WO2017179025A1 - Epitope polypeptides of ragweed pollen allergens - Google Patents

Epitope polypeptides of ragweed pollen allergens Download PDF

Info

Publication number
WO2017179025A1
WO2017179025A1 PCT/IB2017/052172 IB2017052172W WO2017179025A1 WO 2017179025 A1 WO2017179025 A1 WO 2017179025A1 IB 2017052172 W IB2017052172 W IB 2017052172W WO 2017179025 A1 WO2017179025 A1 WO 2017179025A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
amino acid
seq
amb
acid sequence
Prior art date
Application number
PCT/IB2017/052172
Other languages
French (fr)
Other versions
WO2017179025A9 (en
Inventor
Hans-Henrik Ipsen
Lars Harder CHRISTENSEN
Ilka HOOF
Carla OSEROFF
Peter Adler WÜRTZEN
Alessandro Sette
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 WO2017179025A1 publication Critical patent/WO2017179025A1/en
Publication of WO2017179025A9 publication Critical patent/WO2017179025A9/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

Definitions

  • the present invention relates to T cell epitope-containing polypeptides derived from ragweed pollen allergens, in particular polypeptides derived from ragweed allergens belonging to group 3, 4, 8 and 11 allergens, and uses of such polypeptides for modulating an immune response, for treating an allergic response and/or for inducing or promoting immunological tolerance, and for use in diagnostic methods and kits.
  • Ragweed pollen is a major source of pollen allergy and may be treated by allergen-specific immunotherapy using full length allergen(s), modifications thereof (hypoallergens) or more conventionally by administering ragweed pollen extracts.
  • allergen-specific immunotherapy using full length allergen(s), modifications thereof (hypoallergens) or more conventionally by administering ragweed pollen extracts.
  • T cell epitope peptide-based immunotherapy could be one option to provide well-tolerated immunotherapeutic methods.
  • soluble polypeptides containing allergen-derived immunodominant T-cell epitopes are used to treat allergic patients. Due to the use of T cell epitope-conta ining polypeptides without IgE-reactivity the chance of adverse effects are reduced significantly.
  • T cell epitopes of the major allergen of Ambrosia artemisiifolia have been studied extensively and peptide cocktails for the treatment of ragweed pollen allergy by PIT have also been suggested (WO10018378, WO08098749, Jahn-Schmid, the T-cell response to Amb a 1 is characterized by 3 dominant epitopes and multiple MHC restriction elements. J Allergy Clin Immunol, Vol.126, No. 5, 1068-1070, 2010).
  • ragweed pollen allergens of Ambrosia artemisiifolia have been identified based on IgE reactivity, for example those reported in the WHO/IUIS Allergen Nomenclature database (http ⁇ www. allergen. org). However, these are often regarded as minor allergens as the number of ragweed allergic individuals with IgE-reactivity towards these allergens are considerable lower than individua ls with IgE-reactivity towards Amb a 1.
  • Artemisiifolia (Amb a; short ragweed), other ragweed species may be relevant too as the geographic presence of other ragweed species is expanding. Other relevant species may be
  • Ambrosia psilostachya (Amb p; western ragweed) and Ambrosia trifida (Amb t; giant ragweed) . Therefore, there is a desire to provide polypeptides for vaccines against ragweed pollen allergy which not only targets the Amb a species, but also the Amb p and Amp t species. It may also be advantageous to provide polypeptides for use in vaccines against related weed pollen like Artemisia vulgaris (Art v; mugwort).
  • polypeptides capable of providing a T cell- mediated immune response in subjects sensitized to at least either Amb a, Amb p, or Amb t, or combinations of Amb a, Amb p and Amb t.
  • the same polypeptides may be able to provide a T cell-mediated immune response in subjects sensitized to an additional weed species, for example Art v.
  • immunodominant regions and epitope polypeptides consisting of a stretch of contiguous amino acids selected from Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a l l .
  • similar immunodominant polypeptides and regions could be derived from homolog allergens identified in Amb t, Amb p or Art v species.
  • such immunodominant epitope polypeptides have great potential as a vaccine for treatment of weed pollen allergy caused by several weed pollen species, including at least Amb a, Amp, Amb t and optionally Art v
  • a first aspect of the invention relates to immunodominant epitope regions identified in the amino acid sequence of the pollen allergens Amb a 4, Amb a 8 and Amb 11 (SEQ ID NO : 106-125) and any corresponding immunodominant epitope region found in allergens homologous to Amb a 4, Amb 8 and Amb 11 in the species Amb p, Amb t and Art v (SEQ ID NOs: 189-224).
  • immunodominant epitope region is defined by the stretch of amino acid residues formed by overlapping polypeptides that have shown high T cell responder frequency in a donor population. For example, 20-mer polypeptides overlapping each other by 10 amino acid residues may result in an epitopic region of 30 amino acid residues.
  • the allergen Amb a 8 there are identified larger dominant epitopic regions, for example defined by four overlapping polypeptides (SEQ ID NOs : 37- 40 of table 4) resulting in the epitopic region with SEQ ID NO: 112 (table 6) .
  • the immunodominant epitope regions are useful for designing immunodominant epitope polypeptides of various length which have high likelihood of inducing a T cell-mediated immune response in ragweed and/or mugwort pollen allergic subjects.
  • the same immunodominant epitope regions may be used for designing fusion proteins comprising a high density of immunodominant regions in comparison to the wild type allergens and/or comprising immunodominant regions from one allergen or from several different allergens.
  • a first aspect of the invention relates to a polypeptide comprising 9, or at least 9, contiguous amino acids selected from any one of SEQ ID NOs: 106- 125 and 189-224; or a variant polypeptide which comprises an amino acid sequence with at least 65% sequence identity to the 9 or at least 9 contiguous amino acids.
  • a polypeptide comprising 9, or at least 9 contiguous amino acids is meant to define that the polypeptide comprises 9 contiguous amino acids or at least 9 contiguous amino acids.
  • a first aspect relates to a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 106-125 and 189-224, optionally wherein the polypeptide is modified by one, two or three amino acid substitutions.
  • a second aspect relates to polypeptides able to induce a T cell response in ragweed allergic subjects, which are derived from either of the allergens; Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a 11; or from homolog allergens identified in pollen of Amb p, Amb t or Art v.
  • a second aspect relates to a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 1-105 (polypeptides derived from Amb a shown in tables 1 to 5) and SEQ ID NOs : 150-182 (polypeptides derived from homolog allergens shown in tables 9 to 11); or a variant thereof which is 9 to 30 amino acids in length and comprises:
  • a second aspect relates to a polypeptide consisting of an amino acid sequence with SEQ ID NOs : 1- 105, wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or the C-terminal end.
  • Examples of polypeptides having one, two or three amino acid substitutions in comparison to SEQ ID NOs : 1- 105 may be selected from any one of SEQ ID NOs 150-182.
  • a third aspect of the invention relates to novel allergens identified in pollen of Amb p, Amp t or Art v, which are considered homologous to Amb a 3, Amb a 4, Amb a 8 and Amb a l l allergens. It has now been shown that these novel allergens share common epitopes with allergens of Amb a.
  • a third aspect of the invention relates to a polypeptide consisting of an amino acid sequence having 85% to 100% sequence identity (i.e. having at least 85% sequence identity) with an amino acid sequence selected from any one of SEQ ID NOs: 126- 146.
  • a fourth aspect of the invention relates to a chimeric molecule comprising a fusion of two or more of the polypeptides selected from any one of the first, second, or third aspect, or a chimeric molecule comprising a fusion of two or more of the amino acid sequences selected from the group consisting of SEQ ID NOs: 106-125 and 189-224, or a chimeric molecule comprising the fusion of two or more of the amino acid sequences selected from the group consisting of SEQ ID NOs: 126- 149.
  • a fifth aspect of the invention relates to a composition, such as a pharmaceutical composition, which comprises one or more of the polypeptides of the first aspect, second aspect, third aspect and/or fourth aspect of the invention.
  • Such compostions may comprise further polypeptides designed from other allergens, for example designed from the major allergen Amb a 1, or from a homolog allergen to Amb a 1 found in Amb p, Amb t or Art v.
  • a sixth aspect of the invention relates to a method of treating ragweed pollen allergy in a subject, comprising administering to the subject a therapeutically effective amount of a polypeptide of the of the first aspect, second aspect, third aspect and/or fourth aspect of the invention or a composition according to a fifth aspect of the invention.
  • a sixth aspect of the invention relates to a polypeptide of the first aspect, second aspect, third aspect and/or fourth aspect of the invention or a composition according to a fifth aspect of the invention for use in treating ragweed pollen allergy in a subject.
  • a sixth aspect of the invention relates to use of a polypeptide of the first aspect, second aspect, third aspect and/or fourth aspect of the invention or a composition according to a fifth aspect of the invention for the preparation of a medicament for use in treating ragweed pollen allergy in a subject.
  • an “epitope” refers to a region or part of an antigen, such as a peptide disclosed herein, that elicits an immune response when administered to a subject.
  • An epitope may be a T cell epitope, i.e., an epitope that elicits, stimulates, induces, promotes, increases or enhances a T cell activity, function or response. For example, such as a Th2 cell epitope.
  • Any peptide or combination of polypeptides of interest can be analyzed to determine whether they 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.).
  • T cell activity can be analyzed using the methods described in detail herein, for example as described in Example 2.
  • the term "allergen” refers to an antigen which elicits, induces, stimulates, or enhances an immune response by a cell of the immune system of an exposed animal (e.g ., human) .
  • An antigen is an allergen when the specific immune response is the development of enhanced sensitivity or a hypersensitivity to the antigen, but the antigen itself is not typically innately harmful.
  • An allergen is therefore a particular type of antigen that can cause development of enhanced or increased sensitivity or hypersensitivity in a subject.
  • an allergen can elicit production of IgE antibodies in predisposed subjects.
  • T cell response refers to an interleukin or a proliferation response by a T cell. It may be determined as explained in Example 2. It may in some instances be referred to simply as a "response” to a peptide.
  • allergic response is intended to refer to the hypersensitive immune reaction to a normally inocuous environmental substance known as an allergen. The most common mechanism of allergic reactions is the binding of IgE to the surface of mast cells, which causes asthma, hay fever and other common allergic reactions.
  • identity and “identical” and grammatical variations thereof, as used herein, mean that two or more referenced entities are the same (e.g . , amino acid sequences) .
  • polypeptides where two polypeptides are identical, they have the same amino acid sequence.
  • the identity can be over a defined area, e.g. over at least 12, 13, 14, 15 or 16 contiguous amino acids of the original peptide or polypeptide, optionally wherein the alignment is the best fit with gaps permitted .
  • 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.
  • a BLASTP algorithm is typically used in combination with a scoring matrix, such as PAM 100, PAM 250, BLOSUM 62 or BLOSUM 50.
  • 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)) .
  • Figure 1 Shows the fraction of donors producing an in vitro T cell immune response (response frequency) towards polypeptides derived from Amb a 3 allergen, Amb a 4 allergen or homolog polypeptides found in other species.
  • Figure 2A Shows the correlation between T cell response frequency of an Amb a 3 polypeptide
  • Figure 2B Shows the correlation between T cell response frequency of an Amb a 4 polypeptide (B0309* (SEQ ID NO : 163) and the homolog polypeptide of Amb p (B0458*, SEQ ID NO: 167) and Art v (B0385*, SEQ ID NO: 165) .
  • Figure 3 Shows the fraction of donors producing an in vitro T cell immune response (response frequency) towards polypeptides of Amb a 8 allergen and homolog polypeptides found in Amb t and Art v.
  • Figure 4 Shows the correlation between T cell response frequency of an Amb a 8 polypeptide (B0311, SEQ ID NO : 168) and homolog polypeptides (B0387 (SEQ ID NO: 169), B0388 (SEQ ID NO : 170), B0389 (SEQ ID NO : 171), B0393 (SEQ ID NO: 172), B0394 (SEQ ID NO : 173)) .
  • Figure 5 Shows the correlation between T cell response frequency of an Amb a 8 polypeptide (B0312, SEQ ID NO : 174) and homolog polypeptides (B0390 (SEQ ID NO : 175), B0391 (SEQ ID NO : 176), B0392 (SEQ ID NO : 177), B0395 (SEQ ID NO : 178), B0396 (SEQ ID NO : 179))
  • Figure 6A Shows the fraction of donors producing an in vitro T cell immune response (response frequency) towards polypeptides of Amb a 11 allergen and homolog polypeptides found in Amb t and Amb p.
  • Figure 6B Shows the correlation between T cell response frequency of an Amb a l l polypeptide (B0234, SEQ ID NO : 83) and homolog polypeptides (B0397, (SEQ ID NO: 180), B0460 (SEQ ID NO : 181)).
  • Figure 7 Shows sequence alignment between Amb a 3.0101 allergen and homologs identified in Amb p (SEQ ID NO : 126), Amb t (SEQ ID NO: 127) and Art v (SEQ ID NO : 128). The figure also shows T cell-epitope containing polypeptides derived from Amb a 3.0101 and its homologs (SEQ ID Nos : 2, 141 and 142).
  • Figure 8 Shows sequence alignment between Amb a 4.0101 allergen and homologs identified in Amb p (SEQ ID NO : 129), and Art v (SEQ ID NO: 147) .
  • the figure also shows dominant epitope regions identified in Amb a 4 and its homolog allergens (SEQ ID NOs: 108, 189 and 190 (sequences indicated in bolded text) and SEQ ID NOs: 109, 191 and 192 (sequences indicated with underlined text) .
  • Figure 9 Shows sequence alignment between Amb a 4.0101 allergen, epitopic regions with SEQ ID NOs : 108, 109 and 110) and T cell-epitope containing polypeptides derived from Amb a 4.0101 and its homologs (SEQ ID Nos: 11, 153, 155, 15, 159, 161, 19, 163, 165 and 167) .
  • Figure 10 Shows sequence alignment between the first part of the Amb a 8.0101 and Amb a 8.0102 allergen and homologs identified in Amb p (SEQ ID NOs: 130- 135), Amb t (SEQ ID NOs: 136- 141) and Art v (SEQ ID NOs: 142- 144) .
  • the figure also shows dominant epitope regions identified in Amb a 8 (SEQ ID NO: 112) and in its homolog allergens (SEQ ID NOs: 193-207 (sequences indicated in bolded text).
  • Figure 11 Shows sequence alignment between the second part of Amb a 8.0101 and Amb a 8.0102 allergen and homologs identified in Amb p (SEQ ID NOs: 130- 135), Amb t (SEQ ID NOs: 136- 141) and Art v (SEQ ID NOs: 142- 144) .
  • the figure also shows dominant epitope regions identified in Amb a 8 (SEQ ID NO: 116) and in its homolog allergens (SEQ ID NOs: 208-218 (sequences indicated in bolded text).
  • Figure 12 Shows sequence alignment between epitopic region of Amb a 8 having SEQ ID NO: 112, and T cell-epitope containing polypeptides derived from Amb a 8 and its homologs (SEQ ID Nos: 44, 168-173, 55 and 174- 179) .
  • Figure 13 Shows sequence alignment between Amb a 11.0101 and homologs identified in Amb t (SEQ ID NO : 145), and Art v (SEQ ID NO: 146). The figure also shows dominant epitope regions identified in Amb a 11 (SEQ ID NO: 120, 121, 121) and in its homolog allergens (SEQ ID NOs : 219, 220 (sequences indicated with underlined text), SEQ ID NOs: 221 and 222 (sequences indicated with bolded text) and SEQ ID NOs : 223 and 224 (sequences indicated with underlined text).
  • epitopic regions of Amb a 3, 4, 5, 8 and 11 allergens (SEQ ID NOs: 106- 125, shown in table 6) .
  • the corresponding epitopic regions identified in allergens of Amb p, Amp t and Art v shown in table 7 have also been identified by performing sequence alignments between the Amb a epitopic regions and the homolog allergens of table 7. Sequence alignments are shown in figures 7-13 together with the epitopic regions of homolog allergens, which are indicated as bolded or as underlined text.
  • These epitopeic regions have SEQ ID NOs: 189-224.
  • a first aspect of the invention relates to a polypeptide comprising 9 or at least 9 consecutive amino acids selected from an amino acid sequence consisting of any one of SEQ ID NOs : 106 to 125 and 189-224; or a variant thereof which comprises an amino acid sequence with at least 65% sequence identity, such as at least 70%, 75%, 80%, 85%, 90% or 95% sequence identity to the 9 or the at least 9 consecutive amino acids.
  • the polypeptide comprises 12 to 15 continguous amino acids such as 12, 13, 14, 15 contiguous amino acids, or 15 to 30 contiguous amino acids such as 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 contiguous amino acids, or 30 to 40 contiguous amino acids, such as 30, 32, 35, 37 or 40 contiguous amino acids; or the polypeptide comprises at least 12 to 15, such as 12, 13, 14, 15, or at least 15 to 30 such as 15, 16, 17, 18, 19, 20, 21, 22,23, 24, 25, 26,27, 28,29, or 30 contiguous amino acids, or at least 30 to 40 contiguous amino acids, such as 30, 32, 35, 37, or 40 contiguous amino acids.
  • the variant comprises an amino acid sequence with at least 65% sequence identity (for example at least 70%, 75%, 80%, 85%, 90% or 95%) to the 12, 14, 15, 16, 17, 18, 20, 22, 24, 26, 28, 30, 35 or 40 contiguous amino acids or the at least 12, 14, 15, 16, 17, 18 or 20, 22, 24, 26, 28, 30, 35 or 40 contiguous amino acids.
  • the polypeptide comprises 15 or at least 15 contiguous amino acids selected from any one of SEQ ID NOs: 106- 125 and 189-224; or a variant thereof which comprises an amino acid sequence with at least 80% sequence identity (for example at least 85%, 90% or 95%) to the 15 or the at least 15 contiguous amino acids.
  • the polypeptide comprises at most 30 contiguous amino acids, for example at most 28, 26 or 24 contiguous amino acids.
  • the polypeptides is 15- 30 amino acids in length, for example 16-28 amino acids in length, for example 18-26 amino acids in length, wherein the sequence of the polypeptide consists of a portion of a epitopic region selected from any one of SEQ ID NOs: 106- 125 and 189-224.
  • a variant thereof may have at least 80% sequence identity, for example at least 85%, 90% or 95% to the original polypeptide. For a peptide having the length of 20 amino acids, this sequence identity would apply to about one to three amino acid substitutions in the original polypeptide.
  • a first aspect also relates to a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 106- 125 and 189-224, optionally wherein the polypeptide is modified by one, two or three amino acid substitutions.
  • a polypeptide consists of:
  • Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 108, 189 and 190 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 10, 11, 152- 157, 15 and 158- 161.
  • Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 109, 191 and 192 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 18, 19 and 162- 167.
  • Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 112 and 193-207 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 44, 168-173, 55 and 174- 179.
  • Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 116 and 208-218 is SEQ ID NO : 119.
  • Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 121, 221 and 222 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 83, 180 and 81.
  • Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 122, 222 and 223 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 89 and 182. Further polypeptides may be designed from the above-mentioned epitope regions with SEQ ID NOs : 106-125 and 189-224.
  • a polypeptide consists of 16-28 contiguous amino acids of the epitopic region, for example 18-28 contiguous amino acids, 18-26 contiguous amino acids or 18-24 contiguous amino acids, such as 18, 19, 20, 21, 22, 23 or 24 contiguous amino acids, of an epitope region with SEQ ID NOs: 106- 125 and 189-224.
  • the further designs may be made allowing one, two or three amino acid substitutions in the amino acid sequence of the polypeptide.
  • Embodiments of the second aspect have identified epitope polypeptides able to induce proliferation of T cells of ragweed allergic donors, wherein the polypeptides consists of smaller portions of the amino acid sequence of the naturally occurring allergens : Amb a 3, Amb a 4, Amb a 5, Amb a 8 and Amb a 11.
  • the inventors have identified corresponding polypeptides in allergens which are homologous to Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a l l .
  • Such polypeptides likely contain at least one T cell epitope, optionally a Th2 cell epitope.
  • the epitope polypolypeptides comprise about 20 amino acid residues in length and consists of an amino acid sequence selected from any one of SEQ ID NOs: 1- 105 and 150-182. Sequences of polypeptides and T cell responder frequency are shown in the following tables: ⁇ Table 1 : Shows polypeptides of a group 3 allergen of the species Amb a (SEQ ID NOs : 1-9)
  • Table 2 Shows polypeptides of a group 4 allergen of the species Amb a (SEQ ID NOs : 10-26)
  • Table 3 Shoes polypeptides of a group 5 allergen of the species Amb a (SEQ ID NOs: 27-32)
  • Table 4 Shows polypeptides of a group 8 allergen of the species Amb a a (SEQ ID NOs: 33- 67)
  • Table 5 Shows polypeptides of a group 11 allergen of the species Amb a (SEQ ID NOs: 68- 105).
  • Table 8 Shows polypeptides designed from allergens which are homologous to Amb a 3
  • Table 9 Shows polypeptides designed from allergens which are homologous to Amb a 4
  • Table 10 Shows polypeptides designed from allergens which are homologous to Amb a 8
  • Table 11 Shows polypeptides designed from allergens which are homologous to Amb a l l allergen (SEQ ID NOs: 180-182).
  • polypeptides having 20-26 amino acid resides in length SEQ ID NOs : 1-105 and 150- 182
  • Polypeptides of sizes shorter than 30 amino acids in length are thought to be more water soluble and less prone to aggregation than longer ones.
  • Very short polypeptides, for examples having less than 16 amino acids in length may be disadvantageous in terms of containing less epitopes than the longer ones, and prone to losing biological acitivy due to enzymatic degradation.
  • the inventors have found immunodominant polypeptides capable of inducing T cell proliferation in a high fraction of a ragweed allergic population, e.g. in above 25%, 30%, 35%, 40%, 45%, 50%, or above 60% of the allergic population.
  • a polypeptide with SEQ ID NO : 2 polypeptide of Amb_a_3.0101
  • polypeptide of Amb_a_4.0101 optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue
  • a polypeptide with SEQ ID NOs: 30 polypeptides of Amb_a _5.0101, optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue
  • a polypeptide with any one of SEQ ID NOs: 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64 or 66 polypeptide of Amb_a_8.0101, optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue
  • a polypeptide with SEQ ID NOs: 50 or 61 polypeptide of Amb_a_8.0102, optionally wherein a cysteine residue of the wild type sequence has been replaced with a serine residue
  • a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs : 2, 11, 13, 19, 21, 24, 25, 30, 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64, 66, 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105; or a variant thereof which is 9 to 30 amino acids in length and comprises:
  • a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64 and 66, such as SEQ ID NOs : 42, 44, 46 and 55; or a variant thereof which is 9 to 30 amino acids in length and comprises:
  • immunodominant polypeptides could be derived from Amb a l l.
  • polypeptides with any one of SEQ ID NOs: 71, 73, 76, 78, 82, 83, 88, 89, 90, 93, 95, 98, 100 and 105 are considered more immunodominant than the remaining polypeptides of table 5.
  • Polypeptides with any one of SEQ ID NOs: 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105 produced even higher responder frequencies and polypeptides with any one of SEQ ID NOs : 83, 93, 98 and 105 produced the highest responder frequencies.
  • a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 71, 73, 76, 78, 82, 83, 88, 89, 90, 93, 95, 98, 100 and 105, such as SEQ ID NOs: 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105, such as SEQ ID NOs: 83, 93, 98 and 105; or a variant thereof which is 9 to 30 amino acids in length and comprises:
  • a polypeptide consists of an amino acid sequence with SEQ ID NOs : 1- 105, wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at N- and/or C-terminal end.
  • polypeptides with one, two or three amino acids substitutions in the sequence selected from any one of SEQ ID NOs: 1-105 may be designed from the amino acid sequence of homolog allergens present in ragweed pollen of Amb p, Amb t or Art v.
  • homolog allergens may be selected from any one of SEQ ID NOs: 126- 149 (shown in table 7) .
  • a polypeptide with one, two or three amino acids substitutions in the sequence selected from any one of SEQ ID NOs: 1- 105 is selected from the group consisting of SEQ ID NOs : 150- 182.
  • polypeptides capable of inducing T cell-mediated immune responses in a high fraction of ragweed pollen allergic subjects and with high cross reactivity between polypeptides designed from source allergens from Amb a and their homolog counterparts of Amb t, Amb p and Art v Examples of such polypeptides designed from Amb a 4 and homolog allergens are shown in figures 8-9 and table 2.
  • a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 10-15, 18-21 and 24-25, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end .
  • the additional amino acid residues are from a source allergen selected from any one of SEQ ID NO: 184, 129 and 147.
  • a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 11 is a polypeptide having an amino acid sequence of SEQ ID NOs: 10, 153, 155, or 157.
  • a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 11, 10, 153, 155 and 157.
  • a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 15 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NOs : 14, 159 or 161.
  • a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 14, 15, 159 and 161.
  • a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 19 is a polypeptide having an amino acid sequence of SEQ ID NOs: 18, 163, 165, or 167.
  • a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 18, 19, 163, 165 and 167.
  • Examples of immunodominant and cross reactive polypeptides designed from Amb a 8 or homolog allergens are shown in figures 10- 12 and table 4.
  • Examples of T cell responder frequencies are shown in figures 3 and 4 and examples of the cross reactivity between Amb a, Amb p, Amb t and Art v polypeptides are shown in figures 4 and 5.
  • a polypeptide consists of an amino acid sequence selected fro any one of SEQ ID NOs: 37-40, 42-51, 53-56, 61 and 63, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end.
  • the additional amino acid residues are from a source allergen selected from any one of SEQ ID NOs: 186 and 130-144.
  • the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 44 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 168- 173.
  • a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 44 and 168-173.
  • the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 55 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 174- 179.
  • a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 55,174- 179.
  • Examples of immunodominant and cross reactive polypeptides designed from Amb a 11 or homolog allergens are shown in figures 13- 14 and table 5.
  • a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 76-78, 81-83, 88, 89, 92-95, 99, 100 and 105, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues, and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end.
  • the additional amino acid residues are from a source allergen selected from any one of SEQ ID NOs: 188, 145 and 146.
  • the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 83 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 180 or 181.
