WO2015042664A1 - Novel immunotherapeutic composition and uses thereof - Google Patents

Novel immunotherapeutic composition and uses thereof Download PDF

Info

Publication number
WO2015042664A1
WO2015042664A1 PCT/AU2014/050249 AU2014050249W WO2015042664A1 WO 2015042664 A1 WO2015042664 A1 WO 2015042664A1 AU 2014050249 W AU2014050249 W AU 2014050249W WO 2015042664 A1 WO2015042664 A1 WO 2015042664A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
ara
peptides
cell
composition
Prior art date
Application number
PCT/AU2014/050249
Other languages
French (fr)
Inventor
Robyn Elizabeth O'hehir
Sara Rachel PRICKETT
Jennifer May ROLLAND
Original Assignee
Monash University
Alfred Health
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
Priority claimed from AU2013903686A external-priority patent/AU2013903686A0/en
Priority to AU2014328483A priority Critical patent/AU2014328483B2/en
Priority to US15/024,666 priority patent/US11266737B2/en
Priority to BR112016006813A priority patent/BR112016006813A2/en
Priority to RU2016115732A priority patent/RU2689552C2/en
Priority to MX2016003363A priority patent/MX2016003363A/en
Priority to EP14848639.2A priority patent/EP3049098A4/en
Priority to CA2925480A priority patent/CA2925480A1/en
Application filed by Monash University, Alfred Health filed Critical Monash University
Priority to CN201480052398.1A priority patent/CN105992589A/en
Priority to KR1020167010829A priority patent/KR20160075532A/en
Priority to JP2016517506A priority patent/JP7007534B2/en
Publication of WO2015042664A1 publication Critical patent/WO2015042664A1/en
Priority to ZA2016/02043A priority patent/ZA201602043B/en
Priority to AU2020200883A priority patent/AU2020200883B2/en
Priority to US17/672,571 priority patent/US20220280637A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/35Allergens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/577Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 tolerising response

Abstract

The present invention relates generally to an immunotherapeutic composition. More particularly, the present invention relates to an immunotherapeutic composition which interacts immunologically with T lymphocytes in subjects having peanut allergy or allergy to other tree nuts. This composition is preferably immunointeractive with T cells in subjects having an allergy to the Ara h 1 and/or Ara h 2 allergens. The composition of the present invention is useful in the therapeutic or prophylactic treatment of conditions characterised by an aberrant, inappropriate or otherwise unwanted immune response to peanut, Ara h1 and/or Ara h 2 or derivative or homologue thereof.

Description

NOVEL IMMUNOTHERAPEUTIC COMPOSITION AND USES THEREOF
FIELD OF THE INVENTION
The present invention relates generally to an irnmunotherape iic composition.. More particularly, the present invention relates to an iirununotherapeutic composition which interacts itnraunologically with T lymphocytes in subjects having peanut allergy or allerg t other tree nuts. This composition is preferably mnnunointeractive with T cells in subjects havin an allergy to the Ara h 1 and/or Ara h 2 allergens. The composition of the present invention is useful in the therapeutic or prophylactic treatment of conditions characterised, by an aberrant, inappropriate or otherwise unwanted immune response to peanut, Ara hi and or Ara h 2 or derivative or homologue thereof.
BACKGROUND OF THE INVENTION
Bibliographic details of the publications referred to by author in thi specification are collected alphabetically at the end of the description.
The reference to any prior art is this specification is not, and should not he taken as, .an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge.
Peanut allergy is a life-threatening and incurable disorder, affecting approximately 1% of the general population (Husain et at J Am Acad Dermatol. 66(1): 136-43, 2012, Burks, Lancet. 371(9623):1538-46, 2008). It is characterised b the sudden onset of anaphylaxis, which may occur with exposure to minute quantities of peanut proteins (Hoiirihane ei al, J Allergy Clin Immunol 100: 596-600* 1 97; Pumphrey, Current Opinion- in Allergy & Immunology, 4(4): 285-90, 2004). Peanut induced anaphylaxis is that most frequently associated with mortality or with life- threatening features (Bock era!. J Allergy Clin Immunol 119(4): 1016-8. 2007; Burks 2008, supra). Peanut proteins are frequently concealed within apparently safe food sources, such that accidental contact occurs for up to 50% of sufferers over a 5 year period (Sicherer .et at., Paediatrics 102: e6, 1998). .Not -surprisingly, peanut and tree nut allergy is associated, with significant psychological morbidity for sufferers and carers alike, akin to that suffered by those with chronic debilitating illnesses such as rheumatoid arthritis (Primeau et L, Clin Exp Allergy 30: 1135-43, 2000; Kemp et al. Me t J. Ami 1.88(9):503-4, 2008). Cure, while being an imperative to remove peanut and tree nut allergy as a cause of mortality, i also necessary to remove the chronic psychological burden that peanut allergic subjects cany.
To date, efforts at .immunotherapy for peanut allergy have been met by extremely limited success. Nelson et al. have shown that clinical desensitisation of peanut can be induced usin a rush immunotherapy protocol with an imfractioiiated peanut extract, hut that clinical desensitisation is lost in approximately half of the subjects during maintenance dosing and additionally that injections are associated with frequent episodes of anaphylaxis in the majority of subjects during both the buildup and maintenance phases (Nelson et al,, J Allergy Clin hnmunol 99: 744-51 , 1997). Oppenheime.r et al demonstrated similar findings within their study, again showing that active therapy with unfractionated peanut extract is associated with a high rate of systemic anaphylaxis. Data collection in that study was terminated after the admmistration of peanut extract to a placebo randomised subject resulted in their death, highlightin the dangerous nature of this condition (Oppenheimer et al. , J Allergy Clin Immunol 90: 256-62, 1 92). Recent studies of oral immunotherapy with whole peanut flour provide encouragement that (^sensitization is feasible, but the observed adverse reactions highlight major safety concerns ( Hofmann et al, J. Allergy Clin.
Immunol. 124, 286, 2009; Jones et al, J. Allergy Clin, Immunol. 24, 292, 2009; Clark et al Allergy 64, 1218, 2009; Varshney et al. J Allergy Clin Immunol. 1 7(3):654~60, 201 ; Varsfiney et al J Allergy Clin Immimol, 124(6): 1351 -2, 2009; Anagnostou et al Clin Exp Allergy. 41(9): 1273-81, 2011 ; Allen & O'Hehir. Clin Exp Allergy. 41 (9): 1172-4, 2011 : Yu et al, hit Arch Allergy Immunol, 159(2):I79-182, 2012; Thyagarajan ef *. J Allergy Clin Immunol 126(l):3l-2, 2010; Blumehen et al, J Allergy Clin Immimol, 126(1):83-91, 2010). Even with the exclusion of children prone to severe symptoms or asthma, two studies reported an anaphylactic episode, in one case during an initial food challenge (Clark et al, Allergy 64, .1218, 2009) and in the other during treatment of a child who had not previously experienced anaphylaxis (Hofmann et at J. Allergy Clin, Immimol 124, 286, 2009).
Development of novel strategies to overcome the morbidity associated with, allergen immunotherapy depends on an accurate understanding of the immunological basis tp successful immunotherapy, as well as its -side-effects. It has long been established that morbidity due to allergen immunotherapy is due to the .cross-linking of IgE, and that this action is not required for such therapy to be efficacious (Litwtn et aL . Int Arch Allergy Appl Immunol 87: 361 -61, 998). It is also known that one of the critical actions of conventional (subcutaneous injection or sublingual or oral with unfractionated. allergen extract) immunotherapy in producing tolerance is its ability to change the predomin nt specific T cell phenotype from a TH2 to a regulatory phenotype. These regulatory T cells act via production of the anti-inflammatory cytokine IL-1.0 and/or TGFp.
(Akdis & Akdis, Allergy Clin immimol, 123:735-46, 2009; Akdis & Akdis,. Nature Reviews: Drug Discovery, 8:645-60. 2009; Akdis & Akdis, J Allergy Clin Immunol. 127:18-27, 2011).
A key difference in antibody and lymphocyte responses is in antigen recognition, antibodies recognising conformational B ceil epitopes dependent on molecular tertiary structure, while CD4+ T cells recognise short: linear peptides. This difference in antigen recognition is the basis to many novel strategies of immunotherapy, including that using peptides based upon T cell - „1 - epitopes, B cell epitope mutants and altered peptide ligands (Roilaad et al. Pharmacology' _fc Therapeutics 121 :273-284, 2009). Such methods all depend on the alteration or absence of molecular tertiary structure, so that. lgE cros -linking and. effector cell activation is lost. Peptide immunotherapy is a method in respect of which evidence of efficacy exists, being documented for both cat dander allergy and bee venom allergy. Three different studies showed that, in the absence of any systemic side-effects, clinical and immunological tolerance could he achieved for the major bee venom allergen Phospholipase A2 (PLA2) using T cell epitope-containiiig sequences (Mailer etal. J Allergy Clin Immunol 101: 747-54, 1998; Tarzi et al. Clin Exp Allergy. 36: 465-74, 2006; Fellrath et al J A llergy Clin Immunol 1 11 : 854-61 , 2003), while several studies have demonstrated that peptide based on the structure of the major cat allergen Fel d 1 can be used to induce diminished clinical responses (Norman et al. , Am J Respir Crit Care Med 154: 1623-8, 1996; Marcotte etal, J Allergy Clin Immunol 101 : 506- 13, 1998; Pene et al, J Allergy Clin Immunol 102: 571 -8, 1.998; Oldfield et al. Lancet 360:47-53, 002; Alexander et al Clin Exp Allergy 35: 52- 8, 2004; Alexander et al Allergy 60:1269-74, 2005). Most recently, a phase Ila trial confirmed the safety, toler&hility and. potenti l efficacy of a seven-pe.pti.de mixture from Fel d 1 (Tqleromune Cat®, Cicassia Ltd. , Oxford, UK) (Worm et al. J Allergy Clin Immunol. 27: 89-97, 2011) with Phase lib trials now underway (M ldaver & Larehe. Allergy. 66: 784-91 , 2011; Worm et al. Expert Opin. Irwestig. Drugs. 22(1.0): 1 47-1357, 2013). Crucial to the development of such strategies is the retention of T cell epitopes, so that T ceil phenotypic change can be induced.
The ability to bind directly on to MHC class II molecule allows peptides to be presented by .non-professional or immature APC without pro-inflammatory and co-stimulatory signal which promotes induction of tolerance, anergy .and or suppressive activity in respondin T cells
(Moklaver & Larehe, Allergy 66: 784-91, 2011) and/or other CD4+ T cells that express MHC class 11. This also allows -peptides, to he presented at higher frequency than peptides processed from the whole molecule (Santambrogio c¾ al. Proc Natl Acad S i U S A, 1 99, 96: 15056-61), and since they are also safer than whole allergen, peptides can be given at higher concentrations , thus repolarising T cell responses more effectively.
Importan tly, targeting T cells specific for dominan T cell epitopes of major allergens can alter responses to whole allergen extracts (linked suppression). Many studies reporting successful peptide immunotherapy in murine models of allergy demonstrated that administration of dominant T-cell epitope peptides of major allergens induced tolerance not only to those peptides, but also to purified allergen and whole allergen extracts (Yang et al Clin Exp Allergy 40(4)^668-78, 2010; Yoshitomi et al J Pept Sci. 13(8):499-503, 2007; Marazuela et al. Mol Immunol 45(2):438-45, 2008; Rupa et al Allergy, 67(l):74-82, 2012; Hoyne et al J £* Med. 178(5):1783-8, 1993; Hall et al Vaccine. 21 (5-6): 549-61, 2003). Accordingly. there is- a need to both identify the major peanut allergens and, further, to identify the T cell epitopes of these allergens. The identification, characterisation, and analysis of these T cell epitopes is critical to the development of specific immunotherapeutk or prophylactic methodology. To this end, although the Ar h 1 and/or Ara h 2 peanut allergen molecules have previously been the subject of analysis, the identification of the T cell core epitopic regions is essential to the development of aft effective vaccine.
In work leading up to the present invention, dominant, HL -degenerate Ara h 1 and or Ara h 2 core T cell epitopic regions have been identified. This group of core T ceil epitopic regions is unique in terms of its high level of efficacy. Unlike the previously studied 20mer Ara b. 1 and/or Ara h 2 peptides which were identified based only on then- ability to exhibit some level of T ceil reactivity, there have been ide tified a selected set of core T cell epitope regions which are both immunodominant,, relative to other Ara h 1 arid/or Ara h 2 peptide fragments, and are also HLA degenerate in that the bind to two or more HLA types. Still further, many of these T cell epitopic core regions are presented by HLA-DQ molecules. HLA-DQ molecules are more conserved in mixed populations than HLA-DR molecules. Accordingly, peptides presented on HLA-DQ enable broader population coverage.
In a still further finding, it has also been determined that a specific subgroup of seven of these peptides which contai both Ara h 1 and Ara h 2 T cell epitopes provides particularly effective immunological .outcomes. Accordingly, the identification of this uniquely eifective grou of peptides has f cilitated the development of particularl effective therapeutic prophylactic methods for the treatment of conditions characterised fay aberrant, inappropriate or otherwise unwanted immune responses to Ara h 1 and/or Ara h 2 or deri ative or homologue thereof, other tree nut allergies or allergens to a composition, such as foods, containing the Ara h 1 and/or Ara h 2 molecules.
SUMMARY OF THE INVENTION
Throughout this specification and the claims which .follow, unless the context requires otherwise, the word "comprise'", and variations such as. "comprises" and "comprising*', will be understood to imply the inclusion of a stated integer o step or group of integers or steps but not the exclusion of any oth r integer or step or group of integers o steps.
As used herein, the term "derived from" shall, be taken to indicate that a particular integer or group of integer has originated from the species specified, but has not necessarily been obtained directly from the specified source. Further, as used herein the singular forms of a'\ "and" and "the" include plural referents unless the context . clearly dictates otherwise.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the ait to which this invention, belongs.
The subject specification contains amino acid sequence irforrnation prepared using the programme PatentTn Version 3.5, presented herein after the bibliography. Each amino acid sequence is identified in the sequence listing by the .numeric indicator <210> followed by the sequence identifier (eg. <21()>1, <21,0>2, etc). The length, type of sequence (protein, etc) and source organism for each sequence is indicated by information provided in the numeric indicator fields <211>, <212> and <213>, respectively . Amino acid sequences referred to in the specification are identified by the indicator SEQ ID NO: followed by the sequence identifier (eg. SEQ ID NO: 1. SEQ ID NO:2, etc.). The sequence identifier referred to in. the specification correlates to the information provided in numeric indicator field <4QQ> in the sequence listing, which is followed by the sequence identifier (eg. 400>1 , <40G>2, etc). That is SEQ ID NO: 1 as detailed in the specification correlates to the sequence mdicated as <400>1 in the sequence listing.
One aspect of the present i nvention is directed to an immunomodulatory composition comprising at least five of the Ara h 1 and Ara h 2 T cell epitopie regions from the list consisting of:
(i) FQNLQNHR (SEQ ID NO:l)
(«) IVQIEA (SEQ ID O: 2)
(iit) NEGVrVKVSK (SEQ ID NO:3)
(iv) E V KPD PQLQ (SEQ ID N0:4)
(v) EGALML (SEQ ID NO:5)
( i) IMPAAHP (SEQ ID NO:6)
(vii) LRPXEQHLM (SEQ ID NQ:7)
(yiii) EN QRXMXEA (SEQ ID NO: 8)
or functional derivati ves or homologues thereof wherein residue X is cysteine or serine and said .composition comprises at least one T cell epitopie region selected from SEQ ID NQS : 1 -6 and at least one T cell epitopie region selected from SEQ ID NOS: 7-8.
In another aspect, said LRPXEQHLM is LRPSEQHLM (SEQ ID N0;137).
In still another aspect said ENNQRXMXEA is ENNQRSMSEA (SEQ ID NO: 138), In accordanc with these aspects and embodiments of the present invention, said composition comprises at least 6 of said T cell epitopie regions.
In a further aspect, said composition comprises at least 7 of said T cell epitopie regions. In still a further aspect, said composition comprises each of said 8 T cell epitopie regions. In another aspect there is provided an immunomodulatory composition comprising each of the Ara h 1 and Ara h 2 T cell epitopie regions from die list consisting of: (i) FQNLQNHR (SEQ ID NO:. I)
(ii) IVQIEA (SEQ ID NO; 2)
( tin NEGVIVKVSK (SEQ ID NO:3)
(iv) EVKPDKKNPQLQ (SEQ ID NO:4)
(v) EGALML (SEQ ID NO:5
(vi) IMPAAHP (SEQ ID NO:6)
(vii) LRPXEQHLM (SEQ ID NO:7)
(viii) ENNQRXMXEA (SEQ ID NO: 8)
or functional derivatives or homologues thereof wherein said residue X is cysteine or serine, In yet another aspect there is provided an immunomodulatory composition comprising each of the Ara h 1 and Ara h 2 T cell epitopic regions from the list consisting of:
(i) FQNLQNHR (SEQ ID NO : 1 )
(ii) IVQIEA (SEQ ID NO:2)
(iii) NEGVIVKVSK (SEQ ID NO:3)
(iv) EVKPDKKNPQLQ (SEQ ID NO:4)
(v) EGALML (SEQ ID NO:5)
(vi) IMPAAHP (SEQ ID NO: 6)
(vii) LRPSEQHLM (SEQ ID NO: 137)
(viii) ENNQRSMSEA (SEQ ID NO:13S)
or functional derivatives or homologues thereof.
In a related aspect the present invention is directed to an immunomodulatory composition comprising one or more peptides, each of which peptides is up to 60 contiguous amino acids in length and which peptides include each of the Ara h 1 and Ara h 2 T cell epitopic region combinations detailed hereinbefore.
