRYTG
203 A0376 Amb a PMDNQQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFPDR Table 2 - SEQ ID NOS:38-211
SE ID NO: SpeAmino Acid Sequence
Q cies
ID NO:
FKIPSYLGRPWKEFSRLVIMESTIADFIKPEGYMPWNGDFGI TLFYAEYNNRGPGAGTSKRVKWPGFHVITKKDAEQFTAGPFI DGATWLKFTGTPHILGFK
204 A0376 Amb p KFKSIQEAVNA PKGHPGRYVIYVKTGVYDEIVMVPKDKVNI
FMYGDGPKQTRVTGSKSFHDGITTMKTATFSIEASGFICKNM
GFHNTAGAERHQAVALRVQGDLAAFFNCRFDAFQDTLYVHA
RRQFFRNCVISGTIDFIFGNSAAIFQNCLIITRRPMDNQQNSV
TAHGRTDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRP
205 A0376 Que a RDSSVTGRKSFADGVTTMKTATFSIEAAGFICKNMGFHNTA
GAERHQAVALRVQGDLAAFYNCRFDAFQDTLYVHARRQFF
RNCVISGTIDFIFGNSAAIFQNCLIITRRPMDNQQNSVTAHGR
TDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRPWKEFS
RLVIMESTIADFIKPEGYMPWNGDFGLKTLYYAEYANRGPGA
GTSKRVTWPGFRVIGRKEAEQFTAGPFVDGATWLKFTGTPN
YLGFKV
206 A0377 Amb a RIKTGFTHFKTEKYEKNPSLYEELVKEQKPKFMVFACSDSRV
CPSHILDFQPGEAFMVRNIANMVPPYDKTKYSCVGAAIEFAV LTLKVENILVIGHSCCGGIKGLMSIPDDGTTSSDFIEDWVKI
207 A0377 Amb p RIKTGFTHFKTEKYEKNPSLYEELVKEQKPKFMVFACSDSRV
CPSHILDFQPGEAFMVRNIANMVPPYDKTKYSCVGAAIEFAV LNLKVENILVIGHSCCGGIKGLMSIPDDGTTSSDFIEDWVKICS TAKS KVKE E F KE L D FTEQCS KC E VE AVN VS LG N L LTYP FVKS AVMNRTLNIKGGYYNFVNATFDIW
208 A0377 Bet v RIKTGFIHFKKEKYDKNPALYGELAKGQSPKFMVFACSDSRV
CPSHVLDFQPGEAFWRNVANLVPPFDQTKYSGTGAAVEYA
VLHLKVENIWIGHSACGGIKGLLSFPDEGPKSTDFIEDWVKI
GLPA SKVKAEFGNASFPELCGHCEKEAVNVSIGNLLTYPFV
REGLVNKTLALKGGYYDFVKGTFELW
209 A0377 Que a RIKTGFIHFKKEKFEKNPELYGELAKGQKPKFMVFACSDSRV
CPSHILNFQPGEAFMIRNIANMVPPFDKTKYSGGGAAIEYAVL
HLKVEHIWIGHSCCGGIKGLMSIPDDGTTASDFIEHWVQICS
PAKAKVKAEYSGLSFSEQCTNCEKEAVNVSLGNLLTYPFVR
DGLVKKTLALKGAHYDFVKGTFDLWD
210 A0377 Cyn d HLKSGFDKFKADVYDTKPELFEPLKAHQSPKYMVFSCADSR
VCPSVTLGLQPGEAFTVRNIANMVPSYDKTRYASVGSAIEYA
VCALKVEVIWIGHSRCGGIKALLSLKDGAPDTFHFVEDWVRI
GCPAKEKVLAEHAEAPFADQCTILE EAVNLSLENLKSYP-FV
KEGLEKGTIKLVGAHYDFVDGKFETWE
211 A0377 Amb a FIEDWVKICDPARSKIKSVLSCAEFADQCKNCEKEAVNVSLG
NLLTYPFVKEAWNKTLALKGAHYDFVHGAFE
Example 3 - Conservation analysis
This example includes a description of how to identify stretches of amino acid residues (conserved regions) that are conserved across a set of polypeptides found Tables 1 and 2, i.e. which are conserved across a set of polypeptides found in at least two of the pollen species Phi p, Cyn d, Amb a, Amb p, Que a and bet v.
Multiple sequence alignments were generated for each set of homologous sequences using the Phi p sequence of Table 1 as the reference sequence. For each Phi p reference sequence, the degree of conservation of each 15mer contained in this sequence across the other species was determined. For the purpose of this analysis, it was defined that peptides that have a homologous hit with 0, 1 or 2 mismatches are considered as being conserved. Any substitution of an amino acid sequence within the 15mer Phi p peptide is considered to constitute a mismatch. A conserved region was then defined as the region resulting from merging ail conserved 15mer peptides in a Phi p sequence.
A region was defined as conserved across "grass & weed & tree" ("GWT") if the conserved stretches of amino acid residues was detected in the Phi p polypeptide as well as in the homolog polypeptides of at least one weed species (Ambrosia artemisiifolia or Ambrosia psilostachya) and of at least one tree species (Quercus alba or Betula verrucosa). Table 3 shows GWT conserved stretches deriving from Phi p polypeptide sequences shown in Table 1. Table 4 shows GWT conserved stretches deriving from homolog polypeptides shown in Table 2.
Similarly, a region was defined as conserved across "grass & weed" ("GW") if the conserved stretches of amino acid residues was detected in the Phi p polypeptide as well as in the homolog polypeptides of at least one weed species (Ambrosia artemisiifolia or Ambrosia psilostachya) and none of the tree species. Table 5 shows "GW conserved stretches deriving from Phi p polypeptide sequences shown in Table 1. Table 6 shows "GW" conserved stretches deriving from homolog polypeptides shown in Table 2.
Similarly, a region was defined as conserved across "grass & tree" ("GT") if the conserved stretches of amino acid residues was detected in the Phi p polypeptide as well as in the homolog polypeptides of at least one tree species (Quercus alba or Betula verrucosa) and none of the weed species. Table 7 shows "GT" conserved stretches deriving from Phi p polypeptide sequences shown in Table 1. Table 8 shows "GT" conserved stretches deriving from homolog polypeptides shown in Table 2. The conservation analysis was repeated using Amb a as a reference in order to identify amino acid stretches conserved across polypeptides found in in weed pollen (Amb a) and tree pollen (Que a or Bet v), but not in grass pollen (neither Phi p nor Cyn d). Table 9 shows WT conserved stretches of polypeptides shown in Table 1. Table 3. GWT conserved amino acid sequences derived from polypeptides of Table 1
Figure imgf000088_0001
Table 3 - SEQ ID NOS: 212-293
SEQ ID ID NO Spe-cies Amino acid sequences
NO
MDGVLYCKTHFEQLFKETGSFSKNFT
252 A0311 Phi p FSGTQDKCAACQKTVYPLEKL
253 A0311 Phi p ECYHKSCFKCSHGGCILTTSSYAALNGILYCKIHFSQLFKEKGSYNHLIKTAQ
254 A0316 Phi p MADAEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPRHTGVMVGMG
QKDAYVGDEAQSKRGILTLKYPIEHGIVSNWDDMEKIWHHTFYNELRVAPEE HPVLLTEAPLNPKANREKMTQIMFETFNTPAMYVAIQAVLSLYASGRTTGIVL DSGDGVSHTVPIYEGYALPHAILRLDLAGRDLTDYLMKILTERGYSh I I I AER EIVRDVKEKLSYIALD
255 A0317 Phi p GCITTKELGTVMRSLGQNPTEAELQD INEVDADGNGTIDFPEFLNL ARKM
KDTDSEEELKEAFRVFDKD
256 A0325 Phl p VKAKIQD EGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGGMA
KKRKKKNYTTPKKIKH KRKPVK
257 A0326 Phi p WWNGSVDVDIGANPSAEGGCDDEGVDDQAVKWDIVDTFRLQEQPPFDK
KQFL
258 A0326 Phi p DCFKKNIEGATKFLLGKIKDLQFFVGESMHDDCA
259 A0326 Phl p AYYKEGAIDPTFLYF
260 A0327 Phi p EVAAVCKGTPYAELCSATAGKQASHYATVDALAVLG QVDAFSKRASAAS
261 A0327 Phi p YVNVEDNLGAARRAISFKDAVTIRAMMSMAAQDMKNCDEEFRKNAAVNPMT
RFDKSLLDISENCRALSNMI
262 A0334 Phi p KNYNCKILPNTLVLDF
263 A0334 Phi p DVTITAPGDSPNTDGIHIGDSSNI
264 A0334 Phl p TTIGTGDDCISIGGGSS
265 A0334 Phi p VTG VTC G PGQ G I SVG C LG RYKD EKD VS DVTVK
266 A0334 Phi p DVANPIIIDQNYCPEKVCT
267 A0334 Phi p VTVKDVIFRNITGTSSTPAAVSLLCSD
268 A0336 Phi p EQTFIMIKPDGVQRGLIGDII
269 A0336 Phl p KLVDYIISGPWAMVWEGKDW
270 A0349 Phi p KELKDLQRDPPTSCSAGPVAEDMFHWQATIMGPAESPYAGGVFLVTIHFPP
DYPFKPPKVAFKTKVFHPNINSNGSICLDILKEQWSPALTVSKVLLSICSLLTD PNPDDPLVPEIAHMYKSDRV YESTARSWTQKYAMG
271 A0349 Phi p MASKRILKELKDLQKDPPTSCSAGPVGEDMFHWQATI GPSDSPF
272 A0349 Phi p GGLFLVNIHFPPDYPFKPPKVSFRTKVFHPNINSNGSICLDILKEQWSPALTIS
KVLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYESTARSWTQKYAMG
273 A0349 Phi p DMFHWQATIMGPSDSPYSGGVFLVTIHFPPDYPFKPPKVAFKTKVFHPNVN
SNGSICLDILKEQWSPALTiSKVLLSICSLLTDPNPDDPLVPEIAHMYKADRAK YE
274 A0349 Phi p MASKRILKELKDLQKDPPTSCSAGPAGEDMFHWQATIMGPPDSPYAGGVFL
VNIHFPPDYPFKPPKVSFKTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSI CSLLTDPNPDDPLVPEIAHMYKTDRSKYETTARSWTQKYAMG
275 A0356 Phi p EKLHISIWIGHVDSG STTTGHLIYKLGGIDKRVIERFEKEA
276 A0356 Phi p EMNKRSFKYAWVLDKLKAERERGITIDIALWKFETTKYSCTVIDAPGHRDFIK
N M ITGTS Q ADC AVLI I D
277 A0356 Phi p TGGFEAGISKDGQTREHALLAFTLGVKQMICCCNKMDATTPKYSKSRFEEIV
KEVSSYLKKVGYNPDKVPFVPISGFEGDNMIERSTNLDWYKGPTLLEALDQV TEPKRPTDKPLRLPLQDVYKIGGIGTVPVGRVETGLIKPGMLVTFGPTGLTTE VKSVEMHHESLLEAGPGDNVGFNVKNVAVKDIKRGYVASN
278 A0356 Phi p ASFVAQVIIMNHPGQIGNGYAPVLDCHTSHIAVKFAEI
279 A0356 Phi p FPKFLKNGDAGFVKMVPTKPMWETFSQYPPLGRFAVRDMRQTVAVGVIKS
VEKKDPTG
280 A0357 Phl p D ERIFKRFDTNGDGKISLSEL
281 A0357 Phl p TLGSTSADEVQRM AEIDTDGDGFIDFNEFISFCNANPGLMKDVAKVF
282 A0358 Phl p PRIAECLVGDETGCIIFTARNDQVDLLKAG
283 A0358 Phi p ILRNAKIDMFKSSMRLAVDKWGRV
284 A0358 Phi p EENNLSQVEYELVNVAE
285 A0362 Phl p FSPEEISA ILGK KETAEAYLGKKINDAWTVPAYFNDAQRQATKDAGVIAG
LNVARIINEPTAAAIAYGLDKKGSEKNILVFDLGGGTFDVSILTIDNGVFEVLAT NGDTHLGGEDFDHRI DYFIKLIKKKYSKDIS DNRALGKLRREAERAKRALS Table 3 - SEQ ID NOS: 212-293
SEQ ID ID NO Spe-cies Amino acid sequences
NO
NQHQVRVEIESLFDGTDFSEPLTRARFEELNNDLFRKTMGPVKKAMDDAGL
EKSQIHEIVLVGGSTRIPKVQQLLRDYFDGKEPSKGVNPDEAVAFGAAVQGS
ILSGEGGDETKDILLLDVAPLTLGIETVGGVMTKLIPRNTVIPTKKSQVFTTYQ
DQQTTVSIQVFEGERSMTKDCRLLGKFDLNGIPAAPRGTPQIEVTFEVDANGI
LNVKAEDKGTGKSE ITITNEKGRLSQEEIDRMVKEAEEFAEEDKKVK
286 A0366 Phi p EAVLFDIDGTLCDSDPLHHVAF
287 A0366 Phi p GYKRAAVTNAPRINAELML
288 A0376 Phi p PNAWAKDGSGKFKTI
289 A0376 Phi p VTGRKSFKDGITTMKTATFSIEAAGFICKN GFHNTAGAENHQAVALRVQGD
I.AAFYNCRFDAFQDTLYVHARRQFFRNCVISGTIDFIFGNSAAIFQNCLIITRR
PMDNQQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRPW
KEYSRLVIMESTIADFIKPEGYMPWNGDFGIKTLYYAEYANRGPGAGTSKRV
NWPGFRVI
290 A0376 Phi p AEQ FTTG PF VD G ATWLKFTG M P
291 A0377 Phi p PKYMVFACADSRVCPS
292 A0377 Phi p PGEAFTVR IANMVP
293 A0377 Phi p LEKEAVNVSLENLLTYPFVK
Table 4. GWT conserved amino acid sequences derived from polypeptides of Table
2.
Figure imgf000090_0001
Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
327 A0209 Amb a GHKQGPNLNGLFGRQSGTTAGYSYS
328 A0209 Amb a GHKQGPNLNGLFGRQSGTTAGYSYS
329 A0209 Amb p GHKQGPNLNGLFGRQSGTTAGYSYS
330 A0209 Bet v GHKQGPNLNGLFGRQSGTTAGYSYS
331 A0209 Que a GHKQGPNLNGLFGRQSGTTAGYSYS
332 A0209 Cyn d GHKQGPNLNGLFGRQSGTTPGYSYS
333 A0209 Amb a ENTLYDYLLNPKKYIPGTKMVFTGLKKPQERADLISYLK
334 A0209 Amb a EKTLYEYLLNPKKYIPGTKMVFPGLKKPKDRADLIAYLK
335 A0209 Amb p EKTLYEYLLNPKKYIPGTKMVFPGLKKPKDRADLIAYLK
336 A0209 Bet v EKTLYDYLLNPKKYIPGTK VFPGLKKPQDRADLISYLK
337 A0209 Que a EKTLYDYLLNPKKYIPGTKMVFPGLKKPQERADLIAYLK
338 A0209 Cyn d ENTLYDYLLNPKKYIPGTKMVFPGLKKPQERADLIAYLK
339 A0209 Amb a TVGEKIFKTKCAQCHTVEKGAGHKQGPNLNGLFGRQSGTTAGYSYSAGNKNKA
VIWEENTLYDYLLNPKKYIPGTKMVFTGLKKPQERADLISYLKQ
340 A0209 Amb a KVGEKIFKTKCAQCHTWKGAGHKQGPNLNGLFGRQSGTTAGYSYSAANKNMA
VIWEEKTLYEYLLNPKKYIPGTKMVFPGLKKPKDRADLIAYLKE
341 A0209 Amb p KVGEKIFKTKCAQCHTWKGAGHKQGPNLNGLFGRQSGTTAGYSYSAANKNMA
VIWEEKTLYEYLLNPKKYIPGTKMVFPGLKKPKDRADLIAYLKE
342 A0209 Bet v KVGEKIFKTKCAQCHTVEKGAGHKQGPNLNGLFGRQSGTTAGYSYSSANKN A
VNWEEKTLYDYLLNPKKYIPGTKMVFPGLKKPQDRADLISYLKE
343 A0209 Que a SGEKIFKTKCAQCHTVEKGAGHKQGPNLNGLFGRQSGTTAGYSYSTANKNMAV
IWEEKTLYDYLLNPKKYIPGTKMVFPGLKKPQERADLIAYLKS
344 A0209 Cyn d KAGEKIFKTKCAQCHTVDKGAGHKQGPNLNGLFGRQSGTTPGYSYSAANKNRA
VIWEENTLYDYLLNPKKYIPGTKMVFPGLKKPQERADLIAYLKE
345 A0211 Amb a DFDFTTDENCQKSKIFFIAWSPDTSRVRMKMVYASSKD
346 A0211 Amb p DYDFVTEENCQKSRIFFIAWSPDTARVRNKMIYASSKDRFKRELDGIQVELQATD
PTEMDLDVFKSRA
347 A0211 Amb p DFDFTTDENCQKSKIFFIAWSPDTSRVRMKMVYASSK
348 A0211 Amb p DFDFTTDENCQKSKIFFIAWAPDTSKVREKMVYASSKDRFKRELDGIQVEVQAT
DPSEMSLDIVKARA
349 A0211 Amb p DFDFVTDENCQKSKIFFISWAPDIARVRSKMLYASSKDRFKRELDGIQVELQATD
PSEMSMDIVKARA
350 A0211 Bet v DFDFTTDENVQKSKIFFIAWAPDTSRVRSK LYASSKDRFRRELDGVQVELQAT
DPSEMSLDIIKGRA
351 A0211 Bet v DFDFITDENCQKSKIFFIAWSPDTSRVRSKMVYASSKDRFKRELDGIQVELQATD
PSEMSLDIVKGRA
352 A0211 Que a DFDFITDENCQKSKIFFISWSPDTSRVRSKMLYASSKDRFRRELDGVQVELQAT
DPSEMSLDIIKGRA
353 A0211 Cyn d DFDFVTDENCQKSKIFFISWSPDTSRVRSKMLYASSKDRFKRELDGIQVELQATD
PSEMSMDIVKARA
354 A0211 Amb a MANAASGMAVDDECKLKFQELK
355 A0211 Amb p MANAASGMAVHDECKLKFLELK
356 A0211 Amb p MANAASGMAVNDECKLKFSELK
357 A0211 Bet v MANSASGMAVHDECKLKFLELK
358 A0211 Bet v MANAASGMAVSDECKLKFLELK
359 A0211 Que a MANSASGMAVHDDCKLKFLELK
360 A0211 Cyn d MSNSASGMAVCDECKLKFQELK
361 A0211 Amb a DECRY AVFDFDFTTDENCQKSKIFFIAWSPDTSRVRMKMVYASSKD
362 A0211 Amb p AECRYAVFDYDFVTEENCQKSRIFFIAWSPDTARVRNKMIYASSKDRFKRELDGI
QVELQATDPTEMDLDVFKSRA
363 A0211 Amb p DECRYAVFDFDFTTDENCQKSKIFFIAWSPDTSRVRMKMVYASSK
364 A0211 Amb p NECRYAVFDFDFTTDENCQKSKIFFIAWAPDTSKVREKMVYASSKDRFKRELDGI
QVEVQATDPSEMSLDIVKARA
365 A0211 Amb p DECRYAVFDFDFVTDENCQKSKIFFISWAPDIARVRSKMLYASSKDRFKRELDGI
QVELQATDPSEMSMDIVKARA
366 A0211 Bet v NECRYAVFDFDFTTDENVQKSKIFFIAWAPDTSRVRSKMLYASSKDRFRRELDG
VQVELQATDPSEMSLDIIKGRA
367 A0211 Bet v DECRY AVFDFDFITDENCQKSKIFFIAWSPDTSRVRSKMVYASSKDRFKRELDGI
QVELQATDPSEMSLDIVKGRA
368 A0211 Que a NECRYAVFDFDFITDENCQKSKIFFISWSPDTSRVRSKMLYASSKDRFRRELDG
VQVELQATDPSEMSLDIIKGRA
369 A0211 Cyn d DECRYAVFDFDFVTDENCQKSKIFFISWSPDTSRVRSKMLYASSKDRFKRELDGI Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
QVELQATDPSEMSMDIVKARA
370 A0211 Amb a NAASGMAVDDECKLKFQELKAKRSYRFITFKI
371 A0211 Amb p NAASG AVHDECKLKFLELKAKRTFRYIIFKI
372 A0211 Amb p LKFQELKAKRSYRFITFKI
373 A0211 Amb p NAASGMAVNDECKLKFSELKS RNYRFIVFKI
374 A0211 Bet NSASGMAVHDECKLKFLELKAKRNHRFIVFKI
375 A0211 Bet v NAASGMAVSDECKLKFLELKTKRNYRFIIFKI
376 A0211 Que a NSASGMAVHDDCKLKFLELKAKRNYRFIVF I
377 A0211 Cyn d NSASGMAVCDECKLKFQELKAKRSFRFIVFKI
378 A0211 Amb a NSLPADECRYAVFDFDFTTDENCQKSKI
379 A0211 Amb p AALPDAECRYAVFDYDFVTEENCQKSRI
380 A0211 Amb p NSLPADECRYAVFDFDFTTDENCQKSKI
381 A0211 Amb p NSLPANECRYAVFDFDFTTDENCQKSKI
382 A0211 Amb p DECRY AVFDFDFVTDENCQKSKI
383 A0211 Bet ASMPPNECRYAVFDFDFTTDENVQKSKI