  • a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 83, 180 and 181.
  • the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 89 is a polypeptide having an amino acid sequence with SEQ ID NO : 182.
  • a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NO: 2, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end .
  • the additional amino acid residues are from a source allergen selected from any one of SEQ ID NOs: 183, 126- 128.
  • the polypeptide which consists of an amino acid sequence selected from any one of SEQ ID NO : 2 is a polypeptide having an amino acid sequence of SEQ ID NO: 141 or 142.
  • the polypeptide may be elongated with one to five additional amino acid residues added at the N- and/or the C-terminal end of the polypeptide.
  • the elongation may be up to ten amino acids if the polypeptide is elongated with five amino acids at both the N- and C-terminal end.
  • the one to five additional amino acid residues may be contiguous amino acids of an amino acid sequence present in the source allergen from which the polypeptide is designed / derived .
  • the polypeptide may be elongated with one to four additional amino acid residues, for example one, two or three additional amino acid residues, optionally wherein the additional amino acid residues may be contiguous amino acids of an amino acid sequence of the source allergen from which the polypeptide is designed.
  • the polypeptide of SEQ ID NO: 44 which consists of 20 amino acid residues, may be elongated in the C- and/or N- terminal end with one to five amino acid residues selected from the allergen Amb_a_8.0101.
  • it may be elongated by one to five contiguous amino acid residues positioned adjacent to the amino acid sequence of SEQ ID NO : 44, when the sequence of SEQ ID NO : 44 is aligned with the best fit to the sequence of Amb_a_8.0101.
  • polypeptides of the invention may be designed from any one of the polypeptides with SEQ ID NOs: 1- 105 and 150 and 182, by incorporating one, two or three amino acid substitutions in the sequence.
  • further polypeptides of the invention may be designed by elongation of the amino acid sequence of the polypeptide in the N- and/or C-terminal end, including polypeptides with one, two or three amino acid substitutions in the sequence, or by shortening the polypeptide, including those with one, two or three amino acid substitutions in the sequence, with one, two or three amino acid residues from the N- and/or C-termina l end.
  • the inventors have identified homolog allergens of Amb a 3, 4, 8 and 11 in other ragweed pollen species of the genus Ambrosia, i.e. Amb t, Amb p and Art v.
  • the amino acid sequences of these novel full length allergens are shown in table 7. Therefore, some embodiments relate to a polypeptide consisting of an amino acid sequence having 85% to 100% sequence identity with an amino acid sequence selected from any one of SEQ ID NOs : 126-146.
  • the sequence identity is in the range of 90% -100%, for example in the range of 92% - 100%, 95-100%, 97- 100%, 98- 100%, or 99- 100%.
  • the polypeptide has an amino acid sequence which is identical with any one of SEQ ID NOs: 126- 146, for example the sequence identity may be in the range of 85-99%, for example in the range of 90-99%, 95-99%, 96-99%, 97-99%, 98-99% compared to a polypeptide selected from any one of SEQ ID NOs: 126- 146.
  • Polypeptides having a non-identical amino acid sequence may have the same biological activity or functionality as the original polypeptide.
  • the same biological activity or functionality means the same enzymatic functionality; the same, greater or less ability to elicit, stimulate or induce an an in vitro or ex vivo T cell proliferation or T cell cytokine production (e.g. cytokines is selected from IL-4, IL- 5, IL-13 and/or IL- 10) of T cells from subjects allergic to ragweed pollen (e.g . ragweed pollen of the species Amb a, Amp p, Amb t or Art v; the same, greater or less ability to induce
  • polypeptides homologous to Amb a 3 is selected from any one of SEQ ID NOs: 126- 128.
  • polypeptide which is homologous to Amb a 4 is SEQ ID NO : 129
  • polypeptides homologous to Amb a 8 is selected from any one of SEQ ID NOs: 130- 144.
  • polypeptides homologous to Amb a 11 is selected from any one of SEQ ID NOs: 145- 146.
  • polypeptides may be a recombinant polypeptide, thus produced by recombinant manufacturing process and optionally comprising post-translational modifications, such as for example
  • glycosylation phosphorylation
  • lipidation lipidation and/or formation of disulfide bonds.
  • a third aspect of the invention also relates to an isolated nucleic acid molecule, which comprises a nucleotide sequence encoding a polypeptide of the third aspect, for example the nucleic acid molecule is a DNA or an RNA molecule.
  • a vector comprising the nucleic acid molecule, such as a cloning vector or an expression vector, or example a virus, such as an attenuated virus, a bacteriophage, a plasmid, a minichromosome, and a cosmid.
  • a cell which is transformed to carry the vector for example a prokaryotic cell or an eukaryotic cell.
  • a further embodiment relates to a cell line derived from the transformed cell.
  • a further aspect relates to a method for the preparation of a polypeptide of the third aspect comprising
  • Polypeptides of the first and second aspect may be subject to further modifications in order to increase or decrease physical or chemical properties, for example to decrease its ability to resist oxidation, to improve or increase solubility in aqueous solution, to decrease aggregation, to decrease synthesis problems, etc.
  • Non-limiting examples of one or more of the modifications may be selected from the following : a) N-terminal acetylation or methylation
  • a polypeptide according to the first, second or third aspect may elicit, stimulate or induce an in vitro or ex vivo T cell proliferation response or an in vitro or ex vivo T cell cytokine (e.g. IL-4, IL-5, IL- 13 and/or IL- 10) production from T cells of subjects allergic to ragweed pollen (e.g. ragweed pollen of the species Amb a, Amp p, Amb t or Art v) .
  • ragweed pollen e.g. ragweed pollen of the species Amb a, Amp p, Amb t or Art v
  • a polypeptide selected from any one of SEQ ID NOs: 1-105 and 150- 182 is in the present context regarded as an original polypeptide that may be subject to one or more modifications as described herein.
  • a polypeptide designed from the epitope regions of SEQ ID NOs: 106- 125 and 189-224 may also be regarded as original polypeptides subject to modifications as described herein.
  • a polypeptide comprising 9 or at least 9 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 106 to 125 and 189-224 may also be regarded as original polypeptides subject to modifications as described herein.
  • the modification of an original polypeptide may optionally result in greater or less activity or function of the original polypeptide, for example in the ability to elicit, stimulate or induce an in vitro immune response (e.g . T cell proliferation or T cell cytokine production) ; in the ability to bind to Class HLA II alleles (HLA coverages) ; in the ability to induce or enhance immunological tolerance to a relevant allergen, such as Amb a 1, Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a 11 or any combination of the allergens including an allergen extract of ragweed pollen of any species selected from Amb a, Amb t and Amb p; or in the ability to dissolve e.g. in an aqueous solution; or in the ability to aggregate, such as fibrillate.
  • an in vitro immune response e.g . T cell proliferation or T cell cytokine production
  • HLA coverages Class HLA II alleles
  • a relevant allergen such
  • a polypeptide having an amino acid sequence which is non-identical to an original peptide may share about the same functionality or activity as the original polypeptide, e.g . provide slightly poorer functionality or activity, or may have improved functionality or activity.
  • the non-identical polypeptide may bind to at least 70%, such as at least 75%, 80%, 85%, 90% or 95% of the group of Class HLA II alleles that the original peptide binds, optionally wherein the group of HLA Class II alleles is selected from alleles set out in tables 12 or 13.
  • the ability of the original peptide and the non-identical polypeptide to bind HLA Class II alleles may be tested under the same test conditions, for example by use of HLA binding prediction tools or in-vitro binding assay.
  • a non-identical polypeptide may comprise one or more of the same T cell epitopes as the original polypeptide. 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 non-identical polypeptide versus the original polypeptide, 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)
  • cytokines e.g cytokines, IL-5, IL- 13 and/or IL- 10.
  • modifications of the original polypeptide correspond to naturally occurring variations of the wild type a llergens, such as variations between isoforms (intra species variation) of the same species or variations between homologous allergens of other ragweed species (inter species variation). Other modifications are non-natural modifications of the parent peptide.
  • a non-identical polypeptide may for instance include one or more deletions of amino acid residues from the N- and/or C- terminal end of the original polypeptide, one or more additions of amino acid residues to the N- and/or C- terminal of the original peptide and/or one or more amino acid substitutions, additions or deletions within the amino acid sequence of the original peptide, thus resulting in various lengths of the polypeptide.
  • the non-identical polypeptide may be 12 amino acids in length, such as 14, 15, 16, 17, 18, 19, 20, 22, 24, 26, 28 or 30 amino acids in length, or even longer, such as 35, 40, 45, 50, 55 or 60 amino acids in length.
  • the non-identical polypeptide is 15 to 30 amino acids in length, such as 15 to 26, such as 16 to 30, 16 to 28, 16 to 26, or 16 to 24, such as 18 to 30, 18 to 28, 18 to 26, or 18 to 24.
  • a non-identical polypeptide may comprise i) the entire amino acid sequence set forth in any one of SEQ ID NOs : 1- 105 and 150-182 or ii) an amino acid sequence with at least 65% identity to any one of SEQ ID NOs: 1- 105 and 150- 182, such as at least 70%, 75%, 80%, 85%, 90% or 95% sequence identity to any one of SEQ ID NOs: 1- 105 and 150-182.
  • a non-identical polypeptide may comprise 9 or at least 9 contiguous amino acid amino acids selected from any one of SEQ ID NOs : 1-105 and 150- 182, such as 12, 14, 15, 16, 17, 18 or 19 contiguous amino acids selected from any one of SEQ ID NOs: 1-105 and 150- 182, or such as at least 12, 14, 15, 16, 17, 18 or 19 contiguous amino acids selected from any one of SEQ ID NOs: 1-105 and 150- 182.
  • Still further non-identical polypolypeptides may comprise an amino acid sequence with at least 65% sequence identity, such as at least 75%, 80%, 85%, 90% or 95% sequence identity to the 12, 14,
  • the polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 1-105 and 150- 182; or a variant thereof, which has a length of 15 to 30 amino acids and comprises:
  • the variant is 17 to 30 amino acids in length and comprises: i) an amino acid sequence selected from any one of SEQ ID NOs: 1- 105 and 150- 182;
  • a variant or substitutions in the original polypeptide comprises conservative or non-conservative amino acid substitutions.
  • a "conservative substitution” is the replacement of one amino acid by a biologically, chemically or structurally similar residue.
  • Biological similarity means that the substitution does not destroy a biological activity, e.g. T cell reactivity or HLA coverage.
  • Structural similarity 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 both 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, for example 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
  • 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 acid for aspartic acid, 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, He, 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.
  • a polypeptide of the first and second aspect may be provided in the form of a salt, for example as a pharmaceutically acceptable and/or a physiologically acceptable salt, e.g. an acetate salt.
  • Polypeptides of the first, second or third aspect may be produced by peptide synthesis (e.g the polypeptide is synthetic), whereas polypeptides of the third aspect may also be produced by recombinant techniques.
  • a polypeptide of the first, second and third aspect may be provided as a powder, for example as a freeze-dried (lyophilized) powder.
  • polypeptide may be provided in a storage-stable form with low presence of water (e.g. below 10%, for example below 5, 4, 3, 2 or 1% of water) or in a form ready to be administered in dry form (e.g . tablet or capsule) or to be administered in soluble form (e.g. dissolved in aqueous solution) .
  • the concentration of each of the dissolved polypeptides may be in a molar concentration in the range of 1 to 1000 ⁇ , for example in the range of 10 to 800 ⁇ , for example in the range of 20 to 500 ⁇ , for example in the range of 20 to 300 ⁇ .
  • a variant may comprise a fusion (chimeric) sequence of
  • polypolypeptides which optionally may contain an amino acid sequence having one or more molecules not normally present in a reference (wild type, original) sequence covalently attached to the polypeptide 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 second heterologous sequence i.e. a heterologous functional domain
  • a heterologous functional domain is attached to a polypeptide disclosed herein, (covalent or non- covalent binding) that may confer a distinct or complementary function to a polypeptide disclosed herein.
  • Heterologous functional domains are not restricted to amino acid residues.
  • a heterologous functional domain can consist of any one of a variety of different types of small or large functional moieties.
  • moieties include nucleic acid, polypeptide, carbohydrate, lipid or small organic compounds, such as a drug (e.g . an antiviral), a metal (gold, silver), or a
  • a fourth aspect of the invention relates to a chimeric molecule comprising the fusion of two or more of the polypeptides selected from any one of the first, second, or third aspect.
  • the chimeric molecule comprises the fusion of two or more of the amino acid sequences selected from any of SEQ ID NOs: 1- 105 and 150-182. In some embodiments of the invention, the chimeric molecule comprises the fusion of two or more of the amino acid sequences selected from any of SEQ ID NOs: 106-125 and 189-224.
  • the chimeric molecule comprises the fusion of two or more of the amino acid sequences selected from any of SEQ ID NOs: 126-149.
  • the amino acid sequence of the chimeric molecule may comprise one or more molecules covalently attached to the 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.
  • Covalently linking may involve direct linking through the amino acid backbone, in which case two polypeptides are linked by connecting, either directly or by using a spacer amino acid sequence, the N-terminal of one polypeptide to the C-terminal of another polypeptide, which produces a fusion or mosaic protein.
  • Other types of covalent linking may involve the formation of disulfide bonds formed by cysteine amino acids, or other types of covalent bonds between one entity of polypeptide to a second entity of polypeptide.
  • the covalently link is through the amino acid backbone of a polypeptide to a heterologous functional domain.
  • the covalently link is through a cysteine side chain of a polypeptide a heterologous functional domain.
  • Spacers such as amino acid or peptidomimetic sequences, may be inserted between the polypeptide sequence and the addition (e.g., heterologous functional domain) so that the two entities maintain, at least in part, a distinct function or activity.
  • Spacers 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.
  • the polypeptides of the first, second or third aspect are not provided as individual entities, but may be fused together or to a carrier molecule to form an isolated molecule.
  • Such carriers may contribute to presenting the epitopes of the polypeptides of first, second and third aspect to the immune system of the subject to be treated.
  • a fourth aspect of the invention relates to a polypeptide of the first, second or third aspected linked covalently or non-covalently to a component or carrier, for example to various other components, such as proteins capable of forming virus like particles (VLPs), keyhole limpet hemocyanin (KLH), invariant chain or other proteins or polypeptides, which constitute a virus like particles (VLPs), keyhole limpet hemocyanin (KLH), invariant chain or other proteins or polypeptides, which constitute a
  • heterologous functional domain that may or may not be derived from other species than plants.
  • components or carriers are nanoparticles, microparticles or liposomes.
  • one or more amino acid sequence(s) selected from any of SEQ ID NOs : 1- 105 and 150-182 is linked covalently or non-covalently to a component or carrier.
  • one or more amino acid sequence(s) selected any of SEQ ID NOs: 106-125 and 189-224 is linked covalently or non-covalently to a component or carrier.
  • one or more amino acid sequence(s) selected from any of SEQ ID NOs : 126-149 is linked covalently or non-covalently to a component or carrier.
  • polypeptides of the first, second and third aspect are linked to a VLP.
  • the preparation of VLPs is performed by recombinantly expressing the coat protein in a host is within the common knowledge of a skilled artisan.
  • the virus-like particle comprises, or alternatively consists of, recombinant proteins, mutants or fragments thereof, of a virus selected form the group consisting of: a) RNA phages; b)
  • Hepatitis B virus preferably its capsid protein (Ulrich, et al, Virus Res. 50 : 141- 182 (1998)) or its surface protein (WO 92/11291), such as described in WO2012168487; d) measles virus (Warnes, et al., Gene 160: 173- 178 ( 1995)) ; e) Sindbis virus; f) rotavirus (US 5,071,651 and US 5,374,426) ; g) foot-and-mouth-disease virus (Twomey, et al., Vaccine 13 : 1603 1610, (1995)) ; h) Norwalk virus (Jiang, X., et al.
  • the virus-like particle is of Hepatitis B virus.
  • the preparation of Hepatitis B virus-like particles have been disclosed, inter alia, in WO 00/32227, WO 01/85208, WO 02/056905 and WO2012168487.
  • the virus-like particle is a virus of the pi-cornaviridae family such as a species of rhinoviruses, such as rhinovirus 89 and 14, in which case the virus-like particle may be formed by one or more of the proteins selected from VP1, VP2, VP3 and/or VP4 as described in WO2006097530.
  • the virus-like particle is a virus-like (VLP) particle derived from RNA bacteriophage AP205 such as described in WO0603267.
  • fusion proteins are well known in the art and can be found in standard molecular biology references such as Sambrook et al. (Molecular Cloning, 2nd ed. , Cold Spring Harbor Laboratory Press, 1989) and Ausubel et al . (Short Protocols in Molecular Biology, 3rd ed; Wiley and Sons, 1995).
  • a fusion protein is produced by first constructing a fusion gene which is inserted into a suitable expression vector, which is, in turn, used to transfect a suitable hosT-cell.
  • recombinant fusion constructs are produced by a series of restriction enzyme digestions and ligation reactions which result in the desired sequences being incorporated into a plasmid .
  • oligonucleotide adapters or linkers can be used as is known by those skilled in the art and described in the references cited above.
  • the polynucleotide sequences encoding allergens and native proteins can be assembled prior to insertion into a suitable vector or the sequence encoding the allergen can be inserted adjacent to a sequence encoding a native sequence already present in a vector. Insertion of the sequence within the vector should be in frame so that the sequence can be transcribed into a protein. It will be apparent to those of ordinary skill in the art that the precise restriction enzymes, linkers and/or adaptors required as well as the precise reaction conditions will vary with the sequences and cloning vectors used. The assembly of DNA constructs, however, is routine in the art and can be readily accomplished by a person skilled in the art.
  • proteins or polypeptides of the present invention are linked either covalently or non-covalently to nanoparticles, microparticles or liposomes, for example such as described in WO07068747, WO07144150 and WO08071774.
  • Embodiments of the fifth aspect are described in WO07068747, WO07144150 and WO08071774.
  • one or more of the polypolypeptides of the first, second or third aspect may be comprised in a composition, optionally in combination with polypolypeptides designed from other allergens.
  • the composition may further comprise other excipients.
  • the invention relates in a fifth aspect to a composition, such as a pharmaceutical composition, which comprises one or more of the polypeptides of the first, second or third aspect.
  • a pharmaceutical composition typically comprises a pharmaceutically acceptable carrier, excipient and/or adj uvant and may optionally be sterile.
  • composition such as a pharmaceutical composition may comprise one or more of the
  • polypeptides with high T cell responder frequencies and cross reactivity are polypeptides with high T cell responder frequencies and cross reactivity.
  • a compostion may further comprise one or more immunodominant polypeptides from a group 1 allergen of ragweed pollen.
  • Such polypeptides may comprise from 9 to 30 contiguous amino acids of a sequence of Amb a 1, for example of a sequence of selected among any one of the isoforms Amb_a_1.0201(UniProt No : P27760), Amb_a_1.0202(UniProt No : E1XUL3), Amb_a_1.0301(UniProt No : P27761), Amb_a_1.0302 (UniProt No: P27761(variantL48Y)), Amb_a_1.0303(UniProt No: P27761(variantH392R), Amb_a_1.0304(UniProt No : E1XUL4), Amb_a_1.0305(UniProt No:
  • the isoforms have the following sequences (SEQ ID NOs: 225-236:
  • the pharmaceutical composition may be a vaccine, e.g . a product for use in conducting
  • immunotherapy including but not limited to a vaccine for treating an allergic immune response to ragweed pollen, i.e. pollen of the genus Ambrosia such as pollen of Amb a, Amb p and/or Amb t.
  • the vaccine may be formulated for parenteral administration, such as by subcutaneous, intradermal, transcutaneous administration, e.g. formulated as a powder that optionally may be re- dissolved before use.
  • a pharmaceutical composition comprises in addition to the polypeptide combination, therapeutically inactive ingredients, such as pharmaceutically acceptable or physiologically acceptable excipient(s), carrier(s) and/or adjuvant(s), which 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 adj usting agents, buffering agents, pH adjusting agents, isotonicity adj usting agents, mucoadhesive substances, and the like. Examples of formulation strategies are well-known to the person skilled in the art.
  • the polypeptide(s) may be formulated (e.g. mixed together) with immune- modifying agents like adj uvants usually applied in immunotherapy products.
  • 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.
  • a liquid pharmaceutical composition may be formed by dissolving a powder, granulate or lyophilizate of a peptide 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 peptide 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 purpose 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.
  • the concentration of each polypeptide is the same, such as in an equimolar concentration, but each polypeptide of the composition may also 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, i.e. water for injection.
  • 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 pH 5 to 8, such as in the range of pH 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 adj usted with dilutions of strong base (e.g. sodium hydroxide or the like) and/or dilutions of strong acids (e.g . hydrochloric acid) .
  • a pH controlling agent or buffering agent e.g. citrate buffer, phosphate buffer, acetate buffer
  • strong base e.g. sodium hydroxide or the like
  • strong acids e.g . hydrochloric acid
  • the liquid formulation is isotonic, and optionally sterile. Therefore, in some embodiments, the formulation comprises saline or glycerol, such as isotonic saline.
  • the liquid may contain additional excipients, such as another solvent, a solubilizing enhancing agent, ionic and non-ionic emulsifiers, a dispersant, a thickener, a preservative, an anti-microbial agent, and/or an antioxidant.
  • solvents include water, saline, DMSO, glycerol, ethanol, acetonitrile, vegetable or synthetic oils.
  • 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 a pH in the range of 3-8, such as a pH in the range of 4-8.
  • a lyophilizate may contain additional ingredients, e.g. bulking agents and lyoprotectants, buffering, antioxidants, antimicrobial agents, solubilizers.
  • 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 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.
  • 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.,
  • 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 intra lymphatic administration.
  • the pharmaceutical composition comprises an inert gas, e.g. argon or nitrogen.
  • kits comprising a compartment and instructions, wherein the compartment comprises a composition described herein and wherein the instructions are for use in treating allergy to ragweed pollen.
  • 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 WO14064543 Al, WO05049107 A2, WO06054280 A2,
  • a polypeptide of the first, second, third aspect or a chimeric molecule/ polypeptide linked to a carrier of the fourth aspect or a composition of the fifth aspect may be used in treating ragweed pollen allergy in a subject.
  • a sixth aspect of the invention relates to a method for treating ragweed pollen allergy in a subject, for example to treating allergy in a subject where signs and/or symptoms of said allergy are elicited in the subject by exposure to ragweed pollen and/or exposure to at least one allergen present in ragweed pollen, wherein the method comprises administering to the subject a therapeutically effective amount of a polypeptide of the first, second, third aspect or a chimeric molecule/ polypeptide linked to a carrier of the fourth aspect or a composition of the fifth aspect.
  • the treatment of allergy may comprise relieving or reducing (e.g.
  • the treatment of allergy may comprise or consist of relieving one or more signs and/or symptoms of an immune response triggered by exposure to ragweed pollen or at least one allergen of ragweed pollen;
  • the treatment of allergy may consist of or comprises inducing immunological tolerance ragweed pollen or at least one allergen of ragweed pollen; or the treatment of allergy may comprise or consist of relieving one or more signs and/or symptom(s) associated with allergic rhinitis and/or allergic conjunctivitis and/or allergic asthma and/or allergic eczema (e.g. atopic dermatitis).
  • Polypeptides and compositions disclosed herein may produce a therapeutic or beneficial effect, which optionally may be objectively or subjectively measurable.
  • a therapeutic or beneficial effect can but need not be complete ablation of all or any immune responses, or any symptoms caused by or associated with the exposure to ragweed pollen, such as a ragweed pollen allergen.
  • ragweed pollen such as a ragweed pollen allergen.
  • a satisfactory clinical result is achieved when there is an incremental improvement or a partial reduction in an immune response or one or more symptoms and signs, or there is an inhibition, decrease, reduction, suppression, prevention, limit or control of worsening or progression of an immune response or one or more symptoms and signs over a short or long duration (hours, days, weeks, months, etc.) .
  • signs/ symptoms of allergy are those typically associated with the allergies treated according to the present invention, typically signs/symptoms may include one or more of the following; itchy running nose, itchy watery eyes, itchy skin and shortness of breath and the subject may experience that the signs/ symptoms will to some extent be relieved by treatment with antihistamines or steroids.
  • the signs and symptoms may include detectable levels of IgE antibodies against one or more the mites of interest.
  • the treatment entails or consists of relieving one or more signs or symptoms associated with allergic rhinitis, the relief is typically
  • a "sign" of allergy is an objectively observable characteristic of the disease
  • a "symptom” is the subject's subjective experience(s) relative to the disease.
  • Some signs can be symptoms and vice versa, but if a subject for instance experiences dizziness due to a disease, this can only be categorized as a symptom, because it is not objectively observable by anybody else than the subject.