In a further aspect the present invention is directed to an immunomodulatory composition comprising one or more peptides, each of which peptides is up to 60 contiguous amino acids in length and which peptides include each of the Ara h I and Ara h 2 T cell epitopic regions from the list consisting of:
(i) FQNLQNHR (SEQ ID NO: 1)
(ii ) IVQIEA (SEQ ID NO-2)
(iii) NEGVIVKVSK (SEQ ID NO:3>
(iv) EVKPDKKNPQLQ (SEQ ID NQ:4)
(v) EGALML (SEQ ID NO:5)
(vi) IMPAAHP (SEQ ID NO:6)
(vii) LRPSEQHLM (SEQ ID NG:7) (viii) E NQRSMSEA (SEQ ID NO:8)
or functional derivatives or homologiies thereof,
in yet another aspect said peptides or T cell epitopic regions are capable of modifying T cell function when presented to T cells isolated from subjects having a condition characterised by an aberrant, unwanted or otherwise inappropriate immune response to Ara h 1 and/or Ara b 2 or to an allergen present in a composition, such as food, comprising Ara h 1 and/or Ara h 2 but which peptides are unable to hind to Ara h I -specific and or Ara h 2 -specific IgE.
In accordance with these aspects, said, peptides are selected from the list consisting of:
(i) FQNLQNHR1VQ1EAKP T.LV (SEQ ID NO: 1.1 )
i ii t STR SS ENNEG VTVKVSKE (SEQ ID NO: 1.2)
(iii) EVKPDKKNPQLQ (SEQ ID NO:4)
Civ) VEIK EG ALMLPHFNS K A (SEQ ID NO:13>
(v) VFIMPAAHPVAINASS (SEQ ID NO: 14)
(vi) ANLRFXEQHLM (SEQ ID NO:15)
(vii) EFENNQRXMXEALQ (SEQ ID NO: 16)
(viii) NNFG LFEVKPD K .PQLQ (SEQ ID NO: 17)
(ix) GDVFIMPAAHPVAINASSE (SEQ ID NO:18)
( x i SQLE ANLRPX EQHLM (SEQ ID NO: 19)
(xi) ELNEFENNQRXMXEALQ (SEQ ID NQ:20)
( ii) FQNLQNHRIV (SEQ ID NO:2i)
(xiii) RIVQIEAKPNTLV (SEQ ID NO:22)
(xiv) ENNEGVrV K VSKE (SEQ ID MO;23)
(XV) EVKPDKKNPQLQD (SEQ ID NO: 24)
(xvi) EFENNQRXMXEALQQ1 (SEQ ID NO:25)
(xvu) NNFGKLFEVKPDKKNPQLQD (SEQ ID NO:26)
(xviii) ELNEFENNQRXMXEALQQI (SEQ ID NO:27)
(xx) WSTRSSENNEGVIVKVSKE (SEQ ID NQ:28)
(xxi) GD VFIMPAAHPVAIN SS (SEQ ID NO:2<¾
or functional derivatives or homologiies thereof wherei residue is cysteine or serine.
In one embodiment, said residue X is serine.
In a further asepct said peptides are selected from:
(i) FQNLQNHRIV Q1EAKPNTLV (SEQ ID NO: 11 )
(it) STR SS ENNEG VTV VSKE (SEQ ID NO : 12)
(iii) EVKPDKKNPQLQ (SEQ ID NO:4)
(iv) VEIKEGALMLPHFNSKA (SEQ ID NO: 13) (V) VFIMPAAHPVAINASS (SEQ ID NO': 14)
(vi) ANLRPSEQHLM (SEQ ID NO:31 )
fyii) EFENNQRSMSEALQ (SEQ ID NO: 32)
(viii) EVKPDKKNPQLQD (SEQ ID NO:24)
( lx) EFENNQRSMSEALQQI (SEQ ID NO: 33)
or functional derivatives or homologues thereof.
In another aspect, said immunomodulatory composition comprises each of the Ara h 1 and Ara h 2 T cell, peptides from the list consisting of:
(i) FQNLQ HR1VQIEAKP T.LV (SEQ ID NO: 1 1 );
(ii) STR SS ENNEG VTVKVSKE (SEQ ID NO: 12);
(iii) EVKPDKKNPQLQ (SEQ ID NO:34) and/or EVKPDKKMPQLQD (SEQ ID NO:24);
(iv) VEIK EG ALMLPHFNS K A (SEQ ID NO: 13);
(v) VFIMPA HPVAINASS (SEQ ID NO: J 4);
(vi) ANLRPSEQHLM (SEQ ID NO:31); and
(vii) EFENNQRSMSEALQ (SEQ ID NO:32) and/or EFENNQRSMSEALQQI (SEQ ID
NO:33)..
or functional derivatives or homologues thereof.
In a yet a further aspect, the inventors have designed a preferred set of seven peptides, five of which comprise Ara h 1 T cell epitopes and two of which comprise Ara h 2 T ceil epitopes, which function particularly efficaciously, when administered together, to induce desensitisation or tolerance and thereby either prophyiactiealiy or therapeutically treat hypersensitivity t compositions, suc as foods, comprising Ara h 1 and/or Ara h 2< These peptides are:
(i) FQNLQNHRIVQIEAKPNTLV (SEQ ID NO: 11)
iii) STRSSENNEGVJV VSKE (SEQ ID NO: 12)
(iii) EVKPDKKNPQLQ (SEQ ID NO:4) and/or EVKPDKKNPQLQ (SEQ ID NO:24)
(iv) VEIKEGALMLPHFNSKA (SEQ ID NO: 13)
(v) VFIMPAAHPVAINASS (SEQ ID NO: 14)
(vi) ANLRPSEQHLM (SEQ NO:3l)
(vii.) EFENNQRSMSEALQ (SEQ NO: 32) and/or EFENNQRSMSEALQQI (SEQ ID NO:33)
In still yet another aspect there is provided an. immunomodulatory composition comprising each of the Ara h 1 and Ara h 2 T cell peptides from the list consisting of:
ii) FQNLQNHRIVQIEAKPNTLV (SEQ ID NO: 1 1 )
(ii) STR SS ENNEG VI V VS E (SEQ ID NO : 12)
(iii) EVKPDKKNPQLQ (SEQ ID NO:4 and/or EVKPDKKNPQLQD (SEQ ID NO:24) iv) VEIKEGALMLPHFNSKA (SEQ ID NO: 13) (V) VFIMPAAHPVAINASS (SEQ ID NO: 14)
(vi) ANLRPSEQHLM (SEQ NO:31 )
(vii) EFENNQRSMSEALQ (SEQ NO:32) and/or EFENNQRSMSEALQQI (SEQ ID NQ:33) In a yet still, another aspect, there is provided c m osition' comprising each of the Ara h 1. and Ara h 2 T cell peptides from the list consisting of:
(i) FQNLQNHMVQIEAKPNTLV (SEQ ID NO: 1 1)
(ii) ST SSENNEGVrVKVSKE (SEQ ID NO:30)
(hi) EVKPDK NPQLQ (SEQ ID NO:4) and/or EVKPD KNPQLD (SEQ ID NO':24)
(iv) VEI EGALMLPHFNS A (SEQ ID NO: 13)
(v) VFIMPAAHPVAINASS (SEQ ID NO:14)
(vi) ANLRPSEQHLM (SEQ NO:31)
(vii) EFENNQRSMSEALQ (SEQ NO:32) and/or EFENNQRSMSEALQQI (SEQ ID NO:33) which peptides are capable of reducing Ara h. 1 and/or Ara h 2 hypersensitivity or hypersensitivity to a composition comprising Ara h 1 and/or Ara h 2 when administered to a subject having a condition characterised by said hypersensitivity.
The present invention is directed to a composition comprising the peptides hereinbefore defined. It should be understood, though, that the subject composition may comprise additional components, such as additional peptides. Example of other peptides which may be included in the composition include, but are not limited to:
(i) ALMLPHFNSKAMVIVVV (SEQ ID NO: 34)
00 NNFGKLFEV PD KNPQ (SEQ ID NO:35)
( i) SQLERANLRPXEQ (SEQ ID NO:36)
(iv) ELNEFE NQRXM (SEQ ID NO:37)
iv) NNFG LFEV PD KNPQLQD (SEQ ID J O:38)
(vi) NNFG LFEV PDK N.PQL (SEQ ID NO:40)
(vii) SQLERANLRPXEQH (SEQ ID NO:41)
(viii) KAMVlVy VNKGTGNLELVA (SEQ ID NQ:42)
(ix) RELRNLPQQXGLRA (SEQ ID NO:43)
( ) KAMVIVVVN G (SEQ ID NQ:44)
(xi) AMY IV V VNKGTGNLELV (SEQ ID NO:45)
(xii) V VNKGTGNLELV RK (SEQ ID NQ:46)
or functional derivatives or homoiogues thereof wherein residue X is cysteine or serine.
In another aspect, the present invention provides a nucleic acid molecule composition comprisin one or more nucleic acid molecules encoding or complementar t a sequence encoding the T cell epitopes and peptides as hereinbefore defined or a derivative, hoinologue or analogue thereof.
in still another aspect the present invention provides a method, for the treatment and/or prophylaxi of a condition i a subject, which condition is characterised by the aberrant, unwanted or otherwise inappropriate immune response t Ara h 1 and/or Ara h 2 or an allergen in a composition comprising Ara b 1 and/or Ara h 2, said method comprising administering to said subject an effective amount of an immunomodulatory composition as hereinbefore defined for a time and under conditions sufficient to remove or reduce the presence or function in said subject of T cells directed to said Ara h 1 and/or Ara h 2 or other allergen.
In a further aspect said condition is hypersensitivity to peanuts or tree nuts which contain
Ara h 1 and Ara h 2 or Ara h 1 -like or Ara h 2-like molecules, such as hazelnuts, almonds or Brazil nuts.
In another aspect, said method desensitises or induces immunological tolerance to Ara h 1 and/or Ara h 2 or other allergen of said composition.
In still another aspect, said de sensitization or tolerance is achieved by inducing T cell anergy or apoptosis.
In yet still another aspect, said desensitisation or tolerance is achieved by inducing Ara h 1 or Ara h 2-specific Treg cells.
A further aspect of the present invention contemplates the use of an immunomodulatory composition as hereinbefore defined in the manufacture of a medicament for the treatment of a condition in a mammal, which, condition is characterised by an. aberrant, unwanted or otherwise i nappropriate immune response t Ara h 1 and/or Ara h 2.
Preferably said condition is hypersensitivity to peanuts or a tree nut which contains Ara h 1 and/or Ara h 2 or Ara h 1 -like and/or Ara h 2-like molecules, such a a hazelnut.
In yet another further aspect, the present invention contemplates a vaccine comprising the composition as hereinbefore defined together with one or more pharmaceutically acceptable carriers and/or diluents. Said composition is referred to as the active ingredient.
Yet another aspect of the present invention relates to the compositions, as hereinbefore defined, when used in the method of the present invention.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a graphical representation of a CFSE screen for peptide-specific PBMC T ceils: (A) CFSE-labeiled PBMC from a peanut-allergic subject incubated with whole peanut extract or the Vax (7 peptide compilation). Boxes show percent of activated proliferating CD4+ T cells (CD25+CFSEIo). SI indicates fold increase in T cell activation above no antigen control. (B) Confirmation of peanut-allergic donor PBMC CD + T-cell activation and proliferation in response to the Vax (7 peptide compilation) in 7 subjects (all have a positive Si>2,5).
Figure 2 is a graphical representation of a basophil activation test (BAT) (A) FACS plots showing blood from a peanut-allergic subject incubated with, whole peanut extract or the Va (7 peptide compilation). Basophils are identified as IgEhi ceils (box, first plot) and activated basophils as CD63M (boxes, plots 2-4). (B) BAT data and (C) Histamine release data (measured by
commercial kit) from a. peanut-allergic subject following incubation with increasing concentrations ( g/ml) of whole peanut extract or the V ax. Positive controls are anti-lgE and fMLP. Whole peanut extract causes high basophil activation and histamine release but the a does not. Data are representative of 14 peanut-allergic subjects tested.
Figure 3 is a schematic representation of the method employed to identify the dominant epitopes of the major peanut allergens Ara h 1 and Ara h 2.
Figure 4 is a graphical representation of a 7-day CFSE assa designed to detect the ability of dominant 20-mer peptides to induce T cell proliferation in whole PBMC of peanut-allergic donors. The numbers in. the boxes indicate % of dividing (CFSE low) CD4+ T cells SI ~ told increase in dividing cells above unstimulated control.
Figure 5 is a graphical representation demonstrati ng responder frequency of T-cell lines to Ara h I 20-mer peptides. Boxes indicate the 9 dominant 20-rners ultimately selected (based on multiple parameters).
Figure 6 is a graphical representation of PBMC screening with dominant Ara h 1 20-mers.
The ability of dominant 20-rners to target specific CD4+ T-cells in PBMC from peanut-allergic donors was tested.
Figure 7 is a graphical representation demonstrating responder frequency of T-cell lines to Ara h 2 20~mer peptides and number of -specific TCL per 20-mer. Boxes indicate 4 dominant 20- mer peptides ultimately selected based on multiple parameters.
Figure 8 is a graphical representation of the- core T cell epitope mapping results.
Figur 9 is a graphical representation of the HLA restriction of the dominant Ara h .1 and Ara h 2 T cell epi opes.
Figure 10 is -a graphical represen tation of T cell recognition of peptides in which selected cysteine residues were replaced with serine residues. TCL proliferation in response to 'parent'
(cysteine containing) or serine-substituted Ara h 2 peptides as determined by Ή thymidine uptake. Graphs show representative TCL for each epitope (mean cpm replicate wells +SD). A) Ara h 2(32- 44). B) Ara h 2(37-47); C) Ara h 2(9.1 -102); D) Ara.h 2(95-107); E) Ara h 2(128-141).
Figure 11 is a graphical representation of T cell cytokine production in response to peptides in which selected cysteine residues were replaced with serine residues. Cytokine secretion in response t 'parent' or eysteine-substitute Ara h 2 peptides determined by EL1SPOT.
Graphs show representative TCL specific for each epitope (means spots of replicate wells +SD). IL-4, black bars; IL-5, hatched bars; IFN- , white bars; A) Ara h 2(32-44), B) Ara h 2(37-47); C) Ara li 2(91-102); D) Ara h 2(95-107); E) Ara h 2(128-141).
Figure 12 is a graphical representation of PBMC .responses to peptide pools vs whole peanut. Peptides included in each pool are shown in Table 23. P-vahies show Wilcoxon matched pahs signed rank test (for non-parametic data).
Figure 13 is a graphical representation of PBMC responses to peptide pools vs whole peanut. Peptides included in each pool are shown in Table 23. P-values show Wilcoxon matched pairs signed rank test (for non-parametic data).
Figure 14 is a graphical representation comprising PBMC T cell responses to the preferred 7-peptide pool. Stats: Rruskal-Walhs test for non-parametic data with Dunns post-hoc corrections to test for differences between multiple groups. *data normalised to % of response to whole peanut due to variation in magnitudes of responses in different subjects in the different cohorts analysed for the different pools.
Figure 15 is a graphical representation of Ara h 2 peptide -induced inhibition of T cell proliferation.
Figure 16 is a graphical represe tation of Ara h 1 peptide-ihduced inhibitio of T cell proliferation,
DETAILED DESCRIPTION OF THE INVENTION
The present invention is predicated, in part, on the identification of a grou of Ara h 1 and Ara h 2 epitopes which, when administered together, in a group of at least five* produce more efficacious immunological outcomes than any of these epitopes used either alone or together with other combination of these or othe Ara h 1 or Ara 2 peptides, to particular, it ha been determined mat the use of all eight epitopes produces particularly and exceptionally efficacious functional outcomes, most particularly when these eight epitopes are administered in the context of the seven peptides exemplified herein. The design of this composition has enabled the development of significantly more efficacious therapeutic and prophylactic compositions and treatment, approaches, than have been available to date, for conditions such as, but not limited to, peanut allergy.
Accordingly, one aspect of the present invention is directed to a inununomodulatory composition comprising at least five of the Ara h 1 and Ara h 2 T cell epitopie regions from the list consisting of:
(i) FQNLQNHR (SEQ ID NO:l) (it ! IVQIEA (SEQ ID NO:2)
(ill) NEGVIVKVSK (SEQ ID NO:3)
(Iv) E V KPDK K PQLQ (SEQ ID MO:4)
( v) EGALML (SEQ ID N0:5)
( vi ) IMPAAHP (SEQ ID NO:6)
(vii) LRPXEQHLM (SEQ ID N0:7)
(viii) ENNQRXMXEA (SEQ ID NO:8)
or functional derivatives or boraoiogues thereof wherein residue X is cysteine or serine and said composi tion comprises at least one T cell epitopic region, selected from SEQ ID NOS: 1-6 and at least one T cell epitopic region selected from SEQ ID NOS: 7-8.
In one embodiment, said LRPXEQHLM is LRPSEQHLM (SEQ ID NO;137),
In another embodiment, said ENNQRXMXEA is ENNQRSMSEA (SEQ ID NO: 138). In accordance with these aspects and embodiments of the present invention, said composition comprises at least 6 of said T cell epitopic regions,
In a further embodiment, said composition comprises at least 7 of said T c ll epitopic regions.
In still a further embodiment,, said c omposition comprises each of said.8 T cell epitopic regions.
According to this embodiment there is therefore provided an immunomodulatory composition comprising each of the Ara h 1 and Ara h 2 T cell epitopic regions from the list consisting of:
(0 FQNLQNHR (SEQ ID NO: I)
(ri) IVQIEA (SEQ ID NQ:2)
(111 ) NEGVIVKVSK (SEQ ID NO:3)
(iv) EVKPDKKNPQLQ (SEQ ID NO;4)
(v) EGALML (SEQ ID NO:5)
(vi) IMPAAHP (SEQ ID NO:6)
(vii) LRPXEQHL (SEQ ID NO:7)
(viii) ENNQRXMXEA (SEQ ID NO: 8)
or functional deri atives or homologues thereof wherein, said residue X is cysteine .or serine.