384 A0211 Bet ASIPADECRYAVFDFDFITDENCQKSKI
385 A0211 Que a ASMPANECRYAVFDFDFITDENCQKSKI
386 A0211 Cyn d ACLPADECRYAVFDFDFVTDENCQKSKI
387 A0215 Amb p AITVDEACRQYTKHPSYC
388 A0215 Que a AITVDEACRQYTKHPSYC
389 A0215 Cyn d AITVDEACRKYTKHPSYC
390 A0215 Amb p TTLPALAEQAVLLAAESGV
391 A0215 Que a TTLPALAVQAVTLAAESGG
392 A0215 Cyn d TTLPALAVQAVTLAAESGG
393 A0215 Amb p VKNLEKIPGGMPLGCLERCVGRFQAAVAAL
394 A0215 Que a VKNLEKMPGG PLGCLERCVGKFQAAVAEL
395 A0215 Cyn d VKNLEKMPGG PLGCLERCVGKFQAAVAEL
396 A0215 Amb p QSRVAIVEHRDVARVKAWVKTARVDGE
397 A0215 Que a LSRVAIVEHRDVARVKAVWKAARADGE
398 A0215 Cyn d LSRVAIVEHRDVARVKAWVKAARADGE
399 A0219 Amb a LLKVNQIGSVTESIEAVKMSKRAGWGVMASHRSGETEDTFIADLSVGLATGQIKT
GAPCRSERLAKYNQLLRIEEELGAAAVYAGSKFRAP
400 A0219 Amb p LLKVNQIGSVTESIEAVKMSKRAGWGVMASHRSGETEDTFIADLSVGLATGQIKT
GAPCRSERLAKYNQLLRIEEELGAAAVYAGSKFRAP
401 A0219 Bet v LLKVNQIGSVTESIEAVRMSKRAGWGVMASHRSGETEDTFIADLSVGLATGQIKT
GAPCRSERLAKYNQLLRIEEELGSAAVYAGSKYRAP
402 A0219 Que a LLKVNQIGSVTESIEAVKMSKHAGWGVMASHRSGETEDTFIADLSVGLATGQIKT
GAPCRSERLAKYNQLLRIEEELGPAAVYAGSKFRAP
403 A0219 Cyn d LLKVNQIGSVTESIEAVKMSKHAGWGVMTSHRSGETEDTFIADLAVGLATGQIKT
GAPCRSERLAKYNQLLRIEEELGAAAVYAGAKFRAP
404 A0233 Amb a KFPPGPNITTNYNGQWLTARATWYGQPNGAGPDDNGGACGIKGVNLPPYNG
TACGN!PIFKDGKGCGSCYEVRC
405 A0233 Amb p CITKVPPGPNITTKYDTKWLPAKATWYGKPTGAGPKDNGGACGIKDVNLAPYLG
MTACGNVPIFKDGKGCGSCYELKC
406 A0233 Que a CITKVPPGPNITTKYDTKWLPAKATWYGKPTGAGPKDNGGACGIKDVNLAPYLG
MTACGNVPIFKDGKGCGSCYELKC
407 A0233 Cyn d CITKVPPGPNITTKYDTKWLPAKATWYGKPTGAGPKDNGGACGIKDVNLAPYLG
MTACGNVPIFKDGKGCGSCYELKC
408 A0233 Amb a PECSGQPITVFITDMNYEPIAPYHFDFSGKAFGSLAKPGLNDKLRHCGIMDVEFR
RVRCKLP-GQKILFHVEKGCNPNYLAVLVKNVADDG
409 A0233 Amb p EPCSDKPITVFITDKNYEPIAPYHFDLSGKAFGLMALPGKDQALRSVGELELQFR
RVQCKYPAGTKITFHVEKGSNPNYLAVLVKFVANDGDIVQM
410 A0233 Que a APCADKPITIFITDKNYEPIAPYHFDLSGKAFGLMALPGQDQKLRSVGELELQFRR
VQCKYPAGTKITFHVEKGSNPNYLAVLVKFVANDGDIVQM
411 A0233 Cyn d EPCADKPITVFITDKNYEPIAPYHFDLSGHAFGLMALPGKDQALRSVGELELEFR
RVRCKYPPGTKITFHVEKGSN
412 A0233 Amb p IQDSQSAAWKPMTESWGAVWRWDGAPPLKGPLSLRLTSESGKKLIAKDVIPAT
WKADNVYPSNIQF
413 A0233 Que a IQDSQSAAWKPMTESWGAVWRWDGAPPLKGPLSLRLTSESGKKLIAKDVIPAT
WKADNVYPSNIQF
414 A0246 Amb a DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGGMQIFVKTLTG
KTITLEVESSDTIDNVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTL Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
HLVLRLRGG
415 A0246 Amb p NIQKESTLHLVLRLRGGTMIKVKTLTGKEIEIDIEPTDTIDRIKERVEEKEGIPPVQQ
RLIYAGKQLADDKTAKDYNIEGGSVLHLVLALRGG
416 A0246 Bet DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGGTMI VKTLTG
KEIEIDIEPTDTIDRIKERVEEKEGIPPVQQRLIYAGKQLGDDKTAKDYNIEGGSVL HLVLALRGG
417 A0246 Que a QQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGGTMi VKTLTGKEIEIDIEPT
DTIDRIKERVEEKEGIPPVQQRLIYAGKQLGDDKTA DYNIEGGSVLHLVLALRGG
418 A0246 Cyn d DKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGGTMIKVKTLTG
KEIEIDIEPTDTIDRIKERVEEKEGIPPVQQRLIYAGKQLADDKTAKDYNIEGGSVL HLVLALRGG
419 A0260 Amb a LWCFGE LIDFVPSVSGVSLAEAPAFHKAPGGAPANVAV
420 A0260 Amb p LWCFGEMLIDFVPSVSGVSLAEAPAFHKAPGGAPANVAV
421 A0260 Bet LIVSFGEMLIDFVPTVSGVSLAEAPGFVKAPGGAPANVAI
422 A0260 Que a LIVSFGEMLIDFVPTVSGVSLAEAPGFLKAPGGAPANVAI
423 A0260 Cyn d LWSFGEMLIDFVPTVAGVSLAEAPAFVKAPGGAPANVAI
424 A0260 Amb a AFIGKVGDDEFGRMLADIL
425 A0260 Amb p AFIGKVGDDEFGR LADIL
426 A0260 Bet SFVGKLGEDDFGRMLAGIL
427 A0260 Que a AFVGKLGDDEFGHMLAGIL
428 A0260 Cyn d AFVGKLGDDEFGR LAGIL
429 A0260 Amb a FDQKARTALAFVTLRSDGEREFMFFRNPSAD LLH
430 A0260 Amb p FDQKARTALAFVTLRSDGEREFMFFRNPSADMLLH
431 A0260 Bet FDQGARTALAFVTLRADGEREF FYRNPSADMLLR
432 A0260 Que a FDKGARTALAFVTLRADGEREFMFYRNPSADMLLT
433 A0260 Cyn d FDSGARTALAFVTLRADGEREFMFYRNPSADMLLT
434 A0260 Amb a FHYGSISLIEEPCKS
435 A0260 Amb p FHYGSISLIEEPCKS
436 A0260 Bet FHYGSISLIVEPCRS
437 A0260 Que a FHYGSISLIVEPCRT
438 A0260 Cyn d FHYGSISLIAEPCRS
439 A0260 Amb p VKPVDTTGAGDAFVGGIL
440 A0260 Bet v VTQVDTTGAG DSFVGALL
441 A0260 Que a VNTVDTTG AG DS F VG ALL
442 A0260 Cyn d VQQVDTTGAGDAFVGALV
443 A0262 Amb p VEYRC F VG G LAW ATS DQ
444 A0262 Bet v VEYRCFVGGLAWATDDQ
445 A0262 Que a VEFRCFVGGLAWATDDQ
446 A0262 Cyn d VEYRCFVGGLAWATSDD
447 A0262 Amb a TEAKVIRDRETGRSRGFGFVTFAD
448 A0262 Amb p VDT IINDRETGRSRGFGFVTFRD
449 A0262 Bet v LESKIINDRETGRSRGFGFVTFGN
450 A0262 Que a LESKIINDRETGRSRGFGFVTFSN
451 A0262 Cyn d LESKIINDRETGRSRGFGFVTFAN
452 A0265 Amb a MIGLFGVFDGHGGARAAEYVKHHLFN
453 A0265 Amb p MIGLFGVFDGHGGARAAEYVKHHLFN
454 A0265 Bet v IVGLFGVFDGHGGARAAEYVKQNLFS
455 A0265 Cyn d TVGLFGVFDGHGGARAAEFVKQNLFT
456 A0265 Amb a LLKHPKFITDTKAAI
457 A0265 Amb p LLKHPKFITDTKAAI
458 , A0265 Bet v LIRHPKFISDTKSAI
459 A0265 Que a LIKHPKFFTDTKSAI
460 A0265 Cyn d LINHPKLFSDT SAI
461 A0265 Amb a QNRDAGSTASTAILV
462 A0265 Amb p QNRDAGSTASTAILV
463 A0265 Bet v QNRDAGSTASTAILV
464 A0265 Que a HNRDAGSTASTAILV
465 A0265 Cyn d HNRDAGSTASTAILV
466 A0265 Amb a RLLVANVGDSRAVISRGGKAFAVSRDHKPDQSDERQRIEDAGGFVMWAGTWR
VGGVLAVSRAFGDKLLKQYWADPEIQEEKVDNSLEFLILASDGLWDWTNDEA VAMVKPIES Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
467 A0265 Amb p RLLVANVGDSRAVISRGGKAFAVSRDHKPDQSDERQRIEDAGGFVMWAGTWR
VGGVLAVSRAFGDKLLKQYWADPEIQEEKVDNSLEFLILASDGLWDWTNDEA VA VKPIES
468 A0265 Bet v RLLVANVGDSRAVICRGGNAIAVSRDHKPDQTDERQRIEEAGGFV WAGTWRV
GGVLAVSRAFGDRLLKQYWADPEIQEEKIDSSLEFLILASDGLWDWTNEEAVA
MVKPIPE
469 A0265 Que a RLWANVGDSRAVICRGGDAIAVSRDH PDQTDERQRIEDAGGFVMWAGTWR
VGGVLAVSRAFGDKLL QYWADPEI EEWDSSLEFLILASDGLWDWTNEEAV
AMVKPIVD
470 A0265 Cyn d RLWANVGDSRAVICRGGDAIAVSRDHKPDQSDERQRIEDAGGFVMWAGTWR
VGGVLAVSRAFGD LLKQYWADPEIKEEKVDSSLEFLILASDGLWDWTNEEAV
AMVKPIID
471 A0265 Amb a RGSADNITWWRFLE
472 A0265 Amb p RGSADNITWWRFLE
473 A0265 Bet RGSADNITIVWRFL
474 A0265 Que a RGSADNITCVWRFLD
475 A0265 Cyn d RGSADNITCVWRFLD
476 A0270 Amb a lEKHAALLCVLAEDCNQPDYQKLVKALCADHNVSLVTVPSAKTLGEWAGLCKI
477 A0270 Amb p lEKHAALLCVLAEDCNQPDYQKLVKALCADHNVSLVTVPSA TLGEWAGLCKIDS
EGKARKWGCSCLWKDYGEE
478 A0270 Bet v lEKHAAQLCVLAEDCNQADYLKLVKALCADHNVKLMTVPSAKTLGEWAGLCKID
SEGKARKWGCSCVWKDFGEQ
479 A0270 Que a lEKHAAQLCVLAEDCNQPDYVKLVKGLCADHNVSLMTVPSAKTLGEWAGLCKID
SEGKARKWGCSCVWKDFGEV
480 A0270 Cyn d lEKHAAQLCVLAEDCDQPDYVKLVKALCAEHNVHLVTVPSAKTLGEWAGLCKID
SEGKARKWGCSCVWKDYGEE
481 A0300 Amb a IDDAVSRILRV FTMGLFENPLAD
482 A0300 Amb p IDDAVERILRVKFAAGLFEHP TD
483 A0300 Bet v INDAVKRILRVKFSLGLFENPLAD
484 A0300 Que a IDDAVKRILRVKFTMGLFENPLAD
485 A0300 Cyn d VNDAVSRILRVKFAMGLFEDPLPD
486 A0300 Amb a QAHREIAREAVRKSLVLLKNGKTA
487 A0300 Amb a EEHRDLAREAVRKTLVLLKNGKSS
488 A0300 Amb p KPHRELAREAVRKSLVLLKNGKDP
489 A0300 Bet v KEHRELAREAVRKSLVLLKNGKSA
490 A0300 Que a QEHRELAREAVRKSLVLLKNGKSA
491 A0300 Cyn d QNHRAIAREAVRKSLVLLKNGKKG
492 A0300 Amb a TIDALVAAWLPGTEGQGVTDVLFGDYGFTGKLARTWFK
493 A0300 Amb a GTEGQGVTDLLFGDYGFTGKLARTWFK
494 A0300 Amb p TLDALIAAWLPGTEGNGITDVIFGDHEFHGRLPVSWFK
495 A0300 Bet SVDALVAAWLPGSQGEGVADVLFGDYGFTGKLARTWFK
496 A0300 Que a S DALVAAWLPGTEGQGVADVLFGDYGFTGKLARTWFK
497 A0300 Cyn d YMDALVAAWLPGTEGQGVTDVLFGDYGFTGKLPRTWFK
498 A0304 Amb a LVTPPELASVGSNIFSKFIAFLKSKDANDGTEQAL
499 A0304 Amb p LVTPPEKASVGSKIFSTFIAFLKSKDANDGTEQAL
500 A0304 Bet v LGTPPEKASVGSKIFSTFIGFLKSKNPSDGTEQAL
501 A0304 Que a LGTPPEKASVGS IFSTFIGFLKSKDPSDGTEKAL
502 A0304 Cyn d LVTPPEYASVGSKIFSTFVTFLKSKDANDGSEQAL
503 A0311 Amb a MSFIGTQQKCKACGKTVYPVELLSADGIDYHKSCFKCSHCKGTLKLSNYSSMEG
VLYCKPHFEQLFKESGNFNKNFT
504 A0311 Amb a TFSGTQDKCKACDKTVHFIDLLSADGVPYHKTCFKCSHCKGTLSMSGYSSMEG
VLYCKPHFEQLFKETGSFTKKFP
505 A0311 Amb a GFGGTIDKCNACDKTVHFVDLLTVDSVIYHKRCFKCSHCKGTLVMSNYSSMD
GVLYCMPHFEQLFKETGNFSKNFP
506 A0311 Amb a MSFTGTLDKCKACDKTVYFVDLLSVDGVTYHKACFRCSHCKGTLAMSNYSSMD
GVLYCKPHFEQLFKESGNFSKNFH
507 A0311 Amb p MTFTGTQDKCKACDKTVHFIDLLTADSISYHKSCFKCSHCKGTLSMCNYSSMDG
VLYCKTHFEQLFKETGNFNKNFP
508 A0311 Bet MSFTGTQQKCKVCEKTVYPVEQLSADGVIYHKSCFKCSHCKGTLKLSNYSSME
GVLYCKPHFEQLFKETGSFNKNFQ
509 A0311 Que a MTFTGTQDKCKACDKTVHFIDLLTADSISYHKSCFKCSHCKGTLSMCNYSSMDG
VLYCKTHFEQLFKETGNFNKNFP Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
510 A0311 Cyn d MSFTGTQDKCNACDKTVHFIDLLTADGVIYHKTCFKCSHCKGILSMCSYSSMDG
VLYCKTHFEQLFKETGSFSKKFT
511 A0311 Amb a FSGTQEKCATCGKTAYPLEKV
512 A031 Amb a FSGTQDKCSTCKKTV
513 A0311 Amb a FSGTQDKCRLCDKTVYFIDK
514 A031 Amb a FSGTLDKCRQCNKTVYPLEKV
515 A0311 Amb p FCGTQDKCAACKKTVYPLEK
516 A0311 Bet v FSGTQEKCATCGKTAYPLEKV
517 A0311 Que a FCGTQDKCAACKKTVYPLEKM
518 A0311 Cyn d FSGTQDKCAACQKTVYPLEKL
519 A0311 Amb a QAYHKSCFKCSHGGCSLSPSNYAALEGILYCKHHFSQLFKEKGSYNHLIKSAS
520 A0311 Amb a ESYHKQCFRCIHGGCPLTHSSYAALNGVLYCRHHFAQLFLEKGTLSHVLMAAD
521 A0311 Amb a EPYHKSCFRCAHGGCPLTHSSYAALDGILYCKHHFAQLFMEKGNFSHVLEAAN
522 A0311 Amb p EPYHKTCFKCAHGGCLLTTASYAALNGILYCQHHFWQLFKETGSYDNLLKPAK
523 A0311 Bet v QAYHKSCFKCSHGGCPITPSNYAALEGILYCKHHFAQLFKEKGSYNHLIKSAS
524 A0311 Que a EPYHKTCFKCAHGGCLLTTASYAALNGILYCQHHFWQLFKETGSYDNLLKPAK
525 A0311 Cyn d EPYHKSCFKCSHGGCILTTSSYAALNGVLFCKIHFSQLFMEKGSYNHMKKKSE
526 A0316 Amb a MADGEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPRHTGVMVGMGQK
DAYVGDEAQSKRGILTLKYPIEHGIVNNWDDMEKIWHHTFYNELRVAPEEHPVLL
TEAPLNPKANREKMTQIMFETFNSPAMYVAIQAVLSLYASGRTTGIVLDSGDGVS
HTVPIYEGYALPHAILRLDLAGRDLTDSLMKILTERGYSFTTSAEREIVRDMKEKL
AYIALD
527 A0316 Amb p MADGEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPS!VGRPRHTGVMVGMGQK
DAYVGDEAQSKRGILTLKYPIEHGIVNNWDDMEKIWHHTFYNELRVAPEEHPVLL
TEAPLNPKANREKMTQIMFETFNSPAMYVAIQAVLSLYASGRTTGIVLDSGDGVS
HTVPIYEGYALPHAILRLDLAGRDLTDSLMKILTERGYSFTTSAEREIVRDMKEKL
AYIALD
528 A0316 Bet v MAEADDIQPLVCDNGTGMVKAGFAGDDAPRAVFPS!VGRPRHTGVMVG GQK
DAYVGDEAQSKRGILTLKYPIEHGIVSNWDDMEKIWHHTFYNELRVAPEEHPVLL
TEAPLNPKANREKMTQIMFETFNTPAMYVAIQAVLSLYASGRTTGIVLDSGDGVS
HTVPIYEGYALPHAILRLDLAGRDLTDALMKILTERGYSFTTTAEREIVRDMKEKL
AYIALD
529 A0316 Que a MAETEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPSIVGRPRHTGVMVGMGQK
DAYVGDEAQSKRGILTLKYPIEHGIVSNWDDMEKIWHHTFYNELRVAPEEHPVLL TEAPLNPKANREKMTQIMFETFNTPAMYVAIQAVLSLYASGRTTGIVLDSGDGVS HTVPIYEGYALPHAILRLDLAGRDLTDHLMKILTERGYSh I I I AEREIVRDMKEKL AYIALD
530 A0316 Cyn d MADAEDIQPLVCDNGTGMVKAGFAGDDAPRAVFPS!VGRPRHTGVMVGMGQK
DAYVGDEAQSKRGILTLKYPIEHGIVSNWDDMEKIWHHTFYNELRVAPEEHPVLL
TEAPLNPKANREKMTQIMFETFNTPAMYVAIQAVLSLYASGRTTGIVLDSGDGVS
HTVPIYEGYALPHAILRLDLAGRDLTDYLMKILTERGYSFTTTAEREIVRDMKEKL
AYIALD
531 A0316 Amb a QELETSKTSSSVEKSFELPDGQVITIGAERFRCPEVLFQPSMIGMESAGIHETTY
NSIMKCDVDIRKDLYGNIVLSGGTTMFPGIADRMSKEITALAPSSMKIKWAPPER
KYSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
532 A0316 Amb p QELETSKTSSSVEKSFELPDGQVITIGAERFRCPEVLFQPSMIGMESAGIHETTY
NSIMKCDVDIRKDLYGNIVLSGGTTMFPGIADRMSKEITALAPSSMKIKWAPPER
KYSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
533 A0316 Bet v QELETAKTSSSVEKSYELPDGQVITIGAERFRCPEVLFQPSMIGMEAAGIHETTY
NSIMKCDVDIRKDLYGNIVLSGGSTMFPGIADRMSKEITALAPNSMKIKWAPPER
KYSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
534 A0316 Que a QEIETSKTSSSVEKSYELPDGQVITIGAERFRCPEVLFQPSMIGMEAAGIHETTYN
SIMKCDVDIRKDLYGNIVLSGGSTMFPGIADRMSKEITALAPSSMKIKWAPPERK YSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
535 A0316 Cyn d QEMETAKTSSSVEKSYELPDGQVITIGAERFRCPEVLFQPSFIGMEAAGIHETTY
NSIMKCDVDIRKDLYGNIVLSGGTTMFPGIADRMSKEITALAPSSMKIKWAPPER KYSVWIGGSILASLSTFQQMWIAKAEYDESGPSIVHRKCF
536 A0317 Amb a GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFPEFLNLMARKMKD
TDSEEELKEAFRVFDKD
537 A0317 Amb p GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFPEFLNLMARKMKD
TDSEEELKEAFRVFDKD
538 A0317 Bet v GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFPEFLNLMARKMKD Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
TDSEEELKEAFRVFDKD
539 A0317 Que a GCITTKELGTVMRSLGQNPTEAELQDMINEVDADGNGTIDFPEFLNLMARKMKD
TDSEEELKEAFRVFDKD
540 A0317 Cyn d GCITTKELG MRSLGQNPTEAELQDMINEVDADGNGTIDFPEFLNLMARKMKD
TDSEEELKEAFRVFDKD
541 A0325 Amb a AKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG- AKKRKKKNYSTPKKIKHKKKKIK
542 A0325 Amb p VKSKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG AKK
RKKKVYTTPKKIKHKRKKTK
543 A0325 Bet v AGKQLEDGRTLADYNIQKESTLHLVLRLRGG-AKKRKKKQYTTPKKIKHKRKKVK
544 A0325 Bet v VKSKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG AKK
RKKKVYTTPKKIK
545 A0325 Bet v FAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG- AKKRKKKTYTKPKKLKHKKKKVK
546 A0325 Que a VKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLADYNIQKESTLHLVLRLRGG- AKKRKKKTYTKPKKIKHKHKKVK
547 A0325 Cyn d VKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGGMAKK
RKKKNYTTPKKIKHKRKPVK
548 A0326 Amb a DDEGVDDQAVKWDIVDTFRLQEQPPFDKKQFV
549 A0326 Amb p WWQGAVDVDIGANPSAEGADEDEGVDDQAVKWDIVDTFRLQEQPAFNKKQF
V
550 A0326 Bet WWQGAVDVNIGANPSAEGGDEDEGVDDQHVKWDIVDTFRLQEQPPFEKKQF
V
551 A0326 Que a WWQGAVDVDIGANPSAEGGGEDEGVDDQAVKWDIVDTFRLQEQPSFDKKQF
V
552 A0326 Cyn d WVQGPVDVDIGANPSAEGG- DEEGVDDQAVKWDIVDTFRLQEQPAFDKKQFV
553 A0326 Amb a DFFKKNIEAATKFLLSKLSDLQFFVGESMHDDST
554 A0326 Amb p EFFKKNIEAATKFLLGKLSDLQFFVGESMHDDS-