  • increasing levels of for example IgE-antibodies is a "sign", since it cannot be sensed by the subject but it can be objectively measured in an appropriate assay.
  • the relief typically comprises
  • treating the allergy comprises or consists of relieving one or more signs or symptoms associated with allergic asthma
  • the relief typically comprises
  • improving lung function such as increasing Forced Expiratory Volume (FEV), for example by increasing the FEV measured within the first second after full inspiration (FEVl); or increasing FEV1% (the ratio between FEVl and FVCl, where FVC is the forced vital capacity); or increasing FEVl%predicted (FEV1% of the subject divided by the average FEV1% in the population for any person of similar age, sex and body composition) .
  • FEV Forced Expiratory Volume
  • improving lung function can for example be increasing FEVl, FEV1% or FEV 1% predicted to a level that is within a range of normal values of a person of similar height, weight, gender and age which is not suffering from asthma.
  • treating the allergy comprises or consists of relieving one or more signs or symptoms associated with atopic dermatitis
  • the relief typically comprises
  • the method may comprise or consist of reducing the subject's need for concomitant treatment with corticosteroids or H I antihistamines to reduce, relieve, or suppress one or more symptoms of an immune response associated with the allergy. In other words, these embodiments have the long term benefit of reducing the subject's need for medication.
  • 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 ragweed pollen.
  • An increase, improvement, enhancement or induction of "tolerance” may refer to a decrease, reduction, inhibition,
  • the method comprises inducing immunological tolerance in a subject to mites, e.g . to an allergen of ragweed pollen discussed herein to suppress an allergic immune response to the allergen.
  • Immunological tolerance in a subject to an allergen can also be reflected by reducing the occurrence, frequency, severity, progression, or duration of an allergic response of the subject to the allergen.
  • Induction of immune tolerance also referred to as desensitization
  • the relative amount of immune tolerance can be measured by methods disclosed herein or known to the skilled artisan. For example, induction of immune tolerance can be measured by the modulated lymphokine and/or cytokine level in a subject or animal before versus after
  • a modulated cytokine level can be an increase of a cytokine level, for instance an increase of a lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering a polypeptide of the first, second, third or fourth aspect.
  • modulation can be a decrease of the level of a particular cytokine level, for insta nce a decrease of the lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering a polypeptide of the first, second, third or fourth aspect.
  • the lymphokines/cytokines chosen to be measured can be from any relevant lymphokines/cytokines, such as IL-2, IL-5, IL-4, IL-6, IL-10, IL- 12, IL- 13, IL-17, TNF-alfa, IFN-gamma, TGF-beta, MCP- 1, RANK-L and Flt3L. Accordingly, the term "inducing immunological tolerance” may include eliciting, stimulating, promoting, increasing or enhancing immunological tolerance.
  • Immunological tolerance may involve modulation of T cell activity, including but not limited to CD4+ T cells, CD8+ T cells, Th l cells, Th2 cells and regulatory T cells (Tregs), and memory T cells, including inflammatory lymphokines/cytokines produced by T cells.
  • T cell activity including but not limited to CD4+ T cells, CD8+ T cells, Th l cells, Th2 cells and regulatory T cells (Tregs), and memory T cells, including inflammatory lymphokines/cytokines produced by T cells.
  • the subjects subjected to the treatment of the sixth aspect of the invention typically present with an immune response clinically presented as atopic dermatitis, urticaria, contact dermatitis, allergic conj unctivitis, allergic rhinitis, allergic asthma, anapylaxis, and/or hay fever.
  • the treatment thus decreases, reduces, suppresses or inhibits atopic dermatitis, urticaria, contact dermatitis, allergic conjunctivitis, allergic rhinitis, allergic asthma, anaphylaxis, and/or hay fever.
  • the method of the sixth aspect of the invention is capable of inducing or increasing an IgG antibody response in the subject to a ragweed pollen allergen and/or decreasing an IgE antibody response in the subject to a ragweed pollen allergen and/or decreasing a T cell response in the subject against ragweed pollen allergen.
  • the subjects that are subjected to the method of the sixth aspect of the invention are typically sensitized to at least one ragweed pollen allergen. It is to be understood that such patients may exhibit allergy signs or experience symptoms of allergy, but it is not excluded that "patients" that merely exhibit clinical signs of being sensitized against at least one protein allergy of the mites will also benefit from the treatment.
  • the allergy treated according to the invention is in all embodiments of the sixth aspect of the invention allergy towards ragweed pollen of the genus Ambrosia or Artemisiaa (for example selected from the group consisting of Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida and Artemisiia vulgaris.
  • Ambrosia or Artemisiaa for example selected from the group consisting of Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida and Artemisiia vulgaris.
  • the ragweed pollen allergen is in all embodiments of the sixth aspect of the invention selected from one or more allergens in the groups consisting of
  • a group 1 allergen of ragweed pollen for example Amb a 1 and a homolog allergen of Amb a 1 present in pollen of Amb t, Amb p or Art v
  • Amb a 1 and a homolog allergen of Amb a 1 present in pollen of Amb t, Amb p or Art v for example Amb a 1 and a homolog allergen of Amb a 1 present in pollen of Amb t, Amb p or Art v
  • allergen of ragweed pollen for example Amb a 4 and a homolog allergen of Amb a 8 present in pollen of Amb t, Amb p or Art v
  • group 8 allergen of ragweed pollen for example Amb a 4 and a homolog allergen of Amb a 8 present in pollen of Amb t, Amb p or Art v
  • allergen of ragweed pollen for example Amb a 4 and a homolog allergen of Amb a l l present in pollen of Amb t, Amb p or Art v.
  • the allergy is, according to the sixth aspect of the invention and any embodiments thereof, preferably treated by immunotherapy, which may comprise the repeated administration in weekly, bi-weekly, monthly or quarterly intervals.
  • the administration may be selected from any one of subcutaneous, intradermal, epicutaneous, rectal, topical, sublingual, oral, buccal, intranasal, respiratory and intralymphatic route, though subcutaneous, intradermal or sublingual administration are mostly used in current immunotherapies.
  • a pharmacologically effective amount of a single dose of a polypeptide of the first, second, third aspect of the nvention may be in the range of 1 to 1000 nanomole, for example 1 to 500 nanomole, for example 1 to 250 nanomole, for example 5 to 250 nanomole.
  • the single dose may be repeated once daily, twice-weekly, weekly, biweekly or monthly or quarterly.
  • a polypeptide or composition of the invention is administered as a liquid in a volume of about 50 to 150 microliter, such as by intradermal administration.
  • 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.
  • administering may reduce the amount of an adj unct therapy administered to a subject, such as reducing the subject's need for concomitant treatment with fast or long-acting 32-agonists, leukotriene modifiers, theophylline corticosteroids or HI antihistamines (e.g . inhaled or oral) to reduce, relieve, or suppress one or more symptoms of the immune response.
  • an adj unct therapy administered to a subject, such as reducing the subject's need for concomitant treatment with fast or long-acting 32-agonists, leukotriene modifiers, theophylline corticosteroids or HI antihistamines (e.g . inhaled or oral) to reduce, relieve, or suppress one or more symptoms of the immune response.
  • the term “immune response” includes T cell (cellular) mediated and/or B cell (humoral) mediated immune responses, or both cellular and humoral responses.
  • the term “immune response” may include an IgE-mediated immune response (i.e. an allergic immune response) .
  • Exemplary immune responses include T cell responses, such as Th2 responses resulting in cytokine production and/or cellular cytotoxicity.
  • 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+, Th l 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 nonprofessional antigen presenting cells such as keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes) ; natural killer (NK) cells; and myeloid cells, such as macrophages, eosinophils, mast cells, basophils, and granulocytes.
  • T cells CD4+, CD8+, Th l and Th2 cells, memory T cells
  • B cells antigen presenting cells
  • professional antigen presenting cells e.g., professional antigen presenting cells such as dendritic cells, macrophages, B lymphocytes, Langerhans cells, and nonprofessional antigen presenting cells
  • immunoglobulin (Ig) isotypes antibodies or decreasing IgE antibodies.
  • the method comprises inducing or increasing an IgG antibody (e.g . specific IgG) response in a subject to ragweed pollen, such as an allergen of ragweed pollen.
  • the method comprises decreasing an IgE antibody (e.g . specific IgE) response in a subject to to ragweed pollen, such as an allergen of ragweed pollen.
  • the method comprises decreasing a T cell response in a subject to ragweed pollen, such as an allergen of ragweed pollen, for example decreasing the production of Th-2 associated cytokines, like IL-5, IL-4, IL- 13 in response to said ragweed pollen or an allergen of ragweed pollen.
  • modulating an immune response may include to stimulate, induce, promote, increase or enhance an immune response, e.g. a T cell regulatory response, or may include inhibiting, decreasing, suppressing or reducing a T cell response, which may include, but is not limited to a Th2 cell response.
  • a polypeptide or a composition of the invention may be used for conducting immunotherapy against ragweed pollen allergy, optionally wherein a single dose is administered repeatedly within the same day, or with daily, twice-weekly, weekly, bi-weekly, monthly or quarterly intervals until a desirable effect is achieved .
  • Immunotherapy is thought to produce immunological tolerance in the subject undergoing therapy.
  • a polypeptide or a composition of the invention may be used to induce immunological tolerance in a subject in need thereof.
  • the term "immunological tolerance” refers to a) a decreased or reduced level of a specific immunologica l 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 ragweed pollen, i.e. pollen of the genus Ambrosia , such as pollen of Amb a, Amb p or Amb t.
  • an increase, improvement, enhancement or induction of "tolerance” may refer to a decrease, reduction, inhibition, suppression, or limiting or controlling or clearing of specific immunological reactivity to an allergen as compared to reactivity to the allergen in a previous exposure to the same allergen.
  • the method comprises inducing immunological tolerance in a subject to ragweed pollen, e.g. to an allergen of ragweed pollen, such as a group 1, 3, 4, 5, 8 or 11 allergen of Amb a, Amb t and/or Amb p to suppress an allergic immune response to the allergen.
  • Immunological tolerance in a subject to an allergen can also be reflected by reducing the occurrence, frequency, severity, progression, or duration of an allergic response of the subject to the allergen.
  • Induction of immune tolera nce also referred to as desensitization
  • induction of immune tolerance can be measured by methods disclosed herein or known to the skilled artisan.
  • induction of immune tolerance can be measured by the modulated lymphokine and/or cytokine level in a subject or animal before versus after administering a peptide combination described herein for the first time.
  • a modulated cytokine level can be an increase of a cytokine level, for instance an increase of a lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering the peptide combination for the first time.
  • modulation can be a decrease of the level of a particular cytokine level, for instance a decrease of the lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering the peptide combination for the first time.
  • lymphokines/ cytokines chosen to measure can be from any relevant lymphokines/ cytokines, such as IL-2, IL-5, IL-4, IL-6, IL- 10, IL-12, IL-13, IL- 17, TNF-alfa, IFN-gamma, TGF-beta, MCP- 1, RANK- L and Flt3L. Accordingly, the term "inducing immunological tolerance” may include eliciting, stimulating, promoting, increasing or enhancing immunological tolerance.
  • Immunological tolerance may involve modulation of T cell activity, including but not limited to CD4+ T cells, CD8+ T cells, Thl cells, Th2 cells and regulatory T cells (Tregs), and memory T cells, including inflammatory lymphokines/cytokines produced by T cells.
  • T cell activity including but not limited to CD4+ T cells, CD8+ T cells, Thl cells, Th2 cells and regulatory T cells (Tregs), and memory T cells, including inflammatory lymphokines/cytokines produced by T cells.
  • a polypeptide or a composition according to the invention is typically administered to a subject in need thereof by injection, such as by subcutaneous or intradermal administration, but may also include other routes of administration, such as epicutaneous, transcutaneous, topical, rectal, sublingual, oral, buccal, intranasal, respiratory and intralymphatic route of administration.
  • the subject in need thereof is a human, a pet such as a dog or a cat, a domestic animal such as a horse, or a laboratory animal (a mouse, a guinea pig or a rabbit) .
  • the subject may be sensitized to ragweed pollen (e.g. having specific IgE antibodies against an allergen of ragweed pollen and/or having a T cell response against an allergen of ragweed pollen) . Therefore, a subject in need thereof may produce specific IgE antibodies or a T cell response against ragweed pollen allergens.
  • a still other aspect of the invention relates to an in vitro method of determining whether T cells of a subject are responsive to one or more polypeptides of the first, second or third aspect of the invention or to a composition of the fourth aspect.
  • the method comprises contacting T cells obtained from the subject with said polypeptide(s) or composition(s) and determining whether the T cells are stimulated.
  • the in vitro method may be used to determine whether the subject has, or is at risk of developing, an allergy to ragweed pollen allergen.
  • a still other aspect of the invention relates to an in vitro method for determining whether a subject has, or is at risk of developing, an allergy to house dust mites or storage mites, comprising contacting T cells obtained from the subject with one or more polypeptides of the first, second or third aspect of the invention and determining whether the T cells are stimulated .
  • a further aspect of the invention relates to a diagnostic kit comprising one or more of the polypeptides of the first, second or third aspect of the invention.
  • a diagnostic kit comprising one or more of the polypeptides of the first, second or third aspect of the invention.
  • Such a kit will normally also include necessary detection agents, visualisation means, carriers etc. that enable one or more of the above-described diagnostic assays.
  • This example describes the peptide library set out to screen for T cell epitope responses of polypeptides derived from wild type allergens of group 3, 4, 5, 8 and 11 of of Ambrosia artemisiifolia (Amb a ; common ragweed/short ragweed), in short denoted Amb a 3, Amb a 4, Amb a 5, Amb a 8 and Amb a 11, respectively.
  • This example includes a description of the methods used to screen peptide pools and single polypeptides for T cell reactivity.
  • the polypeptides were assembled in pools of 5 polypeptides, on average, and screened for their immunogenicity. All pools were subsequently deconvoluted to make sure that all polypeptides were analysed separately as well.
  • the T cell reactivity of the polypeptides disclosed herein was assessed by measuring T cell proliferation of T cell lines specific to the allergens investigated and/or by measuring the cytokine production of allergen-specific T cells obtained from cultured PBMCs obtained from the donor cohorts.
  • Donor cohort Around 30 donors were recruited in Texas, USA and Slovakia (Europe) on basis of a positive skin prick test to Amb a or Amb t and a clinical history of seasonal allergy with symptoms in the fall. Self reported symptoms in the recent fall confirmed the seasonal responses and sensitization towards Amb a was confirmed by IgE testing (>0.7 kU/L).
  • T-cell lines Isolated PBMCs (peripheral blood mononuclear cells) obtained from the donors were CD8 depleted and T cell lines were established by stimulating the cultures with Amb a allergen extract enriched for allergen content.
  • PBMCs were isolated from freshly drawn heparinized blood by gradient centrifugation on lymphoprep (Nycomed, Norway), washed twice and re-suspended in RPMI 1640 medium with HEPES and ultraglutamine (Cambrex, Belgium) supplemented with 5% v/v human AB-serum
  • PBMCs peripheral blood mononuclear cells
  • the CD8-depleted PBMCs (2 x 10 6 /ml) were stimulated in 1 ml bulk cultures in AB medium in 24 well plates (Nunc, Denmark) with Amb a allergen extract enriched for allergen content (0.5 ug/mL).
  • rIL-2 was added at day 5, 6 and 7 (65U, 30U, 30U, respectively) .
  • ELISpot/ Fluorospot day 14, T- cell line generation 01 (TCLOl)
  • proliferation day 24, TCL02
  • T cells of TCLOl were isolated, washed and counted. The isolated T cells of TCLOl generation were used to perform Fluorospot assay. TCLOl were further re-stimulated as described in the next paragraph for use in T cell proliferation experiment at day 24.
  • T-cells TCL02 were isolated, washed, counted and used for T cell proliferation assay as described later.
  • Fluorospot assays In the Fluorospot assays, release of IL-5 and IFN- ⁇ upon T cell activation (of donor derived lines DKl-TCLOl, and US1-TCL01) with a peptide described herein was measured by Fluorospot assay kit (Mabtech AB) and procedures were followed as per manufacturer's instructions. Briefly anti-human IL-5 and anti-human IFN- ⁇ coated 96 well plates were obtained and blocked with 10% AB serum in RPMI.
  • the plates were washed thoroughly and the isolated T cells were plated and incubated with peptide pools (2ug/mL of each peptide), polypeptides (2ug/ml), full length allergens (2 ug/ml, natural purified or recombinant/synthetic), Amb a allergen extract (5 ug/ml), or medium control) for 20hrs/37°C/5%CO 2 . After the incubation time the cells were removed by emptying the plate. Detection antibodies 7-B6- 1-FS-FITC ( 1 : 200) and 5A10- biotin (2 g/ml) were diluted in PBS-0.1% BSA and were added to the plate and incubated for 2 hours/room temperature (RT) .
  • peptide pools 2ug/mL of each peptide
  • polypeptides 2ug/ml
  • full length allergens (2 ug/ml, natural purified or recombinant/synthetic
  • SI stimulation index
  • P value ⁇ 0.05
  • sfc> 20 (IL-5/IFN-g)/10 6 cells.
  • SI can be defined as [colony forming cells (sfc) stimulation/ sfc medium control] in a Fluorospot assay and [counts per minute (cpm) stimulation/ cpm medium control] in a proliferation assay.
  • Table 1 shows the polypeptides derived from the allergen Amb a 3.0101 which showed positive T cell response according to the criteria set above. More than 20% of the donor cohort responded to the polypeptides with ID number B0182 (33%) and B0185 (20%). In addition to the 20mer polypeptides, longer polypeptides could be tested, for example peptide with start position number 32 having the sequence FKTTDVLWFNFTTGEDSVAEV.
  • Table 1 - Amb a 3 polypeptides SEP ID NOs : 1-91
  • Table 2 shows the polypeptides derived from the allergen Amb a 4.0101 which showed positive T cell response accord ing to the criteria set a bove. More than 20% of the donor cohort responded to the polypeptides with ID number B0188* (33%), B0189* (33%), B0190* (20%) and B0192* (40%), B0193 (27%), B0196 (47%), B0197 (53%) .
  • n immunodominant peptide does not include amino acid residues positioned at the start and end of the defined regions. Therefore, additiona l epitope polypeptides of Amb a 4.0101 may prefera bly be derived from regions covering the position numbers from 6 to 35 and 46- 64, 86- 104.
  • shorter or longer polypeptides could be tested, for example peptide with sta rt position number 1 having the sequence KLSEKPSVTWSGKSKVKQTDK, start position number 39 having the seq uence KRDSKATSFSYFDSDPTKN PGP or start position number 91 having the seq uence GPPPPEGGEGGGDGGGEGGGE
  • Table 3 shows the polypeptides derived from the allergen Amb a 5.0101 which showed positive T cell response according to the criteria set above. More than 20% of the donor cohort responded to the polypeptides with ID number B0199* (20%) and B0200* (27%) . This indicates that the majority of the T cell responses to Amb a 5.0101 derive from the amino acid stretch ranging from amino acid position number 1 to 29, more preferable from 6-25.
  • Table 4 shows the polypeptides derived from the two allergens Amb a 8.0101 and Amb a 8.0102 which showed positive T cell response according to the criteria set above.
  • the table also shows the number of mismatches in the amino acid sequence of polypeptides derived from the allergen Amb a 8.0101 compared to the corresponding peptide derived from allergen Amb a 8.0102 and vice versa .
  • n immunodominant peptide does not include amino acid residues positioned at the start and end of the defined regions. Therefore, addition a l epitope polypeptides of Amb a 8.0101 may prefera bly be derived from regions covering the position numbers from 36 to 63 and 85- 104. For Amb a 8.0102, additional epitope polypeptides may prefera bly be derived from regions covering the position numbers from 38 to 63 and 85- 104.
  • Table 5 shows the polypeptides derived from the allergen Amb a 11.0101 which showed positive T cell response accord ing to the criteria set a bove. More than 20% of the donor cohort responded to the polypeptides with ID number B0227 (21%), B0228* (21%), B0230 (29%), B0231* (36%), B0233* (29%), B0234 (71%), B0237 (29%), B0238 (36%), B0239 (21%), B0241* (50%), B0242* (36%), B0245 (43%), B0246* (36%) and B0250 (43%).
  • an immunodominant peptide does not include amino acid residues of the start and end of the defined regions. Therefore, additional epitope polypeptides may preferably be derived from regions covering the position numbers from 211-230, 250-270, 290-310 and 335-354.
  • Table 6 shows a summary of the dominant epitope regions of each of the allergens tested .
  • polypeptides of the initial screening were re-designed to improve aqueous solubility, in particular to increase the solubility in an aqueous solution having pH in a physiologically acceptable range, e.g. from 4.5 to 8.5.
  • Solubility is closely related to the ratio between hydrophilic and hydrophobic amino acid, the number of charged amino acids and the net charge of the peptide at a given pH.
  • Polypeptides are preferably elongated with amino acid residues naturally encoded within the full length allergen sequence from which the peptide derives. Thus, where a peptide is designed from Amb a 8, the peptide is elongated with amino acid residues from Amb a 8.
  • peptide-solubility is improved by addition of either non-encoding negatively charged amino acids or non-encoded positively charged amino acids.
  • examples where these two combinations are applied i.e. natural elongation and addition of non-encoded amino acids are also shown.
  • polypeptides were provided (by GenScript, USA) as acetate salts, >95 % purity at a 20mg scale.
  • Test samples with a peptide concentration of 1 mg/mL or 3 mg/mL were produced with the following solvents: Buffer A; Water, Buffer B; Acetate buffer, pH 4.5 (25 mM sodium acetate containing 250 mM D-Mannitol), pH 4.5, Buffer C; 25 mM histidine buffer, pH 7.0, Buffer D; Glycine buffer, pH 8.5 (35 mM glycine containing 250 mM D-Mannitol). Test samples were gently mixed and left for one hour at room temperature to allow for dissolution of the polypeptides.
  • the solubility of the polypeptides was initially investigated by visual inspection by the eye and if the sample turned out to be very turbid, no further testing was performed.
  • Test samples having passed the first solubility test were than analysed unfiltered or filtered (500 ⁇ sample through a 0.2 ⁇ PES filter9 by HPLC, DLS, 90°LS and for UV-VIS for detection of particles/aggregates of the test sample and instability of the polypeptides.
  • mRNA was purified from Amb p, Amb t and Art v pollen and subjected to high-throughput sequencing (Illumina HiSeq 2500) .
  • the resulting sequencing reads were assembled using the Trinity software (version 2. 1.1) to obtain the Amb p, Amb t and Art v pollen transcriptomes.
  • Table 7 shows amino acid sequences of homolog proteins found in the transcriptomes of Amb p, Amb t and Art v, which have high percentage sequence identity to the corresponding group 3, 4, 8 or 11 allergens of Amb a and which furthermore could be detected by mass spectrometry in pollen extracts of the species in question. No homologs could be identified for group 5. For each allergen, only those homologs containing sequences with a minimum number of mismatches to the original Amb a sequence are shown. Further is shown the percentage sequence identity between the Amb a sequence and the homolog allergen in the overlapping area (% SEQ ID (I)) and the percentage of the homolog amino acid sequence that covers the Amb a sequence (% SEQ ID (II)). Table 7 - Homoloq allergens (SEP ID NOs : 126-149
  • Tables 8-11 show examples of cross reacting polypeptides.
  • the frequency of T cell responders in a donor population and the cross reactivity between homologous polypeptides designed from each of the allergens of Amb a, Amb p, Amb t and Art v species are shown in figures 1 to 6.
  • This example describes how HLA Class II allele coverage can be determined for individual polypeptides.
  • the peptide binding to a specific HLA molecule can be measured or predicted using in silico algorithms as e.g. NetMHCIIpan-3.1 (Karosiene, Edita, M ichael Rasmussen, Thomas Bö, Ole Lund, S0ren Buus, and Morten Nielsen. "NetMHCIIpan-3.0, a Common Pan-specific M HC Class II Prediction Method Including All Three Human MHC Class II Isotypes, HLA-DR, H LA-DP and HLA- DQ.” Immunogenetics ) available at the internet site ⁇ URL:
  • NetMHCIIpan-3.1 algorithm was used. To assign a given peptide as a binder for a given HLA, a binding threshold of 300 nM was used together with the percentile rank score reported in this algorithm of at most 30.
  • the phenotypic coverage of the binding alleles were calculated using standard methods known in the art. For example, for each HLA locus (DRBl, DRB3, DRB4, DRB5) or locus combination (DQA1-DQB1, DPA1-DPB1), the HLA allele frequencies of all the alleles from the given locus found to bind the given peptide is summed (f sum ,iocus) ⁇ The total phenotypic coverage for each locus, n (C Thread) is calculated as (2 x f slJ m,iocus)-(fsum,iocus) 2 '
  • the total phenotypic coverage of all binding alleles from n loci can be calculated as
  • the predicted worldwide HLA coverage may be predicted using the alleles and allele frequencies shown below in table 12.
  • Table 12 shows the frequencies of 28 alleleles present on loci, HLA-DRBl, HLADRBl, HLA-DRB4, HLA-DRB5, HLA-DP, and HLA-DQ, in a worldwide population ⁇ from Paul et al. Development and validation of a broad scheme for prediction of HLA class II restricted T cell epitopes. Journal of Immunological Methods 422 (2015) 28-34) .
  • Table 13 shows the frequencies of 15 HLA alleles present on loci, HLA-DRB3, HLA-DRB4, HLA- DRB5, HLA-DP, and HLA-DQ, in a worldwide population.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • Public Health (AREA)
  • Microbiology (AREA)
  • Botany (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to T cell epitope-containing polypeptides derived from pollen allergens Ambrosia Artemisiifolia (Amb a), Ambrosia psilostachya (Amb p), Ambrosia trifida (Amb t) and Artemisia vulgaris (Art v) and novel allergens found in pollen of Amb p, Amb t and Art v and which are considered homologous to the allergens Amb a 3, Amb a 4, Amb a 8 and Amb a 11. The invention further relates to the use of such polypeptides and novel allergens for modulating an immune response, for treating an allergic response and/or for inducing or promoting immunological tolerance, and for use in diagnostic methods and kits.