In another embodiment there is provided an immunomodulatory composition comprising each of the Ara h 1 and Ara h 2 T cell epitopic regions from the list consisting of:
(i) FQNLQNHR (SEQ ID NO: l )
(it) IVQIEA (SEQ ID NO:2)
(iii) NEGVIVKVSK (SEQ ID NO:3) (iv i EVKPDK PQLQ (SEQ ID NO;4)
(v) EGALML (SEQ ID NO:5)
fvi) IMPAAHP (SEQ ID NO:6)
(vii) LRPSEQHL (SEQ ED NO: 137)
(viii) ENNQRSMSEA (SEQ ID NO:138)
or functional .derivatives or homologues thereof.
In a related, aspect the present invention is directed to an immunomodulatory composition comprising one or more peptides, each of which peptides is up to 60 contiguous amino acids in length and which peptides include each of the Ara h 1 and Ara h 2 T cell epitopic region combinations detailed hereinbefore.
In accordance with this aspect the present invention is directed to an immunomodulatory composition comprising one or more peptides, each of which peptides is up to 60 contiguous amino acid in length and which peptide include each of the Ara h .1 and Ara h 2 T cell epitopic regions from the list consisting of:
S i) FQNLQNHR (SEQ ID NO':l}
(it) IVQIEA (SEQ ID NO:2)
(iii ) NEGVIV V SK (SEQ ID NO:3)
(iv) EVKPDKKNPQLQ (SEQ ID NO:4
(v) EGALML (SEQ ID NO:5
(yi) IMPAAHP (SEQ ID NO:6)
(vii) LRPSEQHLM (SEQ ID NO:?)
(viii) ENNQRSMSEA (SEQ ID NO: 8 )
or functional derivatives or homologues thereof.
In another embodiment of the preceding aspects of the invention, said peptides or T cell epitopic regions are capable of modifying T cell function when presented to T cell isolated from subjects having a condition characterised by an aberrant, unwanted or otherwise inappropriate immune response to Ara h I and/or Ara h 2 or to an allergen present in a composition, such as food, comprising Ara h 1 and/or Ara h 2 but which peptide are unable to bind to Ara h 1 -specific and/or Ara h 2 -specific IgE.
Without limiting the present invention in n way, peanuts contain many proteins, with the number of distinct bands visible on SDS-PAGE depending on the methodology used. Up to 53 bands are visible following high pressure liquid chromatography (de Jong etttl., Clin Exp Allergy 28: 743-5.1 , .1998). Only tw of these proteins warrant classification as major allergens using standard criteria, whereb IgE reactivity occurs within greater than 50% of the peanut allergic population; these proteins are termed Ara h 1 and Ara h 2 (Burks et at, Allergy 53: 725-30, 1998). Although a number of studies have indicated Ara h 2 to be the more potent of these two allergens (Blanc et al Clin Exp Allergy. 2009; 39(8): 1277-85; Koppelman et al. Clin Exp Allergy. 2004; 34(4):583-90; Palmer et al. Clin Immunol. 2005; 115(3):3Q2-12), Am h I also plays a major role in the pathogenesis of peanut allergy, with numerous studies: reporting strong correlations, between symptom severity and IgE reactivity to both Ara h .1 and Ara h 2 (Glautnann et al. Allergy. 20 2; 67(2):242-7; Chiang et al, Pediatr Allergy Immunol 2009; 21(2 Pt 2):e429-38; Asattt j etal Allergy. 2010, 65(9):1189-95; Moverare et al. i Arch Allergy Immunol 2011 ; 156(3)^82-90; Lin et al. J Microbiol Immunol Infect. 2012; Peeters et al Clia Exp Allergy. 2007; 37(1): 108-15). Ara b t is the most abundant major allergen in peanut, accounting for 12-16% of total peanut protein (Koppelman etal. Allergy. 2001 ; 56(2): 132-7).
Still without limiting the present invention in any way, the Ara h 1 allergen is a 7S seed storage glycoprotein, or vkulm. The concentration of Ara h 1. in peanuts increases with the size of the kernel (4-16 mg extracted Ara h 1/g peanut), so expression of the protein is associated with peanut maturity (Pome's et al. 2006, Clin. Exp. Allergy 36(6) : 824- 30). Ara h 1 is a homotrimer held together through hydrophobic areas at the distal ends of the monomers, where most of the IgE binding B cell epitopes are located. Each 64.5 kD monomer has a ciipin motif which consists of two core β -barrels, each associated to a loop domain of a-helices,
Ara h 2 is a glycoprotein which ha been identified as a member of the eonglutin. seed storage family. 20% of the Ara h 2 molecular mass represents carbohydrate side chains and it migrates as a doublet on SDS-PAGE with an average molecular mass of 17.5 kDa (Burks et al, Int Arch Allergy Immunol 119:165-172, 1992). It has been characterised as a major allergen, on the basis of its reactivity with 6 out of 6 sera tested (B urks et al, 1992, supra) . Others have also confirmed its importance; Clarke demonstrated that 71% of subjects possessed specific IgE to Ara h 2 upon western blotting of crude peanut extract. Kleber-Janke et al. have demonstrated that 85% of subjects possessed IgE towards their recombinant form upon western blotting, and de Jong's group have shown, that, approximately 78% of their group demonstrate specific IgE to purified natural Ara h 2 (Clarke et al, Clin Exp Allergy 28: 1251-7, 1 98; de Jong et al, 1998 supra;
Kleber-Janke et al., Int Arch Allergy Immunol 119: 265-274, 1999),
Reference to 'Ara h 1 " should be understood as a reference to all forms of this molecule including reference to an isoforms which ma arise from alternative splicing of Ara h. 1 mRNA or functional mutant or polymorphic forms of Ara h 1. it should still further be understood to extend to any protein encoded by the Ara h 1 gene, any subunit polypeptide, such as precursor forms which may be generated., whether existing as a monomer, multimer or fusion protein. It also includes reference to analogues or equivalents of Ara h .1 such as may occur, where a product which. naturally comprises Ara h 1 is syntheticall generated for the purpose of generating a product such as a food additive. The present invention thereby provides T cell epitopes and methods for their use in the diagnosis and treatment of any condition characterised by hypersensitivity to an Ara h 1 of Ara h 1 -like molecule, such as peanut allergy or a tree-nut allergy, or an allergy to an antigen present in a composition, such as food, which composition also comprises Ara h. L Preferably, said Ara h 1 comprise the sequence set forth in SEQ ID NO:9 and Ara h 2 comprises the sequence set forth in SEQ ID NO: 10.
Reference to ''T cells" should be understood as a reference to any cell comprising a T cell receptor. In this regard, the T cell receptor may comprise any one or more of the , , or chains. The present invention is not intended to be limited to any particular functional sub-class of T cells although in a preferred embodiment the subject T cell i a T helper cell and still more preferably a Th2-type cell and/or Treg cell. In this regard, reference to "modifying T cell function" should be understood as a reference t modifying any one or more functions which a T cell i capable of performing. For example, the subject function may be proliferation, differentiation or other form of cellular functional activity such as the production of cytokines. In one embodiment the su bjeet functional activity is proliferation.
In terms of "modifying the function" of T cells isolated from subjects having a condition characterised by an aberrant, unw nted or inappropriate immune response to Ara h 1 and/or Ara h 2 or to a composition which comprises Ara h 1 and/or Ara h 2, it should be understood, that this is not necessarily a reference to modifying the function of all the T cells in a given biological sample but is likely, in fact, to reflect the modification of functioning of only some of the T cells in the sample. For example, only a portion of the T helper cells in a given T cell sample may .functionally respond to contact with the subject peptide. Such a partial response should be understood to fail within the scope- of the present invention. It should also be understood that the T cells which are derived from the subject may be freshly harvested T cells or they may have undergone some form of in vitro or in vivo manipulation prior to testing. For example, T cell .lines may have been generated from the cell sample and it is these T cell lines which then form the subject derived T cell population which is tested in accordance with the present invention. To the extent that the subject functional activity is T cell proliferation, the T ceil proliferation assay is preferably performed as disclosed herein. Still more preferably, the subject modification of T cell function is the induction of proliferation. In this regard, reference to "Ara h 1 -reactive" or "Aran 2-reactive" T cells should, be understood as a reference to a T cell which responds functionally to HLA presentation of an Ara h 1 and/or Ara h 2 T cell epitope, respectively. Similarly, reference to "Ara h 1 -specific" or "Ara h 2-specific" lgE should be understood as a reference to lgE directed to Ara h 1 or Ar h 2 B cell epitopes, respectively.
Reference to an "aberrant, unwanted or otherwise inappropriate" immune response should - 1.7 - be understood as a reference to any form of physiological activity which involves the activation and/or functioning of one or more immune cells where that activity is inappropriate in that it is of an inappropriate type or proceeds to an inappropriate degree, it may be aberrant in. that according to known immunological pfincipies it either should not occur when, it does so or else should occur. when it does not do so. In another example, the immune response may be inappropriate in that it is a physiologically normal response but which is unnecessary and/or unwanted, such as occurs with, respect to type-I hypersensitivity responses to innocuous allergens, in the context of the present invention, this immune response may be directed to Ara h 1 and/or Ara h 2 or it may be directed to a different allergen which is present in a composition together with Ara h 1. and/or Ara h 2, Without linn. ting the present invention to any one theory or mode of action, it has been determined that even where the hypersensitivity response is directed to an allergen other than Ara h 1 and/or Ara h 2, which allergen is present in a composition which nevertheless comprises Ara h 1 and/or Ara h 2, treatment via the method of the present invention which is directed to Ara h 1. and/or- Ar h 2 nevertheless induces beneficial modulation of Th2 and Treg functionality such that the hypersensitivit which exists to the unrelated allergen is nevertheless reduced. Preferably said immune response is peanut hypersensitiv ty.
B "peanut hypersensitivi ty" is meant the induction of clinical symptoms of IgE mediated peanut hypersensitivity. However, it should be understood that although clinical symptoms may be evident, not all such individuals would necessarily exhibi t detectable level of peanut specific serum IgE which is measured using the Kaiiestad Allercoat EAST System (Sanofi-Pasteur
Diagnostics, USA), although such, individuals should nevertheless be understood to fall within the scope of the definition of those exhibiting "peanut hypersensitivity". Alternatively, testing may proceed utilising any of the EAST, Pharmacia or the UniCap systems or allergen skin prick testing. Reference to "Ara h 1 and/or Ara h 2 hypersensitivity" should be understood to have a
correspondin meaning in the context of reactivity to the Ara h .1 and or Ara h 2 protein.
In accordance with the preceding aspects, said peptides are selected from the list consisting of:
(i) FQNLQMHRIVQIEA PNTLV (SEQ ID NO: 1.1.)
00 STRSSENNEOVIV VSKE (SEQ ID NO: 12)
( hi ! EV PDKKNPQLQ (SEQ ID NO:4)
(iv) VEIKEGALMLPHFNSKA (SEQ ID NO: .13)
1 v) VFIMPAAHPVAINASS (SEQ ID NO: 14)
(vi) ANLRPXEQHLM (SEQ ID NO:! 5)
(vii) EFENNQRXMXEALQ (SEQ ID NO:I6)
(viii) NNFGKLFEV PDKKNPQLQ (SEQ ID NO: 17) GDVFIMPAAHPVAINASSE (SEQ ID NO: 18)
00 SQLE ANLRPXEQHLM (SEQ ID NO: 19)
f i) ELNEFENNQRXMXEALQ (SEQ ID NO:20)
(xii) .FQNLQNH JV (SEQ ID NO:21 )
(xiii) RIVQIEAKPNTLV (SEQ ID NO:22)
(xiv) BNNEGV IVK VSKE (SEQ ID NO:23)
(xv) EVKPDKKNPQLQD (SEQ ID NO: 24)
(xvi) EFENNQ XMXEALQQI (SEQ ID N :25)
(xvii) NNFGKLFE KPDK NPQLQD (SEQ ID NO:26)
(xviii) ELNEFENNQRXMXEALQQI (SEQ ID NO:27)
f x) WSTRSSENNEGVIVKVSKE (SEQ ID NO:28)
(xxi.) GDVFIMPA A HPVAIN ASS (SEQ ID NO:29)
or functional derivatives or homologues thereof wherein residue X is cysteine or serine.
In one embodiment, said residue is serine.
Preferably, said peptides are selected from:
(0 FQNLQNHRIVQIEAKPNTLV (SEQ ID MOM 1 )
(ii) STRSSENNEGVIVKVSKE (SEQ ID NO: 12)
( iii) EVKFDKKNPQLQ (SEQ ID NO:4
(iv) VEIKEGALMLPHFNSKA (SEQ ID NO: 13)
(v) VFIMPAAHPVAINASS (SEQ ID NO: 14)
(vi) ANLRPSEQHLM (SEQ ID NO:31)
(vii) EFENNQRSMSEALQ (SEQ ID NO: 32)
(viii) EVKPDKKNPQLQD (SEQ ID NO: 24)
i ix ) EFE QRSMSEALQQI (SEQ ID NO: 33)
or functional derivati es or homoiogues thereof.
In another embodiment, said immunomodulatory composition, comprises each of the
Ara h 1 and Ara h 2 T cell peptides from the list consisting of:
(b FQNLQNHRIVQIEAKPNTLV (SEQ ID NO: 11);
(ii) STRSSENNEGVTVK SKE (SEQ ID NO: 12):
(Hi) EVKPDKKNPQLQ (SEQ ID NO:34) and/or EVKPDKKNPQLQD (SEQ ID NO:24)
(iv) VEIKEGALMLPHFNSKA (SEQ ID NO: 13);
1 v) VFIMPAAHPVAINASS (SEQ ID NO: 14);
(vi) ANLRPSEQHLM (SEQ ID MO:31); and
(vii) EFENNQRSMSEALQ (SEQ ID NO:32) and/or EFENNQRSMSEALQQI (SEQ ID NQ:33). - 1.9 - or functional derivatives or hom iogues thereof.
The reduction of peanut, Ara h 1 and Ara h 2 hypersensitivity (and allergen
hypersensitivity more generally) is discussed in more detail hereafter. Briefly, however, this may take (he form of either partially or completely desensi .rising or toierising art individual to Ara h 1 and Ara h 2 specifically .or peanut or other protein more generally.
Reference t a "peptide" includes reference to a peptide, polypeptide or protein or parts thereof. The peptide may be glycosylated or unglycosylated and/or may contain a range of other molecules fused, linked, bound or otherwise associated to the protein such as amino acids, lipids, carbohydrates or other peptides, polypeptides or proteins. Reference hereinafter to a "peptide" includes a peptide comprising a sequence of amino acids as well as a peptide associated with other molecules such as amino acids, lipids, carbohydrates or other- peptides, polypeptides or proteins.
"Derivatives" include fragments, parts, portions and variants from, natural, synthetic or recombinant sources including fusion proteins. Parts or fragments include, for example, active regions of the subject peptide. Derivatives may be derived from insertion, deletion or substitution of amino acids, Amino acid insertional derivatives include amino and/or catboxylte terminal fusions as well as intrasequence insertions of single or multiple amino acids, Insertion al amino acid sequence variants are those in which one or more amino acid residues are introduced into a predetermined site in the protein although random insertion is also possible with suitable screening of the resulting product. Deletional variants are characterized by the removal of one or more amino acids from the sequence.
Substitutional amino acid variants are those in which at least one residue in the sequence has been removed and a different residue inserted in its place. An example of substitutional amino acid variants are conservative amino acid substitutions. Conservative amino acid substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine and leucine; aspartic acid and glutamic acid; asparagine and glutamine; serine and threonine; lysine and arginine; and phenylalanine and tyrosine. Additions to amino acid sequences include fusions with other peptides, polypeptides or proteins, in one embodiment, cysteine residues are substituted with serine, as exemplified herein.
Chemical and functional equivalents of the subject peptide should be understood as molecules exhibiting any one or more of the functional activities of these molecules and may be derived from any source such as bein chemically synthesized or identified via screening processes such as natural product screening.
Homoiogues include peptides derived from varieties other than peanuts, such as peptides derived from other tree nuts.
Analogues contemplated herein include, hut are not limited to, modification to side chains. incorporating of unnatural amino- acids and/or their derivatives during peptide, pol peptide or protein synthesis and the use of crosslinkers and other methods which impose conformational constraints on the pmteinaceous molecules or their analogues. Mutants include molecules which exhibit modified functional, activity (for example, Ara b.1 peptides which, express one or more T cell epitopes but lack B cell reactivity).
Examples of side chain modifications contemplated by the present invention include modifications of amino groups such as by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH^; amidination with methylacetimidate; acylation with acetic anhydride; carbamoyiation of amino groups with cyanate; trinitobenzyiation of amino groups with 2, 4, 6-trinitrobenzen'e sulphonie acid (TNBS); acylation of amino groups with succinic anhydride and tetrahydrophthalic- anhydride; and pyridoxylation of lysine with pyridoxal-5 -phosphate followed by reduction with NaBH^
The guankiine group of arginine residues may be modified by the formation of heterocyclic condensation products with reagents such as 2-3-butanedione, phenylglyoxal and giyoxal. The carboxyl group may be modified by carbodiirtiide activation via O-acylisourea formation followed by subsequent derivatisation, for example, to a corresponding amide, Suiphydryl groups may be modified by methods such as carhoxymethylation with iodoacetic acid or iodoacetamide;
performic acid oxidation to eysteie acid; formation of mixed disulphkles with other thiol compounds; reaction with maleimide, maleic anhydride or other substituted maleimide; formation of mercurial derivatives using- 4-chloroitiercuribenzoate,.4<hloromercuiiphenylsulphonie acid, phenyimercury chloride, 2-chloromereuri-4-nitropbenal and other mercurials; carbamoyiati n with cyanate at alkaline pH. Tryptophan residues may be modified by$ for example, oxidation with N- bromosueciniiiiide or alkylation of the indole rin with 2-hydroxy-5~nitrobe»zyl. bromide or sulphonyl hali es. Tyrosine residues on the other hand, may be altered by nitration with tetranitromethane to form a 3-nitrotyrosine derivative.