555 A0326 Bet ALFKKHIEGAIKYLLPKISDLQFFVGESMHDDGS
556 A0326 Que a DIFKKNIEGATKFLLSKLSDLQFFVGESMHDDGG
557 A0326 Cyn d EEFKKGIEGATKFLLGKLKDLQFFVGESMHDDGT
558 A0326 Amb a AYYKDGASDPTFLYF
559 A0326 Bet v AYYKEGATDPTFIYL
560 A0326 Que a AYYKEG ATD PTF LYF
561 A0326 Cyn d AYYKDGATNPTFLYF
562 A0327 Amb a LAEICKGTAFPDICTSTVGSDPASHGPLDPMAVLR QVEQFLKRTEAAR
563 A0327 Amb p D VAF VC KGTP YAELC I ATAG KQAS H YPTVD ALTVLG MQ VDAFS KR AAAAR
564 A0327 Que a DVAF VC KGTPYAE LC I ATAG KQASH YPTVD ALTVLG MQ VDAFS KRAAAAR
565 A0327 Cyn d I C KATS FPDVC I KTAG KH VKH YSS VD ALS VLH M Q VDAFS KRAAAAR
566 A0327 Amb a YLDVEDNLGACRRAIGFHDAVTIRAT GMAAQDMQNCDEQFRQIGEKNPMEQF
DASLVEMSENCRSLSNMI
567 A0327 Amb p YSNVEDNLGATRRAIGFKDPVTIRA MSMSAQAMKSCDEEFKKNAAVNPLTRF
DQSLLNISENCRALSNMI
568 A0327 Que a YSNVEDNLGATRRAIGFKDPVTIRAMMSMSAQAMKSCDEEFKKNAAVNPLTRF
DQSLLNISENCRALSNMI
569 A0327 Cyn d YLDVEDNLGACRRAMRHRDGVTIRATMSMAAQDMQNCDEQFRQAGENKNPLE
RFNKSLGQMAEVCRSLSNMI
570 A0334 Amb a KSYNCKILPNSLVLDF
571 A0334 Amb a QKYDCKILPNTLVFDF
572 A0334 Amb p HKYDCKKLPTSMELEF
573 A0334 Bet v KDKDCDSLPMNIRFDF
574 A0334 Que a KNYNCKILPNSLVLDY
575 A0334 Cyn d KNYNCKILPNSLVLDY
576 A0334 Amb a KVTVKAPGNSPNTDGIHIGDSSNV
577 A0334 Amb a DLTITAPGDSPNTDGIHIGDSSKV
578 A0334 Amb p GVTVTAPEDSPNTDGIHIGRSSRV
579 A0334 Bet v HFTVRAPGESVNTDGIHIGRSTGI
580 A0334 Que a RVNITAPGDSPNTDGIHIGDSTNV
581 A0334 JDyn d RVNITAPGDSPNTDGIHIGDSTNV
582 A0334 Amb a TTIGVGDDCISIGPGSK
583 A0334 Amb a TTIGTGDDCISIGPGST Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
584 A0334 Amb p ATIGTGDDCVSIGAGTR
585 A0334 Bet SKIGTGDDCISVGDGTE
586 A0334 Que a TNIGTGDDCISIGPGSK
587 A0334 Cyn d TNIGTGDDCISIGPGSK
588 A0334 Amb a IEGVKCGPGHGISIGSLGRYKDEKDVEDVKVK
589 A0334 Amb a ITGVTCGPGHGISIGSLGRYADEKDVTDI
590 A0334 Amb p VTGVTCG PG HG I SVG S LG RYKD ETD VC N V I R
591 A0334 Bet ITG VTCG PG H GISVGSLGKYPNE KP VSG I F VK
592 A0334 Bet ITGVTCGPGHGISVGSLG
593 A0334 Que a VRGVRCGPGHGISVGSLGRYKDEKDVSDVTVR
594 A0334 Cyn d VRGVRCGPGHGISVGSLGRYKDEKDVSDVTVR
595 A0334 Amb a NVSYPIIIDQKYCPNNICV
596 A0334 Amb p DVNNPIIIDMKYCPNKICP
597 A0334 Amb p DVAKPIIIDQYYCPEKVCP
598 A0334 Bet v NVQNPVIIDQVYCPWNQCS
599 A0334 Que a DVAKPIIIDQYYCPEKVCP
600 A0334 Cyn d DVAKPIIIDQYYCPEKVCP
601 A0334 Amb a VAVTDVIFKNIHGTSNTPEAITLNCAN
602 A0334 Amb p VTVRDVTFKHISGTSLNPEWKF CSD
603 A0334 Amb p VAVKDWFRNITGTSSTPEAVSLLCSE
604 A0334 Bet VKISDVSFKSIRGTSATPVWRIACSS
605 A0334 Que a VDVKDWFRNITGTSSTPEAVSLLCSA
606 A0334 Cyn d VAVKDWFRNITGTSSTPEAVSLLCSE
607 A0336 Amb a MEQTFI IKPDGVQRGLVGEII
608 A0336 Amb p MEQTFIMIKPDGVQRGLVGEII
609 A0336 Bet v MEQTFIMIKPDGVQRGLVGEII
610 A0336 Que a MEQTFIMIKPDGVQRGLVGEII
611 A0336 Cyn d MEQTFIMIKPDGVQRGLIGDII
612 A0336 Amb a GLVEYIISGPWAMVWEGKNW
613 A0336 Amb p GLVEYIISGPWAMVWEGKNW
614 A0336 Bet v GLVDYIISGPWATVWEGKNW
615 A0336 Que a GLVDYIISGPWAMIWEGKNW
616 A0336 Cyn d GLVEYIISGPWAMVWEGKDW
617 A0349 Amb a KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYSGGVFLVTIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPEIAHMYKTDRNKYETTARSWTQKYAMG
618 A0349 Amb p KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPSDSPYSGGVFLVNIHFPPDY
PFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDD
PLVPEIAHMYKTDKSKYEATARSWTQKYA
619 A0349 Amb p KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPSDSPYSGGVFLVSIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPEIAHMYKTDKSKYEATARSWTQKYAMG
620 A0349 Amb p KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYSGGVFLVTIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPEIAHMYKTDRNKYETTARSWTQKYAMG
621 A0349 Bet v EDMFHWQATIMGPADSPYAGGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSN
GSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYEATA
RSWTQKYAMG
622 A0349 Bet v KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYAGGVFLVTIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPE I AH M YKTD RG KYETTARSWTQKYAMG
623 A0349 Bet v KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYAGGVFLVTIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPEIAHMYKTDRNKYETTARSWTQKYAMG
624 A0349 Que a KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPADSPYAGGVFLVSIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPEIAHMYKTD R AKYEATARSWTQ KYAMG
625 A0349 Que a KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPTDSPYAGGVFLVTIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP LVPE I AH M YKTD R AKYETTARS WTQKYAMG
626 A0349 Que a KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYAGGVFLVTIHFPPDYP
FKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDP Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
LVPEIAHMYKTDRSKYETTARSWTQKYAMG
627 A0349 Cyn d KELKDLQKDPPTSCSAGPVGEDMFHWQATIMGPSDSPFAGGVFLVNIHFPPDY
PFKPPKVSFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLTDPNPDD
PLVPEIAHMYKTDRAKYESTARSWTQKYAMG
628 A0349 Cyn d KELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPSDSPYAGGVFLVTIHFPPDYP
FKPPKVAFKTKVFHPNVNSNGSICLDILKEQWSPALTVSKVLLSICSLLTDPNPDD PLVPEIAHMYKTDRAKYESTARSWTQKYAMG
629 A0349 Amb a MASKRILKELKDLQ DPPTSCSAGPVAEDMFHWQATI GPPDSPY
630 A0349 Amb p MASKRIIKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPSDSPY
631 A0349 Amb p MASKRILKELKDLQ DPPTSCSAGPVAEDMFHWQATIMGPSDSPY
632 A0349 Amb p MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPY
633 A0349 Bet v ED FHWQATIMGPADSPY
634 A0349 Bet v RILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPY
635 A0349 Bet v MASKRIL ELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPY
636 A0349 Que a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPADSPY
637 A0349 Que a RILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPTDSPY
638 A0349 Que a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATI GPPDSPY
639 A0349 Cyn d MASKRILKELKDLQKDPPTSCSAGPVGEDMFHWQATIMGPSDSPF
640 A0349 Cyn d MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATI GPSDSPY
641 A0349 Amb a GGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAH YKTDRNKYETTARSWTQKYAMG
642 A0349 Amb p GGVFLVNIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKV
LLSICSLLTDPNPDDPLVPEIAHMYKTDKSKYEATARSWTQKYA
643 A0349 Amb p GGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDKSKYEATARSWTQKYA G
644 A0349 Amb p GGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDIL EQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDRNKYETTARSWTQKYAMG
645 A0349 Bet v GGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDRAKYEATARSWTQKYAMG
646 A0349 Bet v GGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDRGKYETTARSWTQKYAMG
647 A0349 Bet v GGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDRNKYETTARSWTQKYAMG
648 A0349 Que a GGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDRAKYEATARSWTQKYAMG
649 A0349 Que a GGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LS I CSLLTD P N P D D P LVP E I AH YKTD R AKYETTARS WTQ KYAM G
650 A0349 Que a GGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVL
LSICSLLTDPNPDDPLVPEIAHMYKTDRSKYETTARSWTQKYAMG
651 A0349 Cyn d GGVFLVNIHFPPDYPFKPPKVSFRTKVFHPNINSNGSICLDILKEQWSPALTISKV
LLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYESTARSWTQKYAMG
652 A0349 Cyn d GGVFLVTIHFPPDYPFKPPKVAFKTKVFHPNVNSNGSICLDILKEQWSPALTVSK
VLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYESTARSWTQKYA G
653 A0349 Amb a DMFHWQATIMGPPDSPYSGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPN!NSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRNKYE
654 A0349 Amb p DMFHWQATIMGPSDSPYSGGVFLVNIHFPPDYPFKPPKVAFRTKVFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAH YKTDKS YE
655 A0349 Amb p DMFHWQATI GPSDSPYSGGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDKSKYE
656 A0349 Amb p DMFHWQATIMGPPDSPYSGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPN!NSNG
S I C LD I LKEQ WS PALTI S VLLS ICSLLTDPNPDDP LVP EIAHMYKTDRNKYE
657 A0349 Bet DMFHWQATIMGPADSPYAGGVFLVSIHFPPDYPFKPPKVAFRT VFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYE
658 A0349 Bet v DMFHWQATI GPPDSPYAGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRGKYE
659 A0349 Bet v D FHWQATI GPPDSPYAGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRNKYE
660 A0349 Que a DMFHWQATIMGPADSPYAGGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSNG
SICLDILKEQWSPALTIS VLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYE
661 A0349 Que a DMFHWQATI GPTDSPYAGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYE
662 A0349 Que a DMFHWQATIMGPPDSPYAGGVFLVTIHFPPDYPFKPPKVAFRTKVFHPNINSNG Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRSKYE
663 A0349 Cyn d DMFHWQATIMGPSDSPFAGGVFLVNIHFPPDYPFKPPKVSFRTKVFHPNINSNG
SICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPE!AHMY TDRAKYE
664 A0349 Cyn d D FHWQATI GPSDSPYAGGVFLVTIHFPPDYPFKPPKVAFKTKVFHPNVNSNG
SICLDILKEQWSPALTVSKVLLSICSLLTDPNPDDPLVPEIAHMYKTDRAKYE
665 A0349 Amb a ASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYSGGVFLVTI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAH YKTDRNKYETTARSWTQKYAMG
666 A0349 Amb p MASKRIIKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPSDSPYSGGVFLVNI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMYKTDKSKYEATARSWTQKYA
667 A0349 Amb p ASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPSDSPYSGGVFLVSI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMYKTDKSKYEATARSWTQKYAMG
668 A0349 Amb p MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYSGGVFLVTI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMYKTDRNKYETTARSWTQKYAMG
669 A0349 Bet v ED FHWQATIMGPADSPYAGGVFLVSIHFPPDYPFKPPKVAFRTKVFHPNINSN
GSICLDILKEQWSPALTISKVLLSICSLLTDPNPDDPLVPEIAH YKTDRAKYEATA
RSWTQKYAMG
670 A0349 Bet RILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYAGGVFLVTIHFPP
DYPFKPPKVAFRTKVFHPNINSNGS!CLDILKEQWSPALTISKVLLSICSLLTDPNP DDPLVPEIAHMYKTDRGKYETTARSWTQKYAMG
671 A0349 Bet v MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYAGGVFLVTI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMY TDRNKYETTARSWTQKYAMG
672 A0349 Que a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPADSPYAGGVFLVSI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMYKTDRA YEATARSWTQKYAMG
673 A0349 Que a RILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPTDSPYAGGVFLVTIHFPP
DYPFKPPKVAFRTKVFHPNINSNGSICLD!LKEQWSPALTISKVLLSICSLLTDPNP
DDPLVPEIAHMYKTDRAKYETTARSWTQ YAMG
674 A0349 Que a MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATIMGPPDSPYAGGVFLVTI
HFPPDYPFKPPKVAFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAH YKTDRSKYETTARSWTQKYA G
675 A0349 Cyn d MASKRILKELKDLQKDPPTSCSAGPVGEDMFHWQATIMGPSDSPFAGGVFLVNI
HFPPDYPFKPPKVSFRTKVFHPNINSNGSICLDILKEQWSPALTISKVLLSICSLLT DPNPDDPLVPEIAHMYKTDRAKYESTARSWTQKYAMG
676 A0349 Cyn d MASKRILKELKDLQKDPPTSCSAGPVAEDMFHWQATi GPSDSPYAGGVFLVTI
HFPPDYPFKPPKVAFKTKVFHPNVNSNGSICLDILKEQWSPALTVSKVLLSICSLL TDPNPDDPLVPEIAHMYKTDRAKYESTARSWTQKYAMG
677 A0356 Amb a EKIHINIWIGHVDSGKSTTTGHLIYKCGGIDKRTIEKFEKEA
678 A0356 Bet EKFHINIWIGHVDSGKSTTTGHLIYKLGGIDKRVIERFE EA
679 A0356 Que a EKVHISIWIGHVDSGKSTTTGHLIY LGGIDKRVIERFEKEA
680 A0356 Cyn d EKSHINIWIGHVDSGKSTTTGHLIYKLGGIDKRVIERFEKEA
681 A0356 Amb a E GKGSFKYAWVLDKLKAERERGITIDIALWKFETSKYYVTIIDAPGHRDFIKNMI
TGTSQADCAVLIVA
682 A0356 Bet E NKRSFKYAVvVLDKLKAERERGITIDIALWKFETTKYYCTVIDAPGHRDFIKNMI
TGTSQADCAILIID
683 A0356 Que a EMNKRSFKYAvWLDKLKAERERGITIDIALWKFETTRYYCTVIDAPGHRDFIKNMI
TGTSQADCAVLI I D
684 A0356 Cyn d EMNKRSFKYAWVLDKLKAERERGITID!ALWKFETTKYYCTVIDAPGHRDFIKNMi
TGTSQADCAVLI ID
685 A0356 Amb a QTREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIVKEVSSYLKKVGYNP
DKIPFVPISGFEGDNMIERSTNLDWYKGPTLLEALDQINEPKRPSDKPLRLPLQD VYKIGGIGTVPVGRVETGVI PGMWTFGPSGLTTEVKSVEMHHEALQEALPGD NVGFNVKNVAVKDLKRGYVASN
686 A0356 Amb a TGEFEAGISKNGQTREHALLAFTLGVKQ
687 A0356 Amb p TREHALLAFTLGVKQ ICCCNKMDATTPKYSKARYDEIVKEVSSYLK VGYNPD
KIPFVPISGFEGDNMIERSTNLDWYKGPTLLEALDQINEPKRPSDKPLRLPLQDV YKIGGIGTVPVGRVETGVIKPGMWTFGPSGLTTEVKSVEMHHEALQEALPGDN VGFNVKNVAVKDLKRGYVASN Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
688 A0356 Bet TGGFEAGISKDGQTREHALLAFTLGVRQMICCCNKMDATTPKYSKARYDEIVKE
VSSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGPTLLEALDLISEPKRP SDKPLRLPLQDVYKIGGIGTVPVGRVETGVIKPGMNA/TFGPTGLTTEVKSVEMHH EALQEALPGDNVGFNVKNVAVKDLKRGFVASN
689 A0356 Que a TGGFEAGISKDGQTREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIIKEV
SSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGPTLLEALDQISEPKRP SDKPLRLPLQDVYKIGGIGTVPVGRVETGIIKPGMWTFGPTGLTTEVKSVEMHH EALLEALPGDNVGFNVKNVAVKDLKRGFVASN
690 A0356 Cyn d TGGFEAGISKDGQTREHALLAFTLGVKQMICCCNKMDATTPKYSKARYDEIVKE
VSSYLKKVGYNPDKIPFVPISGFEGDNMIERSTNLDWYKGPTLLEALDQINEPKR PSDKPLRLPLQDVYKIGGIGTVPVGRVETGVLKPGMWTFGPSGLTTEVKSVEM HHEALQEALPGDNVGFNVKNVAVKDIKRGYVASN
691 A0356 Amb a ASFTSQVIIMNHPGQIGNGYAPVLDCHTSHIAVKFSEL
692 A0356 Amb p ASFTSQVIIMNHPGQIGNGYAPVLDCHTSHIAVKFSEL