Description

EPITOPE POLYPEPTIDES OF RAGWEED POLLEN ALLERGENS FIELD OF THE INVENTION
The present invention relates to T cell epitope-containing polypeptides derived from ragweed pollen allergens, in particular polypeptides derived from ragweed allergens belonging to group 3, 4, 8 and 11 allergens, and uses of such polypeptides for modulating an immune response, for treating an allergic response and/or for inducing or promoting immunological tolerance, and for use in diagnostic methods and kits.
BACKGROUND OF THE INVENTION
Ragweed pollen is a major source of pollen allergy and may be treated by allergen-specific immunotherapy using full length allergen(s), modifications thereof (hypoallergens) or more conventionally by administering ragweed pollen extracts. Unfortunately, the administration of IgE- reactive ragweed allergens to ragweed pollen allergic individuals poses the risk of provoking serious side effects, for example anaphylactic shock. T cell epitope peptide-based immunotherapy (PIT) could be one option to provide well-tolerated immunotherapeutic methods. With this therapy soluble polypeptides containing allergen-derived immunodominant T-cell epitopes are used to treat allergic patients. Due to the use of T cell epitope-conta ining polypeptides without IgE-reactivity the chance of adverse effects are reduced significantly.
T cell epitopes of the major allergen of Ambrosia artemisiifolia (Amb a 1) have been studied extensively and peptide cocktails for the treatment of ragweed pollen allergy by PIT have also been suggested (WO10018378, WO08098749, Jahn-Schmid, the T-cell response to Amb a 1 is characterized by 3 dominant epitopes and multiple MHC restriction elements. J Allergy Clin Immunol, Vol.126, No. 5, 1068-1070, 2010). Several other ragweed pollen allergens of Ambrosia artemisiifolia have been identified based on IgE reactivity, for example those reported in the WHO/IUIS Allergen Nomenclature database (http\www. allergen. org). However, these are often regarded as minor allergens as the number of ragweed allergic individuals with IgE-reactivity towards these allergens are considerable lower than individua ls with IgE-reactivity towards Amb a 1.
Although ragweed pollen allergy is often associated with exposure to pollen of Ambrosia
Artemisiifolia (Amb a; short ragweed), other ragweed species may be relevant too as the geographic presence of other ragweed species is expanding. Other relevant species may be
Ambrosia psilostachya (Amb p; western ragweed) and Ambrosia trifida (Amb t; giant ragweed) . Therefore, there is a desire to provide polypeptides for vaccines against ragweed pollen allergy which not only targets the Amb a species, but also the Amb p and Amp t species. It may also be advantageous to provide polypeptides for use in vaccines against related weed pollen like Artemisia vulgaris (Art v; mugwort). Thus, it is a desire to provide polypeptides capable of providing a T cell- mediated immune response in subjects sensitized to at least either Amb a, Amb p, or Amb t, or combinations of Amb a, Amb p and Amb t. In addition the same polypeptides may be able to provide a T cell-mediated immune response in subjects sensitized to an additional weed species, for example Art v.
SUMMARY OF THE INVENTION
According to the invention, there are identified several immunodominant regions and epitope polypeptides consisting of a stretch of contiguous amino acids selected from Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a l l . Surprisingly, similar immunodominant polypeptides and regions could be derived from homolog allergens identified in Amb t, Amb p or Art v species. In accordance with the present invention, such immunodominant epitope polypeptides have great potential as a vaccine for treatment of weed pollen allergy caused by several weed pollen species, including at least Amb a, Amp, Amb t and optionally Art v A first aspect of the invention relates to immunodominant epitope regions identified in the amino acid sequence of the pollen allergens Amb a 4, Amb a 8 and Amb 11 (SEQ ID NO : 106-125) and any corresponding immunodominant epitope region found in allergens homologous to Amb a 4, Amb 8 and Amb 11 in the species Amb p, Amb t and Art v (SEQ ID NOs: 189-224). An
immunodominant epitope region is defined by the stretch of amino acid residues formed by overlapping polypeptides that have shown high T cell responder frequency in a donor population. For example, 20-mer polypeptides overlapping each other by 10 amino acid residues may result in an epitopic region of 30 amino acid residues. For the allergen Amb a 8, there are identified larger dominant epitopic regions, for example defined by four overlapping polypeptides (SEQ ID NOs : 37- 40 of table 4) resulting in the epitopic region with SEQ ID NO: 112 (table 6) .The immunodominant epitope regions are useful for designing immunodominant epitope polypeptides of various length which have high likelihood of inducing a T cell-mediated immune response in ragweed and/or mugwort pollen allergic subjects. In addition, the same immunodominant epitope regions may be used for designing fusion proteins comprising a high density of immunodominant regions in comparison to the wild type allergens and/or comprising immunodominant regions from one allergen or from several different allergens.
Therefore, a first aspect of the invention relates to a polypeptide comprising 9, or at least 9, contiguous amino acids selected from any one of SEQ ID NOs: 106- 125 and 189-224; or a variant polypeptide which comprises an amino acid sequence with at least 65% sequence identity to the 9 or at least 9 contiguous amino acids. A polypeptide comprising 9, or at least 9 contiguous amino acids is meant to define that the polypeptide comprises 9 contiguous amino acids or at least 9 contiguous amino acids. Furthermore, a first aspect relates to a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 106-125 and 189-224, optionally wherein the polypeptide is modified by one, two or three amino acid substitutions. A second aspect relates to polypeptides able to induce a T cell response in ragweed allergic subjects, which are derived from either of the allergens; Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a 11; or from homolog allergens identified in pollen of Amb p, Amb t or Art v.
Accordingly a second aspect relates to a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 1-105 (polypeptides derived from Amb a shown in tables 1 to 5) and SEQ ID NOs : 150-182 (polypeptides derived from homolog allergens shown in tables 9 to 11); or a variant thereof which is 9 to 30 amino acids in length and comprises:
i) an amino acid sequence set forth in any one of SEQ ID NOs: 1- 105 and 150-182;
ii) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of i); iii) 9 or at least 9 contiguous amino acid amino acids of the sequence of i); or
iv) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of iii).
For example, a second aspect relates to a polypeptide consisting of an amino acid sequence with SEQ ID NOs : 1- 105, wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or the C-terminal end. Examples of polypeptides having one, two or three amino acid substitutions in comparison to SEQ ID NOs : 1- 105 may be selected from any one of SEQ ID NOs 150-182.
A third aspect of the invention relates to novel allergens identified in pollen of Amb p, Amp t or Art v, which are considered homologous to Amb a 3, Amb a 4, Amb a 8 and Amb a l l allergens. It has now been shown that these novel allergens share common epitopes with allergens of Amb a.
Examples of novel allergens are shown in table 7 (SEQ ID NOs: 126- 146). Therefore, a third aspect of the invention relates to a polypeptide consisting of an amino acid sequence having 85% to 100% sequence identity (i.e. having at least 85% sequence identity) with an amino acid sequence selected from any one of SEQ ID NOs: 126- 146.
A fourth aspect of the invention relates to a chimeric molecule comprising a fusion of two or more of the polypeptides selected from any one of the first, second, or third aspect, or a chimeric molecule comprising a fusion of two or more of the amino acid sequences selected from the group consisting of SEQ ID NOs: 106-125 and 189-224, or a chimeric molecule comprising the fusion of two or more of the amino acid sequences selected from the group consisting of SEQ ID NOs: 126- 149.
A fifth aspect of the invention relates to a composition, such as a pharmaceutical composition, which comprises one or more of the polypeptides of the first aspect, second aspect, third aspect and/or fourth aspect of the invention. Such compostions may comprise further polypeptides designed from other allergens, for example designed from the major allergen Amb a 1, or from a homolog allergen to Amb a 1 found in Amb p, Amb t or Art v.
A sixth aspect of the invention relates to a method of treating ragweed pollen allergy in a subject, comprising administering to the subject a therapeutically effective amount of a polypeptide of the of the first aspect, second aspect, third aspect and/or fourth aspect of the invention or a composition according to a fifth aspect of the invention.
Alternatively worded, a sixth aspect of the invention relates to a polypeptide of the first aspect, second aspect, third aspect and/or fourth aspect of the invention or a composition according to a fifth aspect of the invention for use in treating ragweed pollen allergy in a subject.
Still alternatively worded, a sixth aspect of the invention relates to use of a polypeptide of the first aspect, second aspect, third aspect and/or fourth aspect of the invention or a composition according to a fifth aspect of the invention for the preparation of a medicament for use in treating ragweed pollen allergy in a subject. DETAILED DISCLOSURE OF THE INVENTION Definitions
As used herein an "epitope" refers to a region or part of an antigen, such as a peptide disclosed herein, that elicits an immune response when administered to a subject. An epitope may be a T cell epitope, i.e., an epitope that elicits, stimulates, induces, promotes, increases or enhances a T cell activity, function or response. For example, such as a Th2 cell epitope. Any peptide or combination of polypeptides of interest can be analyzed to determine whether they 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.). More particularly, T cell activity can be analyzed using the methods described in detail herein, for example as described in Example 2. The term "allergen" refers to an antigen which elicits, induces, stimulates, or enhances an immune response by a cell of the immune system of an exposed animal (e.g ., human) . An antigen is an allergen when the specific immune response is the development of enhanced sensitivity or a hypersensitivity to the antigen, but the antigen itself is not typically innately harmful. An allergen is therefore a particular type of antigen that can cause development of enhanced or increased sensitivity or hypersensitivity in a subject. For example, an allergen can elicit production of IgE antibodies in predisposed subjects.
If no other meaning is given specifically, the term "T cell response" refers to an interleukin or a proliferation response by a T cell. It may be determined as explained in Example 2. It may in some instances be referred to simply as a "response" to a peptide. The term "allergic response" is intended to refer to the hypersensitive immune reaction to a normally inocuous environmental substance known as an allergen. The most common mechanism of allergic reactions is the binding of IgE to the surface of mast cells, which causes asthma, hay fever and other common allergic reactions. The term "identity" and "identical" and grammatical variations thereof, as used herein, mean that two or more referenced entities are the same (e.g . , amino acid sequences) . Thus, where two polypeptides are identical, they have the same amino acid sequence. The identity can be over a defined area, e.g. over at least 12, 13, 14, 15 or 16 contiguous amino acids of the original peptide or polypeptide, optionally wherein the alignment is the best fit with gaps permitted . 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, WI, U.S.A.). Sequence identity may also be determined using the BLAST algorithm, described in Altschul et a/., 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 . n Im . n ih . gov/) . Such algorithms that calculate percent sequence identity 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 PAM 100, 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)) . Legends to figures
Figure 1 : Shows the fraction of donors producing an in vitro T cell immune response (response frequency) towards polypeptides derived from Amb a 3 allergen, Amb a 4 allergen or homolog polypeptides found in other species. Figure 2A: Shows the correlation between T cell response frequency of an Amb a 3 polypeptide
(B0182, SEQ ID NO : 2) and homolog polypeptide (B0456, SEQ ID NO : 151) derived from of Amb p.
Figure 2B: Shows the correlation between T cell response frequency of an Amb a 4 polypeptide (B0309* (SEQ ID NO : 163) and the homolog polypeptide of Amb p (B0458*, SEQ ID NO: 167) and Art v (B0385*, SEQ ID NO: 165) . Figure 3: Shows the fraction of donors producing an in vitro T cell immune response (response frequency) towards polypeptides of Amb a 8 allergen and homolog polypeptides found in Amb t and Art v.
Figure 4: Shows the correlation between T cell response frequency of an Amb a 8 polypeptide (B0311, SEQ ID NO : 168) and homolog polypeptides (B0387 (SEQ ID NO: 169), B0388 (SEQ ID NO : 170), B0389 (SEQ ID NO : 171), B0393 (SEQ ID NO: 172), B0394 (SEQ ID NO : 173)) .
Figure 5: Shows the correlation between T cell response frequency of an Amb a 8 polypeptide (B0312, SEQ ID NO : 174) and homolog polypeptides (B0390 (SEQ ID NO : 175), B0391 (SEQ ID NO : 176), B0392 (SEQ ID NO : 177), B0395 (SEQ ID NO : 178), B0396 (SEQ ID NO : 179))
Figure 6A: Shows the fraction of donors producing an in vitro T cell immune response (response frequency) towards polypeptides of Amb a 11 allergen and homolog polypeptides found in Amb t and Amb p.
Figure 6B: Shows the correlation between T cell response frequency of an Amb a l l polypeptide (B0234, SEQ ID NO : 83) and homolog polypeptides (B0397, (SEQ ID NO: 180), B0460 (SEQ ID NO : 181)). Figure 7: Shows sequence alignment between Amb a 3.0101 allergen and homologs identified in Amb p (SEQ ID NO : 126), Amb t (SEQ ID NO: 127) and Art v (SEQ ID NO : 128). The figure also shows T cell-epitope containing polypeptides derived from Amb a 3.0101 and its homologs (SEQ ID Nos : 2, 141 and 142).
Figure 8: Shows sequence alignment between Amb a 4.0101 allergen and homologs identified in Amb p (SEQ ID NO : 129), and Art v (SEQ ID NO: 147) . The figure also shows dominant epitope regions identified in Amb a 4 and its homolog allergens (SEQ ID NOs: 108, 189 and 190 (sequences indicated in bolded text) and SEQ ID NOs: 109, 191 and 192 (sequences indicated with underlined text) .
Figure 9: Shows sequence alignment between Amb a 4.0101 allergen, epitopic regions with SEQ ID NOs : 108, 109 and 110) and T cell-epitope containing polypeptides derived from Amb a 4.0101 and its homologs (SEQ ID Nos: 11, 153, 155, 15, 159, 161, 19, 163, 165 and 167) .
Figure 10: Shows sequence alignment between the first part of the Amb a 8.0101 and Amb a 8.0102 allergen and homologs identified in Amb p (SEQ ID NOs: 130- 135), Amb t (SEQ ID NOs: 136- 141) and Art v (SEQ ID NOs: 142- 144) . The figure also shows dominant epitope regions identified in Amb a 8 (SEQ ID NO: 112) and in its homolog allergens (SEQ ID NOs: 193-207 (sequences indicated in bolded text).
Figure 11 : Shows sequence alignment between the second part of Amb a 8.0101 and Amb a 8.0102 allergen and homologs identified in Amb p (SEQ ID NOs: 130- 135), Amb t (SEQ ID NOs: 136- 141) and Art v (SEQ ID NOs: 142- 144) . The figure also shows dominant epitope regions identified in Amb a 8 (SEQ ID NO: 116) and in its homolog allergens (SEQ ID NOs: 208-218 (sequences indicated in bolded text).
Figure 12: Shows sequence alignment between epitopic region of Amb a 8 having SEQ ID NO: 112, and T cell-epitope containing polypeptides derived from Amb a 8 and its homologs (SEQ ID Nos: 44, 168-173, 55 and 174- 179) .
Figure 13: Shows sequence alignment between Amb a 11.0101 and homologs identified in Amb t (SEQ ID NO : 145), and Art v (SEQ ID NO: 146). The figure also shows dominant epitope regions identified in Amb a 11 (SEQ ID NO: 120, 121, 121) and in its homolog allergens (SEQ ID NOs : 219, 220 (sequences indicated with underlined text), SEQ ID NOs: 221 and 222 (sequences indicated with bolded text) and SEQ ID NOs : 223 and 224 (sequences indicated with underlined text).
Embodiments of the first aspect
According to a first aspect of the invention, there is provided epitopic regions of Amb a 3, 4, 5, 8 and 11 allergens (SEQ ID NOs: 106- 125, shown in table 6) . The corresponding epitopic regions identified in allergens of Amb p, Amp t and Art v shown in table 7 have also been identified by performing sequence alignments between the Amb a epitopic regions and the homolog allergens of table 7. Sequence alignments are shown in figures 7-13 together with the epitopic regions of homolog allergens, which are indicated as bolded or as underlined text. These epitopeic regions have SEQ ID NOs: 189-224. The in vitro T cell reactivity of non-limiting exemplary polypeptides designed from these epitopic regions are shown in figures 1 to 6. Therefore, a first aspect of the invention relates to a polypeptide comprising 9 or at least 9 consecutive amino acids selected from an amino acid sequence consisting of any one of SEQ ID NOs : 106 to 125 and 189-224; or a variant thereof which comprises an amino acid sequence with at least 65% sequence identity, such as at least 70%, 75%, 80%, 85%, 90% or 95% sequence identity to the 9 or the at least 9 consecutive amino acids.
In some embodiments, the polypeptide comprises 12 to 15 continguous amino acids such as 12, 13, 14, 15 contiguous amino acids, or 15 to 30 contiguous amino acids such as 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 contiguous amino acids, or 30 to 40 contiguous amino acids, such as 30, 32, 35, 37 or 40 contiguous amino acids; or the polypeptide comprises at least 12 to 15, such as 12, 13, 14, 15, or at least 15 to 30 such as 15, 16, 17, 18, 19, 20, 21, 22,23, 24, 25, 26,27, 28,29, or 30 contiguous amino acids, or at least 30 to 40 contiguous amino acids, such as 30, 32, 35, 37, or 40 contiguous amino acids.
In some embodimens, the variant comprises an amino acid sequence with at least 65% sequence identity (for example at least 70%, 75%, 80%, 85%, 90% or 95%) to the 12, 14, 15, 16, 17, 18, 20, 22, 24, 26, 28, 30, 35 or 40 contiguous amino acids or the at least 12, 14, 15, 16, 17, 18 or 20, 22, 24, 26, 28, 30, 35 or 40 contiguous amino acids.
In some embodiments, the polypeptide comprises 15 or at least 15 contiguous amino acids selected from any one of SEQ ID NOs: 106- 125 and 189-224; or a variant thereof which comprises an amino acid sequence with at least 80% sequence identity (for example at least 85%, 90% or 95%) to the 15 or the at least 15 contiguous amino acids.
In some embodiments, the polypeptide comprises at most 30 contiguous amino acids, for example at most 28, 26 or 24 contiguous amino acids. Thus, in some embodiments, the polypeptides is 15- 30 amino acids in length, for example 16-28 amino acids in length, for example 18-26 amino acids in length, wherein the sequence of the polypeptide consists of a portion of a epitopic region selected from any one of SEQ ID NOs: 106- 125 and 189-224. A variant thereof may have at least 80% sequence identity, for example at least 85%, 90% or 95% to the original polypeptide. For a peptide having the length of 20 amino acids, this sequence identity would apply to about one to three amino acid substitutions in the original polypeptide.
Thus, it should be understood that a first aspect also relates to a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 106- 125 and 189-224, optionally wherein the polypeptide is modified by one, two or three amino acid substitutions. In various embodiments of the first aspect, a polypeptide consists of:
i) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 108, 189 and 190; ) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 109, 191 and 192;
) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 112 and 193-207;
) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 116 and 208-218. v) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 120, 219 and 220; vi) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 121, 221 and 222; or vii) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 122, 223 and 224; optionally wherein any of the polypeptides of i) to vii) is modified by one, two or three amino acid substitutions.
Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 108, 189 and 190 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 10, 11, 152- 157, 15 and 158- 161.
Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 109, 191 and 192 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 18, 19 and 162- 167.
Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 112 and 193-207 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 44, 168-173, 55 and 174- 179.
Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 116 and 208-218 is SEQ ID NO : 119.
Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 121, 221 and 222 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 83, 180 and 81.
Non-limiting examples of a polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 122, 222 and 223 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 89 and 182. Further polypeptides may be designed from the above-mentioned epitope regions with SEQ ID NOs : 106-125 and 189-224. In further embodiments thereof a polypeptide consists of 16-28 contiguous amino acids of the epitopic region, for example 18-28 contiguous amino acids, 18-26 contiguous amino acids or 18-24 contiguous amino acids, such as 18, 19, 20, 21, 22, 23 or 24 contiguous amino acids, of an epitope region with SEQ ID NOs: 106- 125 and 189-224. The further designs may be made allowing one, two or three amino acid substitutions in the amino acid sequence of the polypeptide.
Embodiments of the second aspect As mentioned, the present inventors have identified epitope polypeptides able to induce proliferation of T cells of ragweed allergic donors, wherein the polypeptides consists of smaller portions of the amino acid sequence of the naturally occurring allergens : Amb a 3, Amb a 4, Amb a 5, Amb a 8 and Amb a 11. In addition, the inventors have identified corresponding polypeptides in allergens which are homologous to Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a l l . Such polypeptides likely contain at least one T cell epitope, optionally a Th2 cell epitope. In certain instances, the epitope polypolypeptides comprise about 20 amino acid residues in length and consists of an amino acid sequence selected from any one of SEQ ID NOs: 1- 105 and 150-182. Sequences of polypeptides and T cell responder frequency are shown in the following tables: · Table 1 : Shows polypeptides of a group 3 allergen of the species Amb a (SEQ ID NOs : 1-9)
• Table 2 : Shows polypeptides of a group 4 allergen of the species Amb a (SEQ ID NOs : 10-26)
• Table 3 : Shoes polypeptides of a group 5 allergen of the species Amb a (SEQ ID NOs: 27-32)
• Table 4 : Shows polypeptides of a group 8 allergen of the species Amb a a (SEQ ID NOs: 33- 67)
· Table 5 : Shows polypeptides of a group 11 allergen of the species Amb a (SEQ ID NOs: 68- 105).
• Table 8 : Shows polypeptides designed from allergens which are homologous to Amb a 3
allergen (SEQ ID NOs: 150-151)
• Table 9 : Shows polypeptides designed from allergens which are homologous to Amb a 4
allergen (SEQ ID NOs: 152-167)
• Table 10: Shows polypeptides designed from allergens which are homologous to Amb a 8
allergen (SEQ ID NOs: 168-179)
• Table 11 : Shows polypeptides designed from allergens which are homologous to Amb a l l allergen (SEQ ID NOs: 180-182). According to a second aspect of the invention, there is identified polypeptides having 20-26 amino acid resides in length (SEQ ID NOs : 1-105 and 150- 182), which comprises a T cell epitope, and then suitable for being used in peptide immunotherapy. Polypeptides of sizes shorter than 30 amino acids in length are thought to be more water soluble and less prone to aggregation than longer ones. Very short polypeptides, for examples having less than 16 amino acids in length may be disadvantageous in terms of containing less epitopes than the longer ones, and prone to losing biological acitivy due to enzymatic degradation.
The inventors have found immunodominant polypeptides capable of inducing T cell proliferation in a high fraction of a ragweed allergic population, e.g. in above 25%, 30%, 35%, 40%, 45%, 50%, or above 60% of the allergic population. For example, a polypeptide with SEQ ID NO : 2 (polypeptide of Amb_a_3.0101) ; a polypeptide with any one of SEQ ID NOs : 11, 13, 19, 21, 24 or 25
(polypeptide of Amb_a_4.0101, optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue) ; a polypeptide with SEQ ID NOs: 30 (polypeptides of Amb_a _5.0101), optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue) ; a polypeptide with any one of SEQ ID NOs: 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64 or 66 (polypeptide of Amb_a_8.0101, optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue) ; a polypeptide with SEQ ID NOs: 50 or 61 (polypeptide of Amb_a_8.0102, optionally wherein a cysteine residue of the wild type sequence has been replaced with a serine residue); and a polypeptide with any one of SEQ ID NOs : 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105 (polypeptide of Amb_a_11.0101 optionally wherein a cysteine residue of the wild type sequence is replaced with a serine residue). Therefore, in some embodiments, a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs : 2, 11, 13, 19, 21, 24, 25, 30, 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64, 66, 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105; or a variant thereof which is 9 to 30 amino acids in length and comprises:
i) an amino acid sequence set forth in any one of SEQ ID NOs : 2, 11, 13, 19, 21, 24, 25, 30, 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64, 66, 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105;
ii) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of i);
iii) 9 or at least 9 contiguous amino acid residues of the sequence of i); or
iv) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of iii) . Polypeptides with SEQ ID NOs : 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64 or 66
(derived from Amb a 8.0101 and/or Amb a 8.0102) are considered more immunodominant than the remaining polypeptides shown in table 4 and polypeptides with SEQ ID NOs : 42, 44, 46 or 55 are considered even more immunodominant as these polypeptides produced the highest responder frequencies. Therefore, in some embodiments, a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 37, 38, 40, 42, 44, 46, 49, 50, 51, 55, 58, 60, 61, 62, 63, 64 and 66, such as SEQ ID NOs : 42, 44, 46 and 55; or a variant thereof which is 9 to 30 amino acids in length and comprises:
i) an amino acid sequence set forth in any one of SEQ ID NOs : 37-52, 53-56 and 60-63; ii) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of i);
iii) 9 or at least 9 contiguous amino acid of the sequence of i); or
iv) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of iii) .
Also several immunodominant polypeptides could be derived from Amb a l l. For example, polypeptides with any one of SEQ ID NOs: 71, 73, 76, 78, 82, 83, 88, 89, 90, 93, 95, 98, 100 and 105 are considered more immunodominant than the remaining polypeptides of table 5.
Polypeptides with any one of SEQ ID NOs: 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105 produced even higher responder frequencies and polypeptides with any one of SEQ ID NOs : 83, 93, 98 and 105 produced the highest responder frequencies.
Therefore, in some embodiments, a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 71, 73, 76, 78, 82, 83, 88, 89, 90, 93, 95, 98, 100 and 105, such as SEQ ID NOs: 76, 78, 82, 83, 88, 89, 93, 95, 98, 100 and 105, such as SEQ ID NOs: 83, 93, 98 and 105; or a variant thereof which is 9 to 30 amino acids in length and comprises:
i) an amino acid sequence set forth in any one of SEQ ID NOs : 71, 73, 76, 78, 82, 83, 88, 89, 90, 93, 95, 98, 100 and 105;
ii) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of
0;
iii) 9 or at least 9 contiguous amino acid residues of the sequence of i); or
iv) an amino acid sequence with at least 65% sequence identity to an amino acid sequence of iii) .
In some embodiments of the second aspect, a polypeptide consists of an amino acid sequence with SEQ ID NOs : 1- 105, wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at N- and/or C-terminal end.
Examples of polypeptides with one, two or three amino acids substitutions in the sequence selected from any one of SEQ ID NOs: 1-105 (Amb a polypeptides) may be designed from the amino acid sequence of homolog allergens present in ragweed pollen of Amb p, Amb t or Art v. Non-limiting examples of homolog allergens may be selected from any one of SEQ ID NOs: 126- 149 (shown in table 7) .