Modification of the imidazole ring of a histidine residue may be accomplished by alkylation with iodoacetic acid derivatives or N-carboethoxylation with diethylpyrocarbonate.
Examples of incorporating unnatural amino acids and derivatives during protein synthesis include, but are not limited to, use of notieueine, 4~amino butyric acid, 4-amino-3-hydroxy-5~ plienylpentanoic acid, 6-ami.nohexanoie acid, t-butylglyeine, norvaline, phetwlgiycine, ornithine, sarcosine, 4-amino-3-hydroxy-6-methylheptanoic acid, 2-thienyl alanine and/of D-isomers of amino acids, A list of unnatural amino acids contemplated herein is shown in Table 1 , TABLE 1
Non-conventional Code Non-conventional ( amino acid amino acid
-aminobutyric acid Abu L-N -methylalanine mala
-amino- -methylbutyrate Mgabu L-N -methyl argi nine Nmarg aminocyclopropane- Cpro L-N-methylasparagine Nmasn carboxylate L-N-methy f aspartic acid Nmasp aminoisobutyric acid Aih L-N-methylc ysterne Nmcys aminonorbornyl- Norb L-N-methyiglutainine Nmgln c rboxylate L-N-methylgiutamic acid mglu cyclohexylalanine Chexa L-N-methythistidine Nmhis eyclopentylala ne Cpen L-N - methyl i soil eucine Nmile
D -alanine Dai L-N -methyl leucine Nmleu
D-arginine Darg L-N-methyilysine Nmlys
D-aspartic acid Dasp L-N-methylmethionine Nnmiet
D-eysteine Dcys L-N -rnethyluorieucine Nmnle
D-glutamine Dgln L-N-methylnorvaiine Nmnva
D -glutamic acid Dgiu L-N-methykmii thine Nmorn
D-histidine Dhis L~N~methy {phenylalanine Nmphe
D-isoieucine Dile L-N -methylproline Nmpro
D -leucine Dleu L-N-methylserine Nmser
D -lysine D!ys L-N-methyl threonine Nmthr
D -methionine Drnet L-N-methylf ryptoph an Nmtrp
D-omithine D'orn L-N-niethyltyrosme Nmtyr
D -phenylalanine Dphe L-N-methyivaiiue mval
D-proline Dpro L-N-methykthylglycine Nmetg
D -serine Dser L-N-methyl -t-bu tylglycine Nmtbug
D -threonine Dth L-norleucifie Nie
D-tryptophan Dtrp L-norvaline Nva
D -tyrosine Dtyr -niethyl-aminoisobutyrate Maib
D-valiue Dval -methyl - -aminobuf yrate Mgabu
D- -methyialanme Dmala -methylcyclohexylalanme Mchexa
D- -methylarginine Dmarg -methylcylcopentylalaninc Mcpen D- -methylasparagine Dmasii -methyl- -napthylalanine Manap
D- -methylaspartate Dmasp -me thylpeni cillamine Mpen
D- -methylcysteine Dmcys N-(4-aminobutyi)glycine Nglu
D- -methyl glutamine Dmgln N-{2-amtnoethy1)glycme Naeg
D- -methylhistidine Dmhis N-{3 -ami nopropyDgl yci ne Norn
D- -methylisole cine Dmile N-ami.no- -methylbutyrate Nmaabu
D- -methylleucine Dmleu -napthylalanine Anap
D- -methyilysme Dmlys N-benzylgiycine Nphe
D- -meth ylrneth ionine Dromet N-(2-carbamylet.hyl.)glycine Ngln.
D- -methylornithine Dinorn N-(carbamylmethyl) glycine Nasn
D- -memylphenylalanine Dmphe N -(2-carboxyethyDglycine Nglu
D- -methylproUne Dmpro N-(ca.rboxymetbyi)glycine asp
D- -methyl serine Dmser N-cyelobutylglyeine Ncbut
D- -methyl threonine Dmthr N-cycloheptylgl ye ine Nehep
D- methyl tryptophan Dmtrp N-eyclohexylglycine Nehex
D- -methyityrosine Dmty N-eyclodeeylglyci.n.e Ncdec
D- -methy valine Dmval N-eylcododecylglycine Ncdod
D - N - methylal anine Dnmal N-cyelooety glyeme Ncoct
D -N -methyl arginine Dnmarg N-cyelopropyiglyciiie Ncpro
D -N -methylasparagine Dnmasn N-cyclourrdecylglycine Ncund
D -N -meth l aspartate Dnmasp N -{2 , 2-diphe nyie thyl jglyeine bhm
D-N-methylcysteine Dnmcys N-(3 ,3-di phenyipropyi )gl yc ί ne Nbhe
D -N -methylglu taminc Dnmgln N-(3-guanidinopropyl)glycine Narg
D-N- ethylglutamate Dnmglu N~( 1 -liydroxyethyl)glyciiie Nthr
D- -methylhistidine Dnmhis N-(hydrox.yetbyi))g.iycine Nser
D -N-niethylisoleucine Dnmile N-(iniidazo1ylethyl))g1ycine Nhis
D -N -methylleucine Dnnileu N-(3-tndolylyethyI)glydne Nhtrp
D -N -methyl lysine Dnmlys N-methyl- -aminobutyrate mgahu
N-methylcyclohexylalanine Nmchexa D-N-metbyimethionine Dnmrnet
D -N -meth l ornithine Dnmoin N -riietbyleycl open ty lalanine Nmcpen
N-methylgiycine Nala D-N-methylphenylalantne Dnrnphe
N-methylaminoisobutyrate Nmaib D-N-methy prol me Dnmpro
N -( .1 -methylpropyDglyeii e Nile D-N-methy.lserine Dnmser
N-{ 2-methylpropyt)glycine Nleu D-N-methyUhreonine Dnmthr
D -N-methyltryptophan Dnmtrp N-( 1 -methylcthy ^gl cine Nval D-N-methyltyrosine Dnmtyr N-memyla-naptbylalanine manap
D -N -methyl valine Dnmval N-methylpenicillamme Nmpen
-aminobutyric acid. Gabu -( j-hydrQ yphenyl)glycme Nhtyr
L^but {glycine Tbug N-(t'bi0methyl)glyeine Ncys
L- ethylglycine Etg penicillamine Pen
L-hornophenylalafli,ne Hpbe L- -methylalanine Mala
L- -niethylarginine Marg L- -methylasparagine Masn
L- -methylasparta te Masp L- -methyl-f-butyiglycine. Mtbug
L- -methy.lcysteine Mcys L-metbyiethylgl.yci.ne Metg
L- -methylglutaniine Mgln L- -niethylglutamate Mglu
L- -methylhistidine Mhis L- -metliyihomophenylalaiiine Mbphe
L- -methylisoleucirte Mile N-(2-methylthi( ethyi)glycine met
L- -methylleucine Mleu L- -methyllysine Mlys
L- -methylniethioni ne Mmet L- -methytaorleurine Mnle
L~ -methyliioi-vaime Mijva L- -rnethylornithitse Morn
L- -methylphenylalamne Mphe L- -methylproiine Mpro
L- -methylserine Mser L- -methylthreonine Mthr
L- -methyl tryptophan Mtrp L- -raetliyltyrosine Mtyr
L- -methylvaline Mval L-N-methylhoniophenylalanme mhphe
N-fN-f2,2-diphenyletliyl) Nnbhm N -(N -(3 ,3 -diphenylpropyl) Niibhe earbamylmethy glycine earbatriyimethyi glycine
1 -carboxy- 1 -(2 ,2-diphenyl -Nmbc eth l artiino)cyelopropaae
Cimslinfcers can be used, for example, to stabilise 3D conformations, using homo-biftmetioiial crosslmkers such as the bi&nctional αχύόο- esters having (GH2)J1 spacer groups wi th n=l. to n=6, glutaraldehyde, N-hydroxysuccinimide esters and hetero-bifiiftetioftal reagents which usually contain an ammo-reactive moiety such as N-hydroxysucciaim e and another group specific- reactive moiety.
It is possible to modify the structure of a peptide according to the invention for various purposes such as for increasing solubility., enhancing therapeutic or preventative efficacy, enhancing stability or increasing resistance to proteolytic degradation, A modified peptide may be produced in which the amino acid sequence has been altered, such as by amino acid substitution, deletion or addition, to modify immimogenicity and/or reduce ailergenicity. Similarly components may be added to peptides of the invention to produce the same result.
For example, a peptide can be modified so that it exhibits the ability to induce T cell anergy. In this instance, critical binding residues for the T cell receptor can be determined using known techniques (for example, substitution of each residue and determination of the presence or absence of T cell reactivity) In one example, those residues shown to be essential to interact with the T cell receptor can b modified by replacing the essential amino acid with another, preferably similar amino acid residue (a conservative substitution) whose presence is shown to alter T cell reactivity or T cell functioning. In addition, those amino acid residues which are not essential for T cell receptor interaction can be modified by bein replaced b another amino acid whose incorporation may then alter T cell reactivity or T cell functioning but does not, for example, eliminate binding to relevant MHC proteins. In yet another example, mutant peptides may be created which exhibit normal T cell binding but abrogated IgE binding.
Exemplary conservative substitutions are detailed, below, and include:
Figure imgf000025_0001
Such modifications will result in the production of molecules falling within the, scope of "mutants" of the subject peptide as herein defined, "Mutants" should be understood as a reference to peptides which exhibit one or more structural features or functional activities which are distinct from those exhibited by the non-mutated peptide counterpart. Peptides of the invention ma also be modified to incorporate one or more poiymorphisnis resulting from natural allelic variation and D-amino acids, non-natural amino acids or amino acid analogues may be substituted into the peptides to produce modified peptides which fell, within the scope of (he invention. Peptides may also be modified by conjugation with polyethylene glycol. (PEG) by known techniques. Reporter groups may also be added to facilitate purification and potentially increase solubility of the peptides according to the invention. Other well known types of modification including insertion of specific endoprotease cleavage sites, addition of functional groups or replacement of hydrophobic residue with less hydrophobic residues as well as site- directed mutagenesis of DNA encoding the peptides of the invention may also be used to introduce modifications which could be useful for a wide range of purposes. The various modifications to peptides according to the invention which have been mentioned above are mentioned by way of example only and are merely intended to be indicati ve of the broad range of modifications which can be effected.
As detailed hereinbefore, the present invention provides peptides which retain all or some of their capacit to interact with T cells but exhibit partially or completely inhibited* abrogated or otherwise down-regulated antibody reactivity. Effecting the down-regulation of antibody reactivity can be achieved by any suitable method, which methods would be well known to those skilled in the art. For example, to the extent that a B cell epitope is defined by its linear amino acid sequence, one may add, delete or substitute one or more amino acid residues in order to render the mutated linear sequence distinct from the naturally occurring sequence. To the extent that an epitope may be additionally, or alternatively, defined by a conformational epitope, one may seek to disrupt that conformation by disrupting a 2° or, to the extent that homodimers or heterodimers exist, a 3e structure of the peptide. This may be achieved, for example, by disrupting the formation of bonds, such as disuiphide bonds, which are known to stabilise 2° and/or 3° structures. In terms of the T cell epitopes hereinbefore defined, these T cells epitopie regions do not comprise B cell epitopes.
in a related aspect, the inventors have designed a preferred set of seven peptides, five of which comprise Ara h 1 T ceil epitopes and tw of which comprise Ara h 2 T cell epitopes, which function particularly efficaciously, when administered together, to induce desensitisation or tolerance and thereb eithe prophylacti.cally or therapeutically treat hypersensitivity to compositions, such as foods, compri ing Ara h 1 and/or Ara h 2. These peptides are:
(i) FQNLQNHR1VQIEAKPNTLV (SEQ ID NO: 11 )
(it) ST SS ENNEG VT V VS E (SEQ ID NO: 12)
(iii) EVKPD KNPQLQ (SEQ ID NO:4) and/or EVKPD KNPQLQD (SEQ ID NO:24) (iv) VEIKEGALMLPHFNSKA (SEQ ID NO: 13) (V) VFIMPAAHPVAINASS (SEQ ID NO': 14)
(vi) ANLRPSEQHLM (SEQ NO:31 )
fyii) EFENNQRSMSEALQ (SEQ NO:32) and/or EFENNQRSMSEALQQ1 (SEQ ID NQ:33) Accordingly, in a preferred embodiment there is provided an immunomodulatory
composition comprising each of the Ara h 1 and Ara h 2 T cell peptides from the list consisting of:
(i) FQNLQNHRlVQIEAKPNTLV (SEQ ID NO; 11)
(ii ST SSENNEGVrVKVSKE (SEQ ID NO: 12)
(hi) EVKPDK NPQLQ (SEQ ID NO:4) and/or EVKPD KNPQLQD (SEQ ID NO:24) (iv) VEIKEGALMLPHFNS A (SEQ ID NO: 13)
(v) VFIMPAAHPVAINASS (SEQ ID NO:I4)
(vi) ANLRPSEQHLM (SEQ NQ:31)
(vu) EFENNQRSMSEALQ (SEQ NO:32 and/or EFENNQRSMSE LQQI (SEQ ID NO:33)
In a further aspect, there is provided a composition comprising each of the Ara h 1 and Ara h 2 T cell peptides from the list consisting of:
(i) FQNLQNHRIVQIEAKPNTLV (SEQ ID NO:ll)
(ii) S I SS ENNEGV1 V KE (SEQ ID NO: 12)
(iii) EVKPD KNPQLQ (SEQ ID NO:4) and or E KPDKKNPQLQD (SEQ ID N 0:24)
(iv) VEIKEGALMLPHFNSK A (SEQ ID NO: 13)
(v) VFIMPAAHPVAINASS (SEQ ID NO:14)
(vi ) ANLRPSEQHLM (SEQ NO:31)
(vii) EFENNQRSMSEALQ (SEQ NO:32) and/or EPENNQRSMSEALQQl (SEQ ID NO:33) which peptides are capable of reducing Ara h I and/or Ara h 2 hypersensitivity or hypersensitivity to a composition comprising Ara h 1 and/or Ara h 2 when administered to a subject having a condition characterised by said hypersensitivity.
The peptides of the present invention may be prepared by recombinant or chemical synthetic means. According to preferred aspect, of the present invention, there is provided a recombinant peptide or mutant thereof which is preferentially immunologically reactive with T cells from individuals with peanut hypersensitivity, which is expressed by the expression of a host cell transformed with a vector coding for the peptide sequence of the present invention. The peptide may be fused to another peptide* polypeptide or protein. Alternatively, the peptide may be prepared by chemical synthetic techniques, such as by the Merrifield solid phase synthesis procedure. Furthermore, although synthetic peptides of the sequence given above represent a preferred embodiment, the present inventio also extends to biologically pure preparations of the naturally occurring peptides or fragments thereof. By ''biologically pure" is meant a preparation comprisin at least about 60%, preferably at least about 70%, or preferably at least about 80% and 2? still more preferably at least about 90% or greater as determined by weight, acti vity or other suitable means.
The present invention should therefore be understood to encompass peptides that comprise at least one T cell core epilopie region of Ara h .1 arid/or Ara h 2, as hereinbefore defined, in conjunction with other amino acids (which may or may not be naturally occurring) or other chemical species. In. a preferred aspect of the invention such peptides may comprise one or more epitopes of Ara h 1 and or Ara h 2, which epitopes are T cell core epitopic regions, Peptides with one or more T cell epitopes of Ara h 1 and/or Ara h 2 are desirable for increased therapeutic effectiveness.
As detailed hereinbefore, the present invention is directed to a composition comprising the peptides hereinbefore defined. It should be understood, though, that the subject composition may comprise additional components, such as additional, peptides. These pepttdes ma encompass, for example, partial regions of the core minimal epitope. Al ternatively, they may not comprise any part of a T cell epitope as disclosed herein but may be incorporated for either reasons. Examples of other peptides which ma he included in the composition include, but are not limited to:
(i.) ALMLPHFNSKAMVrVVV (SEQ ID NO:34)
(if) NNFGKLFEVKPDK FQ (SEQ ID NO:35)
(m) SQLER ANLRP EQ (SEQ ID NO:36>
(iv) ELNEFENNQRXM (SEQ ID NO;37)
(v) NNFGKLFEVKPDK NPQLQD (SEQ ID NO:38)
(vi) NNFGKLFEV PD KNPQL (SEQ ID NO:40)
(vii) SQLERANLRPXEQH (SEQ ID NO:4I )
(viii) AM IV VVN GTGNLELVAV (SEQ ID NQ:42)
(ix) RELRNLPQQXGLRA (SEQ ID NO:43)
(x) KAMVrVVVNKG (SEQ ID NO:44)
(xi) AMVrVWNKGTGNLELV (SEQ ID NO:45)
(xii) VVN GTGNLELV AVR (SEQ ID NO:46)
or functional derivatives or homologu.es thereof wherein residue X is cysteine or serine.
One may also include still other peptides or molecules which may he advantageou given the particulars of a specific situation.