693 A0356 Bet ANFTAQVIIMNHPGQIGNGYAPVLDCHTCHIAVKFAEL
694 A0356 Que a ANFTSQVIIMNHPGQIGNGYAPVLDCHTSHIAVKFAEL
695 A0356 Cyn d ASFTSQVIIMNHPGQIGNGYAPVLDCHTSHIAVKFAEL
696 A0356 Amb a EPKFLKNGDAGMVK LPTKPMWETFAEYPPLGRFAVRDMRQTVAVGVIKSVD
K DPT
697 A0356 Amb p EPKFLKNGDAGMVKMLPTKPMWETFAEYPPLGRFAVRDMRQTVAVGVIKSVD
KKDPTG
698 A0356 Bet EPKFLKNGDAGIVKMIPTKPMWETFAEYPPLGRFAVRDMRQTVAVGVIKGVEK
KDPSG
699 A0356 Que a EPKFLKNGDAGMVKMIPTKPMWETFSEYPPLGRFAVRDMRQTVAVGVIKSVEK
KDPSG
700 A0356 Cyn d EPKFLKNGDAGMVKMIPTKPMWETFSQYPPLGRFAVRDMRQTVAVGVIKSVEK
KDPTG
701 A0357 Amb a DRERIFKRFDTNGDGKISSTEL
702 A0357 Bet v ELERIFKRFDLNGDGQISAAEL
703 A0357 Que a DMERIFKRFDTNGDGKISLAEL
704 A0357 Cyn d DMERIFKRFDTNGDGKISLAEL
705 A0357 Amb a TLGSVTSEEIVRMMAEIDTDGDGFISFEEFTDFAGANRGLIKDVAKIF
706 A0357 Amb p TLGSTSADEVQRM AEIDTDGDGFIDFNEFISFCNANPGLMKDVAKVF
707 A0357 Bet v TLGSVTAEEIKRMMAEIDTDGDGF!SFQEFLDFAKANSGLMKDVAKIF
708 A0357 Que a TLGSTSADEVQRMMAEIDTDGDGFIDFNEFISFCNANPGLMKDVAKVF
709 A0357 Cyn d TLGSTSADEVQRMMAEIDTDGDGFIDFDEFISFCNANPGLMKDVAKVF
710 A0358 Amb a TRIAECLVGDETGTILFTARNDQVDLMKAG
711 A0358 Amb p TRIAECLVGDETGTILFTARNDQVDLMKAG
712 A0358 Bet MRIAECLVGDETGMIIFTARNDQVDLMKEG
713 A0358 Que a TRIAECLVGDETATIVFTARNEQVDLMQPG
714 A0358 Cyn d TRIAECLIGDETGCILFTARNEQVDLMKPD
715 A0358 Amb a IIRNAKIDMFKGSMRLAVDKWGRI
716 A0358 Amb p IIRNAKID FKGSMRLAVDKWGRI
717 A0358 Bet ILRNAKIDMFKGSMRLAVDKWGRV
718 A0358 Que a ILRNAKIDMFKGSMRLAVDKWGRV
719 A0358 Cyn d IIRNAKIDMFKGSMRLAVDKWGRI
720 A0358 Amb a EDN N LSLVEYELVN VTE
721 A0358 Amb p ED N N LS LVEYE LVN VTE
722 A0358 Bet EDNNLSLIEYELVNWE
723 A0358 Que a E D N N LS LVEYELVN WE
724 A0358 Cyn d EDNNLSLVEYELVNVSE
725 A0362 Amb p FSPEEISAMILTKMKETAEAFLGKKIKDAWTVPAYFNDAQRQATKDAGVIAGLNV
ARIINEPTAAAIAYGLDKKGGEKNILVFDLGGGTFDVSILTIDNGVFEVLATNGDTH
LGGEDFDQRIMEYFIKLIKKKHGKDISKDHRALGKLRREAERAKRALSSQHQVRV
EIESLFDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMEDAGLEKRQIDEIVLV
GGSTRIPKVQQLLKDYFDGKEPNKGVNPDEAVAYGAAVQGGILSGEGGDETKDI
LLLDVAPLTLG I ETVGGVMTKLI PRNTVI PTKKSQVFTTYQDQQTWSI KVYEGER
SLTKDCRLLGTFDLTGIPPAPRGTPQIEVTFEVDANGILNVKAEDKASGKSEKITIT
NEKGRLSQEEIERMVREAEEFAEEDKKVK
726 A0362 Bet v FSPEEVSAMILTKMKETAEAFLGKKIKDAWTVPAYFNDAQRQATKDAGIIAGLNV
ARIINEPTAAAIAYGLDKKGGEKNILVFDLGGGTFDVSILTIDNGVFEVLATNGDTH LGGEDFDHRIMEYFIKLIKKKHGKDISKDNRALGKLRREAERAKRALSSQHQVRV Table 4 (SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
EIESLFDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMDDAGLEKNQIDEIVLV GGSTRIPKVQQLLKDYFDGKEPNKGVNPDEAVAYGAAVQGSILSGEGGEETKDI LLLDVAPLTLGIETVGGVMTKLIPRNTVIPTKKSQVFTTYQDQQTTVSIQVYEGER SLTKDCRNLGKFDLSGIPPAPRGTPQIEVTFEVDANGILNVKAEDKGTGKSEKITI TNDKGRLSQEEIDRMVREAEEFAEEDKKVK
727 A0362 Que a FSPEEISAMVLTKMKETAEAFLGKKIKDAWTVPAYFNDAQRQATKDAGVIAGLN
VARIINEPTAAAIAYGLDK GGEKNILVFDLGGGTFDVSILTIDNGVFEVLATNGDT
HLGGEDFDHRVMDYFIKLIKKKHSKDISKDNRALG LRREAERAKRALSSQHQV
RVEIESLFDGVDFSEPLTRARFEELNNDLFRKTMGPVKKAMDDAGLEKRQIDEIV
LVGGSTRIPKVQQLLKDFFDGKEPNKGVNPDEAVAYGAAVQGGILSGEGGDET
KDILLLDVAPLTLGIETVGGVMT LIPRNTVIPSKKSQVFTTYQDQQTTVTIQVFEG
ERSLTKDCRLLGNFDLTGIAPAPRGTPQIEVTFEVDANGILNVKAEDKASGKSEKI
TITNDKGRLSQEEIDRMVQEAEEFAEEDKKVK
728 A0362 Cyn d FSPEEISAMILG MKDTAEAYLGKKINDAWTVPAYFNDAQRQATKDAGVIAGLN
VARIINEPTAAAIAYGLDKKGGEKNILVFDLGGGTFDVSILTIDNGVFEVLATNGDT
HLGGEDFDHRIMEYFIKLIKKKYSKDISKDNRALGKLRREAERAKRALSNQHQVR
VEIESLFDGTDFSEPLTRARFEELNNDLFRKTMGPVK AMEDAGLEKSQIHEIVL
VGGSTRIPKVQQLLRDYFDGKEPNKGVNPDEAVAYGAAVQGSILSGEGGEETK
DILLLDVAPLTLGIETVGGVMTKLIPRNTVIPTKKSQVFTTYQDQQTTVSIQVFEGE
RSMTKDCRLLGKFDLSGIPPAPRGTPQIEVTFEVDANGILNVKAEDKGTGKSEKI
TITNEKGRLSQEEIDRMVREAEEFAEEDKKVK
729 A0362 Amb a VDWSKWHMFWVDERWPKDHPDSNYLLA
730 A0362 Amb p VDWSKWHMFWVDERWPKDHPDSNYLLA
731 A0362 Bet v VEWSKWHVFWVDERWPKDHEDSNYKLA
732 A0362 Que a IEWSRWHIFWADERWPKDHEDSNYKLA
733 A0362 Cyn d VDWSKWHVFWVDERWPKDHEDSNYKLA
734 A0366 Que a QAVLFDIDGTICDSDPLHHYAF
735 A0366 Cyn d QAVLFDIDGTLCDSDPLHHVAF
736 A0366 Amb a GL RAAVTNAPRPNAELMI
737 A0366 Amb p GLKRAAVTNAPRPNAELMI
738 A0366 Bet v GLKRAAVTNAPRPNAELMI
739 A0366 Que a GIKRAAVTNAPKANAELMI
740 A0366 Cyn d GYKRAAVTNAPRINAELMI
741 A0376 Amb a VLGHKSNADGLTTQDTATFSVIGQGFICKNMGFSNTAGAIKHQAVALRLQSDFA
AFYNCRMDGYQDTLYVQARRQFFRNCWSGTIDFIFGNAAAVFQNCLIIVRRPM
DNQQNTVTAHGRTDPKMKSGLVIQNCRIVPDQALFPDRFKIPSYLGRPWKEYSR
TVIMESTIGDAIRPEGWMAWNGDFALKTLYYAEYANRGPGANTRSRVRWPGFH
VI
742 A0376 Amb a VTGKKSFADGITTMKTATFSIEAAGFICKNMGFHNTAGAEKHQAVALRVQGDFA
AFYNCRFDAFQDTLYVHARRQFFRNCVISGTIDFIFG
743 A0376 Amb a PMDNQQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRPWKE
FSRLVIMESTIADFIKPEGYMPWNGDFGIKTLFYAEYNNRGPGAGTSKRVKWPG FHVI
744 A0376 Amb p VTGSKSFHDG!TTMKTATFSIEASGFICKNMGFHNTAGAERHQAVALRVQGDLA
AFFNCRFDAFQDTLYVHARRQFFRNCVISGTIDFIFGNSAAIFQNCLIITRRPMDN QQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRP
745 A0376 Bet VTGRKSYRDGITTFKTASFSTMGEGFIAKSMGFQNTAGPEGHQAVALRVQADR
SAFFNCRMDGYQDTLYVQTHRQFFRNCVISGTVDFIFGDAAAVIQNSLIIVRKPM DNQQNTVTAQGRHDKRETTGLVIHNCRIVPEQKLFAQRFTIPTYLGRPWKEYAR TVIMETTLADFIQPVGYMPWAGSFALSTCSYFEYANRGPGARTVRRVRWKGAR VI
746 A0376 Que a VTGRKSFADGVTTMKTATFSIEAAGFICKNMGFHNTAGAERHQAVALRVQGDLA
AFYNCRFDAFQDTLYVHARRQFFRNCVISGTIDFIFGNSAAIFQNCLIITRRPMDN QQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRPWKEFSRLV IMESTIADFIKPEGYMPWNGDFGLKTLYYAEYANRGPGAGTSKRVTWPGFRVI
747 A0376 Cyn d VTGRKSFADGITTMKTATFSIEASGFICKNMGFHNTAGAERHQAVALRVQGDLA
AFFNCRFDAFQDTLYVHARRQFFRNCVISGTIDFIFGNSAAVFQNCLIITRRPMDN QQNSVTAHGRTDPNMKSGLVIQNCRLVPDQKLFPDRFKIPSYLGRPWKEFSRLV IMESTIADFIKPEGYMPWDRDFALKTLYYAEYNNKGPGAGTSKRVNWPGFRVI
748 A0376 Amb a AQQFTVGNFISGNLWLRYTG
749 A0376 Amb a AEQFTAGPFIDGATWLKFTGTP
750 A0376 Bet ALQFTAGPFLQGNLWLKATGFP
751 A0376 Que a AEQFTAGPFVDGATWLKFTGTP Table 4 {SEQ ID NOS: 294-767)
SEQ ID NO SpeAmino acid sequence
ID NO cies
752 A0376 Cyn d AAQ FTAG P F I DG ATWLKFTGTP
753 A0377 Amb a PKFMVFACSDSRVCPS
754 A0377 Amb p PKFMVFACSDSRVCPS
755 A0377 Bet v PKFMVFACSDSRVCPS
756 A0377 Que a PKFMVFACSDSRVCPS
757 A0377 Cyn d PKYMVFSCADSRVCPS
758 A0377 Amb a PGEAFMVRNIANMVP
759 A0377 Amb p PGEAFMVRNIANMVP
760 A0377 Bet v PGEAFWRNVANLVP
761 A0377 Que a PGEAFMIRNIANMVP
762 A0377 Cyn d PGEAFTVRNIANMVP
763 A0377 Amb a CEKEAVNVSLGNLLTYPFVK
764 A0377 Amb p C E VE AVN VS LG N LLTYPFVK
765 A0377 Bet v CEKEAVNVSIGNLLTYPFVR
766 A0377 Que a CEKEAVNVSLGNLLTYPFVR
767 A0377 Cyn d LEKEAVNLSLENLKSYPFVK
Table 5. GW conserved amino acid sequences derived from polypeptides derived from polypeptides of Table 1
Table 5 - SEQ ID NOS: 768-808
Figure imgf000102_0001
Table 5 - SEQ ID NOS: 768-808
Figure imgf000103_0001
Table 6. GW conserved amino acid sequences derived from polypeptides of Table 2
Table 6 - SEQ ID NOS: 809-951
Figure imgf000103_0002
Table 6 - SEQ ID NOS: 809-951
SEQ ID NO Spe-cies Amino acid sequence
ID
NO
853 A0265 Amb p QEASERGSADNITWWRF
854 A0265 Cyn d QEASQRGSADNITCVWRF
855 A0270 Amb a PALGEPMDIMTALQLVL
856 A0270 Amb p PALGEPMDI TALQLVL
857 A0270 Cyn d PVLGEPMDLMTALQLVM
858 A0270 Amb a KLVKALCADHNVSLVTVPSAKTLGEWA
859 A0270 Amb p KLVKALCADHNVSLVTVPSAKTLGEWA
860 A0270 Cyn d KLVKALCAEHNVHLVTVPSAKTLGEWA
861 A0270 Amb p VGCSCLWKDYGEESEGLHIVQEYVKSH
862 A0270 Cyn d VGCSCVWKDYGEESEGLNIVQEYVKSH
863 A0300 Amb a DTI DALVAAWLPGTE
864 A0300 Amb p ETLDALIAAWLPGTE
865 A0300 Cyn d DYMDALVAAWLPGTE
866 A0300 Amb a DYG FTG KLARTWFKS VN QLPM VGDPHYDPLY
867 A0300 Amb a DYGFTGKLARTWFKSVDQLPMNVGDKHYDPLF
868 A0300 Amb p DHEFHGRLPVSWFKSVDQLPMDSNKSSYDPLF
869 A0300 Cyn d DYGFTGKLPRTWFKSVDQLPMNYGD RYDPLF
870 A0304 Amb a NWVSDSDVIVGIIEDKYPHPSLVTPPELASVGSNIFSKFIAFLKSKDANDGTEQA
871 A0304 Amb p KWISDSDVITQTIEEKFPDPSLVTPPEKASVGSKIFSTFIAFLKSKDANDGTEQA
872 A0304 Cyn d KWIADSDVIVQIVEEKYPTPSLVTPPEYASVGSKIFSTFVTFLKSKDANDGSEQA
873 A0304 Amb a AVDLSLGPKLYHLQVALGH
874 A0304 Cyn d AADLSLAPKLYHLHVALEH
875 A0311 Amb a PVELLSADGIDYHKSCFKCS
876 A0311 Amb a FIDLLSADGVPYHKTCFKCS
877 A0311 Amb a FVDLLTVDSVIYHKRCFKCS
878 A0311 Amb a FVDLLSVDGVTYHKACFRCS
879 A0311 Amb p FIDLLTADSISYHKSCFKCS
880 A0311 Cyn d FIDLLTADGVIYHKTCFKCS
881 A0311 Amb a SNYAALEGILYCKHHFSQLFK
882 A0311 Amb a SSYAALNGVLYCRHHFAQLFL
883 A0311 Amb a SSYAALDGILYCKHHFAQLFM
884 A0311 Amb p ASYAALNGILYCQHHFWQLFK
885 A0311 Cyn d SSYAALNGVLFCKI HFSQLF
886 A0316 Amb a EQELETSKTSSSVEK
887 A0316 Amb p EQELETSKTSSSVEK
888 A0316 Cyn d DQEMETAKTSSSVEK
889 A0325 Amb a VDENGKISRLRRECP
890 A0325 Amb p VDGDGKIERLRRECP
891 A0325 Cyn d VDSDGKIKRLRRECP
892 A0326 Amb a RLQEQPPFDKKQFVA
893 A0326 Amb p RLQEQPAFNKKQFVA
894 A0326 Cyn d RLQEQPAFDKKQFVT
895 A0326 Amb a ATKFLLSKLSDLQFFVGE
896 A0326 Amb p ATKFLLGKLSDLQFFVGE
897 A0326 Cyn d ATKFLLGKLKDLQFFVGE
898 A0327 Amb a DAVTIRATMGMAAQDM
899 A0327 Amb p DPVTIRAM SMSAQAM
900 A0327 Cyn d DGVTIRATMSMAAQDM
901 A0334 Amb a LSINGHGTIDGQGALVWS
902 A0334 Amb a LVITGKGTLDGQGKEVWN
903 A0334 Amb p LVITGNGTLDGQGPGVWQ
904 A0334 Cyn d FSINGHGTIDGQGPLVWK
905 A0334 Amb a YNCKILPNSLVLDFV
906 A0334 Amb a YDCKILPNTLVFDFC
907 A0334 Amb p YDCKKLPTSMELEFV
908 A0334 Cyn d YNCKILPNSLVLDYV
909 A0334 Amb a VIDKVTVKAPGNSPNTDGIHIGDSSNVTITGTTIGVGDDCISIGP
910 A0334 Amb a TLKDLTITAPGDSPNTDGIHIGDSSKVTISDTTIGTGDDCISIGP
911 A0334 Amb p TITGVTVTAPEDSPNTDGIHIGRSSRVTISGATIGTGDDCVSIGA
912 A0334 Cyn d LVERVNITAPGDSPNTDGIHIGDSTNVTIAETNIGTGDDCISIGP Table 6 - SEQ ID NOS: 809-951
Figure imgf000105_0002
Table 7. GT conserved amino acid sequences derived from polypeptides derived from polypeptides of Table 1
Figure imgf000105_0001
Figure imgf000106_0001
Table 7 - SEQ ID NOS: 952-1023
Figure imgf000107_0002
Table 8. GT conserved amino acid sequences derived from polypeptides derived from polypeptides of Table 2.
Figure imgf000107_0001
Figure imgf000108_0001
Table 8 - SEQ ID NOS: 1024-1231
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Table 9. WT conserved amino acid sequences derived from polypeptides of Table 2.
Figure imgf000111_0002
Figure imgf000112_0001
Table 9 - (SEQ ID NOS: 1232-1473)
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Example 4 - Release from pollen
This example includes a description of how to examine release patterns of immunogenic molecules from pollen, e.g. the release kinetics of polypeptides of Tables 1 and 2 from pollen and detection of polypeptides in pollen diffusates by Mass Spectrometry.
Materials for Extraction:
Raw pollen or defatted pollen of various pollen sources, Glass bottles (100 ml) for extraction, PD-10 columns with PE bed support combined with 10 ml syringe with silicone tubing, PBS buffer, pH 7.2 containing the following salts: Salt Mw (g/mole) Cone. g/L Cone. mM
Sodium chloride NaCI 58.44 8.0 137
Potassium chloride KCI 74.55 0.2 2.7
Na-phosphate Na2HP04, 175.98 1.44 8.2
2H20
K-phosphate KH2P04 136.09 0.2 1.5
Phosphate cone: 8.2 + 1.5 = 9.7 mM phosphate
NaCI: μ = 1/2 * (137 * 12 + 137 * 12) = 137 mM
KCI: μ = ½ * (2.7 * 12 + 2.7 * 12) = 2.7 mM
Na2HP04: μ = 1/2 * ((8.2 * 2 * 12) + (8.2 * 22)) = 24.6 mM
KH2P04: i = ½ * ((1.5 * 12) + (1.5 * 12)) = 1.5 mM
Total ionic strength: μ = 165.8 mM « 0.17 M
Extraction Procedure (at room temperature, 21-24 °C):
5.0 g of pollen are weighed into a glass bottle and 50 ml of PBS is added and the bottle is immediately rotated, first 5 minutes by hand and thereafter rotated in a sample rotator during the entire extraction.
5 ml of slurry is taken out after 20 sec, transferred to a column with a bed filter and dragged through the filter with a syringe. The syringe is immediately transferred to a filter unit and the extract is pushed through the combined filters into a labelled test tube. The tube is stored in an ice bath until the sample is pipetted in aliquots for further analysis and frozen. About 5 ml of the suspension is taken out at various time points.
Samples are analyzed for presence of a polypeptide of Tables 1 and 2 and, optionally major allergens by MS (Mass Spectrometry).
Mass Spectrometry Buffers/solutions for reduction, alkylation and digestion of the sample:
Sample buffer: 8 M urea in 0.4 M NH4HC03
DTT (45 mM): Make it fresh from the frozen stock 1.0 M: 45 μ1 1 M DTT + 955 μΙ water lodoacetamide (IAA): Make fresh solution, lodoacetamid 100 mM,
Trypsin: Sigma T6567, Dissolve one vial in 20 μΙ of 1 mM HCI. This results in a solution containing 1 Mg/μΙ trypsin. After reconstitution in 1 mM HCI frozen aliquots can be stored for up to 4 weeks.
Enzymatic digestion with trypsin in solution for mass spectrometry: Dilute the dried sample in 5 μΙ of water, add 15 μΙ of sample buffer (8 M Urea in 0.4 M NH4HC03), add 5 μΙ 45 mM DTT, incubate at 56°C for 15 min, cool it to room temperature, add 5 μΙ of 100 mM lodoacetamide, incubate in the dark in room temperature for 15 min, add 90 μΙ of water to lower the concentration of urea <1-2 M, add 1 μg trypsin, incubate at 37°C over night.
Chromatography:
Reverse phase chromatography (Ultimate 3000 HPLC, Dionex) was performed using a C18 pre- and analytical column. The eluting peptides were sprayed directly into an ESI- QTOF mass spectrometer (MaXis, Bruker). After washing the trap column with 0.05% v/v formic acid for 5 min with a flow rate of 30 μΙ/min, the peptides were eluted with an acetonitrile gradient at a flow rate of 2 μΙ/min using solvent A: 0.05% v/v formic acid and solvent B: 80% v/v acetonitrile/0.04% v/v formic acid and the gradient: 4-50% B in 200 minutes; 50-80% B in10 minutes; 100% B in 10 min, 4% B in 5 min. Spectra were acquired in the mass range 50-2599 m/z and a spectra rate of 1.5 Hz. The instrument was tuned and calibrated using ESI-L Low concentration Tunning Mix from Agilent Technology.
Data acquisition and instrument control were carried out with Bruker Compass HyStar 3.2. Data processing was performed using DataAnalysis 4.0 (Bruker). Protein identification was performed using the program Biotools3.2 (Bruker) and two different databases, i.e. Swiss prot and NCBInr. The MS/MS data sets for the tryptic digest were analyzed using the following parameters; peptide tolerance 10 ppm and fragment tolerance 0.05 Da.
Procedure: The extraction samples were all evaporated (50 μΙ) and re-suspended in 5 μΙ of water. The sample is then reduced, alkylated and digested with trypsin. Resulting peptides are separated and identified by reversed phase chromatography followed by MS/MS.
Results: The release of major allergens from the various pollen species investigated is initiated almost instantly after hydration of pollen with buffer and the release continues with high rate within a time range of at least 30 to 60 minutes (data not shown). Table 10 shows polypeptides that starts release from grass pollen (Phi p or Cyn d), weed pollen (Amb a) and/or tree pollen (Que a and/or Bet v) within a period overlapping with the release of major allergens, respectively. In table 10 it is indicated which of the
polypeptides detected in Phi p pollen that can be released from hydrated pollen (pollen extracts) and which of the corresponding homolog polypeptides detected in Cyn d, Amb a, Que a and Bet v pollen that can be released from hydrated pollen of the species Cyn d, Amb a, Que a and Bet v, respectively, within a time period of 10 minutes to 120 minutes. The label "x" means that polypeptides or its homolog is detected in the extracts within the specified time period. No label means no detectable release of the polypeptide within the time period indicated. "GW" means that a given polypeptide is releasable from grass pollen (Phi p and/or Cyn d) and that its corresponding homolog found in Amb a pollen is releasable from weed pollen (Amb a). "GT" means that a given polypeptide is releasable from grass pollen (Phi p and/or Cyn d) and that its corresponding homolog found in Que a or Bet v pollen is releasable from tree pollen (Que a and/or Bet v). "GWT" means that a given polypeptide of Table 1 is releasable from grass pollen (Phi p and/or Cyn d),) that its corresponding homolog found in Amb a pollen is releasable from weed pollen (Amb a) and that its corresponding homolog found in tree pollen is releasable from tree pollen (Que a and/or Bet v).
It was found that the set of polypeptides having the polypeptide ID NOs A0349, A0246, A0209, A0211 , A0325, A0357, A0203 could be released from the grass, weed as well as the tree pollen investigated (herein named GWT polypeptides).
Polypeptide A0262 (herein named GT polypeptide) was released from both grass and tree pollen, but not from weed pollen.
The set of polypeptides having the polypeptide ID NOs A0362, A0316, A0356, A0376, A0336, A0377 and A0366 could be released from grass as well as weed pollen, but not the tree pollen investigated (herein named GW polypeptides).
Table 10. Release of polypeptides from grass, weed and/or tree pollen
Figure imgf000118_0001
Release of polypeptides from pollen species Release from multiple % Identity to pollen species: Human
ID No Phi p Cyn d Amb a Que a Bet v GW GT GWT Homolog
A0233 X X NA
A0246 X X X X X X X X 75.2
A0260 X NA
A0262 X X X X X 51.2
A0265 X NA
A0270 X X NA
A0300 X NA
A0304 X X NA
A0311 X NA
A0316 X X X X NA
A0317 X X X NA
A0325 X X X X X X X X 82.6
A0326 X X X NA
A0327 X NA
A0334 X X NA
A0336 X X X X NA
A0349 X X X X X X X X 75.6-82.6
A0356 X X X X 75.9
A0357 X X X X X X X X 47.2
A0358 X NA
A0362 X X X NA
A0366 X X X X NA
A0376 X X X X NA
A0377 X X X X NA
Example 5 - In silico prediction in vitro and determination of HLA Class II allele binding
To investigate the immunogenic potential of polypeptides of the invention, the binding affinity to a panel of HLA class II molecules may be predicted in silico or determined in vitro, for example as described in PCT application WO 2013/119863 to a set of 15mer or 20-mer overlapping peptides that are subsequences to the polypeptides and conserved sequences set out in Tables 1 to 9.
HLA binding affinity of peptides may be predicted using NetMHC 3.2, for example HLA affinity to the number of commonly found alleles DPA1*01-DPB1*0401 , DPA1*0103-
DPB1*0201 , DPA1*0201-DPB1*0101 , DPA1*0201 , PB1*0501 , DPA1*0301-DPB1*0402, DQA1*0101-DQB1*0501 , DQA1*0102-DQB1*0602, DQA1*0301-DQB1*0302,
DQA1*0401-DQB1*0402, DQA1*0501-DQB1*0201 , DQA1*0501-DQB1*0301 , DRB1*0101 , DRB1*0301 , DRB1*0401 , DRB1*0404, DRB1*0405, DRB1*0701 ,
DRB1*0802, DRB1*0901. Subsequences with predicted binding scores in the top 20% for a given allele may be considered potential binders. Subsequences predicted to bind 13 or more HLA molecules at this threshold may be considered promiscuous binders. Also, the HLA binding of polypeptides or subsequences thereof may be predicted in silico for an estimated worldwide allele distribution. Average haplotype and phenotype frequencies for 63 individual HLA-DRB1 alleles are based on MHC data available at the Allele Frequency Net Database (AFND, http://www.allelefrequencies.net). Frequencies from the four major ethnical groups, Hispanics, Caucasians, African Americans, and Asians, are used as an estimated worldwide allele distribution. The frequencies are calculated as the mean of the frequencies from population IDs, 1514, 1513, 2419, and 2420, respectively. Each ethnical group consists of data from more than 1000 individuals. A set of 15mers peptides, overlapping each other with 14 amino acids, were constructed from each of the wild type sequences of Table 1 as well as the GWT conserved regions of Table 3. The numbers of 15mers peptides that can be derived from these sequences in total are summarized in Table 11a (column 4). For each peptide, the binding was predicted with NetMHCIIpan-3.0 for each of the 63 HLA alleles derived from AFND. Per peptide-allele combination, a binding threshold of 300 nM as well as a percentile rank score, as reported by the algorithm, of at least 30 is applied. Some HLAs bind to the same peptide, and given the estimated worldwide allele distribution, we can estimate the binding capability for each peptide. For each sequence (derived from Table 1 and 3), the number of 15mer peptides predicted to bind to at least half of the estimated worldwide allele distribution are shown in Table 11a, column 5. For example, a total number of 74 15mer peptides could be derived from the sequence representing wild type polypeptide A202, and 13 of those were predicted to be binders in at least half of the worldwide population, as estimated for the four major ethnic groups. This amount to that 18% of the 15mers was predicted to bind DRB1 alleles. Finally, the sum of 15mers with DRB1 allele binding that could be derived from GWT conserved sequences were calculated and summarized in the last column of Table 11a. According to this analysis, the wild type polypeptides may be ranked according to their potential possess several HLA Class II binding sites in the part of the sequence that is conserved across pollen species. For example, some wild type polypeptides have considerable more potential to bind HLA Class alleles in their conserved regions than others, e.g. polypeptides with ID NO: 246, 270, 311 , 316, 325, 356 or 376 versus polypeptides with ID NO: 203, 233, 366 or 377. Table 11a. Number of 15mers peptides with DRB1 HLA allele binding derivable from polypeptides of Table 1 and their GWT sequences of Table 3.
The wording "FL" and "GWT in Table 11 a refer to the wild type sequence of Table 1 and the GWT conserved sequences of Table 3, respectively.
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Table 11b. DRB1 alleles for use in prediction of HLA Class II allele binding
Allele Average Allele Average Allele Average frequency frequency frequency
DRB1 0101 0.04275 DRB1 0804 0.0.1625 DRB1 1312 0.001 DRB1_0102 0.02325 DRB1 0806 0.0015 DRB1 1323 0.00025 Allele Average Allele Average Allele Average frequency frequency frequency
DRB1 0103 0.00475 DRB1 0809 0.0005 DRB1 1331 0.00025
DRB1 0301 0.07725 DRB1 0811 0.0005 DRB1 1401 0.02275
DRB1 0302 0.0175 DRB1 0901 0.0395 DRB1 1402 0.00925
DRB1 0307 0.00025 DRB1 1001 0.017 DRB1 1403 0.00125
DRB1 0401 0.03375 DRB1 1101 0.057 DRB1 1404 0.00525
DRB1 0402 0.00775 DRB1 1102 0.01325 DRB1 1405 0.0045
DRB1 0403 0.01575 DRB1 1103 0.00275 DRB1 1406 0.01
DRB1 0404 0.0275 DRB1 1104 0.0185 DRB1 1407 0.001
DRB1 0405 0.02325 DRB1 1106 0.00075 DRB1 1418 0.00025
DRB1 0406 0.00625 DRB1 1110 0.00075 DRB1 1419 0.00025
DRB1 0407 0.0245 DRB1 1111 0.00025 DRB1 1424 0.00025
DRB1 0408 0.0025 DRB1 1201 0.02325 DRB1 1501 0.074
DRB1 0410 0.0025 DRB1 1202 0.0195 DRB1 1502 0.02575
DRB1 0411 0.0035 DRB1 1301 0.04575 DRB1 1503 0.03125
DRB1 0417 0.00025 DRB1 1302 0.0445 DRB1 1504 0.00025
DRB1 0701 0.1015 DRB1 1303 0.01525 DRB1 1506 0.001
DRB1 0801 0.01125 DRB1 1304 0.004 DRB1 1601 0.00425
DRB1 0802 0.02725 DRB1 1305 0.002 DRB1 1602 0.01725
DRB1 0803 0.014 DRB1 1311 0.00025 DRB1 1607 0.00025
This example also describes how to determine the HLA class II binding in vitro of polypeptides of the invention including subsequences thereof. The assay employed is a competitive MHC class II binding assay, wherein each polypeptide or 15mer or 20mer peptide is analyzed for its ability to displace a known control binder from each of the human MHC class II allotypes shown in Table 11c. Due to the nature of the competitive assay, the data for each polypeptide or subsequence is determined as a ratio of its own IC50 to that of the control peptide. Thus, a polypeptide or subsequence that has an IC50 value that is parity to the control peptide has an identical binding affinity, while
polypeptides or subsequences with a ratio less than one have a higher affinity and those with a ratio greater than one have a lower affinity. The ratio of IC50 may be determined at different cutoff concentrations, for example at 300 or 1000 nM.
Assays to quantitatively measure binding to purified class II MHC molecules are based on the inhibition of binding of a high affinity radiolabeled peptide to purified MHC molecules, and were performed essentially as detailed elsewhere (Sidney et al. 2008; Sidney et al. 2010b, a; Greenbaum et al. 2011 ; Sidney et al. 2001 , Sidney et al 2013, McKinney et al 2013). Briefly, 0.1-1 nM of radiolabeled polypeptide or subsequences may be co- incubated at room temperature or 37°C with purified MHC in the presence of a cocktail of protease inhibitors. Following a two- to four-day incubation, MHC bound radioactivity may be determined by capturing MHC/peptide complexes on monoclonal Ab coated Lumitrac 600 plates (Greiner Bio-one, Frickenhausen, Germany), and may be measuring bound cpm using the TopCount (Packard Instrument Co., Meriden, CT) microscintillation counter. In the case of competitive assays, the concentration of polypeptide or subsequence yielding 50% inhibition of the binding of the radiolabeled peptide may be calculated. Under the conditions utilized, where [label] < [MHC] and IC50 > [MHC], the measured IC50 values are reasonable approximations of the true Kd values (Cheng and Prusoff 1973; Gulukota et al. 1997). Each polypeptide or subsequence may be tested at six different concentrations covering a 100,000-fold dose range in three or more independent experiments. As a positive control, the unlabeled version of the radiolabeled probe may also be tested in each experiment.
Table 11c. HLA alleles used for HLA prediction
Table 11c shows a panel of 25 MHC II molecules (alleles) for which peptide binding affinities may be predicted or determined in vitro.
Figure imgf000125_0001
Example 6 - T cell activity
To investigate the immunogenic potential of a polypeptide of the invention, the