In some embodiments of the invention, a polypeptide with one, two or three amino acids substitutions in the sequence selected from any one of SEQ ID NOs: 1- 105 is selected from the group consisting of SEQ ID NOs : 150- 182. According to the invention, there is identified polypeptides capable of inducing T cell-mediated immune responses in a high fraction of ragweed pollen allergic subjects and with high cross reactivity between polypeptides designed from source allergens from Amb a and their homolog counterparts of Amb t, Amb p and Art v. Examples of such polypeptides designed from Amb a 4 and homolog allergens are shown in figures 8-9 and table 2.
Therefore, in some embodiments, a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 10-15, 18-21 and 24-25, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end . Optionally, the additional amino acid residues are from a source allergen selected from any one of SEQ ID NO: 184, 129 and 147.
In some embodiments, a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 11 is a polypeptide having an amino acid sequence of SEQ ID NOs: 10, 153, 155, or 157. Thus, a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 11, 10, 153, 155 and 157.
In some embodiments, a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 15 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NOs : 14, 159 or 161. Thus, a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 14, 15, 159 and 161.
In some embodiments, a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 19 is a polypeptide having an amino acid sequence of SEQ ID NOs: 18, 163, 165, or 167. Thus, a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 18, 19, 163, 165 and 167.
Examples of immunodominant and cross reactive polypeptides designed from Amb a 8 or homolog allergens are shown in figures 10- 12 and table 4. Examples of T cell responder frequencies are shown in figures 3 and 4 and examples of the cross reactivity between Amb a, Amb p, Amb t and Art v polypeptides are shown in figures 4 and 5.
Accordingly, in some embodiments a polypeptide consists of an amino acid sequence selected fro any one of SEQ ID NOs: 37-40, 42-51, 53-56, 61 and 63, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end. Optionally, the additional amino acid residues are from a source allergen selected from any one of SEQ ID NOs: 186 and 130-144. In some embodiments, the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 44 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 168- 173. Thus, a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 44 and 168-173. In some embodiments, the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 55 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 174- 179. Thus, a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 55,174- 179. Examples of immunodominant and cross reactive polypeptides designed from Amb a 11 or homolog allergens are shown in figures 13- 14 and table 5.
Accordingly, in some embodiments a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 76-78, 81-83, 88, 89, 92-95, 99, 100 and 105, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues, and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end. Optionally, the additional amino acid residues are from a source allergen selected from any one of SEQ ID NOs: 188, 145 and 146.
In some embodiments, the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 83 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 180 or 181. Thus, a polypeptide capable of inducing a T cell-mediated immune response in a ragweed and/or mugwort pollen allergic subject may consist of an amino acid sequence selected from any one of SEQ ID NO: 83, 180 and 181.
In some embodiments, the polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 89 is a polypeptide having an amino acid sequence with SEQ ID NO : 182.
Examples of immunodominant and cross reactive polypeptides deriving from Amb a 3 or homolog allergens are shown in figure 7 and table 1.
Accordingly, in some embodiments a polypeptide consists of an amino acid sequence selected from any one of SEQ ID NO: 2, wherein optionally the amino acid sequence of the polypeptide may be modified by substitution of one, two or three amino acid residues and wherein the polypeptide optionally may be further modified by the addition of one to five amino acid residues at the N- and/or the C-terminal end or by deletion of one, two or three amino acid residues at the N- and/or C-terminal end . Optionally, the additional amino acid residues are from a source allergen selected from any one of SEQ ID NOs: 183, 126- 128.
In some embodiments, the polypeptide which consists of an amino acid sequence selected from any one of SEQ ID NO : 2 is a polypeptide having an amino acid sequence of SEQ ID NO: 141 or 142.
As mentioned, in some embodiments, the polypeptide may be elongated with one to five additional amino acid residues added at the N- and/or the C-terminal end of the polypeptide. Thus, in some embodiments the elongation may be up to ten amino acids if the polypeptide is elongated with five amino acids at both the N- and C-terminal end. In further embodiments thereof, the one to five additional amino acid residues may be contiguous amino acids of an amino acid sequence present in the source allergen from which the polypeptide is designed / derived . In some embodiments, the polypeptide may be elongated with one to four additional amino acid residues, for example one, two or three additional amino acid residues, optionally wherein the additional amino acid residues may be contiguous amino acids of an amino acid sequence of the source allergen from which the polypeptide is designed.
For example, the polypeptide of SEQ ID NO: 44, which consists of 20 amino acid residues, may be elongated in the C- and/or N- terminal end with one to five amino acid residues selected from the allergen Amb_a_8.0101. For example, it may be elongated by one to five contiguous amino acid residues positioned adjacent to the amino acid sequence of SEQ ID NO : 44, when the sequence of SEQ ID NO : 44 is aligned with the best fit to the sequence of Amb_a_8.0101.
Further polypeptides of the invention may be designed from any one of the polypeptides with SEQ ID NOs: 1- 105 and 150 and 182, by incorporating one, two or three amino acid substitutions in the sequence. As mentioned, further polypeptides of the invention may be designed by elongation of the amino acid sequence of the polypeptide in the N- and/or C-terminal end, including polypeptides with one, two or three amino acid substitutions in the sequence, or by shortening the polypeptide, including those with one, two or three amino acid substitutions in the sequence, with one, two or three amino acid residues from the N- and/or C-termina l end.
Embodiments of the third aspect
As mentioned, the inventors have identified homolog allergens of Amb a 3, 4, 8 and 11 in other ragweed pollen species of the genus Ambrosia, i.e. Amb t, Amb p and Art v. The amino acid sequences of these novel full length allergens are shown in table 7. Therefore, some embodiments relate to a polypeptide consisting of an amino acid sequence having 85% to 100% sequence identity with an amino acid sequence selected from any one of SEQ ID NOs : 126-146. According to some further embodiments, the sequence identity is in the range of 90% -100%, for example in the range of 92% - 100%, 95-100%, 97- 100%, 98- 100%, or 99- 100%. Thus, in some embodiments, the polypeptide has an amino acid sequence which is identical with any one of SEQ ID NOs: 126- 146, for example the sequence identity may be in the range of 85-99%, for example in the range of 90-99%, 95-99%, 96-99%, 97-99%, 98-99% compared to a polypeptide selected from any one of SEQ ID NOs: 126- 146. Polypeptides having a non-identical amino acid sequence may have the same biological activity or functionality as the original polypeptide. In some embodiments, the same biological activity or functionality means the same enzymatic functionality; the same, greater or less ability to elicit, stimulate or induce an an in vitro or ex vivo T cell proliferation or T cell cytokine production (e.g. cytokines is selected from IL-4, IL- 5, IL-13 and/or IL- 10) of T cells from subjects allergic to ragweed pollen (e.g . ragweed pollen of the species Amb a, Amp p, Amb t or Art v; the same, greater or less ability to induce
immunological tolerance against ragweed pollen, a ragweed allergen; or the same, greater or less ability to bind or interact with IgE, IgG or IgA antibodies raised against the original polypeptide.
Examples of polypeptides homologous to Amb a 3 is selected from any one of SEQ ID NOs: 126- 128.
An example of a polypeptide which is homologous to Amb a 4 is SEQ ID NO : 129 Examples of polypeptides homologous to Amb a 8 is selected from any one of SEQ ID NOs: 130- 144.
Examples of polypeptides homologous to Amb a 11 is selected from any one of SEQ ID NOs: 145- 146.
The polypeptides may be a recombinant polypeptide, thus produced by recombinant manufacturing process and optionally comprising post-translational modifications, such as for example
glycosylation, phosphorylation, lipidation and/or formation of disulfide bonds.
Therefore, a third aspect of the invention, also relates to an isolated nucleic acid molecule, which comprises a nucleotide sequence encoding a polypeptide of the third aspect, for example the nucleic acid molecule is a DNA or an RNA molecule. Further embodiments, relates to a vector comprising the nucleic acid molecule, such as a cloning vector or an expression vector, or example a virus, such as an attenuated virus, a bacteriophage, a plasmid, a minichromosome, and a cosmid. Further embodiments relates to a cell which is transformed to carry the vector, for example a prokaryotic cell or an eukaryotic cell. A further embodiment relates to a cell line derived from the transformed cell. A further aspect relates to a method for the preparation of a polypeptide of the third aspect comprising
- culturing a transformed cell as mentioned above or the cell line as mentioned above under conditions that facilitate that the transformed cell expresses the nucleic acid molecule and subsequently recovering said polypeptide, or
- preparing said polypeptide by means of solid or liquid phase peptide synthesis.
Further embodiments of the first, second and third aspects
Polypeptides of the first and second aspect may be subject to further modifications in order to increase or decrease physical or chemical properties, for example to decrease its ability to resist oxidation, to improve or increase solubility in aqueous solution, to decrease aggregation, to decrease synthesis problems, etc.
Non-limiting examples of one or more of the modifications may be selected from the following : a) N-terminal acetylation or methylation
b) C terminal amidation
c) C-terminal amination
d) addition of one to three charged amino acid residues selected from any one of lysine (K), arginine (R), aspartic acid (D), and glutamic acid (E) in the N-terminal or C-terminal e) addition of one to three lysine residues in the N-terminal or C-terminal
f) one or more cysteine residues replaced with serine or 2-aminobutyric acid
g) one or more serine residues replaced with cysteine
h) one or more hydrogen on the side chain amines of arginine and/or lysine replaced with a methylene group
i) glycosylation, and
j) phosphorylation
When designing further examples of polypeptides according to the invention, it might be considered to designt polypeptides which have similar or greater biological activity. Thus, a polypeptide according to the first, second or third aspect may elicit, stimulate or induce an in vitro or ex vivo T cell proliferation response or an in vitro or ex vivo T cell cytokine (e.g. IL-4, IL-5, IL- 13 and/or IL- 10) production from T cells of subjects allergic to ragweed pollen (e.g. ragweed pollen of the species Amb a, Amp p, Amb t or Art v) .
A polypeptide selected from any one of SEQ ID NOs: 1-105 and 150- 182 is in the present context regarded as an original polypeptide that may be subject to one or more modifications as described herein. Likewise, a polypeptide designed from the epitope regions of SEQ ID NOs: 106- 125 and 189-224 may also be regarded as original polypeptides subject to modifications as described herein. Finally, a polypeptide comprising 9 or at least 9 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 106 to 125 and 189-224 may also be regarded as original polypeptides subject to modifications as described herein. The modification of an original polypeptide may optionally result in greater or less activity or function of the original polypeptide, for example in the ability to elicit, stimulate or induce an in vitro immune response (e.g . T cell proliferation or T cell cytokine production) ; in the ability to bind to Class HLA II alleles (HLA coverages) ; in the ability to induce or enhance immunological tolerance to a relevant allergen, such as Amb a 1, Amb a 3, Amb a 4, Amb a 5, Amb a 8 or Amb a 11 or any combination of the allergens including an allergen extract of ragweed pollen of any species selected from Amb a, Amb t and Amb p; or in the ability to dissolve e.g. in an aqueous solution; or in the ability to aggregate, such as fibrillate.
As mentioned, a polypeptide having an amino acid sequence which is non-identical to an original peptide may share about the same functionality or activity as the original polypeptide, e.g . provide slightly poorer functionality or activity, or may have improved functionality or activity. For example, the non-identical polypeptide may bind to at least 70%, such as at least 75%, 80%, 85%, 90% or 95% of the group of Class HLA II alleles that the original peptide binds, optionally wherein the group of HLA Class II alleles is selected from alleles set out in tables 12 or 13. The ability of the original peptide and the non-identical polypeptide to bind HLA Class II alleles may be tested under the same test conditions, for example by use of HLA binding prediction tools or in-vitro binding assay.
Furthermore, a non-identical polypeptide may comprise one or more of the same T cell epitopes as the original polypeptide. 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 non-identical polypeptide versus the original polypeptide, 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)
Some modifications of the original polypeptide correspond to naturally occurring variations of the wild type a llergens, such as variations between isoforms (intra species variation) of the same species or variations between homologous allergens of other ragweed species (inter species variation). Other modifications are non-natural modifications of the parent peptide.
A non-identical polypeptide may for instance include one or more deletions of amino acid residues from the N- and/or C- terminal end of the original polypeptide, one or more additions of amino acid residues to the N- and/or C- terminal of the original peptide and/or one or more amino acid substitutions, additions or deletions within the amino acid sequence of the original peptide, thus resulting in various lengths of the polypeptide. For example, the non-identical polypeptide may be 12 amino acids in length, such as 14, 15, 16, 17, 18, 19, 20, 22, 24, 26, 28 or 30 amino acids in length, or even longer, such as 35, 40, 45, 50, 55 or 60 amino acids in length. In some instances, the non-identical polypeptide is 15 to 30 amino acids in length, such as 15 to 26, such as 16 to 30, 16 to 28, 16 to 26, or 16 to 24, such as 18 to 30, 18 to 28, 18 to 26, or 18 to 24.
A non-identical polypeptide may comprise i) the entire amino acid sequence set forth in any one of SEQ ID NOs : 1- 105 and 150-182 or ii) an amino acid sequence with at least 65% identity to any one of SEQ ID NOs: 1- 105 and 150- 182, such as at least 70%, 75%, 80%, 85%, 90% or 95% sequence identity to any one of SEQ ID NOs: 1- 105 and 150-182. Furthermore, a non-identical polypeptide may comprise 9 or at least 9 contiguous amino acid amino acids selected from any one of SEQ ID NOs : 1-105 and 150- 182, such as 12, 14, 15, 16, 17, 18 or 19 contiguous amino acids selected from any one of SEQ ID NOs: 1-105 and 150- 182, or such as at least 12, 14, 15, 16, 17, 18 or 19 contiguous amino acids selected from any one of SEQ ID NOs: 1-105 and 150- 182. Still further non-identical polypolypeptides may comprise an amino acid sequence with at least 65% sequence identity, such as at least 75%, 80%, 85%, 90% or 95% sequence identity to the 12, 14,
15, 16, 17, 18 or 19 contiguous amino acids selected from any one of SEQ ID NOs: 1-105 and 150- 182, or such as at least 75%, 80%, 85%, 90% or 95% sequence identity to the at least 12, 14, 15,
16, 17, 18 or 19 contiguous amino acids selected from any one of SEQ ID NOs: 1-105 and 150- 182.
For example, in some embodiments, the polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 1-105 and 150- 182; or a variant thereof, which has a length of 15 to 30 amino acids and comprises:
i) an amino acid sequence selected from any one of SEQ ID NOs: 1- 105 and 150- 182;
ii) an amino acid sequence with at least 80% sequence identity to an amino acid sequence of i); iii) at least 15 contiguous amino acid amino acids of the sequence of i) ; or
iv) an amino acid sequence with at least 80% sequence identity to an amino acid sequence of iii). In still other embodiments, the variant is 17 to 30 amino acids in length and comprises: i) an amino acid sequence selected from any one of SEQ ID NOs: 1- 105 and 150- 182;
ii) an amino acid sequence with at least 85% sequence identity to an amino acid sequence of i); iii) at least 17 contiguous amino acid amino acids of the sequence of i) ; or
iv) an amino acid sequence with at least 85% sequence identity to an amino acid sequence of iii).
In some embodiments of the first, second or third aspect, a variant or substitutions in the original polypeptide comprises conservative or non-conservative amino acid substitutions. A "conservative substitution" is the replacement of one amino acid by a biologically, chemically or structurally similar residue. Biological similarity means that the substitution does not destroy a biological activity, e.g. T cell reactivity or HLA coverage. Structural similarity 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 both 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, for example 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 acid for aspartic acid, 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, He, 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.
A polypeptide of the first and second aspect may be provided in the form of a salt, for example as a pharmaceutically acceptable and/or a physiologically acceptable salt, e.g. an acetate salt.
Polypeptides of the first, second or third aspect may be produced by peptide synthesis (e.g the polypeptide is synthetic), whereas polypeptides of the third aspect may also be produced by recombinant techniques. A polypeptide of the first, second and third aspect may be provided as a powder, for example as a freeze-dried (lyophilized) powder. Thus, polypeptide may be provided in a storage-stable form with low presence of water (e.g. below 10%, for example below 5, 4, 3, 2 or 1% of water) or in a form ready to be administered in dry form (e.g . tablet or capsule) or to be administered in soluble form (e.g. dissolved in aqueous solution) . The concentration of each of the dissolved polypeptides may be in a molar concentration in the range of 1 to 1000 μΜ, for example in the range of 10 to 800 μΜ, for example in the range of 20 to 500 μΜ, for example in the range of 20 to 300 μΜ.
In particular embodiments, a variant may comprise a fusion (chimeric) sequence of
polypolypeptides, which optionally may contain an amino acid sequence having one or more molecules not normally present in a reference (wild type, original) sequence covalently attached to the polypeptide 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 variant is one in which a second heterologous sequence, i.e. a heterologous functional domain, is attached to a polypeptide disclosed herein, (covalent or non- covalent binding) that may confer a distinct or complementary function to a polypeptide disclosed herein. Heterologous functional domains are not restricted to amino acid residues. Thus, a heterologous functional domain can consist of any one of a variety of different types of small or large functional moieties. Such moieties include nucleic acid, polypeptide, carbohydrate, lipid or small organic compounds, such as a drug (e.g . an antiviral), a metal (gold, silver), or a
radioisotope.
Embodiments of fourth aspect
A fourth aspect of the invention relates to a chimeric molecule comprising the fusion of two or more of the polypeptides selected from any one of the first, second, or third aspect.
In some embodiments of the invention, the chimeric molecule comprises the fusion of two or more of the amino acid sequences selected from any of SEQ ID NOs: 1- 105 and 150-182. In some embodiments of the invention, the chimeric molecule comprises the fusion of two or more of the amino acid sequences selected from any of SEQ ID NOs: 106-125 and 189-224.
In some embodiments of the invention, the chimeric molecule comprises the fusion of two or more of the amino acid sequences selected from any of SEQ ID NOs: 126-149.
In particular embodiments thereof, the amino acid sequence of the chimeric molecule may comprise one or more molecules covalently attached to the 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.
Covalently linking may involve direct linking through the amino acid backbone, in which case two polypeptides are linked by connecting, either directly or by using a spacer amino acid sequence, the N-terminal of one polypeptide to the C-terminal of another polypeptide, which produces a fusion or mosaic protein. Other types of covalent linking may involve the formation of disulfide bonds formed by cysteine amino acids, or other types of covalent bonds between one entity of polypeptide to a second entity of polypeptide. In one embodiment, the covalently link is through the amino acid backbone of a polypeptide to a heterologous functional domain. In another specific embodiment, the covalently link is through a cysteine side chain of a polypeptide a heterologous functional domain.
Spacers, such as amino acid or peptidomimetic sequences, may be inserted between the polypeptide sequence and the addition (e.g., heterologous functional domain) so that the two entities maintain, at least in part, a distinct function or activity. Spacers 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 another aspect of the invention, the polypeptides of the first, second or third aspect are not provided as individual entities, but may be fused together or to a carrier molecule to form an isolated molecule. Such carriers may contribute to presenting the epitopes of the polypeptides of first, second and third aspect to the immune system of the subject to be treated.
Therfore, a fourth aspect of the invention relates to a polypeptide of the first, second or third aspected linked covalently or non-covalently to a component or carrier, for example to various other components, such as proteins capable of forming virus like particles (VLPs), keyhole limpet hemocyanin (KLH), invariant chain or other proteins or polypeptides, which constitute a
heterologous functional domain that may or may not be derived from other species than plants. Further examples of components or carriers are nanoparticles, microparticles or liposomes.
In some embodiments of the invention, one or more amino acid sequence(s) selected from any of SEQ ID NOs : 1- 105 and 150-182 is linked covalently or non-covalently to a component or carrier. In some embodiments of the invention, one or more amino acid sequence(s) selected any of SEQ ID NOs: 106-125 and 189-224 is linked covalently or non-covalently to a component or carrier.
In some embodiments of the invention, one or more amino acid sequence(s) selected from any of SEQ ID NOs : 126-149 is linked covalently or non-covalently to a component or carrier.
In one embodiment of the present invention, polypeptides of the first, second and third aspect, e.g. selected from any one SEQ ID NOs : 1-105, 150-182, 106-125 and 189-224 and SEQ ID NOs: 126- 149 are linked to a VLP. The preparation of VLPs is performed by recombinantly expressing the coat protein in a host is within the common knowledge of a skilled artisan. In one embodiment, the virus-like particle comprises, or alternatively consists of, recombinant proteins, mutants or fragments thereof, of a virus selected form the group consisting of: a) RNA phages; b)
bacteriophages; c) Hepatitis B virus, preferably its capsid protein (Ulrich, et al, Virus Res. 50 : 141- 182 (1998)) or its surface protein (WO 92/11291), such as described in WO2012168487; d) measles virus (Warnes, et al., Gene 160: 173- 178 ( 1995)) ; e) Sindbis virus; f) rotavirus (US 5,071,651 and US 5,374,426) ; g) foot-and-mouth-disease virus (Twomey, et al., Vaccine 13 : 1603 1610, (1995)) ; h) Norwalk virus (Jiang, X., et al. , Science 250: 1580 1583 ( 1990); Matsui, S. M ., et al., J . Clin. Invest. 87: 1456 1461 ( 1991)); i) Alphavirus; j) retrovirus, preferably its GAG protein (WO 96/30523) ; k) retrotransposon Ty, preferably the protein pi; I) human Papilloma virus (WO 98/15631); m) Polyoma virus; n) Tobacco mosaic virus; o) Flock House Virus; and p) Cucumber mosaic virus (CMV), such as described in WO201704224 or WO16062720; q) a virus of the pi- cornaviridae family such as a species of rhinoviruses, such as rhinovirus 89 and 14, in which case the virus-like particle may be formed by one or more of the proteins selected from VP1, VP2, Vp3 and/or VP4 as described in WO2006097530; r) as papaya mosaic virus such as mentioned in WO08058369; and s) enterovirus as described in WO08092996.
In one embodiment, the virus-like particle is of Hepatitis B virus. The preparation of Hepatitis B virus-like particles have been disclosed, inter alia, in WO 00/32227, WO 01/85208, WO 02/056905 and WO2012168487.
In another embodiment, the virus-like particle is a virus of the pi-cornaviridae family such as a species of rhinoviruses, such as rhinovirus 89 and 14, in which case the virus-like particle may be formed by one or more of the proteins selected from VP1, VP2, VP3 and/or VP4 as described in WO2006097530. In another embodiment, the virus-like particle is a virus-like (VLP) particle derived from RNA bacteriophage AP205 such as described in WO0603267.
Methods for the production of fusion proteins are well known in the art and can be found in standard molecular biology references such as Sambrook et al. (Molecular Cloning, 2nd ed. , Cold Spring Harbor Laboratory Press, 1989) and Ausubel et al . (Short Protocols in Molecular Biology, 3rd ed; Wiley and Sons, 1995). In general, a fusion protein is produced by first constructing a fusion gene which is inserted into a suitable expression vector, which is, in turn, used to transfect a suitable hosT-cell. In general, recombinant fusion constructs are produced by a series of restriction enzyme digestions and ligation reactions which result in the desired sequences being incorporated into a plasmid . If suitable restriction sites are not available, synthetic oligonucleotide adapters or linkers can be used as is known by those skilled in the art and described in the references cited above. The polynucleotide sequences encoding allergens and native proteins can be assembled prior to insertion into a suitable vector or the sequence encoding the allergen can be inserted adjacent to a sequence encoding a native sequence already present in a vector. Insertion of the sequence within the vector should be in frame so that the sequence can be transcribed into a protein. It will be apparent to those of ordinary skill in the art that the precise restriction enzymes, linkers and/or adaptors required as well as the precise reaction conditions will vary with the sequences and cloning vectors used. The assembly of DNA constructs, however, is routine in the art and can be readily accomplished by a person skilled in the art.
In one embodiment, the proteins or polypeptides of the present invention are linked either covalently or non-covalently to nanoparticles, microparticles or liposomes, for example such as described in WO07068747, WO07144150 and WO08071774. Embodiments of the fifth aspect.
According to a fifth aspect, one or more of the polypolypeptides of the first, second or third aspect, such as polypeptides of the first and second aspect, may be comprised in a composition, optionally in combination with polypolypeptides designed from other allergens. The composition may further comprise other excipients. Accordingly, the invention relates in a fifth aspect to a composition, such as a pharmaceutical composition, which comprises one or more of the polypeptides of the first, second or third aspect. A pharmaceutical composition typically comprises a pharmaceutically acceptable carrier, excipient and/or adj uvant and may optionally be sterile.
A composition, such as a pharmaceutical composition may comprise one or more of the
polypeptides with high T cell responder frequencies and cross reactivity.
A compostion may further comprise one or more immunodominant polypeptides from a group 1 allergen of ragweed pollen. Such polypeptides may comprise from 9 to 30 contiguous amino acids of a sequence of Amb a 1, for example of a sequence of selected among any one of the isoforms Amb_a_1.0201(UniProt No : P27760), Amb_a_1.0202(UniProt No : E1XUL3), Amb_a_1.0301(UniProt No : P27761), Amb_a_1.0302 (UniProt No: P27761(variantL48Y)), Amb_a_1.0303(UniProt No: P27761(variantH392R), Amb_a_1.0304(UniProt No : E1XUL4), Amb_a_1.0305(UniProt No:
E1XUL5), Amb_a_1.0401 (UniProt No : P28744), Amb_a_1.0402(UniProt No : E1XUL9),
Amb_a_1.0501(UniProt No : P27762) and/or Amb_a_1.0502(UniProt No: E1XUM 1), optionally wherein any one of cysteine residues in the wild type allergen is replaced with a serine residue. The isoforms have the following sequences (SEQ ID NOs: 225-236:
Amba l .0101_P27759 (SEQ ID NO : 225)
MGIKHCCYILYFTLALVTLLQPVRSAEDLQEILPVNETRRLTTSGAYNIIDGCWRGKADWAENRKALADCAQGFG KGTVGGKDGDIYTVTSELDDDVANPKEGTLRFGAAQNRPLWIIFERDMVIRLDKEMVVNSDKTIDGRGAKVEIIN AGFTLNGVKNVIIHNINMHDVKVNPGGLIKSNDGPAAPRAGSDGDAISISGSSQIWIDHCSLSKSVDGLVDAKL GTTRLTVSNSLFTQHQFVLLFGAGDENIEDRGMLATVAFNTFTDNVDQRMPRCRHGFFQVVNNNYDKWGSYAI GGSASPTILSQGNRFCAPDERSKKNVLGRHGEAAAESMKWNWRTNKDVLENGAIFVASGVDPVLTPEQSAGMI PAEPGESALSLTSSAGVLSCQPGAPC
Amba l .0201_P27760 (SEQ ID NO : 226)
MGIKHCCYILYFTLALVTLLQPVRSAEDVEEFLPSANETRRSLKACEAHNIIDKCWRCKADWANNRQALADCAQG FAKGTYGGKHGDVYTVTSDKDDDVANPKEGTLRFAAAQNRPLWIIFKRNMVIHLNQELVVNSDKTIDGRGVKVN IVNAGLTLMNVKNIIIHNINIHDIKVCPGGMIKSNDGPPILRQQSDGDAINVAGSSQIWIDHCSLSKASDGLLDIT LGSSHVTVSNCKFTQHQFVLLLGADDTHYQDKGMLATVAFNMFTDHVDQRM PRCRFGFFQVVNNNYDRWGTY AIGGSSAPTILSQGNRFFAPDDIIKKNVLARTGTGNAESMSWNWRTDRDLLENGAIFLPSGSDPVLTPEQKAGMI PAEPGEAVLRLTSSAGVLSCHQGAPC Amba l .0202_ElXUL3 (SEQ ID NO: 227)
MGIKHCCYILYFTLALVTLLQPVRSAEDVEEFLPSANETRRSLKACEAHNIIDKCWRCKADWANNRQALADCAQG FAKGTYGGKHGDVYTVTSDKDDDVANPKEGTLRFAAAQNRPLWIIFKRNMVIHLNQELVVNSDKTIDGRGVKVN IVNAGLTLMNVKNIIIHNI IHDIKVCPGGMIKSNDGPPILRQQSDGDAINVAGSSQIWIDHCSLSKASDGLLDIT LGSSHVTVSNCKFTQHQFVLLLGADDTHYQDKGMLATVAFNMFTDHVDQRMPRCRFGFFQVVNNNYDRWGTY AIGGSSAPTILSQGNRFFAPDDIIKKNVLARTGTGNAESMSWNWRTDKDLLENGAIFLPSGSDPVLTPEQKAGMI PA E PG EAVLR LTS SAG V LSC H QGA PC
Amba l .0301_P27761 (SEQ ID NO : 228)
MGIKQCCYILYFTLALVALLQPVRSAEGVGEILPSVNETRSLQACEALNIIDKCWRGKADWENNRQALADCAQGF AKGTYGGKWGDVYTVTSNLDDDVANPKEGTLRFAAAQNRPLWIIFKNDMVINLNQELVVNSDKTIDGRGVKVEI INGGLTLMNVKNIIIHNINIHDVKVLPGGMIKSNDGPPILRQASDGDTINVAGSSQIWIDHCSLSKSFDGLVDVTL GSTHVTISNCKFTQQSKAILLGADDTHVQDKGM LATVAFNMFTDNVDQRMPRCRFGFFQVVNNNYDRWGTYAI GGSSAPTILCQGNRFLAPDDQIKKNVLARTGTGAAESMAWNWRSDKDLLENGAIFVTSGSDPVLTPVQSAGMIP AEPGEAAIKLTSSAGVFSCHPGAPC Amba l .0302_P27761(variantL48Y) (SEQ ID NO : 229)
MGIKQCCYILYFTLALVALLQPVRSAEGVGEILPSVNETRSLQACEAYNIIDKCWRGKADWENNRQALADCAQGF AKGTYGGKWGDVYTVTSNLDDDVANPKEGTLRFAAAQNRPLWIIFKNDMVINLNQELVVNSDKTIDGRGVKVEI INGGLTLMNVKNIIIHNINIHDVKVLPGGMIKSNDGPPILRQASDGDTINVAGSSQIWIDHCSLSKSFDGLVDVTL GSTHVTISNCKFTQQSKAILLGADDTHVQDKGM LATVAFNMFTDNVDQRMPRCRFGFFQVVNNNYDRWGTYAI GGSSAPTILCQGNRFLAPDDQIKKNVLARTGTGAAESMAWNWRSDKDLLENGAIFVTSGSDPVLTPVQSAGMIP AEPGEAAIKLTSSAGVFSCHPGAPC
Amba l .0303_P27761(variantH392R) (SEQ ID NO: 230)
MGIKQCCYILYFTLALVALLQPVRSAEGVGEILPSVNETRSLQACEALNIIDKCWRGKADWENNRQALADCAQGF AKGTYGGKWGDVYTVTSNLDDDVANPKEGTLRFAAAQNRPLWIIFKNDMVINLNQELVVNSDKTIDGRGVKVEI INGGLTLMNVKNIIIHNINIHDVKVLPGGMIKSNDGPPILRQASDGDTINVAGSSQIWIDHCSLSKSFDGLVDVTL GSTHVTISNCKFTQQSKAILLGADDTHVQDKGM LATVAFNMFTDNVDQRMPRCRFGFFQVVNNNYDRWGTYAI GGSSAPTILCQGNRFLAPDDQIKKNVLARTGTGAAESMAWNWRSDKDLLENGAIFVTSGSDPVLTPVQSAGMIP AEPGEAAIKLTSSAGVFSCRPGAPC
Amba l .0304_ElXUL4 (SEQ ID NO: 231) MGIKQCCYILYFTLALVALLQPVRSAEGVGEILPSVNETRSLQACEALNIIDKCWRGKADWENNRQALADCAQGF AKGTYGGKWGDVYTVTSNLDDDVANPKEGTLRFAAAQNRPLWIIFKNDMVINLNQELVVNSDKTIDGRGVKVEI INGGLTLMNVKNIIIHNINIHDVKVLPGGMIKSNDGPPILRQASDGDTINVAGSSQIWIDHCSLSKSFDGLVDVTL GSTHVTISNCKFTQQSKAILLGADDTHVQDKGM LATVAFNMFTDNVDQRMPRCRFGFFQVVNNNYDRWGTYAI GGSSAPTILCQGNRFLAPDDQIKKNVLARTGTGAAESMAWNWRSDKDLLENGAIFVTSGSDPVLTPVQSAGMIP AEPGEAAIKLTSSAGVLSCRPGAPC
Amba l .0305_ElXUL5 (SEQ ID NO: 232)
MGIKQCCYILYFTLALVALLQPVRSAEGVGEILPSVNETRSLQACEAYNIIDKCWRGKADWENNRQALADCAQGF AKGTYGGKWGDVYTVTSNLDDDVANPKEGTLRFAAAQNRPLWIIFKNDMVINLNQELVVNSDKTIDGRGVKVEI INGGLTLMNVKNIIIHNINIHDVKVLPGGMIKSNDGPPILRQASDGDTINVAGSSQIWIDHCSLSKSFDGLVDVTL GSTHVTISNCKFTQQSKAILLGADDTHVQDKGM LATVAFNMFTDNVDQRMPRCRFGFFQVVNNNYDRWGTYAI GGSSAPTI LCQGNRFLAPDDQIKKNVLARTGTGAAESMAWNWRSDKDLLENGAIFVTSGSDPVLTPVQSAGMIP AEPGEAAIKLTSSAGVLSCRPGAPC Amba l .0401_P28744 (SEQ ID NO : 233)
MGIKHCCYILYFTLALVTLLQPVRSAEDLQQILPSANETRSLTTCGTYNIIDGCWRGKADWAENRKALADCAQGF AKGTIGGKDGDIYTVTSELDDDVANPKEGTLRFGAAQNRPLWIIFARDMVIRLDRELAINNDKTIDGRGAKVEIIN AGFAIYNVKNIIIHNIIMHDIVVNPGGLIKSHDGPPVPRKGSDGDAIGISGGSQIWIDHCSLSKAVDGLIDAKHGS THFTVSNCLFTQHQYLLLFWDFDERGMLCTVAFNKFTDNVDQRMPNLRHGFVQVVNNNYERWGSYALGGSAGP TILSQGNRFLASDIKKEVVGRYGESAMSESINWNWRSYM DVFENGAIFVPSGVDPVLTPEQNAGMIPAEPGEAV LRLTSSAGVLSCQPGAPC
Amba l .0402_ElXUL9 (SEQ ID NO: 234)
AEDLQQILPSVNETRSLTTCGTYNIIDGCWRGKADWAENRKALADCAQGFAKGTIGGKDGDIYTVTSELDDDVA NPKEGTLRFGAAQNRPLWIIFARDMVIRLDRELAINNDKTIDGRGAKVEIINAGFAIYNVKNIIIHNIIM HDIVVNP GGLIKCNDGPPAPRKGSDGDAIGISGGSQIWIDHCSLSKAVDGLIDAKHGSTHFTVSNCLFTQHQYLLLFWDFD ERGMLCTVAFNKFTDNVDQRM PNLRHGFVQVVNNNYERWGSYALGGSAGPTILSQGNRFLASDIKKEVVGRYG ESAMSESINWNWRSYMDVFENGAIFVPSGVDPVLTPEQNAGMIPAEPGEAVLRLTSSAGVLSCQPGTPC
Amba l .0501_P27762 (SEQ ID NO : 235)
MGIKHCCYILYFTLALVTLVQAGRLGEEVDILPSPNDTRRSLQGCEAHNIIDKCWRCKPDWAENRQALGNCAQG FGKATHGGKWGDIYMVTSDQDDDVVNPKEGTLRFGATQDRPLWIIFQRDMIIYLQQEMVVTSDKTIDGRGAKV ELVYGGITLM VKNVIIH NIDI HDVRVLPGGRIKSNGGPAIPRHQSDGDAIHVTGSSDIWIDHCTLSKSFDG LVD VNWGSTGVTISNCKFTHHEKAVLLGASDTHFQDLKM HVTLAYNIFTNTVHERMPRCRFGFFQIVNNFYDRWDKY AIGGSSNPTILSQGNKFVAPDFIYKKNVCLRTGAQEPEWMTWNWRTQNDVLENGAIFVASGSDPVLTAEQNAG MMQAEPGDMVPQLTMNAGVLTCSPGAPC Amba l .0502_ElXUM l (SEQ ID NO : 236)
MGIKHCCYILYFTLALVTLVQAGRLGEEVDILPSPNDTRRSLQGCEAHNIIDKCWRCKPDWAENRQALGDCAQG FGKATHGGKWGDIYMVTSDQDDDVVNPKEGTLRFGATQDRPLWIIFQRDMIIYLQQEMVVTSDTTIDGRGAKV ELVYGGITLM VKNVIIH NIDI HDVRVLPGGRIKSNGGPAIPRHQSDGDAIHVTGSSDVWIDHCTLSKSFDG LVD VNWGSTGVTISNCKFTHHEKAVLLGASDTHFQDLKM HVTLAYNIFTNTVHERMPRCRFGFFQIVNNFYDRWDKY AIGGSSNPTILSQGNKFVAPDFIYKKNVCLRTGAQEPEWMTWNWRTQNDVLENGAIFVASGSDPVLTAEQNAG MMQAEPGDMVPQLTMNAGVLTCSPGAPC T cell epitope-containing polypeptides of Amb a 1 have been described by others and may be added to a composition of one or more of the polypolypeptides describe herein. Non-limiting examples are the epitope polypeptides described in the international patent applications WO10018378 and WO08098749.
The pharmaceutical composition may be a vaccine, e.g . a product for use in conducting
immunotherapy, including but not limited to a vaccine for treating an allergic immune response to ragweed pollen, i.e. pollen of the genus Ambrosia such as pollen of Amb a, Amb p and/or Amb t. The vaccine may be formulated for parenteral administration, such as by subcutaneous, intradermal, transcutaneous administration, e.g. formulated as a powder that optionally may be re- dissolved before use. A pharmaceutical composition comprises in addition to the polypeptide combination, therapeutically inactive ingredients, such as pharmaceutically acceptable or physiologically acceptable excipient(s), carrier(s) and/or adjuvant(s), which 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 adj usting agents, buffering agents, pH adjusting agents, isotonicity adj usting agents, mucoadhesive substances, and the like. Examples of formulation strategies are well-known to the person skilled in the art.
In some embodiments, the polypeptide(s) may be formulated (e.g. mixed together) with immune- modifying agents like adj uvants usually applied in immunotherapy products. 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. A liquid pharmaceutical composition may be formed by dissolving a powder, granulate or lyophilizate of a peptide 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 peptide 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 purpose 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. Typically the concentration of each polypeptide is the same, such as in an equimolar concentration, but each polypeptide of the composition may also 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, i.e. water for injection. 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 pH 5 to 8, such as in the range of pH 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 adj usted 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 or glycerol, such as isotonic saline. The liquid may contain additional excipients, such as another solvent, a solubilizing enhancing agent, ionic and non-ionic emulsifiers, 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.
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 a pH in the range of 3-8, such as a pH in the range of 4-8. A lyophilizate may contain additional ingredients, e.g. bulking agents and lyoprotectants, buffering, antioxidants, antimicrobial agents, solubilizers.
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). 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 intra lymphatic administration.
Polypeptides 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 composition described herein and wherein the instructions are for use in treating allergy to ragweed pollen. 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 WO14064543 Al, WO05049107 A2, WO06054280 A2,
WO07066341 A3 and W014188429 Al .
Embodiments of the sixth aspect
In a further aspect of the invention, a polypeptide of the first, second, third aspect or a chimeric molecule/ polypeptide linked to a carrier of the fourth aspect or a composition of the fifth aspect may be used in treating ragweed pollen allergy in a subject. Therfore a sixth aspect of the invention relates to a method for treating ragweed pollen allergy in a subject, for example to treating allergy in a subject where signs and/or symptoms of said allergy are elicited in the subject by exposure to ragweed pollen and/or exposure to at least one allergen present in ragweed pollen, wherein the method comprises administering to the subject a therapeutically effective amount of a polypeptide of the first, second, third aspect or a chimeric molecule/ polypeptide linked to a carrier of the fourth aspect or a composition of the fifth aspect. According to the sixth aspect of the invention, the treatment of allergy may comprise relieving or reducing (e.g. treating) an immune response triggered by exposure to ragweed pollen or at least one allergen of ragweed pollen in a subject; the treatment of allergy may comprise or consist of relieving one or more signs and/or symptoms of an immune response triggered by exposure to ragweed pollen or at least one allergen of ragweed pollen; the treatment of allergy may consist of or comprises inducing immunological tolerance ragweed pollen or at least one allergen of ragweed pollen; or the treatment of allergy may comprise or consist of relieving one or more signs and/or symptom(s) associated with allergic rhinitis and/or allergic conjunctivitis and/or allergic asthma and/or allergic eczema (e.g. atopic dermatitis). Polypeptides and compositions disclosed herein may produce a therapeutic or beneficial effect, which optionally may be objectively or subjectively measurable. A therapeutic or beneficial effect can but need not be complete ablation of all or any immune responses, or any symptoms caused by or associated with the exposure to ragweed pollen, such as a ragweed pollen allergen. Thus, a satisfactory clinical result is achieved when there is an incremental improvement or a partial reduction in an immune response or one or more symptoms and signs, or there is an inhibition, decrease, reduction, suppression, prevention, limit or control of worsening or progression of an immune response or one or more symptoms and signs over a short or long duration (hours, days, weeks, months, etc.) .
The signs/ symptoms of allergy mentioned above are those typically associated with the allergies treated according to the present invention, typically signs/symptoms may include one or more of the following; itchy running nose, itchy watery eyes, itchy skin and shortness of breath and the subject may experience that the signs/ symptoms will to some extent be relieved by treatment with antihistamines or steroids. In a clinical setting, the signs and symptoms may include detectable levels of IgE antibodies against one or more the mites of interest. In the event that the treatment entails or consists of relieving one or more signs or symptoms associated with allergic rhinitis, the relief is typically
- reduction of the intensity of itchy nose and/or
- reduction of the number of sneezes within a given period (e.g. daily, weekly, monthly) and/or
- reduction of the intensity of blocked nose (congestion) and/or
- reduction of the amount of nasal fluid and/or
- reduction of the eosinophilic count in nasal fluid and/or
- reduction of specific IgE antibody level (titre) in nasal fluid or in serum and/or
- reduction of basophil histamine release in blood.
It is to be noted that a "sign" of allergy is an objectively observable characteristic of the disease, whereas a "symptom" is the subject's subjective experience(s) relative to the disease. Some signs can be symptoms and vice versa, but if a subject for instance experiences dizziness due to a disease, this can only be categorized as a symptom, because it is not objectively observable by anybody else than the subject. On the other hand, increasing levels of for example IgE-antibodies is a "sign", since it cannot be sensed by the subject but it can be objectively measured in an appropriate assay.
Where treating the allergy comprises or consists of relieving one or more signs or symptoms associated with allergic conjunctivitis, the relief typically comprises
- reducing the intensity of itchy eyes, redness in the white of the eyes and/or watery eyes; and/or
- reducing the eosinophilic count in conj unctival tissue scrapings; and/or
- reducing specific IgE antibody level (titer) in conj unctiva l tissue scrapings or in serum; and/or -reducing basophil histamine release in blood.
Where treating the allergy comprises or consists of relieving one or more signs or symptoms associated with allergic asthma, the relief typically comprises
- reducing the intensity and/or number of coughs within a given period (e.g. daily, weekly, monthly) ; and/or
- reducing the intensity of wheezes; and/or
- improving being short of breath; and/or
- improving lung function; and/or
- reducing specific IgE antibody level (titre) in lung fluid or in serum; and/or
- reducing basophil histamine release in blood; and/or improving lung function, such as increasing Forced Expiratory Volume (FEV), for example by increasing the FEV measured within the first second after full inspiration (FEVl); or increasing FEV1% (the ratio between FEVl and FVCl, where FVC is the forced vital capacity); or increasing FEVl%predicted (FEV1% of the subject divided by the average FEV1% in the population for any person of similar age, sex and body composition) . Thus, improving lung function can for example be increasing FEVl, FEV1% or FEV 1% predicted to a level that is within a range of normal values of a person of similar height, weight, gender and age which is not suffering from asthma. Where treating the allergy comprises or consists of relieving one or more signs or symptoms associated with atopic dermatitis, the relief typically comprises
- reducing itch intensity of the skin; and/or
- reducing eczema score; and/or
- reducing number of (peripheral) blood eosinophils. In all embodiments of the sixth aspect of the invention, the method may comprise or consist of reducing the subject's need for concomitant treatment with corticosteroids or H I antihistamines to reduce, relieve, or suppress one or more symptoms of an immune response associated with the allergy. In other words, these embodiments have the long term benefit of reducing the subject's need for medication. 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 ragweed pollen. An increase, improvement, enhancement or induction of "tolerance" may refer to a decrease, reduction, inhibition,
suppression, or limiting or controlling or clearing of specific immunological reactivity to an allergen as compared to reactivity to the allergen in a previous exposure to the same allergen. Thus, in certain embodiments, the method comprises inducing immunological tolerance in a subject to mites, e.g . to an allergen of ragweed pollen discussed herein to suppress an allergic immune response to the allergen. Immunological tolerance in a subject to an allergen can also be reflected by reducing the occurrence, frequency, severity, progression, or duration of an allergic response of the subject to the allergen. Induction of immune tolerance (also referred to as desensitization), and the relative amount of immune tolerance, can be measured by methods disclosed herein or known to the skilled artisan. For example, induction of immune tolerance can be measured by the modulated lymphokine and/or cytokine level in a subject or animal before versus after
administering a polypeptide of the first, second, third or fourth aspect. A modulated cytokine level can be an increase of a cytokine level, for instance an increase of a lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering a polypeptide of the first, second, third or fourth aspect. Alternatively, modulation can be a decrease of the level of a particular cytokine level, for insta nce a decrease of the lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering a polypeptide of the first, second, third or fourth aspect. The lymphokines/cytokines chosen to be measured can be from any relevant lymphokines/cytokines, such as IL-2, IL-5, IL-4, IL-6, IL-10, IL- 12, IL- 13, IL-17, TNF-alfa, IFN-gamma, TGF-beta, MCP- 1, RANK-L and Flt3L. Accordingly, the term "inducing immunological tolerance" may include eliciting, stimulating, promoting, increasing or enhancing immunological tolerance. Immunological tolerance may involve modulation of T cell activity, including but not limited to CD4+ T cells, CD8+ T cells, Th l cells, Th2 cells and regulatory T cells (Tregs), and memory T cells, including inflammatory lymphokines/cytokines produced by T cells.
The subjects subjected to the treatment of the sixth aspect of the invention typically present with an immune response clinically presented as atopic dermatitis, urticaria, contact dermatitis, allergic conj unctivitis, allergic rhinitis, allergic asthma, anapylaxis, and/or hay fever. In particular advantageous versions of any one of the embodiments of the sixth aspect of the invention, the treatment thus decreases, reduces, suppresses or inhibits atopic dermatitis, urticaria, contact dermatitis, allergic conjunctivitis, allergic rhinitis, allergic asthma, anaphylaxis, and/or hay fever.
Without being bound to any theory, it is believed that the method of the sixth aspect of the invention is capable of inducing or increasing an IgG antibody response in the subject to a ragweed pollen allergen and/or decreasing an IgE antibody response in the subject to a ragweed pollen allergen and/or decreasing a T cell response in the subject against ragweed pollen allergen.
It will be understood that the subjects that are subjected to the method of the sixth aspect of the invention are typically sensitized to at least one ragweed pollen allergen. It is to be understood that such patients may exhibit allergy signs or experience symptoms of allergy, but it is not excluded that "patients" that merely exhibit clinical signs of being sensitized against at least one protein allergy of the mites will also benefit from the treatment.
The allergy treated according to the invention is in all embodiments of the sixth aspect of the invention allergy towards ragweed pollen of the genus Ambrosia or Artemisiaa (for example selected from the group consisting of Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida and Artemisiia vulgaris.
Consequently, the ragweed pollen allergen is in all embodiments of the sixth aspect of the invention selected from one or more allergens in the groups consisting of
- a group 1 allergen of ragweed pollen (for example Amb a 1 and a homolog allergen of Amb a 1 present in pollen of Amb t, Amb p or Art v),
- a group 3 allergen of ragweed pollen (for example Amb a 3 and a homolog allergen of Amb a 3 present in pollen of Amb t, Amb p or Art v),
- a group 4 allergen of ragweed pollen (for example Amb a 4 and a homolog allergen of Amb a 4 present in pollen of Amb t, Amb p or Art v), -
- a group 5 allergen of ragweed pollen (f for example Amb a 5 and a homolog allergen of Amb a 5 present in pollen of Amb t, Amb p or Art v),
- a group 8 allergen of ragweed pollen (for example Amb a 4 and a homolog allergen of Amb a 8 present in pollen of Amb t, Amb p or Art v), and
- a group 11 allergen of ragweed pollen (for example Amb a 4 and a homolog allergen of Amb a l l present in pollen of Amb t, Amb p or Art v) .
As will be understood, the allergy is, according to the sixth aspect of the invention and any embodiments thereof, preferably treated by immunotherapy, which may comprise the repeated administration in weekly, bi-weekly, monthly or quarterly intervals. The administration may be selected from any one of subcutaneous, intradermal, epicutaneous, rectal, topical, sublingual, oral, buccal, intranasal, respiratory and intralymphatic route, though subcutaneous, intradermal or sublingual administration are mostly used in current immunotherapies.
A pharmacologically effective amount of a single dose of a polypeptide of the first, second, third aspect of the nvention may be in the range of 1 to 1000 nanomole, for example 1 to 500 nanomole, for example 1 to 250 nanomole, for example 5 to 250 nanomole. The single dose may be repeated once daily, twice-weekly, weekly, biweekly or monthly or quarterly. Typically, a polypeptide or composition of the invention is administered as a liquid in a volume of about 50 to 150 microliter, such as by intradermal administration. 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 and signs caused by or associated with an allergen. For example, administration of a peptide combination described herein may reduce the amount of an adj unct therapy administered to a subject, such as reducing the subject's need for concomitant treatment with fast or long-acting 32-agonists, leukotriene modifiers, theophylline corticosteroids or HI antihistamines (e.g . inhaled or oral) to reduce, relieve, or suppress one or more symptoms of the immune response.
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. In particular, the term "immune response" may include an IgE-mediated immune response (i.e. an allergic immune response) . Exemplary immune responses include T cell responses, such as Th2 responses resulting in cytokine production and/or cellular cytotoxicity. 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+, Th l 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 nonprofessional antigen presenting cells such as keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes) ; natural killer (NK) cells; and myeloid cells, such as macrophages, eosinophils, mast cells, basophils, and granulocytes. A particular immune response is production of
immunoglobulin (Ig) isotypes antibodies or decreasing IgE antibodies.
Therefore, in some embodiments, the method comprises inducing or increasing an IgG antibody (e.g . specific IgG) response in a subject to ragweed pollen, such as an allergen of ragweed pollen. In still some embodiments, the method comprises decreasing an IgE antibody (e.g . specific IgE) response in a subject to to ragweed pollen, such as an allergen of ragweed pollen. In still some embodiments, the method comprises decreasing a T cell response in a subject to ragweed pollen, such as an allergen of ragweed pollen, for example decreasing the production of Th-2 associated cytokines, like IL-5, IL-4, IL- 13 in response to said ragweed pollen or an allergen of ragweed pollen.
The term "modulating an immune response" or "modulate an immune response" may include to stimulate, induce, promote, increase or enhance an immune response, e.g. a T cell regulatory response, or may include inhibiting, decreasing, suppressing or reducing a T cell response, which may include, but is not limited to a Th2 cell response.
In some embodiments, a polypeptide or a composition of the invention may be used for conducting immunotherapy against ragweed pollen allergy, optionally wherein a single dose is administered repeatedly within the same day, or with daily, twice-weekly, weekly, bi-weekly, monthly or quarterly intervals until a desirable effect is achieved . Immunotherapy is thought to produce immunological tolerance in the subject undergoing therapy. Thus, in still other embodiments, a polypeptide or a composition of the invention may be used to induce immunological tolerance in a subject in need thereof.
As used herein, the term "immunological tolerance" refers to a) a decreased or reduced level of a specific immunologica l 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 ragweed pollen, i.e. pollen of the genus Ambrosia , such as pollen of Amb a, Amb p or Amb t. An increase, improvement, enhancement or induction of "tolerance" may refer to a decrease, reduction, inhibition, suppression, or limiting or controlling or clearing of specific immunological reactivity to an allergen as compared to reactivity to the allergen in a previous exposure to the same allergen. Thus, in certain embodiments, the method comprises inducing immunological tolerance in a subject to ragweed pollen, e.g. to an allergen of ragweed pollen, such as a group 1, 3, 4, 5, 8 or 11 allergen of Amb a, Amb t and/or Amb p to suppress an allergic immune response to the allergen. Immunological tolerance in a subject to an allergen can also be reflected by reducing the occurrence, frequency, severity, progression, or duration of an allergic response of the subject to the allergen. Induction of immune tolera nce (also referred to as desensitization), and the relative amount of immune tolerance, can be measured by methods disclosed herein or known to the skilled artisan. For example, induction of immune tolerance can be measured by the modulated lymphokine and/or cytokine level in a subject or animal before versus after administering a peptide combination described herein for the first time. A modulated cytokine level can be an increase of a cytokine level, for instance an increase of a lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering the peptide combination for the first time. Alternatively, modulation can be a decrease of the level of a particular cytokine level, for instance a decrease of the lymphokine and/or cytokine level of at least 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 20, 50 times or more relative to before administering the peptide combination for the first time. The
lymphokines/ cytokines chosen to measure can be from any relevant lymphokines/ cytokines, such as IL-2, IL-5, IL-4, IL-6, IL- 10, IL-12, IL-13, IL- 17, TNF-alfa, IFN-gamma, TGF-beta, MCP- 1, RANK- L and Flt3L. Accordingly, the term "inducing immunological tolerance" may include eliciting, stimulating, promoting, increasing or enhancing immunological tolerance. Immunological tolerance may involve modulation of T cell activity, including but not limited to CD4+ T cells, CD8+ T cells, Thl cells, Th2 cells and regulatory T cells (Tregs), and memory T cells, including inflammatory lymphokines/cytokines produced by T cells.
A polypeptide or a composition according to the invention is typically administered to a subject in need thereof by injection, such as by subcutaneous or intradermal administration, but may also include other routes of administration, such as epicutaneous, transcutaneous, topical, rectal, sublingual, oral, buccal, intranasal, respiratory and intralymphatic route of administration.
Typically, the subject in need thereof is a human, a pet such as a dog or a cat, a domestic animal such as a horse, or a laboratory animal (a mouse, a guinea pig or a rabbit) . The subject may be sensitized to ragweed pollen (e.g. having specific IgE antibodies against an allergen of ragweed pollen and/or having a T cell response against an allergen of ragweed pollen) . Therefore, a subject in need thereof may produce specific IgE antibodies or a T cell response against ragweed pollen allergens.
Other aspects
A still other aspect of the invention relates to an in vitro method of determining whether T cells of a subject are responsive to one or more polypeptides of the first, second or third aspect of the invention or to a composition of the fourth aspect. The method comprises contacting T cells obtained from the subject with said polypeptide(s) or composition(s) and determining whether the T cells are stimulated. The in vitro method may be used to determine whether the subject has, or is at risk of developing, an allergy to ragweed pollen allergen.
A still other aspect of the invention relates to an in vitro method for determining whether a subject has, or is at risk of developing, an allergy to house dust mites or storage mites, comprising contacting T cells obtained from the subject with one or more polypeptides of the first, second or third aspect of the invention and determining whether the T cells are stimulated .
A number of assay formats are available for the purpose of determining T cell stimulation and are well known for the person skilled . For instance ELISPOT/ Fluorospot, simple proliferation assays as well as the assay disclosed in Example 2 are all useful for the purpose of determining T cell responsiveness. A further aspect of the invention relates to a diagnostic kit comprising one or more of the polypeptides of the first, second or third aspect of the invention. Such a kit will normally also include necessary detection agents, visualisation means, carriers etc. that enable one or more of the above-described diagnostic assays.