In another a pects the present invention provides a nucleic acid molecule composition comprising one or more nucleic acid molecules encoding or complementary to a sequence encoding the T cell epitopes and peptides as hereinbefore defined or a derivative, homologue or analogue thereof. Ir should be understood that reference to "peptides" includes reference t peptides comprising one or more T cell epitopes. A nucleic acid molecule encoding the subject peptide is preferably a sequence of deoxyribonucleic acids such as cDNA or a genomic sequence. A genomic sequence may comprise exons and introns. A genomic sequence may also include a promoter region or other regulatory regions.
The nucleic acid molecule may be ligated to an expression vector capable of expression in a prokaryotic cell (eg. E. cpli) or a eukaryotic cell (eg. yeast cells, fungal, cells, insect cells, mammalia cell or plant cells). The nucleic acid molecule may be ligated or fused or otherwise associated with a nucleic acid molecule encoding another entit such as, for example, a signal peptide. It may also comprise additional nucleotide sequence information fused, linked or
otherwise associated with it cither, at the 3' or 5* terminal portions or at both the 3' and 5' terminal portions. The nucleic acid molecule may also be part of a vector, such as an expression vector. The latter embodiment, facilitates production of recombinant forms of the subject peptide which forms are encompassed by the present invention.
Such nucleic acids may be useful for recombinant production of T cell epitopes of Ara h 1 and/or Ara h 2 or proteins comprising them by insertion into an appropriate vector and transfection into a suitable cell line. Such expression vectors and host ceil lines also form an aspect of the invention.
In producing peptides by recombinant techniques, host cells transformed wi th a nucleic acid having a sequence encoding a peptide according to the invention or a functional equivalent of the nucleic acid sequence are cultured in a medium, suitable, for the particular cells concerned. Peptides can then be purified from cell culture medium, the host cells or both using techniques well known in the art such as ion exchange chromatography, gel filtration chromatography,
ultrafiltration , electrophoresis or Immunopurification with antibodie specific for the peptide,
Nucleic acids encoding Ara h 1 and/or Ara fi 2 or peptides comprising T cell core epitopic regions of Ara h 1 and/or Ara h 2 may be expressed in bacterial cells such as E. coli, insect cells, yeast, or mammalian cells such as Chinese hamster ovary cells (CHO). Suitable expression vectors, promoters, enhancers and other expression control elements are referred to in Sambruck et al (1989). Other suitable expression vectors, promoters, enhancers and other expression elements: are well known to those skilled in the art. Examples of suitable expression vectors in yeast include Yep Sec 1 (Balderi et at, 1987, E bo J.s 6V229-234); pMFa (Kurjan and Herskowitz., 1982, Cell, 50.-933-943); JRY8.8 (Schultz et l, 1987, Gene., 5 113-123) and pYES2 (Invitrogen Corporation, San Diego, CA). These vectors are freely available as are haeulovirus and mammalian expression systems. For example, a baculovirus system is commercially -available .(ParMingen, San Diego, CA) for expression in insect cells while the pMsg vector is commercially available (Pharmacia, Piscataway, NJ) for expression in mammalian cells.
For expression in E. caU suitable expression vectors include among others, pTrc (Amann et ah, 1.998, Gene,. <59;301-31S) pGex (Arorad Corporation, Melbourne, Australia); pMal (N.B. Biolabs, Beverley, MA); pRitS (Pharmacia, , Piscataway, NJ); pEt-1 Id (Novagen, Maddison, WI) Clameei et L 1990, J. Virol, 64:3963-3966} and Sem ( napp *i 1990, Bio Techniques., 5:280-281). The use of pTRC, and pEt-1 Id, for example, will lead to the expression of unfused protein. The use of pMal, pRitS, pSem and pGex will lead to the expression of allergen fused to maltose E binding protein (pMal), protein A (p it5), truncated -gaiactosidase (PSEM) or glutathione S-transferase (pGex). When a T cell epitope of Ara h 1 or a peptide comprisin it is expressed as a fusion protein, it is particularly advantageous to introduce an enzymatic cleavage site at the fusion junction between the carrier protein and the peptide concerned. The peptide of the invention may then be recovered from the fusion protein through enzymatic cleavage at the enzymatic site and biochemical purification usin conventional techniques for purification of proteins and peptides. The different vectors also have different promoter regions allowing constitutive or inducible expressio or temperature induction. It ma additionally be appropriate t express recombinant peptides in different E. eoli hosts that have an altered capacity to degrade recombinantly expressed proteins. Alternatively, it may be advantageous t alter the nucleic acid sequence to use codons preferentially utilised by E. coil, where such nucleic acid alteration would not effect the amino acid sequence of the expressed proteins.
Host cells ca be transformed to express the nucleic acids of the invention using
conventional techniques such as calcium phosphate or calcium chloride co-precipitation, DEAE- dextraii-mediated transfection or electroporation. Suitable methods for transforming the host cells may be found in Sanibruck et al. (1989), and other laboratory texts. The nucleic acid sequence of the invention may also be chemically synthesized using standard techniques.
In addition to recombinant production of peptides according to the invention, the nucleic acids may be utilised as probes for experimental or purification purposes.
Identif ication and synthesis of the peptides as di closed herein now facilitates the development of a range of prophylactic and therapeutic treatment protocols for use with respect t peanut related immune conditions. Also facilitated is the development of reagents for us therein. Accordingly, the present invention should be understood to extend to the use of the peptides or functional deri vatives, homologues or analogues thereof in the therapeutic and/or prophylactic treatment of patients. Such methods of treatment include, but are not limited to;
(i) Administration of the subject peptides to a patient as a means of desensitising or inducing immunological tolerance to peanut, Ara h 1 and/or Ara h 2 or Ara h 1-like; and/or Ara h 2- like molecules. This may be achieved, for example, by inducing Ara h 1 and/or Ara h 2 directed Th2 anergy or apoptosis. In a preferred embodiment, such an outcome is achieved by th use of peptides which maintain T cell epitope reactivity but which are unable to undergo IgE binding. Alternati ely, one may utilise treatment protocols which are based on the administration of specific concentrations of a given peptide in accordance with a specific regimen in order to induce tolerance. Such methodolog may eliminate Ara h 1 and/or Ara h 2 hypersensitivity or it may reduce the severity of Ara h 1 and/or Ara h 2 hypersensitivity or sensitivity to an allergen present in a composition comprising Ara h 1 and/or Ara h 2, such as a peanut allergy. Reference herein, to the treatment of Ara h .1 and/or Ara h 2 sensitivity should be understood to encompass within its scope the treatment of conditions characterised by sensitivity to compositions which comprise Ara h I and/or Ara h 2, such as peanuts generally , even if the sensitivity is directed to an allergen, other than Ara h 1 and/or Ara h 2.
Preferably such treatment regimens are capable of modifying the T cell response or both the B and T ceil response of the individual concerned. As used herein, modification of the allergic response of the individual suffering from peanut hypersensitivity can be defined as inducing either non-responsiveness or diminution in symptoms to the Ara h 1 molecule a determined b standard clinical procedures (Varney et a .1991 British Medical Journal 302:265-269). Diminution in the symptoms includes any reduction in an allergic response in an individual to Ara h 1 after a treatment regimen has been completed. This diminution may be subjective or .clinically determined, for example by using standard food challenge tests or standard skin tests known in the art.
Exposure of an individual to the peptides of the present, invention may tolerise or anergise appropriate T cell subpopuiations such that they become unresponsive to Ara h 1 and/or Ara h 2 and do not. participate in stimulating an immune response upon such exposure.
Preferabl the peptides according to the invention will retain immunodominant T cell epitopes but possess abrogated IgE binding. Still further, even if the allergen in issue is not Ara b 1 and/or Ara h 2, but is directed t a different allergen which is present in the same composition as Ara h 1 and/or Ara h 2 (such as a different peanut allergen') immunisation with Ara h 1 and/or Ara h 2 may nevertheless induce a bystander suppressiv effect which acts to reduce the degree of hypersensitivity to that allergen.
Administration of a peptide of the invention may modify the cytokine secretion profile as compared with exposure to naturally occurring Ara h I and/or Ara h 2 allergen. This exposure may also influence T cell subpopuiations which normally participate in the allergic response to .migrate away from the site or sites of normal exposure to the allergen and towards the site or sites of therapeutic administration. This redistribution of T cell subpopulations may ameliorate or reduce the ability of an individual's immune system to stimulate the usual immune response at the site of normal exposure to the allergen, resulting in diminution of the allergic symptoms.
Modification of the B cell response may be achieved, for example, via modulation of the cytokine profile produced by T cells, as detailed above. Specifically, decreasing T cell derived IL-4 and IL-13 production thereby decreasing IgE synthesis,
(it ) The peptides of the present invention may be used in the capacity of an adsorbent to
remove Ara h 1 and/or Ara b. 2 directed T ceils from a biological, sample or from a patient. Accordingly, in another aspect the present invention provides a method for the. treatment and/or prophylaxis of a condition in a subject, which condition is characterised b the aberrant, unwanted or otherwise in appropriate immune response to Ara h 1 and/or Ara h 2 or an allergen in a composition compri sing Ara h 1 and/or Ara h 2, said method comprising administering to said subject an effective amount of an immunomodulatory composition as hereinbefore defined for a time and under conditions sufficient to remove or reduce the presence or function in said subject o T cells directed to said Ara h 1 and or Ara h 2 or other allergen.
Preferably said condition is hypersensitivity to peanuts or tree nuts which contain Ara h. 1 and Ara b 2 or Ara h 1 -like or Ara h 2-lrke molecules, such as hazelnuts, almonds or Brazil nuts.
In one embodiment, said method desensitises or induces immunological tolerance to Ara h 1 and/or Ara h 2 or other allergen of said composition.
In anothe embodiment, said desensitization or tolerance is achieved by inducing T cell anergy or apoptosis.
In still another embodiment, said desensitisation or tolerance is achieved by inducing Ara h 1 or Ara h 2 -specific Treg cells.
An "effective amount" means an amount necessary at least partly to attain the desired immune response, or to delay the onset or inhibit progression or halt altogether, the onset or progression of a particular condition being treated. The amount varies depending upon the health and physical condition of the individual to be treated, the taxonomie group of individual to be treated, the degree of protec tion desired, the formulation of the composition, the assessment of the medical, situation, and. other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
The. subject of the treatment or prophylaxis is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, eat), laboratory test animal (e.g.. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Preferably the mammal is a human or primate. Most preferably the mammal is a human.
Reference herein to "treatment" and "prophylaxis" is to be considered in its broadest context. The term, "treatment" does not necessaril imply that a subject is treated until total recovery. Similarly, "prophylaxis" does not necessarily mean that Ac subject will not eventu lly contract a disease condition. Accordingly, treatment and prophylaxis include amelioration of the symptoms of a particular condition or preventing or otherwise reducing the risk of developin a particular condition. The term ''prophylaxis" may be considered as reducing the severity or onset of a particular condition. "Treatment"' may also reduce the severity of an. existing condition,
Administration of the composition of the present invention (herein referred to as "agent") in the form of a pharmaceutical composition, may be performed by any convenient means. The agent of the pharmaceutical composition is contemplated to exhibit, therapeutic activity when administered in an amount which depends on the particular case. The variation, depends, for example, on the human or animal and the agent chosen. A broad range of doses may be applicable. Considering a patient, for example, from about 0,01 g to about 1 mg of an agent maybe administered per dose. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily, weekly, monthly or other suitable time intervals or the dose may be proportionaily reduced as indicated by the exigencies of the situation. In another example, said composition is administered initially to induce tolerance and then, if necessary, booster administrations of the composition are administered to maintain tolerance. These boosters may be administered monthly, for example, and may be administered for any per iod of time, including the life of the patient.
The agent may be administered in a convenient manner such as by the oral, intravenous (where water soluble), intraperitoneal, intramuscular, subcutaneous, intradermal (with or without using a Ixaditional needle or other transdermal delivery device), transdermal, intranasal, sublingual or suppository routes or implanting (e.g. using slow release molecules). Preferably, said composition is administered intradermally. The agent may be administered in the form .of pharmaceutically acceptable nontoxic salts, such as acid addition salts or metal complexes, e.g. with zinc, iron o the like (which are considered as salts for purposes of this application).
Illustrative of such acid addition salts are hydrochloride.,, hydrobr mide, sulphate, phosphate, maleate, acetate, citrate, henzoate, succinate, roalate, ascorbate, tartrate and the like. If the active ingredient is to be administered in tablet form, the tablet may contain a binder such as tragacanth, corn starch or gelatin; a disintegratin agent, such as algimc acid; and a lubricant, such as magnesium, stearate. In accordance with these methods, the agent defined in accordance with the present, invention. may be coadministered with one or more other compounds or molecules. By
"coadministered" is meant simultaneous admi istration in the same formulation or in two different formulations via the same or different routes or sequential administration by the same or different routes. By "sequential" administration is meant a time difference of from seconds, minutes, hours or days between the administration of the tw types of molecules. These molecules may be;
administered in any order. It should also be understood- that the peptides of the present invention may be themselves administered simultaneously or sequentially. They may be administered as one or more compositions, either simultaneously or sequentially. For example, one may formulate some of the peptides in one formulation and the others in a separate formulation; with these two formulations being given one in each arm. Alternatively, additional separate formulations eouid be generated and administered, either simultaneously to different sites or sequentially. It i well within the skill of the person in the art to design and generate the production of an appropriate formulation or mix of formulations.
Another aspect of the present invention contemplates the use of an immunomodulatory composition as hereinbefore defined in the manufacture of a medicament for the treatment of a condition in a mammal, which condition is characterised by an aberrant, unwanted or otherwise inappropriate immune response to Ara h 1 and/or Ara h 2.
Preferably said condition is hypersensitivity to peanuts or a tree nut which contains Ara h 1 and/or Ara h 2 or Ara h 1-like and/or Ara h 2-Iike molecules, such as a hazelnut.
In yet another further aspect, the present inventio contempl tes a vaccine comprising the composition as hereinbefore defined together with one of more pharmaceutically acceptable carriers and or diluents. Said composition is referred- to as the active ingredient.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion or may be- in the form of a cream or other form suitable for topical application. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by th us of coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of superfactants. The preventions of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens. chlorobutanol, phenol, sorbic acid, thimerosal. and the like. Tonicity adjusting agents are useful to keep the preparation isofonie with human plasma and thus avoid tissue damage. Commonly used tonicity agents include Dextrose, Trehalose, Glycerin and annttol. Glycerol and sodium chloride are other options but are less commonly used. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption, of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilisation. Generally, dispersions are prepared by incorporating the various sterilised active ingredient into a sterile vehicle which contains the basic dispersion medium and .the required other ingredients from those enumerated above, hi the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from previously sterile -filtered solution thereof,
When the active ingredients are suitably protected they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it ma be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions arid preparations should contain at least 1 % by weight of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 5 to about 80% of the weight of the unit. The amount of active compound in such therapeutically u eful compositions in such that a suitable dosage will be obtained. Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 0.1 pg and 1000 pg of active compound.
The tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: a binder such as gum* acacia, corn starch or gelatin; excipients such as diealeium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin may be added or flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring. When the dosage uni t form is a capsule, it may eontain, in addition to materials of the above type, a liquid carrier. Various other materials ma be present as coatings or to otherwise modify the physical form of the dosage unit For instance, tablets, pills, or capsules may be coated wi th shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour. Of course, any material used in preparing any dosage unit form should he
pharmaceutically pure and substantially non-toxic in the amounts employed, in addition, the active compound(s) may be incorporated into sustained-release preparations and formulations.
The pharmaceutical composition may also comprise genetic molecules such as a vector capable of transf ecting target cells where the vector carries a nucleic acid molecule encodin a modul tory agent. The vector may, for example, be a viral vector.
Routes of administration include, but are not limited to, respiratoralfy (eg. intranasaily or orally via aerosol), intratra heall , nasopharyngeally, intravenously, intraperitoneaily, subcutaneously, intracranially, intradermally, transdermal ly, intramuscularly, intraoecul rly, intratheeally, intracereberally, intranasaily, infusion, orally, rectally, via. IV drip patch, implant and sublingual. Preferably, said route of administration is subcutaneously, intradermally, txansdermally or intranasaily.
Yet another aspect of the present invention relates to the compositions, as hereinbefore defined , when used in the method of the present invention.
The present invention, is further described by reference to the following non-limiting examples.
EXAMPLE 1
Ara h 1 and Ara h 2 are the most allergenic and abundant proteins in peanut, making peptides comprising their dominant T-cell epitopes essential for inclusion in a therapy. Another important consideration when selecting peptides for immunotherapy, is whether they can be presented by different MHC class II molecules (HLA molecules in humans) and therefore be suitable for treating a genetically diverse human population. The HLA-res friction of peptide presentation to T cells was tested using blocking antibodies and HLA-genotypmg and showed that every T cell epitope identified could be presented on two or more different HLA-moleeul.es,
Furthermore, it was demonstrated that the identified T cell epitopes were collectively presented on a combination of HLA.-DR, HLA-DQ. and HLA-DP molecules (Table 2), Inclusion of HLA-DQ and -DP-restrieted T cell epitopes is particularly advantageous for a therapeutic since these HLA- types are more conserved in mixed population than HLA-DR molecules, enabling broader population coverage with fewer T cell epitope sequences.
Adjacent or overlapping T cell epitopes were combined into single peptides (<20 aa long) to minimise the number of peptides in the final therapeutic set resulting in three candidate peptides from Ara h 2 and seven from Ara h 1. Since cysteine residues can be problematic for peptide stability and biological reactivity, cysteine residues were substituted with structurally conserved but less reactive serine residues. Minor changes were also made to two Ara h 1 peptides to improve stability and/or solubility (Table 2). In all eases it has been confirmed that T-cell reactivity to the variant peptide has been re tained.
Table 2: Therapeutic candidate peptides of Ara ft / and Am h 2
Figure imgf000037_0001
Ara h J peptides shaded; Are h 2 peptides unshaded, HLA column shows HLA types known to present T ce lt epitope(s) within the peptide. ^Peptides altered to improve properties; 1 'W omitted from Nterminus * Έ* (from native sequence) added to C-terminus. 'Bolded serin replaced a
cysteine.