polypeptide may be tested for T cell immune reactivity, which may be determined by the production of IL-5 or IFN-gamma from PBMCs f allergic donors, wherein the PBMC's are cultured under the presence of the polypeptide. The cytokines IL-5 and IFNg may be measured by known methods in the art, for example as described in PCT application WO 2013/119863.
Briefly, specific T cells may be obtained in the following manner; PBMCs may be thawed and cultured with 5% human AB serum at a density of aboutn 2 x 106 cells/ml in 24-well plates and stimulated with the polypeptide or a pollen extract comprising the polypeptide or a subsequence thereof (concentration may be about i g/ml). Cells may be kept at 37°C, 5% C02 and IL-2 may be added every 3 days after the initial stimulation with the polypeptide. On day 14, cells may be harvested and screened for reactivity to the polypeptide or a subsequence thereof. The production of IL-5 and IFN-γ from the cultured PBMCs described above may be measured in response to stimulation with the to the polypeptide or a subsequence thereof using ELISpot as described in Oseroff C et al, 2010. The criteria for positive response may be 20 SFCs/10 PBMCs, p< 0.05 of stimulated cells compared to background, and a stimulation index (SI) > 2.
Alternative to the ELISpot assay, the cytokine production of T cells can be measured in cell culture supernatants harvested at day 1 of the above procedure by MesoScale or MagPix analysis. The design of both assays resemble ELISA assays with the capture antibodies of several different specificities spotted in each well of a 96 well plate
(Mesoscale, #144259) or the individual catching antibodies attached to beads that can be mixed to the final assay reagent (MagPix #230608) and both assays are developed to analyze various different cytokines or cellular mediators in a single sample. The cytokines IL-2, IL-4, IL-5, IL-10, IL-12, IL-13, and IFN-g may be measured by Mesoscale, whereas IL-4, IL-5, IL-9, IL-10, IL-12, IL-13, IL-17, IL-22, IL-31 may bemeasured by MagPix.
The T cell reactivity may also be tested by Ex-Vivo FluoroSpot or EliSpot analysis of cytokines induced in response to polypeptides added to PBMCs that have not previously been stimulated with the polypeptides or pollen extract.
Example 7 - In vivo activity
This example describes how to determine the ability of a polypeptide of the invention to relieve, e.g. treat, an immune response in mice.
Initially, the sensitization pattern of a molecule (e.g. an immunogenic molecule) of the invention (A0349) was investigated in BALB/c mice sensitized to Phi p extract (Figure 2). For the purposes of these studies, the immunogenic molecule were expressed in E. coli using standard expression protocols.
Mice were sensitized by one intraperitoneal injection with Phi p extract adsorbed to aluminium hydroxide. Eleven days later the mice were euthanized and splenocytes were stimulated in vitro with Phi p pollen extract, the allergen Phi p 5 or A0349. The cells were incubated for 6 days at 37°C under 5% C02 and incorporated 3H-thymidine was counted and used as a measure for T cell proliferation.
The results show that the in vitro T-cell response towards A0349 is much weaker compared to the response to Phi p 5 (Figure 2). This correlates well with the human situation, where Phi p 5 is considered to be a major T-cell allergen. In line with this, the results also show that the response towards A0349 is much weaker compared to the response towards the Phi p extract that was used for intraperitoneal sensitization. Then the tolerance inducing potential of A0349 was investigated in a prophylactic mouse model using sublingual administration.
The ability of A0349 to induce prophylactic tolerance was investigated by SLIT treatment of naive BALB/c mice with A0349for two weeks (Monday-Friday) followed by one intraperitoneal Phi p extract sensitization. Eleven days after the sensitization, splenocytes were harvested and stimulated in vitro with Phi p extract or the allergen Phi p 5.
Figure 3 shows that prophylactic SLIT treatment with A0349 is capable of inducing tolerance towards the Phi p extract as shown by the reduced Phi p extract specific proliferation in splenocytes from the A0349-treated mice compared to buffer (sham) treated mice. In addition, it was shown that A0349 was capable of inducing tolerance toward the allergen Phi p 5 (Figure 4), which amino acid sequence is different from A0349. Thus, the treatment with A0349 resulted in the suppression of an immune response caused by an unrelated antigen, i.e. Phi p 5, possibly via bystander tolerance induction. Further polypeptides identified in the same manner and reported in patent application PCT/US2014/066577 also shows low immunogenicity of the polypeptide compared to major allergens, but nevertheless they were able to reduce an immune response caused by an allergen unrelated to the polypeptide itself (In vivo data of the polypeptides NTGA 86/51 and NTGA 6 shown in Example 5 of PCT/US2014/066577). Whether a molecule (e.g. an immunogenic molecule) of the invention can relieve, e.g. treat, an immune response triggered by a pollen allergen in mice that are sensitized to the pollen allergen when starting SLIT treatment can be investigated in a therapeutic mouse model. For example, BALB/cJ mice or HLA-transgenic mice may be IP sensitized with model allergen adsorbed to aluminium hydroxide (e.g. an extract of a grass pollen species, e.g. Cyn d, Poa p, Phi p or a model allergen like OVA). Subsequently, the mice might be treated by sublingual immunotherapy (SLIT) with a molecule (e.g. an immunogenic molecule) of the invention for a period of about 4-8 weeks, followed by about 2-3 weeks of intranasal challenge with model allergen together with the
immunogenic molecule or model allergen alone to induce an allergic immune response in the airways. Mice are then sacrificed one day after the last challenge and blood, Broncho alveolar fluid (BAL), spleen and cervical lymph nodes may be collected for analysis. Clinically relevant readouts, such as sneezes, airway hyper-reactivity and presence of eosinophils, might be obtained on the last day of intranasal challenge. For example, sneezes may be observed in an 8 min-period after intranasal administration of model allergen and the numbers of sneezes be counted during this period. Airway hyperreactivity may be determined using a whole body pletysmograph; airflow obstruction might be induced by increasing concentrations of aerosolized metacholine. Pulmonary airflow obstruction may be measured by enhanced pause (penh) in a period of 6 minutes after administration of metacholine. Differential counting of bronchial fluid (BAL) is performed after centrifugation of BAL fluid and removal of supernatant. The pellet was re- suspended in PBS and the fraction of eosinophils might be determined by an automated cell counter (Sysmex).
The results may show that a molecule (e.g. an immunogenic molecule) of the invention is able to reduce the number of sneezes, number of eosinophils, airway obstruction, T cell proliferation of spleen cells or cervical lymph nodes and may be shown to depend on the co-exposure of model allergen and immunogenic molecule at the target organ
(e.g. airways).
Whether SLIT treatment with pan pollen immunogenic molecules is capable of inducing tolerance that can be re-activated by a non-identical, but highly conserved immunogenic molecule from a different pollen source can be addressed in several different in vivo models, as outlined below.
Experiment 1 :
1. SLIT treatment with immunogenic molecule A 2. IP Sensitization with immunogenic molecule B (contains conserved regions
overlapping with A)
3. in vitro stimulation with immunogenic molecule B
Where results verify that the specific in vitro proliferation to immunogenic molecule B is down-regulated in mice SLIT-treated with immunogenic molecule A, then cross-species tolerance induction has been demonstrated for this immunogenic molecule, since the two immunogenic molecules are sufficiently similar in order for cross-species tolerance induction to occur.
Experiment 2:
1. SLIT treatment with immunogenic molecule A 2. IP Sensitization with extract of pollen source containing immunogenic molecule B (pollen extract containing the homologous immunogenic molecule B)
3. In vitro stimulation with extract of pollen source containing immunogenic molecule B and immunogenic molecule B Where results verify that the specific in vitro proliferation to immunogenic molecule B extract is down-regulated in mice SLIT-treated with immunogenic molecule A, then cross- species tolerance induction has been demonstrated for this immunogenic molecule.
Furthermore, it has been demonstrated that pollen source B contains sufficient amounts of immunogenic molecule B to re-activate the tolerance induced by SLIT treatment with immunogenic molecule A.
In the above-mentioned mouse model, Balb/cJ mice have been suggested. However, in vivo studies may instead be carried out in humanized mice models using transgenic mice, e.g. "HLA-DRB1*0401 transgenic mice" that may be obtained from Taconic. Also, in the above-mentioned mouse models, the immune response against an allergen of a grass pollen (Phi p grass extract) have been investigated, but other models may investigate the immune response against non-grass pollen allergens, e.g. allergens of weed or tree pollen, or there may be used model allergens like OVA protein.
Furthermore, the T cell responses in mice or humans may be evaluated by in-vitro T cell proliferation assays or ELISPOT assays. The production of IL-5 and IFN-γ from cultured PBMCs (Peripheral blood monocytes) obtained from humans or splenocytes obtained from mice in response to stimulation with a molecule (e.g. an immunogenic molecule) disclosed herein. Such assays are well known in the art. The assays may be able to analyze various different cytokines or cellular mediators associated with the immune response, e.g. the cytokines IL-2, IL-4, IL-5, IL-9, IL-10, IL-12, IL-13, IL-17, IL-22, IL-31 and IFN-gamma.
Example 8 - Analysis of cross reactivity
This example describes how to determine that T cells responding to a particular polypeptide containing a conserved sequence, e.g. GWT, GT, GW sequence derived from Phi p polypeptides (Tables 3, 5 and 7) also recognizes the corresponding
polypeptide identified in another pollen species.
In the present set-up, the cross reactivity was investigated using model polypeptides derived from Phi p pollen, for example the known allergens (Phi p 1 and Phi p 5) or polypeptides described in international patent application WO2013/119863: PBMCs from Phi p reactive donors were expanded with conserved peptides derived from the model polypeptidesfor 14 days (the specific peptides used for the analysis is shown in Table 12). For each peptide, the mismatch to a corresponding sequence in a non-grass pollen species or a pollen species other than Phi p were determined. Cytokine IL-5 responses were measured in response to the peptide itself, Phi p extract and extracts of the other pollen species. Reponses to extracts and peptide pools were expressed as the relative fraction of the response to the peptide itself and plotted as a function of conservation of the peptide in the different extracts (Fig 1). The data points for each peptide are contained in Table 12. A clear hierarchy of responses was observed, with non-Phi p extracts in which the peptide is completely conserved (zero mismatches) showing the highest response, followed by non-Phi p extracts with 1-2 mismatches, and lowest responses with non-Phi p extracts with 3 or more mismatches. The exact same hierarchy was observed when analyzing peptides from the major allergens and the NTGA-derived peptides separately. Thus, Phi p epitopes conserved in other pollen species, including pollen of Amb a and Que a and other non-grass pollen, were indeed able to induce cross- reactive T cell immune responses.
Table 12. Cross reactivity data
Table 12 shows individual peptide data for the cross reactivity experiments and the peptide used. Each peptide was used to stimulate cells and cross reactivity was tested for extracts from each pollen species. The number of mismatches (# of mm) for each peptide compared to the pollen species and the reactivity of the extracts as a percentage of the reactivity compared to the peptide are shown.
Figure imgf000130_0001
CD
Figure imgf000131_0001
Figure imgf000132_0001
O
Figure imgf000133_0001