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.
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
Jahn-Schmid, the T-cell response to Amb a 1 is characterized by 3 dominant epitopes and multiple MHC restriction elements. J Allergy Clin Immunol, Vol.126, No. 5, 1068- 1070, 2010).
Jiang, X., et al., Science 250 : 1580 1583 ( 1990) . Patens and patent applications; WO 92/11291, WO 96/30523, WO 98/15631, WO 00/32227, WO 01/85208, WO 02/056905, WO 06/097530, WO 06/03267, WO 07/068747, WO 07/144150 and WO 08/071774, WO 08/058369, WO 08/098749, WO 08/092996, WO 10/018378, WO 12/168487, WO 16/062720, WO 17/04224, US 5,071,651 and US 5,374,426.
Twomey, et al. , Vaccine 13 : 1603 1610 (1995) . Ulrich, et al, Virus Res. 50: 141- 182 ( 1998) .
Warnes, et al., Gene 160 : 173-178 ( 1995) .
EXAMPLE 1
Initial epitope mapping of pollen allergens of Ambrosia artemisiifolia
This example describes the peptide library set out to screen for T cell epitope responses of polypeptides derived from wild type allergens of group 3, 4, 5, 8 and 11 of of Ambrosia artemisiifolia (Amb a ; common ragweed/short ragweed), in short denoted Amb a 3, Amb a 4, Amb a 5, Amb a 8 and Amb a 11, respectively.
Similar peptide libraries can be established using the the homolog sequences of corresponding allergens of Ambrosia psilostachya (Amb p; western ragweed) and Ambrosia trifida (Amb t; giant ragweed) or related species Artemisia vulgaris (Mugwort) as shown in table 7
Sequences of the full length allergens Amb a 3, 4, 5, 8 and 11 were obtained from the WHO/IUIS Allergen Nomenclature database (htttp\www. allergen. org) and where more than one isoform was reported, all isoforms were included in the library. The underlined part of the sequence (shown below) was used to design the peptide library as the leader sequence or starting methionine was removed .
Amb a 3.0101 (Unitprot: P00304) - (SEQ ID NO: 183) :
GKVYLVGGPELGGWKLOSDPRAYALWSAROOFKTTDVLWFNFTTGEDSVAEVWREEAYHACDIKDPIRLEPGG
PDRFTLLTPGSHFICTKDOKFVACVPGR Amb a 4.0101 (Unitprot: D4IHC0) (SEQ ID NO : 184) :
MAAGLLVFVLAI5EIASVKGKLCEKPSVTW5GKCKVKOTDKCDKRCIEWEGAKHGACHKRDSKATCFCYFDCDP TKNPGPPPGAPKGKAPAPSPPSGGGAPPPSGGEGGDGPPPPEGGEGGGDGGGEGGGEGGGDGGGEGGGEGG GDGGGDGGGEGGGEGGGER Amb a 5.0101 (Unitprot: P02878) -(SEQ ID NO: 185) :
LVPCAWAGNVCGEKRAYCCSDPGRYCPWOVVCYESSEICSKKCGK
Amb a 8.0101(Unitprot: Q2KN24) - (SEQ ID NO : 186) :
MSWOTYVDEHLMCDIEGTGOHLASAAIFGTDGNVWAKSSSFPEFKPDEINAIIKEFSEPGALAPTGLFLAGAKYM VIOGEPGAVIRGKKGAGGICIKKTGOAMVFGIYEEPVNPGOCNMVVERLGDYLVDOGM Amb a 8.0102 (Unitprot: Q2KN23) - (SEQ ID NO : 187) :
MSWOTYVDEHLMCDIDGSGHHLSSAAIFGTDGAVWAKSGSFPEFKPDEINAIIKEFDAAGTLAPTGLFLAGAKY MVIOGEPGAVIRGKKGAGGICIKKTGOAMVFGIYEEPVAPGOCNMVVERLGDYLVDOGM
Amb a 11.0101 (Unitprot:V5LU01) - (SEQ ID NO : 188) :
MEINKLVCFSFSLVLILGLVESFHYHERELESEEGFMGMYDRWREOHNIEMRSPERFNVFKYNVRRIHESNKMDK PYKLKVNEFADMTNLEFVNTYANSKISHFOALRGSAPGSIDTDPNKDFIYANVTKIPDKVDWREKNAVTDVKGO GGCGSCWAFAAVVALEGINAIRTGKLVKFSEOOLVDCDMTNAGCDGGLMEPAFTYVIKHGGIAPEASYPYVGKR ETCDKAKIKDVLKIDGRONVPGLDEEALRKAVAHOPVATGIOLSGHGLOFYSEGVYTGDCGTEPNHGVGIVGYG ENEKGIKFWTVKNSWGPTWGEKGYIHLORGARKEGLCGVAM HSSFPIM NDPNPPKDDPNGPKDDPDAPKDPK FKTTORLOGIRTKLLEL. Sets of polypeptides of 20 amino acids in length, overlapping by 10 residues, were generated to cover the allergen protein sequences (underlined part as shown above), with few exceptions with overlap of 9 or 11 residues. To avoid dimerization and polymerization of polypeptides by intra- and intermolecular disulfide bond formation between cysteine residues, this amino acid was consistently substituted by a serine residue in the polypeptides for screening . Such polypeptides are herein marked with an asterisk (*) .
EXAMPLE 2
Initial Screening for T cell reactivity
This example includes a description of the methods used to screen peptide pools and single polypeptides for T cell reactivity. The polypeptides were assembled in pools of 5 polypeptides, on average, and screened for their immunogenicity. All pools were subsequently deconvoluted to make sure that all polypeptides were analysed separately as well.
The T cell reactivity of the polypeptides disclosed herein was assessed by measuring T cell proliferation of T cell lines specific to the allergens investigated and/or by measuring the cytokine production of allergen-specific T cells obtained from cultured PBMCs obtained from the donor cohorts.
Donor cohort: Around 30 donors were recruited in Texas, USA and Slovakia (Europe) on basis of a positive skin prick test to Amb a or Amb t and a clinical history of seasonal allergy with symptoms in the fall. Self reported symptoms in the recent fall confirmed the seasonal responses and sensitization towards Amb a was confirmed by IgE testing (>0.7 kU/L).
Peptide synthesis: Polypeptides were purchased from Innovagen in the form of triflouric acetic (TFA) salts, which were synthezised in an approximately 10 mg scale with a purity above 95% w/w. Polypeptides were resuspended in DMSO or DMSO 80% at a final concentration of about 2 ug/mL. Establishing cultured PBMCs and T-cell lines (TCP : Isolated PBMCs (peripheral blood mononuclear cells) obtained from the donors were CD8 depleted and T cell lines were established by stimulating the cultures with Amb a allergen extract enriched for allergen content.
In detail, PBMCs were isolated from freshly drawn heparinized blood by gradient centrifugation on lymphoprep (Nycomed, Norway), washed twice and re-suspended in RPMI 1640 medium with HEPES and ultraglutamine (Cambrex, Belgium) supplemented with 5% v/v human AB-serum
(Cambrex, Belgium), 100 units/ml penicillin, and 0.1 mg/ml streptomycin (Sigma, St Louis, USA) (referred to as 5% AB-medium) . Aliquots of the isolated PBMCs were frozen and were thawed later to generate the T cell lines. Additionally, thawed PBMCs were used as antigen presenting cells (APCs) in the proliferation assay as described laterFor eneration of T-cell lines PBMCs were CD8 depleted by MACS magnetic depletion according to the manufacturer's instructions (Miltenyi Biotec, Bergisch Gladbach, Germany) . On day 0 the CD8-depleted PBMCs (2 x 106/ml) were stimulated in 1 ml bulk cultures in AB medium in 24 well plates (Nunc, Denmark) with Amb a allergen extract enriched for allergen content (0.5 ug/mL). rIL-2 was added at day 5, 6 and 7 (65U, 30U, 30U, respectively) .To measure the immunogenicity of the polypeptides, ELISpot/ Fluorospot (day 14, T- cell line generation 01 (TCLOl)) and/or proliferation (day 24, TCL02) experiments were carried out.
On day 14 the T cells of TCLOl were isolated, washed and counted. The isolated T cells of TCLOl generation were used to perform Fluorospot assay. TCLOl were further re-stimulated as described in the next paragraph for use in T cell proliferation experiment at day 24.
Restimulation of T cell lines were performed in AB-medium with thawed autologous PBMCs
( 106/ ell, irradiated 2500 Rad), Amb a allergen extract enriched for allergen content (0.5 ug/mL), and PHA-P (0.5 pg/ml, Difco, Detroit, MI, USA) . rIL-2 was added at day 17, 18 and 19 (65U, 30U, 30U, respectively) . On day 24 T-cells (TCL02) were isolated, washed, counted and used for T cell proliferation assay as described later.
Fluorospot assays: In the Fluorospot assays, release of IL-5 and IFN-γ upon T cell activation (of donor derived lines DKl-TCLOl, and US1-TCL01) with a peptide described herein was measured by Fluorospot assay kit (Mabtech AB) and procedures were followed as per manufacturer's instructions. Briefly anti-human IL-5 and anti-human IFN-γ coated 96 well plates were obtained and blocked with 10% AB serum in RPMI. The plates were washed thoroughly and the isolated T cells were plated and incubated with peptide pools (2ug/mL of each peptide), polypeptides (2ug/ml), full length allergens (2 ug/ml, natural purified or recombinant/synthetic), Amb a allergen extract (5 ug/ml), or medium control) for 20hrs/37°C/5%CO2. After the incubation time the cells were removed by emptying the plate. Detection antibodies 7-B6- 1-FS-FITC ( 1 : 200) and 5A10- biotin (2 g/ml) were diluted in PBS-0.1% BSA and were added to the plate and incubated for 2 hours/room temperature (RT) . There-after the plates were washed and anti-FITC-green (1 : 200) and SA-Red (1 : 200) diluted in PBS+0.1% BSA were added and incubated for 1 hour/RT/dark. After the incubation period the plates were washed, emptied and fluorescence enhancer solution was added for 15 minutes/RT. Plates were emptied, fluorescence enhancer solution was firmly tapped out against a clean paper, soft plastic under drain was removed and the plates were left in the dark to dry. The counts were inspected in a fluorospot reader. Hit criteria for peptide were 20 SFCs/10s PBMCs, p< 0.05 and poisson< 0.05 stimulated cells compared to background, and a stimulation index (SI) > 2.
T cell proliferation assay: The capacity of the different polypeptides, peptide mixes and wild type allergens to stimulate allergen specific T cells was analyzed in a standard 72 h T cell proliferation assay, as described in Henmar H et al., Clin Exp Immunol 2008; 153(3) : 316-23. Criteria for positive T cell response: The polypeptides were identified as hits if they fulfilled the criteria as mentioned below where stimulation index (SI) value of 2 or above, Poisson value = < 0.05 and P value = < 0.05, sfc> =20 (IL-5/IFN-g)/106 cells. SI can be defined as [colony forming cells (sfc) stimulation/ sfc medium control] in a Fluorospot assay and [counts per minute (cpm) stimulation/ cpm medium control] in a proliferation assay. Table 1 shows the polypeptides derived from the allergen Amb a 3.0101 which showed positive T cell response according to the criteria set above. More than 20% of the donor cohort responded to the polypeptides with ID number B0182 (33%) and B0185 (20%). In addition to the 20mer polypeptides, longer polypeptides could be tested, for example peptide with start position number 32 having the sequence FKTTDVLWFNFTTGEDSVAEV. Table 1 - Amb a 3 polypeptides (SEP ID NOs : 1-91
Figure imgf000042_0001
Table 2 shows the polypeptides derived from the allergen Amb a 4.0101 which showed positive T cell response accord ing to the criteria set a bove. More than 20% of the donor cohort responded to the polypeptides with ID number B0188* (33%), B0189* (33%), B0190* (20%) and B0192* (40%), B0193 (27%), B0196 (47%), B0197 (53%) .
This ind icates that the majority of the T cell responses to Amb a 4.0101 derive from the amino acid stretch ranging from amino acid position number 1-40 (epitope reg ion A), from a mino acid position number 41 to 70 (epitope reg ion B) and from a mino acid position number 81- 110 (epitope region C) .
It is considered that a n immunodominant peptide does not include amino acid residues positioned at the start and end of the defined regions. Therefore, additiona l epitope polypeptides of Amb a 4.0101 may prefera bly be derived from regions covering the position numbers from 6 to 35 and 46- 64, 86- 104. In addition to the 20mer polypeptides, shorter or longer polypeptides could be tested, for example peptide with sta rt position number 1 having the sequence KLSEKPSVTWSGKSKVKQTDK, start position number 39 having the seq uence KRDSKATSFSYFDSDPTKN PGP or start position number 91 having the seq uence GPPPPEGGEGGGDGGGEGGGE
Table 2- Amb a 4 polypeptides (SEP ID NOs : 10-26)
Figure imgf000042_0002
SEQ ID Peptide ID Start Sequence Allergen % donors
NO Pos. with T cell
response
13 B0189* 11 SGKSKVKQTDKSDKRSIEWE Amb_a_4.0101 33%
14 B0190 21 KCDKRCIEWEGAKHGACHKR Amb_a_4.0101
15 B0190* 21 KSDKRSIEWEGAKHGASHKR Amb_a_4.0101 20%
16 B0191 31 GAKHGACHKRDSKATCFCYF Amb_a_4.0101
17 B0191* 31 GAKHGASHKRDSKATSFSYF Amb_a_4.0101 13%
18 B0192 41 DSKATCFCYFDCDPTK PGP Amb_a_4.0101
19 B0192* 41 DSKATSFSYFDSDPTKNPGP Amb_a_4.0101 40%
20 B0193 51 DCDPTKNPGPPPGAPKGKAP Amb_a_4.0101
21 B0193* 51 DSDPTKNPGPPPGAPKGKAP Amb_a_4.0101 27%
22 B0194 61 PPGAPKGKAPAPSPPSGGGA Amb_a_4.0101 13%
23 B0195 71 APSPPSGGGAPPPSGGEGGD Amb_a_4.0101 13%
24 B0196 81 PPPSGGEGGDGPPPPEGGEG Amb_a_4.0101 47%
25 B0197 91 GPPPPEGGEGGGDGGGEGGG Amb_a_4.0101 53%
26 B0198 101 GGDGGGEGGGEGGGDGGGEG Amb_a_4.0101 7%
Table 3 shows the polypeptides derived from the allergen Amb a 5.0101 which showed positive T cell response according to the criteria set above. More than 20% of the donor cohort responded to the polypeptides with ID number B0199* (20%) and B0200* (27%) . This indicates that the majority of the T cell responses to Amb a 5.0101 derive from the amino acid stretch ranging from amino acid position number 1 to 29, more preferable from 6-25.
Table 3 - Amb a 5 polypeptides (SEP ID NOs : 27-32)
Figure imgf000043_0001
Table 4 shows the polypeptides derived from the two allergens Amb a 8.0101 and Amb a 8.0102 which showed positive T cell response according to the criteria set above. The table also shows the number of mismatches in the amino acid sequence of polypeptides derived from the allergen Amb a 8.0101 compared to the corresponding peptide derived from allergen Amb a 8.0102 and vice versa . More than 20% of the donor cohort responded to the polypeptides with ID number B0206 (27%), B0252* (27%), B0260* (33%) and B0254* (33%), B0262 (20%), B0255 (33%), B0263 (20%), B0207 (40%), B0256 (20%), B0264 (20%), B0257(27%), B0265 (27%), B0258 (27%), B0259 (20%), B0267 (20%), B0208 (33%), B0209 (20%), B0212 (27%) and B0213 (27%) .
This ind icates that the majority of the T cell responses to Amb a 8.0101 derive from the amino acid stretch ranging from amino acid position number 31 to 68 (epitope reg ion A), with most responders found in the reg ion of from position number 37 to 59 (epitope region Aa), 43-64 (epitope reg ion Ab), 49-68 (epitope reg ion Ac) and from position n umber 85- 1 13 (epitope reg ion B) .
With respect to allergen Amb a 8.0102, the majority of T cell responses derive from the amino acid stretch ranging from amino acid position number 33 to 68 (epitope reg ion A) with most responders found in the reg ion of from position number 43 to 62 (epitope region Aa) a nd from position number 85- 104(epitope region B) .
It is considered that a n immunodominant peptide does not include amino acid residues positioned at the start and end of the defined regions. Therefore, additiona l epitope polypeptides of Amb a 8.0101 may prefera bly be derived from regions covering the position numbers from 36 to 63 and 85- 104. For Amb a 8.0102, additional epitope polypeptides may prefera bly be derived from regions covering the position numbers from 38 to 63 and 85- 104.
Examples of further epitope peptide of Amb a 8.0101 that likely will produce a T cell response in many donors sta rt with position number 39 (SFPEFKPDEINAIIKEFSEPGA) or 46
(DEINAIIKEFSEPGALAPTGLFL) . Examples of further epitope polypeptides of Amb a 8.0102 that have been shown to produce a T cell response in many donors start with position number 25 (AIFGTDGAVWAKSGSF), 33 (VWAKSGSFPEFKPDEI) a nd 41 (PEFKPDEINAIIKEFD) .
The domina nt epitope reg ion from position number 37 to 59 a ppears to be unique to the Amb a 8.0101 isoform as this region contains several mismatches to the Amb a 8.0102 isoform and that fa r more donors respond to the T cell responses aga inst Amb a 8.0101 polypeptides of this reg ion versus Amb a 8.0102 polypeptides of the same reg ion . Table 4 - Amb a 8 polypeptides (SEP ID NOs : 33-67)
Figure imgf000044_0001
SEQ Peptide Start Sequence Pa rent a llergen No of mis% donors ID NO ID Pos. matches to with T cell other response isoform
40 B0253 35 AKSSSFPEFKPDEINAIIKE Amb_a_8.0101 1 27%
41 B0261 35 AKSGSFPEFKPDEINAIIKE Amb_a_8.0102 1 13%
42 B0254 37 SSSFPEFKPDEI NAN KEFS Amb_a_8.0101 2 33%
43 B0262 37 SGSFPEFKPDEINAIIKEFD Amb_a_8.0102 2 20%
44 B0255 39 SFPEFKPDEINAIIKEFSEP Amb_a_8.0101 3 33%
45 B0263 39 SFPEFKPDEINAIIKEFDAA Amb_a_8.0102 3 20%
46 B0207 40 FPEFKPDEINAIIKEFSEPG Amb_a_8.0101 3 40%
47 B0256 41 PEFKPDEINAIIKEFSEPGA Amb_a_8.0101 4 20%
48 B0264 41 PEFKPDEINAIIKEFDAAGT Amb_a_8.0102 4 20%
49 B0257 43 FKPDEINAIIKEFSEPGALA Amb_a_8.0101 4 27%
50 B0265 43 FKPDEINAIIKEFDAAGTLA Amb_a_8.0102 4 27%
51 B0258 45 PDEINAIIKEFSEPGALAPT Amb_a_8.0101 4 27%
52 B0266 45 PDEINAIIKEFDAAGTLAPT Amb_a_8.0102 4 13%
53 B0259 47 EINAIIKEFSEPGALAPTGL Amb_a_8.0101 4 20%
54 B0267 47 EINAIIKEFDAAGTLAPTGL Amb_a_8.0102 4 20%
55 B0208 49 NAIIKEFSEPGALAPTGLFL Amb_a_8.0101 4 33%
56 B0209 58 PGALAPTGLFLAGAKYMVIQ Amb_a_8.0101 2 20%
57 B0210 67 FLAGAKYMVIQGEPGAVIRG Amb a 8.0101/ 0 13%
8.0102
58 B0211 76 IQGEPGAVIRGKKGAGGICI Amb a 8.0101/ 0
8.0102
59 B0211* 76 IQGEPGAVIRGKKGAGGISI Amb a 8.0101/ 13%
8.0102
60 B0212 85 RGKKGAGGICIKKTGQAMVF Amb a 8.0101/ 0
8.0102
61 B0212* 85 RGKKGAGGISIKKTGQAMVF Amb a 8.0101/ 27%
8.0102
62 B0213 94 CIKKTGQAMVFGIYEEPVN P Amb_a_8.0101 1
63 B0213* 94 SIKKTGQAMVFGIYEEPVN P Amb_a_8.0101 27%
64 B0214 103 VFGIYEEPVNPGQCNMVVER Amb_a_8.0101 1
65 B0214* 103 VFGIYEEPVNPGQSNMVVER Amb_a_8.0101 13%
66 B0215 113 PGQCNMVVERLGDYLVDQGM Amb a 8.0101/ 0
8.0102
67 B0215* 113 PGQSNMVVERLGDYLVDQGM Amb a 8.0101/ 7%
8.0102
Table 5 shows the polypeptides derived from the allergen Amb a 11.0101 which showed positive T cell response accord ing to the criteria set a bove. More than 20% of the donor cohort responded to the polypeptides with ID number B0227 (21%), B0228* (21%), B0230 (29%), B0231* (36%), B0233* (29%), B0234 (71%), B0237 (29%), B0238 (36%), B0239 (21%), B0241* (50%), B0242* (36%), B0245 (43%), B0246* (36%) and B0250 (43%).
This indicates that the majority of the T cell responses to Amb a 11.0101 derive from the amino acid stretch ranging from amino acid position number 138- 167 (epitope region A), 167- 195 (epitope region B), 206-235 (epitope region C), 245-274 (epitope region D), 285-314 (epitope region E), 335-354 (epitope region F). It is considered that an immunodominant epitope peptide of the region from 138 to 167 and 167 to 195 most likely may be derived from positions numbers 143 to 162 and 172 to 191, respectively, as the intermittently overlapping peptide of region A and B show considerably lower responder frequency. It is also considered that an immunodominant peptide does not include amino acid residues of the start and end of the defined regions. Therefore, additional epitope polypeptides may preferably be derived from regions covering the position numbers from 211-230, 250-270, 290-310 and 335-354.
For example, it has been shown that the following 15mer polypeptides also produce a T cell response in many donors: peptide with position number 141 (GSCWAFAAVVALEGIN),
179(AFTYVIKHGGIAPEAS) and 227 (EALRKAVAHQPVATGI) and 299 (YIHLQRGARKEGLCGV) .
Table 5 - Amb a 11 polypeptides (SEP ID NOs : 68- 105)
Figure imgf000046_0001
SEQ ID NO Peptide ID Start Sequence Parent allergen % donors with Pos. T cell response
87 B0236 196 PYVGKRETSDKAKIKDVLKI Amb_a_11.0101 7%
88 B0237 206 KAKIKDVLKIDGRQNVPGLD Amb_a_11.0101 29%
89 B0238 216 DGRQNVPGLDEEALRKAVAH Amb_a_11.0101 36%
90 B0239 225 DEEALRKAVAHQPVATGIQL Amb_a_11.0101 21%
91 B0240 235 HQPVATGIQLSGHGLQFYSE Amb_a_11.0101 14%
92 B0241 245 SGHGLQFYSEGVYTGDCGTE Amb_a_11.0101
93 B0241* 245 SGHGLQFYSEGVYTGDSGTE Amb_a_11.0101 50%
94 B0242 255 GVYTGDCGTEPNHGVGIVGY Amb_a_11.0101
95 B0242* 255 GVYTGDSGTEPN HGVGIVGY Amb_a_11.0101 36%
96 B0243 265 PN HGVGIVGYGEN EKGIKFW Amb_a_l 1.0101 7%
97 B0244 275 GENEKGIKFWTVKNSWGPTW Amb_a_11.0101 7%
98 B0245 285 TVK SWG PTWG E KG YI H LQR Amb_a_11.0101 43%
99 B0246 295 GEKGYIHLQRGARKEGLCGV Amb_a_11.0101
100 B0246* 295 GEKGYIHLQRGARKEGLSGV Amb_a_l 1.0101 36%
101 B0247 305 GARKEGLCGVAMHSSFPIMN Amb_a_11.0101
102 B0247* 305 GARKEGLSGVAMHSSFPIMN Amb_a_11.0101 14%
103 B0248 315 AMHSSFPIM DP PPKDDPN Amb_a_11.0101 14%
104 B0249 325 DPNPPKDDPNGPKDDPDAPK Amb_a_11.0101 14%
105 B0250 335 GPKDDPDAPKDPKFKTTQRL Amb_a_11.0101 43%
Table 6 shows a summary of the dominant epitope regions of each of the allergens tested .
Table 6 - Epitopic reaionsfSEO ID NOs : 106- 1251
Figure imgf000047_0001
SEQ Allergen Epitope Position Sequence
ID region
NO
114 Amb a 8.0101 Ab 43-64 FKPDEINAIIKEFSEPGALAPT
115 Amb a 8.0101 Ac 49-68 NAIIKEFSEPGALAPTGLFL
116 Amb a 8.0101 B 85-113 RGKKGAGGICIKKTGQAMVFGIYEEPVN P
117 Amb a 8.0102 A 33-68 VWAKSGSFPEFKPDEINAIIKEFDAAGTLAPTGL
118 Amb a 8.0102 Aa 43-62 FKPDEINAIIKEFDAAGTLA
119 Amb a 8.0102 B 85-104 RGKKGAGGICIKKTGQAMVF
120 Amb a 11.0101 A 138-167 AVVALEGINAIRTGKLVKFSEQQLVDCDMT
121 Amb a 11.0101 B 167-195 TNAGCDGGLMEPAFTYVIKHGGIAPEASY
122 Amb a 11.0101 C 206-235 KAKIKDVLKIDGRQNVPGLDEEALRKAVAH
123 Amb a 11.0101 D 245-274 SGHGLQFYSEGVYTGDCGTEPNHGVGIVGY
124 Amb a 11.0101 E 285-314 TVKN SWG PTWG E KGYI H LQRGARKEG LCGV
125 Amb a 11.0101 F 335-354 GPKDDPDAPKDPKFKTTQRL
EXAMPLE 3
Solubility optimization of T cell epitope-containing polypeptides
This example describes how polypeptides of the initial screening were re-designed to improve aqueous solubility, in particular to increase the solubility in an aqueous solution having pH in a physiologically acceptable range, e.g. from 4.5 to 8.5. Solubility is closely related to the ratio between hydrophilic and hydrophobic amino acid, the number of charged amino acids and the net charge of the peptide at a given pH. Polypeptides have zero net charge at pH = pi, and would be expected to show the lowest solubility at this pH. Polypeptides are preferably elongated with amino acid residues naturally encoded within the full length allergen sequence from which the peptide derives. Thus, where a peptide is designed from Amb a 8, the peptide is elongated with amino acid residues from Amb a 8.
In cases where polypeptides are not easily improvable by elongation by the natural allergen- encoding sequence (as there are no charged amino acids in close proximity to the peptide within the allergen sequence) peptide-solubility is improved by addition of either non-encoding negatively charged amino acids or non-encoded positively charged amino acids. Finally, examples where these two combinations are applied (i.e. natural elongation and addition of non-encoded amino acids) are also shown. To evaluate aqueous solubility, polypeptides were provided (by GenScript, USA) as acetate salts, >95 % purity at a 20mg scale. Test samples with a peptide concentration of 1 mg/mL or 3 mg/mL (dry matter) were produced with the following solvents: Buffer A; Water, Buffer B; Acetate buffer, pH 4.5 (25 mM sodium acetate containing 250 mM D-Mannitol), pH 4.5, Buffer C; 25 mM histidine buffer, pH 7.0, Buffer D; Glycine buffer, pH 8.5 (35 mM glycine containing 250 mM D-Mannitol). Test samples were gently mixed and left for one hour at room temperature to allow for dissolution of the polypeptides.
The solubility of the polypeptides was initially investigated by visual inspection by the eye and if the sample turned out to be very turbid, no further testing was performed. Test samples having passed the first solubility test, were than analysed unfiltered or filtered (500 μΙ sample through a 0.2 μιτι PES filter9 by HPLC, DLS, 90°LS and for UV-VIS for detection of particles/aggregates of the test sample and instability of the polypeptides.
Finally, it was concluded in which buffer the polypeptides had a solubility of at least 3 mg/mL ( > 3 mg/ml) or about 1 mg/mL (~ 1 mg/mL) or whether the polypeptides were not soluble in any of the buffers. Results are shown in the tables of Example 4.
EXAMPLE 4
Cross reactivity - Identifying allergens homologous to Amb a allergens in Ambrosia psilostachya (Amb p; western ragweed), Ambrosia trifida (Amb t; giant ragweed) and Artemisia vulgaris (Art v; mugwort) To investigate cross reactivity between homologous T cell epitope-containing polypeptides derived from the three ragweed species, Amb a, Amb p and Amb t, and the related weed Art v, the wild type full length amino acid sequence of allergens of these species must be provided. For that purpose, mRNA was purified from Amb p, Amb t and Art v pollen and subjected to high-throughput sequencing (Illumina HiSeq 2500) . The resulting sequencing reads were assembled using the Trinity software (version 2. 1.1) to obtain the Amb p, Amb t and Art v pollen transcriptomes.
Homology searches for the Amb a allergens in these three transcriptomes as well as among the Amb p, Amb t and Art v sequences available in the UniProt database were performed by Blast.
Table 7 shows amino acid sequences of homolog proteins found in the transcriptomes of Amb p, Amb t and Art v, which have high percentage sequence identity to the corresponding group 3, 4, 8 or 11 allergens of Amb a and which furthermore could be detected by mass spectrometry in pollen extracts of the species in question. No homologs could be identified for group 5. For each allergen, only those homologs containing sequences with a minimum number of mismatches to the original Amb a sequence are shown. Further is shown the percentage sequence identity between the Amb a sequence and the homolog allergen in the overlapping area (% SEQ ID (I)) and the percentage of the homolog amino acid sequence that covers the Amb a sequence (% SEQ ID (II)). Table 7 - Homoloq allergens (SEP ID NOs : 126-149
Figure imgf000050_0001
Figure imgf000051_0001
SEQ Amb a Homolog Identifier % SEQ % SEQ Protein sequence
ID allergen species ID (I) ID (II)
148 Amb a Art v Art v 4.010 89.39 100 MSWQTYVDDHLMCDIEGTGQHLTSAAIFGTD 8.0101 1 (Q8H2C9) GTVWAKSASFPEFKPNEIDAIIKEFNEAGQLAP
TGLFLGGAKYMVIQGEAGAVIRGKKGAGGICI KKTGQAMVFGIYDEPVAPGQCNMVVERLGDY LLDQGM
149 Amb a Art v Art v 4.020 81.82 100 MSWQTYVDDHLMCDIEGTGQHLTAAAILGLD 8.0101 1 (Q8H2C8) GTVWAKSDKFPEFKPEEMKGIINEFNEVGTLA
PTGLFLGGAKYMVLQGEAGAVIRGKKGAGGIC
IKKTGQAMVMGIYDEPVAPGQCNMIVERLGDY
LVDQNM
Determination of cross-reactivitv: Homologous polypeptides designed from the homolog allergens of Amb t, Amb p and/or Art v and which correspond to a selection of the polypeptides with high T cell responses of tables 1 to 5 were purchased at Innovagen in a purity of >95% (TCA salt). For determining cross-reactivity, it was investigated if a donor responds to both the Amb a peptide and the corresponding Amb t, Amb p or Art v peptide and XY correlation plots were made. Cross- reactivity was considered to be present if this comparison revealed a correlation between the Amb a polypeptides and the homolog polypeptides when depicted graphically.
Tables 8-11 show examples of cross reacting polypeptides. The frequency of T cell responders in a donor population and the cross reactivity between homologous polypeptides designed from each of the allergens of Amb a, Amb p, Amb t and Art v species are shown in figures 1 to 6.
Table 8 - homolog polypeptides to selected Amb a 3 polypeptides (SEP ID NOs: 150 and 151)
Table 9 - homolog polypeptides to selected Amb a 4 polypeptides (SEP ID NOs : 152- 167)
Figure imgf000052_0002
Figure imgf000053_0001
167 B0458* 60 QHESIYMSFSYFDSDPKNQGP Amb p homolog
Table 10 - homolog polypeptides to selected Amb a 8 polypeptides fSEO ID NOs: 168- 179)
Figure imgf000053_0002
Table 11 - homolog polypeptides to selected Amb a 11 polypeptides (SEP ID NOs: 180- 182")
Figure imgf000053_0003
180 B0397 198 MEPAFTYVIKHGGIAPEANY Amb t homolog
181 B0460 198 MEPAFIYI IKHGGIAPEAAY Amb p homolog
89 B0238 216 DGRQNVPGLDEEALRKAVAH Amb_a_11.0101
182 B0398 238 DGRQNVPGLDEELLRKAVAH Amb t homolog
EXAMPLE 5 HLA coverage
This example describes how HLA Class II allele coverage can be determined for individual polypeptides.
The peptide binding to a specific HLA molecule can be measured or predicted using in silico algorithms as e.g. NetMHCIIpan-3.1 (Karosiene, Edita, M ichael Rasmussen, Thomas Blicher, Ole Lund, S0ren Buus, and Morten Nielsen. "NetMHCIIpan-3.0, a Common Pan-specific M HC Class II Prediction Method Including All Three Human MHC Class II Isotypes, HLA-DR, H LA-DP and HLA- DQ." Immunogenetics ) available at the internet site < URL:
http:/ www,cb¾,dtu,dk/service¾ Neti 1HCIIpan-3, l >.
The exact affinity necessary for an immune response is not known, but it is generally assumed to be below ΙΟΟΟηΜ . For predicting the affinities of the polypeptides disclosed herein, the
NetMHCIIpan-3.1 algorithm was used. To assign a given peptide as a binder for a given HLA, a binding threshold of 300 nM was used together with the percentile rank score reported in this algorithm of at most 30.
Assuming that alleles at different loci are independently distributed in the worldwide population (a reasonable approximation), the phenotypic coverage of the binding alleles were calculated using standard methods known in the art. For example, for each HLA locus (DRBl, DRB3, DRB4, DRB5) or locus combination (DQA1-DQB1, DPA1-DPB1), the HLA allele frequencies of all the alleles from the given locus found to bind the given peptide is summed (fsum,iocus)■ The total phenotypic coverage for each locus, n (C„) is calculated as (2 x fslJm,iocus)-(fsum,iocus)2 '
The total phenotypic coverage of all binding alleles from n loci can be calculated as
Figure imgf000054_0001
, where K0 = 0 The predicted worldwide HLA coverage may be predicted using the alleles and allele frequencies shown below in table 12. Table 12 shows the frequencies of 28 alleleles present on loci, HLA-DRBl, HLADRBl, HLA-DRB4, HLA-DRB5, HLA-DP, and HLA-DQ, in a worldwide population {from Paul et al. Development and validation of a broad scheme for prediction of HLA class II restricted T cell epitopes. Journal of Immunological Methods 422 (2015) 28-34) .
Table 12
Figure imgf000055_0001
Table 13 shows the frequencies of 15 HLA alleles present on loci, HLA-DRB3, HLA-DRB4, HLA- DRB5, HLA-DP, and HLA-DQ, in a worldwide population.
Table 13
Figure imgf000055_0002