Preclinical screening of these peptides confirms PBMC T-cell reacti vity (Fig 1), lack of inflammatory cell activation (Fig 2) and serum stability in an additional peanut-allergic cohort (n = 40), It has been confirmed that PBMC T-cell recognition of one or more of these ten peptides in 100% of subjects (n=20) analysed, with 50-90% responding to each peptide. The analyses to date clearly demonstrate that, the ten peptides in Table 2 above, provide a sufficient, feasible and suitable mixture.
EXAMPLE 2
Brief overview of steps
1) Identification of dominant T cell epitopes of major peanut allergens Ara h 1 and Ara h 2
(Figures 3 and 4)
Isolated CD4+ T cells specific for Ara h 1 or Ara h 2 from PBMC of peanut-allergic subjects
Determined T cell specificity to overlapping 20-mer peptides spanning full Ara b 1 (Figures 5 and 6) or 2 (Figure 7) sequence
Selected dominant 20-m.ers and mapped core T cell, epitope sequences within them (Figure 8)
2) Determining HLA-restriction of core T cell epitopes
Blocked T cell epitope presentation to specific T cells using anti-HLA antibodies (Figure 9)
HLA -genotype*! subjects used for T cell epitepe-mapping
Further assessed HLA-binding degeneracy of T cell epitopes with algorithms
3) Design of therapeutic candidate peptides Replaced cysteine residues with serine residues
Combined, overlapping T cell epitopes into single peptides 20 aa long (10 peptides) (Tables 16-18)
Designed shorter peptide variants based on single T cell epitopes (13 peptides)
Synthesized all 23 peptides to >95% GLP-grade purity & determined solutions lor solubility
4) Selection and testing of final therapeutic mixture
Compared PBMC T cell reactivity to all 23 peptides in peanut-allergic cohort and selected final 7- peptide therapeutic
Assessed PBMC T cell response to 7-peptide mix at 2 therapeutic doses in peanut-allergic cohort
Tested basophil response to 7-peptide mi at 4-log dose range i peanut-allergic cohort
Materials and Methods
Subjects: Peanut-allergic adult subjects were recruited from The Alfred Allergy Clinic, Melbourne, Australia. Peanut-allergic subjects had clinical symptoms of IgE -mediated peanut allergy and peanut-specific IgE CAP score .2 ( .1.16 kUA/1; Pharmacia CAP SystemTM, Pharmacia
Diagnostics, Uppsala, Sweden) and many had a history of anaphylaxis. Some subject were gefiotyped (HLA-DRB 1 , -DQBl and -DPB 1, exon 2) by the Victorian Transplantation and
Immu oge ics Service. The study was approved by The Alfred and Monash University Ethics Committees and. informed written consent obtained from each subject.
Antigens: Crude peanut extract (CPE) was prepared from commercial unsalted, dry-roasted
peanuts as described elsewhere, (de-Leon et al. Clin Exp Allergy. 2003;33(9):1273-80) dialyzed against phosphate-buffered saline (PBS) and filter-sterilized. (0.2 m). Natural Ara h 1 and Am h 2 were enriched fr m CPE based on published methodology , (de Jong EC- et al Clin Exp Allergy. 1998;28(6):743~51 ) Briefly, CPE was buffer exchanged into 20 niM TRIS-bis-propane (TBP), pH. 7.2, using Vivaspin columns (Sartorius Stedim Biotech S.A., Aubagne, France) and applied onto a 5 niL Mono-Q 10/10 column (Pharmacia FPLC System, St Albans* UK) equilibrated, with TBP. After washing with TBP, a linear gradient of 30 mL 0-1 M NaCl/TBP was applied to. elate bound proteins (1 mL min). Fractions, 0.5 m'L, were analyzed by SDS-PAGE and those containing Ara h .1 or Ara h 2 with minimal other proteins pooled and dialyzed against PBS. Endotoxin contents were 1.7, 4.0 and 78,0 EU/rag for CPE,. Ara b 1 and Ara h 2 respectively (Endpoint Chromogenic LAL assay, Lonza, Walkersville, USA). Peptides (Mimotopes, Victoria, Australia and GenScript USA Inc., New Jersey, USA) were reconstituted -at 1-4 mg ml in 1.0% dimethyl sulfoxide/PBS (20- niefs and truncated peptide sets) or PBS, .1 -2% acetic acid or 0, 1 M atninonium bicarbonate buffers as specified (custom-synthesized core epitope peptides). All antigens were confirmed to be neither mitogenic nor toxic as described (Eusebius NP etal. Int Arch Allergy Immunol, 2002; 127{3):234-
44),
QeneratifM of Ara h ./ and Am h 2-specific CD4+ T-ce'tt lines (TCL): Ara ft 1 or Ara h 2-speeiftc oligoclonal TCL were generated from peripheral blood mononuclear cells (PBMC) of peanut- allergic subjects using 5,6-carboxyfl.uorescein diacetate succtnimidyl ester (CFSE)-based
methodology. (Mannering SI et al.. / Immunol Methods. 2005;298(ί-2):83-92; Priekett SR., et ol, J Allergy Clin Immunol* 2011 ;127(3):608-15 el -5). Briefly, cuituring was performed in RPMI- .1640 containing 2 mM L-glutamine, 100 lU/rnL pen icillio -streptomycin and 5% human AB serum (Sigma-Aldrieh, St Louis, USA) (cRPMI), PBMC were labelled with 0. J μΜ CFSE (Molecular Probes, Eugene, USA) and cultured (2.5 x 106/mL) with cRPMI alone, CPE (100 μ§/ητΕ), Ara b 1 or Ara b. 2 (TO g/mLi. Ara h 1 or Ara h 2 20-mer-peptide pools (10 g/mL peptide) or as a control, tetanus toxoid (TT; 1.0 LftJ/mL; Statens Serum Institute, Copenhagen, Denmark) for 7 days at 3 C. After staining with CD4-PE and 7AAD (BD Pharmingen, San Diego, USA), CD4+CFSEdirn7AAD- cells were sorted (10 eells/weH) into 96-U-well plates containing irradiated allogeneic feeder-cells, anti-CD3 (OKT-3), rIL-2 (Cetus, Emeryville, USA) and Fungizone (invitrogen, Carlsbad, USA) as described. Cells were fed with rJL-2 as .required, and after 10-14 days, transferred to 48-well plates and tested for proliferation to Ara h 1 or Ara h 2 (.10 g/iriL). Ara h 1 or Ara h 2-positive TCL were expanded with anti-CD3 and rIL-2 in T25 culture flasks (BD, Franklin Lakes, USA) for 10-12 days then tested for specificity (proliferation) to overlapping 20-mer peptide spanning the respective sequence (1.0 g/ruL). Core epitope sequences were mapped within selected 20-mers using peptide sets truncated from the N- or C -terminus of the 20- mer as described (Priekett SR, et ale, J Allergy Clin Immunol 2011;127(3):608-15 el -5),
T-cel assays: Ml cuituring was performed in RPMI-1640 containing 2 mM L-glutamine, 00 lU/mL peniciliin-strepfom cin and 5% heat-inactivated human AB serum (Sigma- Aldrich, St Louis, USA) (cRPMI). Antigen-induced TCL proliferation was assessed by 3H-thymidine ("Ή- Tc!R) uptake assays as follows: assays were performed, on 72-hour duplicate or triplicate cultures in 96-U-we 1.1 plates containing 1 x 104 T cells/well, 1 x 1.04 irradiated (5000 rads) autologous E.B V- ti'ansformed PBMC (EB'V-B cells as antigen, presenting cells and antigens as specified. Negative control was cRPMI alone. Cells were pulsed with 3H-thymidme (¾-TdR; 0,5 Ci/well) for the last 16 hour and uptake recorded as mean counts per minute (cpm) of replicate cultures, A stimulation index (SI; cpm antigen-stimulated T cells/cpm unstimulated T cells) 2.5 was considered positive and all positive responses confirmed in 2 assa s. To allow detection of peptMe-indueed CD4+ T- cell proliferation within whole PBMC, 7-day cultures of CFSE-tabefled PBMC were set up as described, for TCL generation, with addition of anti-CD25 antibodies (BD). to assess T cell activatio in. addition to proliferation. At least 10,000 CD4+ T cells were analyzed per sample and SI calculated as percentage of CD4+CFSElo (proliferated), CD4+CD25+ (activated) or
CD4+CD254-CPSE.O (activated and proliferated) cells with antigen divided by the percentage of the same population, without antigen (background). Analysing CD4+CD25+CFSE1.0 (activated and proliferated) cells provided the most sensitive method for detection of T cell responses with designation of an SI. 1.5 as positive.
HLA class II blocking assays: T cells and irradiated EBV-B cells (1 x 1.04of each) were incubated with 0.1-10 g mL blocking monoclonal antibody (mAb) against HLA-DR (L243, BD
Pharmingen), HLA-DQ (SVP-L3) or HLA-DP (B7/21) or isotype-contTol antibodies (IgG2a: BD Pharmingen; IgGl : BioLegend, San Diego, USA) far 1 hour at 37"C prior to addition of peptides {2-10 g/mL) or CPE (100 g/mL) and testing proliferative response as above.
Cytokine EUSPOT assays: ΜΑ.ΪΡ. EOS POT plates (MiUi ote, Billerica, USA) were coated overnight at 4"'C with 10 g/mL IL.-4, IPN- or IL-5 antibodies; (eBioscience, San Diego, USA) in. PBS. Wells were blocked (cRPMI, 1 hour, 37°C) then PBMC (3.5 x 1.0s) or T cells and irradiated EBV-B cells (1 x 104 of each) added in duplicate 100 L cultures with CPE (100 g/mL), nAra h 2 {] g mL) or peptides (10 g/mL). Controls were cRPMI alone, TT (10 IfU/ml) and
phytohaemiigglutmin (1 g/mL; Sigma-Aldrich). After 48 hours culture at 37°C, plates were incubated with hiotinylated IL-4, IL-5 or JFN- antibodies (eBioscience) (.1 g/ml PBS, 2 hours) followed by ExtrAvidin®-alkaline phosphatase (Sigma- Aidrieh) (1/3,000 PBS, 2 hours) before developing with alkaline phosphatase substrate (Bio-Rad). When spots appeared in positive- control wells, plates were washed, air dried and read (AID ELISPOT 4.0 h reader, Autoimmun Diagnostika, Strassberg, Germany).
Basophil- activation lest: Basophil activation was assessed by CD63 upregulatioii detected by flow cytometry as described (Drew AC, el l., J Immunol. 2004;173(9):5872-9). Positive controls were rabbit anti-human IgE antibody (7.5 μg mL; DA O Corporation, CA, USA), N-formyl- methwnine-leueine-phenylalanine (iMLP) (0,4 pg/tnL; Sigma) and CPE, CPE was tested over a 3 log concentration range (50, 5 and 0.5 pg/mL) and the peptide pool was tested over a 4-1 og concentration range (50, 5, 0.5 and 0.05 }ig/niL). Histamine release was assessed using Histamine Release and the Histamine EL!SA kits (IBL International GmbH, Hamburg, Germany) as per manufacturer's instructions.
Results
The factors considered in dominant 20-mer selection included:
« Responder f equency
• Number of specific TC.L generated per patient/prevalence of specific T ceils in patient PBMC · Magnitude of T cell response * Patterns of T cell responses (peptide combinations recognised within and between subjects)
• Ability to directly target specific T cells amongst the whole PBMC population with peptide (CFSE screening)
• Consistency of T cell responses
♦ Identificatio of core T cell epitope(s) within the 20-mer peptide
Am k 1 dominant 20-mer selection
145 Ara h 1 -specific T cells lines (TCL) were generated from 18 peanut-allergic donor and 65/69 overlapping 20-mer peptides spanning Ara h 1 were recognised by these TCL (see Table 3 and Figure 5). 14 of these 65 20-mers were selected as most frequen tly recognised (4-6 responders of 18; 22-33%) (peptide numbers 23. 24. 26, 38, 40, 44-51 and 57), Of these 14 peptides, 9 were selected for further analysis (peptide numbers 23, 24, 40, 6, 47, 49, 50, 1 and 57) (Table 4).
These selections were, made based on number of specific TCL per subject, magnitude of TCL response, reproducibilit of TCL response and ability to target specific T cells in PBMC, The 9 20-mers which were selected were:
• collectively recognised by TCL from 16 of 18 subjects (89%) in this cohort
• typically induced strong and consistent responses in specific TCL
» each recognised by multiple TCL from, many responders
• each able to target specific T cells in donor PBMC (collectively inducing detectib!e PBMC T cell responses in 18/20 additional subjects with 8-16 responders (40-80%) per 20-mer,
One or more of the nine 20-mers was recognised by T cells in 35 (92%) of 38 subjects analysed by TCL isolation and/or CFSE-screening (Table 3).
Table 3
Pmliferative responses (thymidine uptake) of TCL to Arah 1 20-mer peptides. Table shows SI values (-fold increase in TCL proliferation with peptide above proliferation in unstimulated TCL). Only stimulation indices (SI) 2.5 are shown. For subjects with multiple TCL specific for a given 20-mer, the highest SI is shown. Dark grey, SI 2.5<5.0; SI 5.0. The dominant peptide ultimately selected, are shown in Table 4, Table 3
Figure imgf000042_0001
Table 4: --Summary of dominant Ara h J 20-raer selection
Figure imgf000043_0001
PBMC screening with dominant- Am h 1 20-mers
Table 5 shows: that 1 dominant 20-mer peptide is recognised by 18/20 (or 22/24 of all data). Each 20-mer peptide is recognised by at least 7 subjects. CFSE and TCL data combined; recognition confirmed in 43/45 subjects.
Table 5
Sis of peptk!e-induced proliferation for 24 subjects. Upper panel shows new peanut-allergic donor cohort (distinct to cohort used for TCL). Lower panel shows 4 subjects from cohort used for TCL generation wi th combined totals -from upper and lower panels.
CPE, crude peanut extract; +ye, positive; nt, not tested (peptide stocks not available at time of testing); Grey, stimulation indices 1 ,1< 2.5; Black, stimulation indices 2.5. Stimulation Indices > Si ¾ -t-ve 20-mers
Sub No Ara h 1 20-mers SI>1.1 SI>1.5 ject Antfften 23 24 40 46 47 49 SO 51 57 No. No.
Figure imgf000044_0001
Respcniders # 20 20 1 /20 10/20 8/20 7/14 10/14 1 1/14 8/20 1 1/20 11/20
w SI>1.1 % 100 55 50 40 50 71 79 40 55 55
Responders # 17/20 10/20 7/20 4/20 6/14 7/14 9/14 7/20 8/20 10/20
w SI>1,5 % 85 50 35 20 43 50 64 35 40 50
Figure imgf000044_0002
Responders # 24/24 15/24 14/24 12/24 9/1 11/16 13/16 1 /24 14/24 15/24
w SI> % 100 63 58 50 56 69 81 46 58 63
Responders # 21/24 1 /24 1 /24 8/24 7/16 8/16 11/16 10/24 11/24 13/24
w SI>1.S % 88 58 46 33 44 50 69 42 46 54
Ara h 2 dominant 20-mer selection
.69 Ara h 2-specific T cells lines (TCL) were generated from 16 peanut-allergic donors and 16/17 overlapping 20-mer peptides spanning Ara h 2 were recognised by these TCL (Table 6), 4 of these .16 20-mer were selected .as most frequently recognised (each with 7-9 responders of 16; 44-46%) (peptide numbers 4, 5, 11, 15) (Figure 7). These selections were made based on number of specific TCL per subject, magnitude of TCL response, reproducibility of TCL response and ability to target specific T cells in PBMC. The 420-mer peptides which were selected were,:
• collectively recognised, by TCL from all .16 subjects (100%) in this cohort
• typically induced strong and consistent responses in specific TCL
• each recogni ed by multiple TCL from many responders
• collectively recognised by --80% of all 69 TCL • each able to target specific T cells in donor PBMC (detectible PBMC T cell responses demonstrated in 6 subjects tested).
One or more of the four 20-mers. was recognised by T cells in 16/16 (100%) of subjects analysed by TCL isolation and/or CFSE- creening (Table 6).
Table 6
Proliferative responses (thymidine uptake) of TCL to Ara h 2 20-mer peptides. Table shows SI values (=fok! increase in. TCL proliferation with peptide above proliferation in unstimulated TCL). Only positive stimulation indices ( 2.5) are shown: Grey, 2.5 5.0; Black, >5.0. A) allergen- driven TCL; B) peptide-driven TCL, The dominant 20-mers are indicated in Table 7.
Table 6
Figure imgf000046_0001
Ta le 7: Summary of dominant Ara h 2 20-mer selection
Figure imgf000047_0001
Core T-cell Epitope Mapping
Technical approach
TCL from different subjects were used to ma each core T cell epitope. Exact T cell epitopes varied between TCL and subjects. The minimum T-cell stimulatory sequence (core epitope) within each selected 20-mer was determined by testing proliferation of reactive TCL from different subjects to truncated peptide sets (Figure 8) . The number of residues required to induce maximal T-cell proliferation vailed from 6-19 aa between different TCL and/or subjects (Tables 8 and 9). Due to variation in the number of flankuig-residues required for optimal epitope recognition, TCL were considered to recognize the same epitope if peptides containing a common core sequence induced recognition. Based on this criterion, ten distinct Ara h 1 and 5 distinct Ara h 2 CD4* T-ceil epitopes were identified
(.'consolidated epitopes' . Tables 8 and 9), with common 'minimal core epitope' sequences varying from 5-12 aa (underlined sequences, Tables 8 and 9). ''Consolidated epitope' sequences were the minimum sequences encompassing all residues required for optimal T ceil reactivity across different subjects to ensure broadest possible recognition.