Claims

Claims
1. A molecule comprising or consisting of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293; SEQ ID NOS: 617-676, 294-616 and 677-767; SEQ ID NOS: 768- 808; SEQ ID NOS: 809-951 , SEQ ID NOS: 952-1023, SEQ ID NOS: 1024-1231 ; and SEQ ID NOS: 1232-1473; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293; SEQ ID NOS: 617-676, 294-616 and 677-767; SEQ ID NOS: 768-808; SEQ ID NOS: 809-951 ; SEQ ID NOS: 952-1023; SEQ ID NOS: 1024-1231 and SEQ ID NOS: 1232-1473; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2
mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 274, 270-273, 212-269 and 275-293; SEQ ID NOS: 617-676, 294-616 and 677-767; SEQ ID NOS: 768-808; SEQ ID NOS: 809-951 ; SEQ ID NOS: 952-1023; SEQ ID NOS: 1024-1231and SEQ ID NOS: 1232-1473; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 30, 1-29, 31-37 set out in Table 1 and SEQ ID NOS: 161-172, 38-160 and 173-211.
2. The molecule according to claim 1 , wherein the polypeptide of option d) comprises an amino acid sequence having at least 75% similarity or identity to a sequence selected from any one of SEQ ID NOS: 30, 1-29, 31-37 and SEQ ID NOS: 161-172, 38-160 and 173-211.
3. The molecule according to claim 1 , wherein the polypeptide of option a) comprises an amino acid sequence having at least 75% similarity or identity to a sequence selected from any one of SEQ ID NOS: 270-274; 212-269; 275-293; SEQ ID NOS: 617- 676, 294-616 and 677-767; SEQ ID NOS: 768-808; SEQ ID NOS: 809-951 ; SEQ ID NOS: 952-1023; SEQ ID NOS: 1024-1231and SEQ ID NOS: 1232-1473.
4. The molecule according to claim 1 , wherein the polypeptide of option b) comprises an amino acid sequence having at least 75% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274; 212-269; 275-293;SEQ ID NOS: 617-676, 294-616 and 677-767; SEQ ID NOS: 768-808; SEQ ID NOS: 809-951 ; SEQ ID NOS: 952-1023; SEQ ID NOS: 1024-1231 and SEQ ID NOS: 1232-1473.
5. The molecule according to claim 1 , wherein the immunogenic molecule comprises or consists of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 270-274 and SEQ ID NOS: 617-676; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 and SEQ ID NOS: 617-676; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 270-274 and SEQ ID NOS: 617-676; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 27-30 or SEQ ID NOS: 161-172.
6. The molecule according to claim 1 , wherein the immunogenic molecule comprises or consists of a) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 217-220 and SEQ ID NOS: 321-344; or b) a polypeptide comprising an amino acid sequence having at least 65% similarity or identity to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 217-220 and SEQ ID NOS: 321-344; or c) a polypeptide, which includes at least one amino acid sequence with 0, 1 or 2 mismatches to a subsequence of at least 15 contiguous amino acid residues of a sequence selected from any one of SEQ ID NOS: 217-220 and SEQ ID NOS: 321-344; or d) a polypeptide comprising an amino acid sequence having at least 65% sequence similarity or identity to a sequence selected from any one of SEQ ID NOS: 4 and 5 and SEQ ID NOS: 49-54.
7. The molecule according to any one of claims 1 to 3, 5 and 6 wherein the polypeptide of option a) or of option d) has a length of no more than 800 amino acid residues.
8. The molecule according to any one of claims 1 and 4 to 6, wherein the polypeptide of option b) or option c) has a length of 15-800 amino acid residues.
9. The molecule according to any one of the preceding claims, comprising one or more T cell epitope-containing amino acid sequence(s).
10. The molecule according to any one of claims 1 , 5 and 6 wherein the subsequence comprises a T cell epitope.
11. The molecule according to any one of the preceding claims, wherein the polypeptide of option a), b), c) or d) modifies a cytokine response by a cell from an allergic subject.
12. The molecule according to claim 11 , wherein the cytokine response comprises one or more of IL-5, IFN gamma, IL-4, IL-10, IL-13 and IL-17.
13. A composition comprising one or more molecules according to any one of claims 1 to 12.
14. The composition according to claim 13 that is lyophilized and optionally sterile.
15. A pharmaceutical composition comprising one or more molecules according to any one of claims 1 to 12 or a composition according to any one of claims 13 or 14 and a pharmaceutically acceptable carrier, excipient and/or adjuvant.
16. A method for relieving an immune response in a subject in need thereof, comprising administering a therapeutically effective amount of a molecule according to any one of claims 1 to 12 or a composition according to any one of claims 13 to 15.
17. A method for relieving one or more symptoms of an immune response in a subject in need thereof, comprising administering a therapeutically effective amount of a molecule according to any one of claims 1 to 12 or a composition according to any one of claims 13 to 15.
18. A method for bystander suppression of an immune response in a subject in need thereof, comprising administering a therapeutically effective amount of a molecule according to any one of claims 1 to 12 or a composition according to any one of claims 13 to 15.
19. The method according to any one of claims 16 to 18, wherein the immune response is triggered by pollen from one or more plant families.
20. The method according to claim 19, wherein the one or more plant families are selected from the group consisting of Poaceae, Asteraceae, Fagaceae, Betulaceae,
Oleaceae, and Plantaginaceae.
21. The method according to claim 20, wherein the immune response is triggered by pollen of the plant family Poaceae and pollen of a plant family selected from the group consisting of Asteraceae, Fagaceae and Betulaceae.
22. The method according to claim 20, wherein the immune response is triggered by pollen of the plant genus Phleum and pollen from one or more of a plant genus selected from the group consisting of Conydon, Ambrosia, Betula and Quercus or combinations thereof.
23. The method according to any one of claims 16 to 22, wherein the immune response is triggered by one or more pollen allergens.
24. The method according to claim 23, wherein the one or more pollen allergens are from a plant family selected from the group consisting of Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae.
25. The method according to claim 24, wherein the one or more pollen allergens are from pollen of the plant family Poaceae and from pollen of a plant family selected from the group consisting of Asteraceae, Fagaceae and Betulaceae.
26. The method according to claim 25, wherein the one or more pollen allergens are from pollen of the plant genus Phleum and from pollen of a plant genus selected from the group consisting of Conydon, Ambrosia, Betula and Quercus and combinations thereof.
27. The method according to any one of the preceding claims, wherein the subject is sensitized to one or more pollen species from one or more plant families.
28. The method according to claim 27, wherein the one or more plant families are selected from the group consisting of Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae.
29. The method according to any one of the claims 16 to 28, wherein the subject is sensitized to a pollen species of the plant family Poaceae and a pollen species of a plant family selected from the group consisting of Asteraceae, Fagaceae and Betulaceae.
30. The method according to any one of the claims 16 to 29, wherein the subject is sensitized to a pollen species of the plant genus Phleum and a pollen species from one ore more plant genera selected from the group consisting of Conydon, Ambrosia, Betula and Quercus and combinations thereof.
31. The method according to any one of claims 16 to 30, wherein the subject is sensitized to one or more pollen allergens from one or more plant families.
32. The method according to claim 31 , wherein the one or more plant families are selected from the group consisting of Poaceae, Asteraceae, Fagaceae, Betulaceae, Oleaceae, and Plantaginaceae.
33. The method according to any one of claims 16 to 32, wherein the subject is sensitized to a pollen allergen of the plant family Poaceae and a pollen allergen from one or more of plant families selected from the group consisting of Asteraceae, Fagaceae and Betulaceae and combinations thereof.
34. The method according to any one of claims 16 to 33, wherein the subject is sensitized to a pollen allergen of the plant genus Phleum and a pollen allergen from one or more plant genera selected from the group consisting of Conydon, Ambrosia, Betula and Quercus and combinations thereof.
35. The method according to any one of claims 16 to 34, wherein the immune response is a specific T-cell response to the immunogenic molecule being administered to the subject.
36. The method according to claim 35, wherein the specific T-cell response is determined by way of contacting a sample of PBMCs obtained from the subject with said immunogenic molecule polypeptides and measuring the IL-5 secretion or IL-5 mRNA gene expression.
37. A molecule according to any one of claims 1-12 or a composition according to any one of claims 13-15 for use in a method according to any one of claims 16 to 36.
38. Use of a molecule according to any one of claims 1-12 or a composition according to any one of claims 13-15 for the preparation of a medicament for use in a method according to any one of claims 16 to 36.
PCT/US2015/018026 2014-02-28 2015-02-27 Polypeptides derived from phl p and methods and uses thereof for immune response modulation WO2015131053A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461946370P 2014-02-28 2014-02-28
US61/946,370 2014-02-28