Claims

1. A polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NOs: 112- 119, 193-218, 106-111; 119- 125 and 189- 192, 219-224, optionally wherein the polypeptide is modified by one, two or three amino acid substitutions.
2. The polypeptide according to claim 1, wherein the polypeptide consists of: i) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 108, 189 and 190; ii) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 109, 191 and 192; iii) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 112 and 193-207; iv) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 116 and 208-218. v) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 120, 219 and 220; vi) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 121, 221 and 222; or vii) 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO: 122, 223 and 224; optionally wherein any of the polypeptides of i) to vii) is modified by one, two or three amino acid substitutions.
3. The polypeptide according to claim 2, wherein the polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO : 108, 189 and 190 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs : 10, 11, 152-157, 15 and 158-161.
4. The polypeptide according to claim 2, wherein the polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO : 109, 191 and 192 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs : 18, 19 and 162- 167.
5. The polypeptide according to claim 2, wherein the polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO : 112 and 193-207 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs : 44, 168- 173, 55 and 174- 179.
6. The polypeptide according to claim 2, wherein the polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO : 116 and 208-218 is a polypeptide with SEQ ID NO : 119.
7. The polypeptide according to claim 2, wherein the polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO : 121,
221 and 222 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs : 83, 180 and 81.
8. The polypeptide according to claim 2, wherein the polypeptide consisting of 15-30 contiguous amino acids of an amino acid sequence selected from any one of SEQ ID NO : 122,
222 and 223 is a polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs : 89 and 182.
9. The polypeptide according to any one of the preceding claims, wherein the one, two or three amino acid substitutions comprises conservative substitutions.
10. The polypeptide according to any one of the preceding claims, wherein the polypeptide has one or more of the modifications selected from the following : a) N-terminal acetylation or methylation
b) C terminal amidation
c) C-terminal amination
d) addition of one to three charged amino acid residues selected from any one of lysine (K), arginine (R), aspartic acid (D), and glutamic acid (E) in the N- terminal or C-terminal
e) addition of one to three lysine residues in the N-terminal or C-terminal f) one or more cysteine residues replaced with serine or 2-aminobutyric acid g) one or more serine residues replaced with cysteine
h) one or more hydrogen on the side chain amines of arginine and/or lysine
replaced with a methylene group
i) glycosylation, and
j) phosphorylation.
11. The polypeptide according to any one of the preceding claims, wherein the polypeptide elicits, stimulates or induces an in vitro or ex vivo T cell proliferation immune response, or an in vitro or ex vivo T cell cytokine production response by T cells from subjects allergic to ragweed pollen.
12. The polypeptide according to any one of the preceding claims, wherein the polypeptide is a syntetic peptide.
13. The polypeptide according to any one of the preceding claims, wherein the polypeptide is provided as a powder.
14. The polypeptide according to any one of the preceding claims, wherein the polypeptide is linked to a carrier.
15. A polypeptide consisting of an amino acid sequence selected from any one of SEQ ID NOs: 46-55, 1-45, 55- 105, optionally wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end, or by deletion of one, two or three amino acid residues at N- and/or C-terminal end .
16. The polypeptide according to claim 15, wherein a polypeptide modified with one, two or three amino acid substitutions in an amino acid sequence selected from any one of SEQ ID NOs : 46-55, 1-45, 55-105 is selected from the group consisting of SEQ ID NOs : 150- 182.
17. The polypeptide according to any one of claims 15 or 16, wherein the polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 10-15 and 18-21, optionally wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end, or by deletion of one, two or three amino acid residues at N- and/or C-terminal end.
18. The polypeptide according to claim 17, wherein the one to five additional amino acid residues is from a source allergen selected from any one of SEQ ID NO : 184, 129 and 147.
19. The polypeptide according to claim 17, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 11 is a polypeptide having an amino acid sequence of SEQ ID NOs : 10, 153, 155, or 157.
20. The polypeptide according to claim 17, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 15 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NOs: 14, 159 or 161.
21. The polypeptide according to claim 17, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 19 is a polypeptide having an amino acid sequence of SEQ ID NOs : 18, 163, 165, or 167.
22. The polypeptide according to any one of claims 15 or 16, wherein the polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 37-40, 42-51, 53- 56, 61 and 63, optionally wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at N- and/or C- terminal end.
23. The polypeptide according to claim 22, wherein the one to five additional amino acid residues is from a source allergen selected from any one of SEQ ID NO : 186 and 130- 144.
24. The polypeptide according to claim 22, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 44 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 168- 173.
25. The polypeptide according to claim 22, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 55 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 174- 179.
26. The polypeptide according to any one of claims 15 or 16, wherein the polypeptide consists of an amino acid sequence selected from any one of SEQ ID NOs: 76-78, 81-83, 88, 89, 92-95, 99, 100 and 105, optionally wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at N- and/or C- terminal end.
27. The polypeptide according to claim 26, wherein the one to five additional amino acid residues is from a source allergen selected from any one of SEQ ID NO : 188, 145 and 146.
28. The polypeptide according to claim 26, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 83 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 180 or 181.
29. The polypeptide according to claim 26, wherein a polypeptide with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 89 is the polypeptide having an amino acid sequence of SEQ ID NO: 182.
30. The polypeptide according to any one of claims 15 or 16, wherein the polypeptide consists of an amino acid sequence of SEQ ID NO: 2, optionally wherein the amino acid sequence of the polypeptide may be modified by one, two or three amino acid substitutions and wherein the polypeptide optionally is further modified by the addition of one to five amino acid residues at the N- and/or C-terminal end or by deletion of one, two or three amino acid residues at N- and/or C-terminal end.
31. The polypeptide according to claim 30, wherein the one to five additional amino acid residues is from a source allergen selected from any one of SEQ ID NOs: 183, 126- 128.
32. The polypeptide according to claim 30, wherein a polypeptide modified with one, two or three substitutions in the amino acid sequence of SEQ ID NO : 2 is a polypeptide having an amino acid sequence selected from any one of SEQ ID NO: 141 or 142.
33. The polypeptide according to any one of claims 15 to 32, wherein the polypeptide elicits, stimulates or induces an in vitro or ex vivo T cell proliferation immune response, or an in vitro or ex vivo T cell cytokine production response by T cells from subjects allergic to ragweed pollen.
34. The polypeptide according to any one of claims 15 to 33, wherein the polypeptide has one or more of the modifications selected from the following : a) N-terminal acetylation or methylation
b) C terminal amidation
c) C-terminal amination
d) addition of one to three charged amino acid residues selected from any one of lysine (K), arginine (R), aspartic acid (D), and glutamic acid (E) in the N- terminal or C-terminal
e) addition of one to three lysine residues in the N-terminal or C-terminal f) one or more cysteine residues replaced with serine or 2-aminobutyric acid g) one or more serine residues replaced with cysteine
h) one or more hydrogen on the side chain amines of arginine and/or lysine replaced with a methylene group
i) glycosylation, and
j) phosphorylation
35. The polypeptide according to any one of claims 15 to 34, wherein the polypeptide is a syntetic peptide.
36. The polypeptide according to any one of claims 15 to 35, wherein the polypeptide is provided as a powder, for example as a freeze-dried powder.
37. The polypeptide according to any one of claims 15 to 36, wherein the polypeptide is linked to a carrier.
38. A polypeptide consisting of an amino acid sequence having 85% to 100% sequence identity with an amino acid sequence selected from any one of SEQ ID NOs: 131- 144, 126- 130, 145 and 146.
39. The polypeptide according to claim 38, wherein the sequence identity is in the range of 85% -99%.
40. The polypeptide according to claim 38, wherein the sequence identity is in the range of 90% - 99%.
41. The polypeptide according to claim 38, wherein the polypeptide having 85% to 100% sequence identity with an amino acid sequence selected from any one of SEQ ID NOs: 126- 146 has the same biological activity or functionality as the original polypeptide, and wherein said biologica l activity or functionality is selected from the group consisting of the same enzymatic functionality; the same, greater or less ability to elicit, stimulate or induce an an in vitro or ex vivo T cell proliferation or T cell cytokine production of T cells from subjects allergic to ragweed pollen; the same, greater or less ability to induce immunological tolerance against ragweed pollen, a ragweed allergen; and to bind or interact with IgE, IgG or IgA antibodies raised against the original polypeptide.
42. The polypeptide according to claim 41, wherein the cytokines is selected from IL- 4, IL-5, IL- 13 and/or IL-10.
43. The polypeptide according to claim 41, wherein ragweed pollen is of the species Amb a, Amp p, Amb t or Art v.
44. The polypeptide according to any one of claims 38 to 41, wherein the polypeptide is a recombinant polypeptide, optionally comprising post-translational modifications.
45. The polypeptide according to any one of claims 38 to 44, wherein the polypeptide is provided as a powder, for example as a freeze-dried powder.
46. The polypeptide according to any one of claims 38 to 45, wherein the polypeptide is linked to a carrier.
47. A composition comprising one or more of the polypeptides according to any one of the claims 1 to 37.
48. The composition according to claim 47, wherein the composition further comprises one or more epitope polypeptides from a group 1 allergen of Amb a, for example from any of the isoforms: Amb_a_1.0201(UniProt No : P27760), Amb_a_1.0202(UniProt No : E1XUL3), Amb_a_1.0301(UniProt No : P27761), Amb_a_1.0302 (UniProt No : P27761(variantL48Y)), Amb_a_1.0303(UniProt No : P27761(variantH392R), Amb_a_1.0304(UniProt No: E1XUL4), Amb_a_1.0305(UniProt No : E1XUL5), Amb_a_1.0401(UniProt No: P28744),
Amb_a_1.0402(UniProt No : E1XUL9), Amb_a_1.0501(UniProt No: P27762) and/or
Amb_a_1.0502(UniProt No : E1XUM 1).
49. A composition comprising one or more of the polypeptides according to any one of the claims 38 to 46.
50. The composition according to any one of claims 47 to 49, wherein the composition is a pharmaceutical composition.
51. The pharmaceutical composition according to claim 50, further comprising a pharmaceutically acceptable carrier, excipient and/or adj uvant, optionally sterile.
52. The pharmaceutical composition according to claim 51 formulated as a vaccine for subcutaneous or intradermal injection or for sublingual administration.
53. The pharmaceutical composition according to any one of claims 50 to 52, wherein the pharmaceutical composition is a powder, optionally formulated to be re-dissolved before use.
54. The pharmaceutical composition according to any one of claims 50 to 53, wherein the pharmaceutical composition is a tablet.
55. A method of treating ragweed pollen allergy in a subject, comprising
administering to the subject a therapeutically effective amount of a polypeptide according to any one of claims 1 to 46 or a composition according to any one of claims 47 to 54.
56. A method of treating allergy in a subject, where signs and/or symptoms of said allergy are elicited in the subject by exposure to ragweed pollen and/or exposure to at least one allergen present in ragweed pollen, the method comprising administering to the subject a therapeutically effective amount of a polypeptide according to any one of claims 1 to 46 or a composition according to any one of claims 47 to 54.
57. The method according to any one of claims 55 and 56, wherein treating the allergy comprises or consists of relieving or reducing an immune response triggered by exposure to ragweed pollen or at least one allergen of ragweed pollen.
58. The method according to any one of claims 55 to 57, wherein treating the allergy comprises or consists of relieving one or more signs and/or symptoms of an immune response triggered by exposure to ragweed pollen or at least one allergen of ragweed pollen.
59. The method according to any one of claims 55 to 58, wherein treating the allergy consists of, or comprises, inducing immunological tolerance ragweed pollen or at least one allergen of ragweed pollen.
60. The method according to any one of claims 55 to 59, wherein treating the allergy comprises, or consists of, relieving one or more signs and/or symptom(s) associated with allergic rhinitis and/or allergic conjunctivitis and/or allergic asthma and/or allergic eczema (e.g . atopic dermatitis) .
61. The method according to any one of claims 55 to 60, wherein treating the allergy comprises, or consists of, relieving one or more signs or symptoms associated with allergic rhinitis, such as
- reducing the intensity of itchy nose and/or
- reducing the number of sneezes within a given period (e.g . daily, weekly, monthly) and/or
- reducing the intensity of blocked nose (congestion) and/or
- reducing the amount of nasal fluid and/or
- reducing the eosinophilic count in nasal fluid and/or
- reducing specific IgE antibody level (titre) in nasal fluid or in serum and/or
- reducing basophil histamine release in blood.
62. The method according to any one of claims 55 to 61, wherein treating the allergy comprises, or consists of, relieving one or more signs or symptoms associated with allergic conj unctivitis, such as - reducing the intensity of itchy eyes, redness in the white of the eyes and/or watery eyes; and/or
- reducing the eosinophilic count in conj unctival tissue scrapings; and/or
- reducing specific IgE antibody level (titer) in conj unctiva l tissue scrapings or in serum; and/or
-reducing basophil histamine release in blood.
63. The method according to any one of claims 55 to 62, wherein treating the allergy comprises, or consists of, relieving one or more signs or symptoms associated with allergic asthma, such as
- reducing the intensity and/or number of coughs within a given period (e.g . daily, weekly, monthly) ; and/or
- reducing the intensity of wheezes; and/or
- improving being short of breath; and/or
- improving lung function; and/or
- reducing specific IgE antibody level (titre) in lung fluid or in serum; and/or
- reducing basophil histamine release in blood.
64. The method according to any one of claims 55 to 63, wherein treating the allergy comprises, or consists of, relieving one or more signs or symptoms associated with atopic dermatitis, such as
- reducing itch intensity of the skin; and/or
- reducing eczema score; and/or
- reducing number of (peripheral) blood eosinophils.
65. The method according to any one of claims 55 to 64, comprising, or consisting of, reducing the subject's need for concomitant treatment with corticosteroids or H I
antihistamines to reduce, relieve or suppress one or more symptoms of an immune response associated with the allergy.
66. The method according to claim 65, wherein the immune response is clinically presented as atopic dermatitis, urticaria, contact dermatitis, allergic conjunctivitis, allergic rhinitis, allergic asthma, anapylaxis, and/or hay fever.
67. The method according to claims 64 or 65, wherein the method decreases, reduces, suppresses or inhibits atopic dermatitis, urticaria, contact dermatitis, allergic conj unctivitis, allergic rhinitis, allergic asthma, anaphylaxis, and/or hay fever.
68. The method according to any one of claims 55 to 67, comprising inducing or increasing an IgG antibody response in the subject to a ragweed pollen allergen and/or decreasing an IgE antibody response in the subject to a ragweed pollen allergen and/or decreasing a T cell response in the subject against ragweed pollen allergen .
69. The method according to any one of claims 55 to 68, wherein the subject is sensitized to at least one ragweed pollen allergen.
70. The method according to any one of claims 55 to 69, wherein the ragweed pollen is of the genus is of the genus Ambrosia or Artemisiaa .
71. The method according to any one of claims 55 to 70, wherein the ragweed pollen is of a species selected from any one of Ambrosia artemisiifolia, Ambrosia psilostachya, Ambrosia trifida and Artemisiia vulgaris.
72. The method according to any one of claims 55 to 71, wherein the ragweed pollen allergen is Amb a 1.
73. The method according to any one of claims 55 to 72, wherein the ragweed pollen allergen is Amb a 8.
74. The method according to any one of claims 55 to 73, wherein the treatment comprises repeated administration of the composition in weekly, bi-weekly, monthly or quarterly intervals.
75. The method according to any one of claims 55 to 74, wherein the treatment method is by immunotherapy.
76. The method according to any one of claims 55 to 75, wherein the administration is by a route of administration selected from any one of subcutaneous, intradermal, epicutaneous, rectal, topical, sublingual, oral, buccal, intranasal, respiratory and
intralymphatic route.
77. The method according to any one of claims 55 to 76, wherein the subject is a human, a pet such as a dog or a cat, or a domestic animal such as a horse.
78. A polypeptide according to any one of claims 1 to 46 for use in a method according to any one of claims 55 to 77.
79. A composition according to any one of claims 47 to 54 for use in a method according to any one of claims 55 to 77.
80. Use of a polypeptide according to any one of claims 1 to 46 for the preparation of a medicament for use in a method according to any one of embodiments 55 to 77.
81. Use of a composition according to any one of claims 47 to 54 for the preparation of a medicament for use in a method according to any one of embodiments 55 to 77.
82. An in vitro method of determining whether T cells of a subject recognize a polypeptide as defined in any one of claims 1 to 46, or a composition as defined in any one of claims 47 to 54, comprising a step of contacting T cells obta ined from the subject with said polypeptide or composition and detecting whether the T cells are stimulated .
83. An in vitro method of diagNOsing a subject for sensitization to ragweed pollen, comprising a step of contacting T cells obtained from the subject with a polypeptide as defined in any one of claims 1 to 46, or a composition as defined in any one of claims 47 to 54, and detecting whether the T cells are stimulated.
84. An in vitro method for determining whether a subject has, or is at risk of developing, an allergy to ragweed pollen, comprising a step of contacting T cells obtained from the subject with a polypeptide as defined in any one of claims 1 to 46, or a composition as defined in any one of claims 47 to 54, and detecting whether the T cells are stimulated.
85. A diagnostic kit comprising a polypeptide as defined in any one of claims 1 to 46, or a composition as defined in any one of claims 47 to 54.
86. An isolated nucleic acid molecule, which comprises a nucleotide sequence encoding a polypeptide according to any one of claims 38-46.
87. The nucleic acid molecule according to claim 86, which is a DNA or an RNA fragment.
88. A vector comprising the nucleic acid molecule according to any one of claims 86 and 87.
89. The vector according to claims 88, which is selected from the group consisting of a virus, such as an attenuated virus, a bacteriophage, a plasmid, a minichromosome, and a cosmid .
90. A cell which is transformed to carry the vector according to any one of claims 88 and 89.
91. The transformed cell according to claim 90, which is selected from a prokaryotic cell and a eukaryotic cell.
92. A cell line derived from the transformed cell according to any one of claims 88 and 89.
93. A method for the preparation of the polypeptide according to any one of claims 38-46 comprising
- culturing a transformed cell according to any of claims 90 and 91 or the cell line according to claim 92 under conditions that facilitate that the transformed cell expresses the nucleic acid fmolecule according to claim 86 and subsequently recovering said polypeptide, or
- preparing said polypeptide by means of solid or liquid phase peptide synthesis.
PCT/IB2017/052172 2016-04-15 2017-04-14 Epitope polypeptides of ragweed pollen allergens WO2017179025A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662322888P 2016-04-15 2016-04-15
US62/322,888 2016-04-15

Publications (2)

Publication Number Publication Date
WO2017179025A1 true WO2017179025A1 (en) 2017-10-19
WO2017179025A9 WO2017179025A9 (en) 2017-12-14

Family

ID=58640941

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2017/052172 WO2017179025A1 (en) 2016-04-15 2017-04-14 Epitope polypeptides of ragweed pollen allergens

Country Status (1)

Country Link
WO (1) WO2017179025A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109939227A (en) * 2018-03-23 2019-06-28 中国医学科学院北京协和医院 A kind of ragweed pollen allergen extract, its immersion liquid and preparation method thereof

Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5071651A (en) 1986-09-03 1991-12-10 University Of Saskatchewan Rotavirus nucleocapsid protein VP6 as a carrier in vaccine compositions
WO1992011291A1 (en) 1990-12-20 1992-07-09 Smithkline Beecham Biologicals (S.A.) Vaccines based on hepatitis b surface antigen
WO1993021321A2 (en) * 1992-04-09 1993-10-28 Immulogic Pharmaceutical Corporation T CELL EPITOPES OF THE MAJOR ALLERGENS FROM $i(AMBROSIA ARTEMISIIFOLIA)
WO1996030523A2 (en) 1995-03-31 1996-10-03 Hans Wolf Antigen presentation system based on retrovirus-like particles
WO1998015631A1 (en) 1996-10-09 1998-04-16 Btg International Limited Attenuated microorganism strains expressing hpv proteins
WO2000032227A2 (en) 1998-11-30 2000-06-08 Cytos Biotechnology Ag Ordered molecular presentation of antigens, method of preparation and use
WO2001085208A2 (en) 2000-05-05 2001-11-15 Cytos Biotechnology Ag Molecular antigen arrays and vaccines
WO2002056905A2 (en) 2001-01-19 2002-07-25 Cytos Biotechnology Ag Molecular antigen array
WO2005049107A2 (en) 2003-11-18 2005-06-02 Nanapass Technologies Ltd. Enhanced penetration system and method for sliding microneedles
WO2006003267A1 (en) 2004-06-03 2006-01-12 France Telecom Method for determining a list of elements potentially visible by region for ultra large 3d scenes of virtual cities
WO2006054280A2 (en) 2004-11-18 2006-05-26 Nanopass Technologies Ltd. System and method for delivering fluid into flexible biological barrier
WO2006097530A2 (en) 2005-03-18 2006-09-21 Cytos Biotechnology Ag Cat allergen fusion proteins and uses thereof
WO2007066341A2 (en) 2005-12-08 2007-06-14 Nanopass Technologies Ltd. Microneedle adapter for dosed drug delivery devices
WO2007068747A1 (en) 2005-12-14 2007-06-21 Cytos Biotechnology Ag Immunostimulatory nucleic acid packaged particles for the treatment of hypersensitivity
WO2007144150A1 (en) 2006-06-12 2007-12-21 Cytos Biotechnology Ag Processes for packaging oligonucleotides into virus-like particles of rna bacteriophages
WO2008058369A1 (en) 2006-11-15 2008-05-22 Folia Biotech Inc. Immunogenic affinity-conjugated antigen systems based on papaya mosaic virus and uses thereof
WO2008071774A1 (en) 2006-12-14 2008-06-19 Cytos Biotechnology Ag Purification process for coat protein of rna bacteriophages
WO2008092996A1 (en) 2007-02-01 2008-08-07 Vactech Oy Prevention of allergic sensitization
WO2008098749A2 (en) 2007-02-13 2008-08-21 Biomay Ag Peptides derived from the major allergen of ragweed (ambrosia artemisiifolia) and uses thereof
WO2010018378A2 (en) 2008-08-15 2010-02-18 Circassia Limited Ragweed peptides for vaccine
US20120100167A1 (en) * 2003-03-14 2012-04-26 Anergis S.A. Allergen peptide fragments and use thereof for treatment of dust mite allergies
WO2012168487A1 (en) 2011-06-09 2012-12-13 Biomay Ag Peptide carrier fusion proteins as allergy vaccines
WO2013156467A1 (en) * 2012-04-16 2013-10-24 Alk-Abelló A/S Plant profilin polypeptides for use in non-specific allergy immunotherapy
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
WO2016062720A1 (en) 2014-10-22 2016-04-28 Saiba Gmbh Modified virus-like particles of cmv
WO2017004224A1 (en) 2015-06-29 2017-01-05 Herman Miller, Inc. Attachment structure for suspension seating

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5071651A (en) 1986-09-03 1991-12-10 University Of Saskatchewan Rotavirus nucleocapsid protein VP6 as a carrier in vaccine compositions
US5374426A (en) 1986-09-03 1994-12-20 University Of Saskatchewan Rotavirus nucleocapsid protein VP6 in vaccine compositions
WO1992011291A1 (en) 1990-12-20 1992-07-09 Smithkline Beecham Biologicals (S.A.) Vaccines based on hepatitis b surface antigen
WO1993021321A2 (en) * 1992-04-09 1993-10-28 Immulogic Pharmaceutical Corporation T CELL EPITOPES OF THE MAJOR ALLERGENS FROM $i(AMBROSIA ARTEMISIIFOLIA)
WO1996030523A2 (en) 1995-03-31 1996-10-03 Hans Wolf Antigen presentation system based on retrovirus-like particles
WO1998015631A1 (en) 1996-10-09 1998-04-16 Btg International Limited Attenuated microorganism strains expressing hpv proteins
WO2000032227A2 (en) 1998-11-30 2000-06-08 Cytos Biotechnology Ag Ordered molecular presentation of antigens, method of preparation and use
WO2001085208A2 (en) 2000-05-05 2001-11-15 Cytos Biotechnology Ag Molecular antigen arrays and vaccines
WO2002056905A2 (en) 2001-01-19 2002-07-25 Cytos Biotechnology Ag Molecular antigen array
US20120100167A1 (en) * 2003-03-14 2012-04-26 Anergis S.A. Allergen peptide fragments and use thereof for treatment of dust mite allergies
WO2005049107A2 (en) 2003-11-18 2005-06-02 Nanapass Technologies Ltd. Enhanced penetration system and method for sliding microneedles
WO2006003267A1 (en) 2004-06-03 2006-01-12 France Telecom Method for determining a list of elements potentially visible by region for ultra large 3d scenes of virtual cities
WO2006054280A2 (en) 2004-11-18 2006-05-26 Nanopass Technologies Ltd. System and method for delivering fluid into flexible biological barrier
WO2006097530A2 (en) 2005-03-18 2006-09-21 Cytos Biotechnology Ag Cat allergen fusion proteins and uses thereof
WO2007066341A2 (en) 2005-12-08 2007-06-14 Nanopass Technologies Ltd. Microneedle adapter for dosed drug delivery devices
WO2007068747A1 (en) 2005-12-14 2007-06-21 Cytos Biotechnology Ag Immunostimulatory nucleic acid packaged particles for the treatment of hypersensitivity
WO2007144150A1 (en) 2006-06-12 2007-12-21 Cytos Biotechnology Ag Processes for packaging oligonucleotides into virus-like particles of rna bacteriophages
WO2008058369A1 (en) 2006-11-15 2008-05-22 Folia Biotech Inc. Immunogenic affinity-conjugated antigen systems based on papaya mosaic virus and uses thereof
WO2008071774A1 (en) 2006-12-14 2008-06-19 Cytos Biotechnology Ag Purification process for coat protein of rna bacteriophages
WO2008092996A1 (en) 2007-02-01 2008-08-07 Vactech Oy Prevention of allergic sensitization
WO2008098749A2 (en) 2007-02-13 2008-08-21 Biomay Ag Peptides derived from the major allergen of ragweed (ambrosia artemisiifolia) and uses thereof
WO2010018378A2 (en) 2008-08-15 2010-02-18 Circassia Limited Ragweed peptides for vaccine
WO2012168487A1 (en) 2011-06-09 2012-12-13 Biomay Ag Peptide carrier fusion proteins as allergy vaccines
WO2014064543A1 (en) 2011-10-26 2014-05-01 Nanopass Technologies Ltd. Microneedle intradermal drug delivery with auto-disable functionality
WO2013156467A1 (en) * 2012-04-16 2013-10-24 Alk-Abelló A/S Plant profilin polypeptides for use in non-specific allergy immunotherapy
WO2014188429A1 (en) 2013-05-22 2014-11-27 Nanopass Technologies Ltd. Intradermal delivery of drugs, pharmaceuticals and other therapeutic agents via microneedles
WO2016062720A1 (en) 2014-10-22 2016-04-28 Saiba Gmbh Modified virus-like particles of cmv
WO2017004224A1 (en) 2015-06-29 2017-01-05 Herman Miller, Inc. Attachment structure for suspension seating

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
"Pharmaceutical Principles of Solid Dosage Forms", 1993, TECHNONIC PUBLISHING CO., INC.
"Remington: The Science and Practice of Pharmacy", 2003, MACK PUBLISHING CO.
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO.
"The Merck Index", 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
ANSEL; SOKLOSA: "Pharmaceutical Calculations", 2001, LIPPINCOTT WILLIAMS & WILKINS
AUSUBEL ET AL.: "Short Protocols in Molecular Biology", 1995, WILEY AND SONS
BOSTICK ET AL., BIOCHEM BIOPHYS RES COMMUN., vol. 304, 2003, pages 320
HENMAR H ET AL., CLIN EXP IMMUNOL, vol. 153, no. 3, 2008, pages 316 - 23
JAHN-SCHMID: "the T-cell response to Amb a 1 is characterized by 3 dominant epitopes and multiple MHC restriction elements", J ALLERGY CLIN IMMUNOL, vol. 126, no. 5, 2010, pages 1068 - 1070
JIANG, X. ET AL., SCIENCE, vol. 250, 1990, pages 1580 - 1583
MATSUI, S.M. ET AL., J. CLIN. INVEST, vol. 87, 1991, pages 1456 - 1461
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
PAUL ET AL.: "Development and validation of a broad scheme for prediction of HLA class II restricted T cell epitopes", JOURNAL OF IMMUNOLOGICAL METHODS, vol. 422, 2015, pages 28 - 34
PEARSON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PEARSON, METHODS MOL. BIOL., vol. 132, 2000, pages 185
PEARSON; LIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
POZNANSKY ET AL.: "Drug Delivery Systems", 1980, pages: 253 - 315
SAMBROOK ET AL.: "Molecular Cloning", 1989, COLD SPRING HARBOR LABORATORY PRESS
SMITH ET AL., J. MOL. BIOL., vol. 147, 1981, pages 195
SMITH; WATERMAN, ADV. APPL. MATH, vol. 2, 1981, pages 482
TWOMEY ET AL., VACCINE, vol. 13, 1995, pages 1603 - 1610
ULRICH ET AL., VIRUS RES., vol. 50, 1998, pages 141 - 182
WARNES ET AL., GENE, vol. 160, 1995, pages 173 - 178

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109939227A (en) * 2018-03-23 2019-06-28 中国医学科学院北京协和医院 A kind of ragweed pollen allergen extract, its immersion liquid and preparation method thereof
CN109939227B (en) * 2018-03-23 2023-04-28 中国医学科学院北京协和医院 Ragweed pollen allergen extract, extract liquid and preparation method thereof

Also Published As

Publication number Publication date
WO2017179025A9 (en) 2017-12-14

Similar Documents

Publication Publication Date Title
EP3086806B1 (en) Peptide combinations and uses thereof in treating dust mite allergy
KR101699554B1 (en) Peptides for vaccine
JP5807994B2 (en) Nucleic acids for allergy treatment
JP5399895B2 (en) Vaccine carrier
JP5705113B2 (en) Allergen-derived T cell antigen for stimulation of IL-10 production
BR112013031505B1 (en) POLYPEPTIDE, USE OF A POLYPEPTIDE, VACCINE FORMULATION AND USE OF A VACCINE FORMULATION
US9017689B2 (en) Peptides for vaccine against birch allergy
EP3030259B1 (en) Peptides
EA034692B1 (en) Improved modular antigen transportation molecules and uses thereof
Moldaver et al. Induction of bystander tolerance and immune deviation after Fel d 1 peptide immunotherapy
US11013781B2 (en) Peptide combinations and uses thereof for treating grass allergy
WO2017179025A1 (en) Epitope polypeptides of ragweed pollen allergens
DK2838556T3 (en) Plant profile polypeptides for use in non-specific allergy immunotherapy
WO2016192788A1 (en) Methods and compositions for preventing and treating cat or dog dander allergy
WO2015077442A9 (en) Grass pollen immunogens and methods and uses for immune response modulation
WO2016209959A2 (en) Peptide combinations and use thereof in treating cedar pollen allergy
WO2015131053A1 (en) Polypeptides derived from phl p and methods and uses thereof for immune response modulation
US20160287696A1 (en) Pan pollen immunogens and methods and uses for immune response modulation
JP2017079742A (en) Nucleic acids for treatment of allergies
JPWO2003020306A1 (en) Desensitizer
JP6088584B2 (en) Nucleic acids for allergy treatment
WO2015054217A2 (en) Methods and uses for reducing an allergic response in a subject
Weiss et al. DNA and RNA vaccines for prophylactic and therapeutic treatment of type I allergy

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17720230

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17720230

Country of ref document: EP

Kind code of ref document: A1