(i) Core T cell epitopes found in domi.n ant Ara b J 20-me rs
Core T-eell epitope sequences were mapped within the dominant Ara h 1 2Q-mer peptides, 10 Arab 1 T cell epitopes were identified ('consolidated T ceil epitope'), including 4 pairs of overlapping T cell epitopes, (Table 8) Table 8
Figure imgf000048_0001
40 (352-37). ) (353-371) WSTT?SSE .NEGVIVKVSKE(SHQ
(SKQ « NO-:!43p59-37i) ENNEGVIVKVSKE 1 WSTRSSE KEGV1VKVSKE*
<SBQ IB NO:67¾361-370; Nt'i iVIVKVHK
Figure imgf000048_0002
57 (505-524) (507-524) .GDVFiMPAAHPVATN'ASS (SEQ ΪΒΝΟ-.88)
(SEQro O :S¾ (509-524; VFIM AHPVAINASS ,,
tSJEQ n> NO.-90) (5.10-521) FI PAAHFVAJ io .GDVHMPAAHPVAINASS* 12 iSEQiDNO:9l)(511-517) IMPAAHP 33
<SBQ ID O:*»2)(5 J 1-521 J tVlPAAttPVAlN
(ii) Core T cell epitopes found in dominant Ara h 2 20-mers
Core T-eel.l epitope sequences were mapped within dominant Ara h 220~mer peptides. 5 Ara h 2 T cell epitopes were identified ('consolidated T cell epitopes'), including 2 pairs of overlappin T cell epi topes (Table 9)
Table 9
Figure imgf000049_0001
HLA -restriction of Ara h 1 and Ara h 2 T cell epitopes
T cell recognition of dominant T cell epitopes was blocked with monoclonal antibodies agamst HLA-DP, HLA-DQ or HLA-DR (Figure 9). Some T cell epitopes presented on both HLA-DR and
HLA-DQ niolecules while the T ceil epitopes were collectively presented on HLA-DP, HLA-DQ and HLA-DR (Tabl 10).
Table 10
Figure imgf000049_0002
Figure imgf000050_0001
HLA-pestrietion of Ara h 1 and Ara h 2 T cell epitope presentation
HLA-typing was performed on subjects, with TCL recognising dominant T cell epitopes, in order to assess HLA-subtypes potentially able to T cell present epitopes. The absence of shared HLA. alleles for subjects recognising a T cell epitope with confirmed HLA-DR/DQ/DP restriction indicated T cell epitope HLA -binding degeneracy. The Ara h 1 results are shown in Table 11 and the Ara h 2 results in Table 12.
Table 11
Grey shading indicates T cell epitopes included in current 7~peptide mix
Figure imgf000050_0002
Figure imgf000051_0001
HLA-restrlction of Ar i h 2 epitope presentation
Table 12
Figure imgf000051_0002
Figure imgf000052_0001
The absence of a shared HLA-DQB allele between, all subjects from whom recognition of Ara h 2 T cell epitope (95-107) was blocked by anti-HLA-DQ indicated that this T cell epitope must he presented by multiple HLA-DQB 1 molecules. Similarly, the diversity in HLA-DRB alleles between subjects for whom recognition of Ara h 2 T ceil epitopes (127- 141) or (37-47) was blocked by anti-HLA-DR indicated binding-degeneracy of both T-ceil epitopes for 'multiple HLA- DRB 1 molecules.
In addition to presentation by at least 2 HLA-DR molecules., Ara h 2 T cell epitope (37-47) was also presented by HLA-DQB 1 *06:09 as both subjects who recognised this T cell epitope in the context of HLA-DQ had this allele, and for subject 9 it was the only DQB 1 allel present.
As DFBJ *04:0.1 or DRB /*./ ' 5:0-1 alleles were present to all subjects recoguising Ara h 2 T cell epitopes (32-44) (blocked by antt-HLA-DP) or (95- 107) (blocked by anti-HLA-DR) respectively, degeneracy o these T cell epitopes could not be determined. However, as
DPB1*0401 and. i¾RB1 *1501 are prevalent in populations worldwide, T cell epitopes presented by these HL -molecules would still be broadly recognized, There were no shared alleles between two or mote subjects recognising the dominant consolidated Ara h 1 T cell epi topes on a gi ven HLA-type, thus demonstrating that each of the identified Ara h 1 T cell epitopes was also presented by 2 or more different HLA-molecuIes.P ' redicting LA-binding motifs: Am h I 20-mer peptides
Table 13 provides a results summary for an HLA-DR prediction algorithm for binding motifs within dominant Ara h 1 20-mers.
able 13
Figure imgf000054_0001
Bsaa Predkting HLA-binding motifs: Ara h 2 20-mer peptides
Table 14 provides a results summary for 2 HLA-DR prediction algorithms for binding motifs within 3 dominant Am h 2 20-mers (NB dominant '20-mer 5' not shown as predicted and actual epitopes fall in overl p with '20-mer 4').
Table 14
20-mer name Sequence Residues
Ara h 2 pep 4 RRCQSQLERANLRPCEQHLM (SEQ ID NO: 122) 12:8-47
Ara h 2 pep 11 ELNEFENNQRCMCEALQQIM (SEQ ID NO: 123) 91-110
Ara n 2 pep 15 RELRNLPQQCGLRAPQRCD (SEQ ID NO: 124) 1127-146
Table IS
Shading indicates particularly f requent alleles in Caucasian populations.
Figure imgf000056_0001
Re mtng peptides for therapeutic delivery
Potentially pi'oblematic cysteine residues were replaced with structurally conserved, but less chemically reactive serine residues. Retained T ceil reactivity was confirmed (Figures 10 and 11 ). Serme-contaimng T cell epitope peptide showed comparable T cell responses to native cysteine- containing peptides.
Combining overlappmg Am h 1 T cell epitopes into single peptides 20 aa long
Table 16
2(Mtne.r Minimum sequence required for T- ceil recog«fri< »i Respond?-'
Residues/ Sub-
Figure imgf000057_0001
49 (433-452) (436-445) VE1KEGALML
(436-452)
(436-449 VEIKEGAWLPHFN VEIKEOAL LPHF SKA*
Figure imgf000057_0002
O verlapping epitopes combined ¾ ' liisi M
57 (505-524) (507-524) GDVHMPAAHPVAINASS
(509-524) VFTMPAAHPVAINASS
(507-524)
(510-521) PIMPAAHPVAfN GDVPt P.AAHPVALVASS*
IS aa
(511-517) IMPAAHP
( 11 -52 J ) TMPAAHPVA!N
Grey shading indicates overlapping consolidated T cell epitope pairs combined into single peptides for further analyses as outlined, in the text, * Asterisks and boxes indicate the seven Ara h 1 candidate peptides proposed- for a therapeutic. Co bining overlapping Ara h 2 T cell epitopes into single peptides 20 aa long
Table 17
Dominant 20-mers Core Epitopes Candidate Peptides
Residues Sequence 1 Re itees 1 Sequenc Residues Sequence
2S-47 RRCQSQIJiRA L PCEQHlJvl 32-44 1 SQLERA LRPCBQ SQLER A LKPSEQHLM
(' ¾f:0
57-17 A .LRPCK;QHLM
82-10.1 SQHQKRC NELNEFEH. QR 91-102 EL EE&N QRCM 91407 ELNEFllNNQRSMSEALC
91 -110 tLL Hl¾ 0.R MCEALO01M j 95-107 ! EFEN QRCMCEALQ C[i01+l03jS
127-146 KRBLRNLFQQrx APQRC 128- 141 R.ELR NLFQQCGLR A 128- 141 Cn /.S REERNLFQQSGLR A.
Am h j and Am h 2 candidate peptides summarised
There are 10 candidate peptides: 7 from Ara n 1 and 3 from. Ara h 2 (Table 18)
Table 18
Figure imgf000058_0001
Also designed are 13 additional shorte peptid variants (based, on single T cell epitopes) for comparison. Some sequences have been leisgthened or shortened (in line with native sequences and critical residues for T cell recognition) to improve peptide properties for production and solubility. This resulted in a panel of 23 candidate peptides for comparison. Peptide details are summarised in Tabl 1 .
All of the peptides in Table 18 were produced at 95-99.9% purity and solutions determined for solubility. T ceil responses were then compared to each of these peptides at 2 doses in PBMC from 25 peanut-allergic subjects in order to select a final therapeutic combination*
Figure imgf000059_0001
• B Buffers: all peptides tried in PBS first; then O. IM NH4HC03 if sequence suggested preference for high pfi, or 1 % acetic acid for low pH; if not soluble in 1% acetic acid, increased to 2, 5, 10% etc.
• N -terminal SW omitted from 'peptide 2 to improve stability and ease of synthesis
• C-terminal Έ' added to 'peptide 7 to improve solubility (otherwise peptide insoluble, except in toxic buffers)
Considerations for selecting final peptides
Aims:
Maximise population coverage and/or T cell reactivit whilst minimising sequence number and/or length.
Broad considerations for peptide selection:
• Comparison of T cell responses in 23-peptk!e screen
« Prior T cell reactivity data (for individual T cell epitopes/peptides)
• Sequence (ease of production/solubility)
• HLA -restriction, (most degenerate and HLA-DQ-restricted T cell epitopes)
Considerations for selection b ased on data from 23 -peptide screen:
• .Main assessment criteria based on SI values for CD25+CFSE-iow ceils
• Donor responder frequency per peptide at one or both concentrations
• Compare responses to long versus short variants
• Strength/consistency of response (i.e. those subjects who respond t both, concentrations vs those who respond to just one concentration)
• Patterns of responses
Table 20 shows the analysis of PBMC T cell responses to the full set of 23 candidate peptides in 34 peanut-allergic subjects. These data show SI values for % CD25-i-CFSElow CD4+ T cells with peptide/unstimul ted.
Data are grouped into a long and short version of each T ceil epitope-coiitainiii region (see column borders; e.g. Is1 'group' = peptides 1 , 23 and 24 [Pep 1 combines overlapping peps 23 and 24 which, each contain a separate T cell, epitope]). The summary at the bottom of each group comments on the optimal peptide selected from that group. (Data show SI values for % CD25+CFSE10W CD4+ T cells with peptide/unstimulated).
Data are sorted by descending value for 50 g sample of long version for each peptide 'group' (or 1.0 g sample where responses are better to this dose). Each row within in a. peptide group shows deita for a single subject, but the order of subjects varies in each peptide group.
Figure imgf000061_0001
-61 -
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Summary of responses to 23 -peptide panel in cohort of 34
The data indicate that the selected 7 peptides are the best combination, but boxes indicate groups containing other viable peptides as substitutions, (or additions) to the current pool:
For example:
1) Peptide 3 could replace peptide 15
2) Peptide § could replace peptide 21
3) Peptide 9 could replace peptide 23
Figure imgf000067_0001
Summar of responses to each peptide of 7 -peptide- mix in cohort of 39
• All recognised 1 or more peptides
« 13/39 (33%) recognise 100% of peptides
• 21/39 (.54%) recognise >85% (6 or more) peptides
• 31/39 (79%) recognise >70% (5 or more) peptides.
• Each peptide recognised by at least 25/39 subjects (64%)
Of the 74 subjects tested, all reacted to at least .1 peptide of the 7 selected peptides.
Responses to different peptide pools
Table 23
Figure imgf000068_0001
NB; Table 19 provides the sequences for each of peptides 1-23.
Pool .1 = 7 x original Ara h 1 'candidates'
Pool 2 = 3 original Ara h 2 'candidates'
Pool 3 = 10 x mix of above 2 pools ( Ara h I & 2)
Pool 4 = 3 x Ara, h 1 'candidates* .+ 5 x shorter variants (equivalent Ara h I sequence coverag to pool 1)
Pool 5 ~ 5 x shorter (single epitope) variants of Ara h 2 candidates (equivalent Ara h 2 sequence coverag to pool 2) Table 24: Refined peptid pool
Peptide Va
Original # HLA Sequence Residues aa Notes
name
Cores 1 DR FQNLQNHRlvqiEAKPNTLV Ara h 1 [206- 20 Contains 2 major Ara h 1 23+24 225] epitopes Present in Ara In 1
T cell patent
Core 40 2 DQ STRSSENNEGVIVKVSKE Ara h i [354- 18 *key sequence flagged in
371 ] Ara h 1 patent, hut one residue shorter to facilitate stability; contains one major Ara h 1 T celt epitope
Core 47 15 DR EVKFDKKNPQLQ Asa h i [416- 12 Shortened version of a
427] candidate peptide in Ara. h
1 patent; contains 1 Ara h .1 T eell epitope
Induced equivalent T cell responses to longer version containing an additional T celt epitope in screen of 25 new subjects
Core 49 4 DR/DQ VEIKEGALMLPHFNSKA Ara h 1 [436- 17 *key sequence flagged in
452] Ara h 1 patent
Core 57 19 DR/DQ VPIM'PAAHPVAINASS Ara h 1 [509- 16 Shortened version of ke (short) 524] sequence flagged in Ara h
1 patent;
Easier to produce and more soluble
Induced equivalent T cell responses to longer version containin an additional epitope in screen of 25 new subjects.
B 21 DR/DQ A LRPSEQHLM Ara h 2 [37-47] 11 Induced equi valent T cell
C42S responses to longer
version containing an additional T celt epitope in scree of 25 new subjects D 23 Q EFENNQRSMSEALQ Ara h 2 [94- 14 Induced equivalent T cell
107] responses to longer iC[10f+103]S version containing an additional T cell epitope in screen of 25 new subjects; One residue longer than core T cell epitope reported in Ara h 2 paper.
Pool 7a = 5 x Ara h 1 + 3 x Ara h 2 for 'refined' pool (same as final pool but with additional Ara h
2 T cell epitope)
Pool 7b = 5 x Ara h 1 + 3 Ara h 2 for 'refined' pool of 7
Basophil responses to 7 -peptide pool (Pool 7b}
Basophil reactivity data was collected from 14 peanut-allergic subjects following incubation with peanut (CPE) or the 7 -peptide pool, (pool 7b) over a 3-4 log concentration range ( g/m1)(Figure 2), In these subjects, basophil activation and histamine release was induced by whole peanut and positive controls, but not b the 7-peptide mixture.
Prior to selection of pool 7a and 7b. Pools 7a and7b were subsequently designed and tested,
PBMC T cell responses were compared to whole peanut and peptide pools 1-5 of Table 23. (Figures 12 and 13). In relation to pools 1-5, none of the pools were able to induce a positive T cell response in all subjects tested. Nearly all responses were considerably lower to the peptide pool than to whole peanut (at the conce trations tested) and. only one subject of each of pools 2. 3 and 4 showed a greater or equal response to the peptides as to whole peanut, to relation to pools 7a and 7b, 100% response was described to pools 7a and 7b (SI > 1.5). Pools 7a and 7b induced comparable or greater responses to whole peanut in many subjects 6/30 = 100 CPE response, 6/30 - 50-80% of CPE response.
When comparing PBMC T cell responses to the 7-peptide pool (Figure 14), there was n significant difference between pools 7a and 7b (comparing paired data; n = 15 per group; no advantage adding 3rd Ara h 2 peptide), There was still no significant difference when comparing full data set for pool 7b (n=30) with cohort for pool 7 using non-paired Mann. Whitney test for non-parametric data; p = 0.9).