Publications (1)

Publication Number Publication Date
WO2015131053A1 true WO2015131053A1 (en) 2015-09-03

Family

ID=52633721

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/018026 WO2015131053A1 (en) 2014-02-28 2015-02-27 Polypeptides derived from phl p and methods and uses thereof for immune response modulation

Country Status (1)

Country Link
WO (1) WO2015131053A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023242436A1 (en) * 2022-06-17 2023-12-21 Danmarks Tekniske Universitet Allergy vaccines based on consensus allergens

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US5422109A (en) 1989-07-03 1995-06-06 Societe D'exploitation De Produits Pour Les Industries Chimiques (S.E.P.P.I.C.) Fluid vaccines and active principle vehicles containing a metabolizable oil
WO1995017210A1 (en) 1993-12-23 1995-06-29 Smithkline Beecham Biologicals (S.A.) Vaccines
WO1996002555A1 (en) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
WO1996033739A1 (en) 1995-04-25 1996-10-31 Smithkline Beecham Biologicals S.A. Vaccines containing a saponin and a sterol
WO1998016247A1 (en) 1996-10-11 1998-04-23 The Regents Of The University Of California Immunostimulatory polynucleotide/immunomodulatory molecule conjugates
WO1998056414A1 (en) 1997-06-11 1998-12-17 Smithkline Beecham Biologicals S.A. Oil in water vaccine compositions
WO1999011241A1 (en) 1997-09-05 1999-03-11 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins
WO1999012565A1 (en) 1997-09-05 1999-03-18 Smithkline Beecham Biologicals S.A. Vaccines
US6087329A (en) 1991-10-25 2000-07-11 Immunex Corporation CD40 ligand polypeptide
EP1403280A1 (en) * 2002-09-27 2004-03-31 BIOMAY Produktions- und Handels- Aktiengesellschaft Hypoallergenic allergy vaccines based on the timothy grass pollen allergen Phl p 7
WO2005049107A2 (en) 2003-11-18 2005-06-02 Nanapass Technologies Ltd. Enhanced penetration system and method for sliding microneedles
WO2006054280A2 (en) 2004-11-18 2006-05-26 Nanopass Technologies Ltd. System and method for delivering fluid into flexible biological barrier
US20070044171A1 (en) * 2000-12-14 2007-02-22 Kovalic David K Nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
WO2007066341A2 (en) 2005-12-08 2007-06-14 Nanopass Technologies Ltd. Microneedle adapter for dosed drug delivery devices
US20110167514A1 (en) * 2007-07-05 2011-07-07 Ceres, Inc. Nucleotide sequences and corresponding polypeptides conferring modulated plant characteristics
WO2012049310A1 (en) 2010-10-15 2012-04-19 Alk-Abelló A/S Suppression of a hypersensitivity immune response with unrelated antigen derived from allergen source material
WO2012168487A1 (en) 2011-06-09 2012-12-13 Biomay Ag Peptide carrier fusion proteins as allergy vaccines
US20130058978A1 (en) * 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Induced tolerogenic dendritic cells for inducing regulatory b cells
WO2013119863A1 (en) 2012-02-07 2013-08-15 La Jolla Institute For Allergy And Immunology Timothy grass allergens and methods and uses for immune response modulation
WO2014064543A1 (en) 2011-10-26 2014-05-01 Nanopass Technologies Ltd. Microneedle intradermal drug delivery with auto-disable functionality
WO2014188429A1 (en) 2013-05-22 2014-11-27 Nanopass Technologies Ltd. Intradermal delivery of drugs, pharmaceuticals and other therapeutic agents via microneedles

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US5422109A (en) 1989-07-03 1995-06-06 Societe D'exploitation De Produits Pour Les Industries Chimiques (S.E.P.P.I.C.) Fluid vaccines and active principle vehicles containing a metabolizable oil
US6087329A (en) 1991-10-25 2000-07-11 Immunex Corporation CD40 ligand polypeptide
WO1995017210A1 (en) 1993-12-23 1995-06-29 Smithkline Beecham Biologicals (S.A.) Vaccines
WO1996002555A1 (en) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
WO1996033739A1 (en) 1995-04-25 1996-10-31 Smithkline Beecham Biologicals S.A. Vaccines containing a saponin and a sterol
WO1998016247A1 (en) 1996-10-11 1998-04-23 The Regents Of The University Of California Immunostimulatory polynucleotide/immunomodulatory molecule conjugates
WO1998056414A1 (en) 1997-06-11 1998-12-17 Smithkline Beecham Biologicals S.A. Oil in water vaccine compositions
WO1999011241A1 (en) 1997-09-05 1999-03-11 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins
WO1999012565A1 (en) 1997-09-05 1999-03-18 Smithkline Beecham Biologicals S.A. Vaccines
US20070044171A1 (en) * 2000-12-14 2007-02-22 Kovalic David K Nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
EP1403280A1 (en) * 2002-09-27 2004-03-31 BIOMAY Produktions- und Handels- Aktiengesellschaft Hypoallergenic allergy vaccines based on the timothy grass pollen allergen Phl p 7
WO2005049107A2 (en) 2003-11-18 2005-06-02 Nanapass Technologies Ltd. Enhanced penetration system and method for sliding microneedles
WO2006054280A2 (en) 2004-11-18 2006-05-26 Nanopass Technologies Ltd. System and method for delivering fluid into flexible biological barrier
WO2007066341A2 (en) 2005-12-08 2007-06-14 Nanopass Technologies Ltd. Microneedle adapter for dosed drug delivery devices
US20110167514A1 (en) * 2007-07-05 2011-07-07 Ceres, Inc. Nucleotide sequences and corresponding polypeptides conferring modulated plant characteristics
WO2012049310A1 (en) 2010-10-15 2012-04-19 Alk-Abelló A/S Suppression of a hypersensitivity immune response with unrelated antigen derived from allergen source material
WO2012168487A1 (en) 2011-06-09 2012-12-13 Biomay Ag Peptide carrier fusion proteins as allergy vaccines
US20130058978A1 (en) * 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Induced tolerogenic dendritic cells for inducing regulatory b cells
WO2014064543A1 (en) 2011-10-26 2014-05-01 Nanopass Technologies Ltd. Microneedle intradermal drug delivery with auto-disable functionality
WO2013119863A1 (en) 2012-02-07 2013-08-15 La Jolla Institute For Allergy And Immunology Timothy grass allergens and methods and uses for immune response modulation
WO2014188429A1 (en) 2013-05-22 2014-11-27 Nanopass Technologies Ltd. Intradermal delivery of drugs, pharmaceuticals and other therapeutic agents via microneedles

Non-Patent Citations (40)

* Cited by examiner, † Cited by third party
Title
"Archiv. fur die gesamte Virusforschung", vol. 44, SPRINGER VERLAG, article "Saponin adjuvants", pages: 243 - 254
"Pharmaceutical Principles of Solid Dosage Forms", 1993, TECHNONIC PUBLISHING CO., INC.
"Remington's Pharmaceutical Sciences 18th ed.,", 1990, MACK PUBLISHING CO.
"The Merck Index 12th ed.,", 1996, MERCK PUBLISHING GROUP
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL, S.F.; GISH, W.; MILLER, W.; MYERS, E.W.; LIPMAN D.J.: "Basic local alignment search tool", J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ANSEL; SOKLOSA: "Pharmaceutical Calculations 11th ed.,", 2001, LIPPINCOTT WILLIAMS & WILKINS
BOSTICK D; VAISMAN: "A new topological method to measure protein structure similarity", BIOCHEM BIOPHYS RES COMMUN, vol. 304, 2003, pages 320 - 325
BOSTICK ET AL., BIOCHEM BIOPHYS RES COMMUN., vol. 304, 2003, pages 320
C. EBNER ET AL: "Immunological changes during specific immunotherapy of grass pollen allergy: reduced lymphoproliferative responses to allergen and shift from TH2 to TH1 in T-cell clones specific for Phi p 1, a major grass pollen allergen", CLINICAL & EXPERIMENTAL ALLERGY, vol. 27, no. 9, 1 September 1997 (1997-09-01), pages 1007 - 1015, XP055188787, ISSN: 0954-7894, DOI: 10.1111/j.1365-2222.1997.tb01252.x *
C. OSEROFF ET AL: "Molecular Determinants of T Cell Epitope Recognition to the Common Timothy Grass Allergen", THE JOURNAL OF IMMUNOLOGY, vol. 185, no. 2, 15 July 2010 (2010-07-15), pages 943 - 955, XP055023641, ISSN: 0022-1767, DOI: 10.4049/jimmunol.1000405 *
DAVID B. TROY; PAUL BERINGER: "Remington: The Science and Practice of Pharmacy", 2006, LIPPINCOTT WILLIAMS & WILKINS
DR BJOERN PETERS ET AL: "800 A Systematic Analysis Of Pollen Transcriptomes From Plant Allergens Reveals Conserved Targets Of Immune Responses", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 133, no. 2, 28 February 2014 (2014-02-28), pages AB231, XP055188731 *
FEDEROV ALEXANDER A ET AL: "The molecular basis for allergen cross-reactivity: Crystal structure and IgE-epitope mapping of birch pollen profilin", STRUCTURE, CURRENT BIOLOGY LTD., PHILADELPHIA, PA, US, vol. 5, no. 1, 15 January 1997 (1997-01-15), pages 33 - 45, XP002506728, ISSN: 0969-2126, [retrieved on 20040605], DOI: 10.1016/S0969-2126(97)00164-0 *
GREENBAUM J; SIDNEY J; CHUNG J; BRANDER C; PETERS B; SETTE A: "Functional classification of class II human leukocyte antigen (HLA) molecules reveals seven different supertypes and a surprising degree of repertoire sharing across supertypes", IMMUNOGENETICS, vol. 63, 2011, pages 325 - 335
KAROSIENE, EDITA; MICHAEL RASMUSSEN; THOMAS BLICHER; OLE LUND; SOREN BUUS; MORTEN NIELSEN: "NetMHCllpan-3.0, a Common Pan-specific MHC Class II Prediction Method Including All Three Human MHC Class 11 lsotypes, HLA-DR, HLA-DP and HLA-DQ.", IMMUNOGENETICS, vol. 65, no. 10, 2013, pages 711 - 724
MCKINNEY DM; SOUTHWOOD S; HINZ D; OSEROFF C; ARLEHAMN CS; SCHULTEN V ET AL.: "A strategy to determine HLA class II restriction broadly covering the DR, DP, and DQ allelic variants most commonly expressed in the general population", IMMUNOGENETICS, vol. 65, 2013, pages 357 - 70
NEEDLEMAN SB; WUNSCH CD: "A general method applicable to the search for similarities in the amino acid sequence of two proteins", J. MOL. BIOL., vol. 48, 1970, pages 443
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
OSEROFF C; SIDNEY J; KOTTURI MF; KOLLA R; ALAM R; BROIDE DH ET AL.: "Molecular determinants of T cell epitope recognition to the common Timothy grass allergen", JOURNAL OF IMMUNOLOGY, vol. 185, 2010, pages 943 - 55
PEARSON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PEARSON WR; LIPMAN DJ: "Improved tools for biological sequence comparison", PROC. NATL. ACAD. SCI. USA, vol. 85, no. 8, 1988, pages 2444 - 2448
PEARSON WR1: "Flexible sequence similarity searching with the FASTA3 program package", METHODS MOL BIOL., vol. 132, 2000, pages 185 - 219
PEARSON, METHODS MOL BIOL., vol. 132, 2000, pages 185
PEARSON; LIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
POWELL, M. F.; NEWMAN M.: "Pharmaceutical Biotechnology", 1995, PLENUM PRESS, ISBN: 0-306-44867-X, article "Vaccine Design--the subunit and adjuvant approach"
POZNANSKY ET AL.: "Drug Delivery Systems", 1980, pages: 253 - 315
REMINGTON: "The Science and Practice of Pharmacy 20th ed.", 2003, MACK PUBLISHING CO.
SAMBROOK ET AL.: "Molecular Cloning: a Laboratory Manual, 3rd ed.,", 2001, COLD SPRING HARBOUR LABORATORY PRESS
SIDNEY J; ASSARSSON E; MOORE C; NGO S; PINILLA C; SETTE A; PETERS B: "Quantitative peptide binding motifs for 19 human and mouse MHC class I molecules derived using positional scanning combinatorial peptide libraries", IMMUNOME RES., vol. 4, no. 2, 2008
SIDNEY J; SOUTHWOOD S; GREY HM; MOORE C; OSEROFF C; PINILLA C; SETTE A ET AL.: "Current protocols in immunology", 2013, article "Measurement of MHC/peptide interactions by gel filtration or monoclonal antibody capture"
SIDNEY J; SOUTHWOOD S; OSEROFF C; DEL GUERCIO MF; SETTE A; GREY HM: "Curr Protoc Immunol.", 2001, article "Measurement of MHC/peptide interactions by gel filtration"
SIDNEY J; STEEN A; MOORE C; NGO S; CHUNG J; PETERS B; SETTE A: "Divergent motifs but overlapping binding repertoires of six HLA-DQ molecules frequently expressed in the worldwide human population", J IMMUNOL, vol. 185, no. 7, 2010, pages 4189 - 4198
SIDNEY J; STEEN A; MOORE C; NGO S; CHUNG J; PETERS B; SETTE A: "Five HLA-DP molecules frequently expressed in the worldwide human population share a common HLA supertypic binding specificity", J IMMUNOL, vol. 184, no. 5, 2010, pages 2492 - 2503
SMITH ET AL., J. MOL. BIOL., vol. 147, 1981, pages 195
SMITH; WATERMAN, ADV. APPL. MATH, vol. 2, 1981, pages 482
SMITH; WATERMAN: "Comparison of biosequences", ADV. APPL. MATH, vol. 2, 1981, pages 482
V. SCHULTEN ET AL: "Previously undescribed grass pollen antigens are the major inducers of T helper 2 cytokine-producing T cells in allergic individuals", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 110, no. 9, 26 February 2013 (2013-02-26), pages 3459 - 3464, XP055164674, ISSN: 0027-8424, DOI: 10.1073/pnas.1300512110 *
VÉRONIQUE SCHULTEN ET AL: "Association between specific timothy grass antigens and changes in TH1- and TH2-cell responses following specific immunotherapy", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 134, no. 5, 16 July 2014 (2014-07-16), pages 1076 - 1083, XP055164673, ISSN: 0091-6749, DOI: 10.1016/j.jaci.2014.05.033 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023242436A1 (en) * 2022-06-17 2023-12-21 Danmarks Tekniske Universitet Allergy vaccines based on consensus allergens

Similar Documents

Publication Publication Date Title
AU2013216993C1 (en) Timothy Grass allergens and methods and uses for immune response modulation
EP3086806B1 (en) Peptide combinations and uses thereof in treating dust mite allergy
KR20100068252A (en) Peptides for vaccine
KR20120120965A (en) Peptides for vaccine against birch allergy
US9103835B2 (en) Hypoallergenic hybrid polypeptides for the treatment of allergy
US11013781B2 (en) Peptide combinations and uses thereof for treating grass allergy
WO2007065633A1 (en) Modulation of the immune response by administration of intralymphatic transduction allergen (itag) -molecules
WO2015131053A1 (en) Polypeptides derived from phl p and methods and uses thereof for immune response modulation
EP2838556B1 (en) Plant profilin polypeptides for use in non-specific allergy immunotherapy
US11421006B2 (en) T cell epitopes from Cockroach and methods of making and using same
WO2015077442A9 (en) Grass pollen immunogens and methods and uses for immune response modulation
US20160287696A1 (en) Pan pollen immunogens and methods and uses for immune response modulation
US20180291071A1 (en) Novel antigens and t cell epitopes from cockroach and methods of making and using same
WO2016209959A2 (en) Peptide combinations and use thereof in treating cedar pollen allergy
WO2017179025A1 (en) Epitope polypeptides of ragweed pollen allergens
WO2015054217A2 (en) Methods and uses for reducing an allergic response in a subject

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15709065

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15709065

Country of ref document: EP

Kind code of ref document: A1