In summary,, pools 7a and 7b were both significantly better than the other 5 pools tested. There was no significant difference in pool 7a over pool 7b. Pool 7b was recognised by 100% of subjects tested and induced comparable or greater PBMC T cell responses than peanut in over 33% of subjects. Pools .1 -5 were not recognised by 100% of subjects and very rarely induced responses equal to whole peanut. TABLE 25: More detailed summary of steps and data
Approach Results
1) Identification of dominant T cell epitopes of major peanut allergens Ara h 1 and Ara h 2
Isolated CD4+ T cells specific for Am h 1 or 145 Ara h 1 -specific T cell lines (TCL) (18 Ara h 2 from PBMC of peanut-allergic subjects)
subjects 69 Ara h 2 -specific TCL (16 subjects)
Total = 214 TCL from 20 subjects
Determined specificity to overlapping 20-mer 9 (of 69) dominant Ara h 1 20-mers peptides spanning whole Ara b .1 or 2 4 (of ί 7) dominant Ara h 2 20-mers sequence
& selected dominant 20-mers
Confirmed dominant 20-mers could target 1 Ara h 1 20-mer detected in 17/1
PBMC T cells in peanut-allergic subjects 1 Ara h 2 20-mer detected in 6/6
(CFSE screens)
Assessed total .frequency of rssponders 1. dominant Ara h 1 20-mer recognised by (combined data from TCL data and CFSE 43/45. 1 dominant Ara h 2 20-mer screens) recognised by 16/16
Mapped core T cell epitope sequences within .10 dominant Ara h .1 core T cell epitopes dominant 20-mers 5 dominant Ara h 2 core T cell epitopes
2) Determining HLA- restriction of core T ceil epitopes
Blocked T cell epitope presentation to specific Ara h 1 T ceil epitopes HLA-DR &/or -DQ TCL using anti-HLA antibodies restricted
HLA-genotyped subjects used for TCL Ara h 2 T cell epitope HLA-DR, -DQ &or - generation DP restricted
All T cell epitopes presented by 1 Hl .A- noleeule
Assessed HLA-binding degeneracy with Strong & degenerate binding motifs in. all T algorithms cell epi topes
3) Design of therapeutic candidate peptides
Replaced cysteine residues with serine All serine variants still T cell reactive residues Combined overlapping T cell epitopes into 7 candidate Ara h 1 peptides (Prickett et al peptides 20 aa long 2013)
Designed shorter variants with single T cell 3 candidat Ara h 2 peptides (Prickett et al epitopes 201 .1)
13 shorter variants of above candidate peptides
Synth esised all 23 peptides to GLP-grad AH. peptides obtained at 95-99.9% purity purity. Determined- suitable solutions for 22/23 peptides soluble in PBS, 0.1M solubility. NH4HC03, or 1 -2% acetic acid (1 insoluble peptide redesigned and new version now soluble; peptide 7 in Table 18)
4) Selection and testing of final therapeutic m ixture
Compared PBMC T cell reactivity to ail 23 34 subjects screened with all 23 peptides peptides (2 doses) i new peanut-allergic Optimal peptide combinations determined cohort
Peptides selected for final therapeutic 7 peptides selected (5xAra h 1 and 2xAr'a h 2)
(considered T cell responses, peptid Presented on. HLA-D (5/7) and/or HLA-DQ properties, responder frequency, HLA~ (5/7)
restriction, patents) 1.-7 of peptides recognised by 56/56 donors
Assessed PBMC T cell response t 7 -peptide T cell response seen in 24/25 subjects (often mix at 2 therapeutic doses in peanut-allergic comparable or greater than response to peanut) cohort
EXAMPLE 3
Ara h 2 peptide-induced. T cell an rgy in an Ar h 2 epitope specific TCC ( igure 15)
T cells (1 x 1 O'Yml) of an Ara h 2 peptide specific human T cell clone were cultured tor 16 hours in the presence of Ara h 2 peptide ( ANLRPSEQHLM (SEQ ID NO:31; hatched) at 100 pg/ral in the absence of accessory ceils or in complete medium* alone (No Ag). The T cells were then washed thoroughly and rechallenged (l04/well) with complete medium alone, IL-2 SOU/rnl or an immunogenic concentration of the Ara h 2 peptide (10 pg/ml) in the presence of irradiated autologous PBMC ( IQVwell) as accessory cells. Proliferation as correlated with tritiated thymidine incorporation was determined at 72 hours. Results are expressed as mean cpm +SD for tripl icate cultures. ^Complete medium: RPMT + 5% AB serum + Pen/Strep L-g utamine + lOU/mL of lL-2, Ara h I p ptide-induced energy in an Ara h 1 epiwpe-specific TCC (Figure 16)
T cells (1 x 10°/ml) of an Ara h 1 peptide specific human T cell clone were cultured for 16 hours in the presence of Ara h I peptide (ST.RSSENNEGVIVKVS E (SEQ ID' NO: 12; hatched) or an irrelevant Bahta grass Pas n 1 peptide (stippled) at 100 pg l in the absence of accessory cells or in complete medium" alone (No Ag). The T cells were then washed thoroughly and reehallenged (10 /well.) with .complete medium alone, IL-2 50U/ml or an immunogenic concentration of the Ara h I peptide (10
Figure imgf000073_0001
in the presence of irradiated autologous PBMC ( J (//well) as accessory cells. Proliferation as correlated with tritiated thymidine incorporation was determined at 72 hours. Results are expressed as mean cpm for triplicate cultures. ^Complete medium: RPMi + 5% AB serum + Pen/Strep/L-glutamine + 10U/mL o lL-2.
Those skilled in the art will appreciate that the invention described herein is .susceptible to vari tions and modifications other than those specifically described, it is to be understood that the invention includes all such variations and modifications. The i nvention also includes all of the Steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and ail combinations of any two or more of said steps or features.
BIBLIOGRAPHY
Akdis & Akdis. J Allergy Clin Immunol. 123:735-46. 2009
Akdis & Akdis, Nature Reviews: Drug Discovery. 8:645-60. 2009
Akdis & Akdis, J Allergy Clin Immunol. 127: 8-27, 201 1
Alexander et al Clin Exp Allergy 35: 52-8, 2004
Alexander et al. Allergy 60:1269-74, 2005
Alien & O'Hehk. Clin Exp Allergy. 41(9): 172 , 2011
Amann et l., 1998, Gem., <59:301-315
Anagnostou «< «£ Clin Exp Allergy, 4.1.(9): 1.273-81, 201 1
Apostolou E etal 2006, Anaphylaxis to Geioiiisine confirmed by an in vitro basophil activation test: a case series. Anaesthesia', 61(3):264
Asarnoj et al Allergy. 2010. 65(9): 1 189-95
Baldcri et al., 1987, Embo J., 6:229-234
Blanc et al. Clin Exp Allergy. 2009; 39(8):1277-85
Blumehen et al J Allergy Clin Immunol 126(1): 83-91 , 2010
Boek et at. J Allergy Clin Immunol 119(4): J 016-8, 2007
Burks et at, hit Arch Allergy Immunol 119:165-172, 1992
Burks et al, Allergy 53: 725-30, 1998
Burks AW. 2008. Peanut allergy. Lancet;371(9623): 1538
Chiang et al, Pediatr Allergy Immunol. 2009; 21 (2 Pt 2):e429-38
Clarke er a*., Clin Exp Allergy 28: 1251-7, 1998
Clark et al Allergy 64, .1218, 2009
de Jong et al, Clin Exp Allergy 28: 743-51 , 1998
de Leon MP et al, Suphioglu, C. 2003, Immunological analysis of allergenic cross-reactivity between peanut and tree nuts. Clin Exp Allergy; 33(9): 1273
Drew AC et al 2004. Hypoa!ler genie variants of the major latex allergen Hev h 6,01 retaining human T lymphocyte reactivity. J Immunol: 173(9):5872-9
Eusebius P, Papalia L, Suphioglu C. McLellan SC. Varney M, Rolland JM, et al. Oligocioiiai analysis of the atopic T cell response to the group 1 allergen of Cynodon dactylon (benmida grass) pollen: pre- and post-allergen-specific immunotherapy, hit Arch Allergy Immunol,
2002; 127 (3):234-44
Fellrath et al J Allergy Clin Immunol 111 : 854-61, 2003
Giaumann et al Allergy. 2012; 67(2): 242-7
Hall et al. Vaccine. 21(S-6):.549-61, 2003 Hofmann et al. J. Allergy Clin. Immunol 124, 286, 2009
Hourihane et at., J Allergy Clin Immunol 100: 596-600, 1997
Emm et al J Exp Med. J 78(5): 1783-8, 1993
Husain Z, Schwartz RA, 2012. Peanut allergy: an increasingly common life-threatenmg disorder, J Am Acad Dermatol ;66(1): 36
Jameel et a!., 1990, . Virol, 64:3963-3966
Jones et al. J. Allergy Clin. Immunol 24, 292, 2009
Kemp et al. Med. J. Attst. 188(9):503-4, 2008
Kleber-Janke et al, int Arch Allergy Immunol 119: 265-274, 1999
Krwpp et l., 1990, . Bio Techniques,, &280-281
Koppelman etal Allergy. 2001; 56(2): 132-7
Koppelman et al. Clin Exp Allergy. 2004; 34(4 ):583-90
Kurjan and Herskowitz., 1982, ell, _?0:933-943
Larch M. 2008. Of cats and men: immynodominance and the role of HLA-DP/DQ. Clin Exp Allergy; 38(11):1709
Lin. et l J Microbiol Immunol Infect. 2012
Litwin et al.. hit Arch Allergy Ap.pl Immunol 87: 361-61, 998
Mannering SI, Dromey J A, Mon is JS, Theade DJ, Jensen KP, Han ison LC. An efficient method, for cloning huma autoantigen-speci tk T cells. J Immunol Methods. 2O05;298( 1-2): 83-92
Marazuela et al. Mol Immunol 45(2):438-45, 2008
Marcotte et al, J Allergy Clin Immunol 101 : 506-13, 1998
Mittag D, et al 201.0. The effector T cell response to ryegrass pollen is counter-regulated by simultaneous induction of regulatory T cells. Immunol; 184(9);4708
Moldaver & Larche, Allergy 66: 784-9 ! , 201 1
M verare et al Int Arch Allergy Immunol 2011 ; 156(3):282-90
MuUer et al J Allergy Clin Immunol 101: 747-54, 1998
Nelson et al, J Allergy Clin Immunol 99: 744-51, 1997
No p A, et al. 2006. Basophil allergen threshold sensitivity: a useful approach to anti-IgE treatment efficacy evaluation. Allergy; 61(3):298
Norman et al, Am J Respir Crit Care Med 1 4: 1623-8, 1996
O'Hehir RE, et al, 2009. House dust mite sublingual immunotherapy: the role of TGF-beta and functional regulatory T cells. Am J Mespir Crit Care Med; 180(10):936
Oldficld etal Lancet 360:47-53, 2002
.O penhejiner et l. .1 Allergy Clin Immunol 90: 256-62, 1992 Palmer et al Clin Immunol 2005; 115{3):302-12
Peeters et al Clm Exp Allergy. 2007; 37( 1 ): 108- J 5
Pene et al.. J Allergy Clin Immunol 102; 571-8, 1998
Pomes et at. 2006, Clm Exp. Allergy 36(6): 824-30
Pricfcett SR et al 201.1. Ara h 2 peptides containing dominant CD4+ T-cell epitopes: candidates for a peanut allergy therapeutic. J Allergy Clin Immunol; 127(3):60S
Primeau et al, Clin Exp Allergy 30: 1135-43, 2000
Pumphrey, Current Opinion in Allergy & Immunology. 4(4);2S5-90, 2004
RolIaiK r til. Pharmacology & Therapeutics 121 :273-284, 2009
Rupa et al. Allergy. 67(l):74-82, 2012
Sambnick i/ ii/ (19S9)
Santambrogio et al. P roc Natl Acad ' Set U SA, 1999, 96:15056-61
SchultZ f?i «/„ 1987, Gene., 54-113-123
Sicherer et al. Paediatrics 102: e§, 1998
Tarzi et al. Clin Exp Allergy. 36: 465-74, 2006
Thyagarajan et al. J Allergy Clm Immunol, i 26( 11: 1 -2. 2010
Varney et al. 1991. British Medical Journal 302:265-269
Varshney et al. J Allergy Clin Immunol. .124(6):.1351-2, 2.009
Varshney et al. J Allergy Clin Immunol 127(3):654-·60, 20.11
Worm el al. J Allergy Clin Immunol. 127: 89-97, 201 1
Worm et al. Expert Optn, Investig. Drugs. 22(10): 1347-1357, 2013
Yang .et al Clin. Exp Allergy 40(4):668-78, 2010
Yoshitoini et al J Pept Sci. 13(8) '499-503. 2007
Yu et al, hit Arch Allergy Immunol, 1 9(2): 179-1.82, 2012
Zauviders JJ, et al 2009. High level of human antigen-specific CD4+ T cells in peripheral blood revealed by stimulated coexpression of CD25 and CD.1 4 (OX40). / Immunol; .183(4):2827

Claims

CLAI S:
1. An immunomodulatory composition comprising at least five of the Ara h 1 and Ara h 2 T cell epitopic regions from the list consisting of:
(i) FQNLQNHR (SEQ ID O:l)
(it * IVQ1EA (SEQ ID NO:2)
i iii ) NECVIV VX (SEQ ID NO:3)
(iv) EVKPDKKNPQLQ (SEQ ID NO:4)
f ) EGALML (SEQ ID NQ:5)
(vi) lMPAAHP (SEQ ID NG:6)
(vii) LRPXEQHLM (SEQ ID NO:?)
(viii) ENNQRXMXEA (SEQ ID NQ:8)
or functional derivatives or homoiogues thereof wherein residue X is cysteine or serine and said composition comprises at least one epitopic region selected from SEQ ID NOS: l-6 and at least one epitopic region selected from SEQ ID NOS:7-8.
2. The composition according to claim 1 wherein epitopic region LRPXEQHLM is
LRPSEQHLM (SEQ ID NO: 137).
3. The composition according to claim 1 or 2 wherei epitopic region ENNQRXMXEA is ENNQRSMSEA (SEQ ID NO: 138),
4. The composition according to any one of claims 1 to 4 wherein said composition comprises at least 6 of said epitopic regions,
5. The composition according to any one of claims 1 to 4 wherein said composition comprises at least 7 epitopic regions.
6. The composition according to any one of claims 1 to 4 wherein said composition comprises each of said 8 epitopic regions.
7. The composition according to any one of claims 1 to 6 wherein said composition comprises one or more peptides, each of which peptides is up to 60 contiguous amino acid in length and which peptides include one or more of the Ara h 1 and Ara h 2 T cell epitopic regions defined by SEQ ID NOS: l-8.
8. The composition according to any one of claims 1 to 7 wherein said composition comprises Ara h. 1 and Ara h 2 T cell peptides selected from the list consisting of:
(i) FQNLQNHRIVQIE AKPNTL (SEQ ID NO: 1 1)
(ii) STRS ENN EG VI V KVSKE (SEQ ID NO: 12)
(iii) EVKPDKKNPQLQ .(SEQ I'D NO:4)
( iv) VEIKEGALMLPHFNSKA (SEQ ID NO: i 3) (v) VF1MPAAHPVA1NA8S (SEQ ID NO: 14)
(vi) ANLRPXEQHLM (SEQ ID NO:l 5)
(yii) EFENNQRXMXEALQ (SEQ ID NO: 16)
(viii) NNFGKLFEVKPDKKNPQLQ (SEQ ID NO: 17)
(ix) GD VFIMPAAHPV AIN AS SE (SEQ ID NO: 18)
(x) SQLERANLRPXEQHLM (SEQ ID NO: 19)
(xi) ELNEFENNQRXMXE ALQ (SEQ ID NO:20)
(xii) FQNLQNHKIY (SEQ ID NO:21 )
xiii) RIVQIEAKPNTLV (SEQ ID NO:22)
(xi V) ENNEGVIY VS E (SEQ ID NO: 23)
( XV I EVKPDKKNPQLQD (SEQ ID NO:24)
(xvi i EFENNQRXMXEALQQl (SEQ ID NO:25)
(xvii) NNFG LFEV PD KNPQLQD (SEQ ID NO:26)
(xvjii) ELNEFENNQRXMXEALQQI (SEQ ID NO:27)
(xx) WSTRSSENNEGVIVKVSKE (SEQ ID NO:28)
ixxi) GDVFIMPAAHPVA1NASS (SEQ ID NO:29)
or functional derivatives or homologues thereof wherein residue X is cysteine or serine,
9. The composition according to claim 8 wherein said residue X is serine,
10. The composition according to claim 8 wherein, said composition compri es Ara h I and Ara h 2 T cell peptides selected from the list consisting of:
(i) FQNLQNHRIVQIEA PNTLV (SEQ ID NO: 11 )
(ii) STRSSENNEGVIVKVSKE (SEQ ID NO:12)
(iii) EVKPDKKNPQLQ (SEQ ID NO:4)
(iv) VEIKEGALMLPHFNS A (SEQ ID NO: 13)
(V) VPI P A AHP V A IN ASS (SEQ D NO: .1 )
(vi) ANLRPSEQHLM (SEQ ID NO:31)
(vii) EFENNQRSMSEALQ (SEQ ID NO: 32)
(viii) EVKPDKKNPQLQD (SEQ ID NO: 24)
(ix) EFENNQRSMSEALQQI ( SEQ ID N :33)
or functional derivative or homologue thereof.
11. The composition according to claim 10 wherein said compositio comprises each of the Ara h .1 and Ara h 2 T cell, peptides selected from, the list consistin of:
((i) FQNLQNHRIVQIE KPNTLV (SEQ ID NO: 11 ) ;
(ii) STRSSENNEGVIVKVSKE (SEQ ID NO: 12);
(iii) EVKPDKKNPQLQ (SEQ ID NO:4) and/or EVKPDKKNPQLQD (SEQ ID NO:24); (i v) V EIKEGALMLPHFNSK A (SEQ ID NO: 13);
(v) VFIMPAAHPV AIMASS (SEQ ID NO: 14);
(yi) ANLRPSEQHLM (SEQ ID NO:31); and
(vii) E FE Q SMS E ALQ (SEQ ID NO:32) and/or EFENNQRS MSEALQQ! (SEQ ID
NO:33)
or functional derivative or homologue thereof.
12. The composition according to any one of claims 1 to 11 wherein said peptides are capable of reducing Ara h 1 and/or Ara h 2 hypersensitivity or hypersensitivit to a composition comprisin Ara h 1 and/or Ara h i when administered to a subject having a condition characterised by said hypersensitivity,
13. The composition according to any one of claims 1 to 12 said composition additionally including one or more peptides selected from the list consisting of:
( ! ) ALMLPHFNSKAMVIVVV (SEQ ID NO:34)
(it) NNFG LFEVKPDKKNPQ (SEQ ID NQ:35)
(iii) SQLERANLRPXEQ (SEQ ID NO:36)
(iv) ELNEFENNQRXM (SEQ ID NO:3?)
(vj NNFGKLFEVKPDKKMPQLQD (SEQ ID NO:38)
(vi) NNFG LFEVKPD NPQL (SEQ ID NQ:4Q)
(vii) SQLER ANLRPXEQH (SEQ ID NO:41 )
(viii) AMVIVVVNKGTGNLELVAV (SEQ ID NO:42)
(ix) RELRNLPQQXGLRA (SEQ ID NO:43)
(K) KAMVIVVVNKG (SEQ ID NO:44)
(xi) AMVIVVVN GTGNLELV (SEQ ID NO:45)
(xii) VVNKGTGNLELVAV K (SEQ ID NG:46)
or functional derivatives or homologues thereof wherein, residue X is cysteine or. serine.
14. A composition comprising one or more nucleic acid molecules encoding or complementary to a sequence encoding the epitopes and peptides according to any one. of claims 1 to 10 or 13.
.15. A method for the treatment and/or prophylaxi of a condition in a subject, which condition is characterised by the aberrant, unwanted or otlierwise inappropriate immune response to Ara h 1 and/or Ara h 2 or an allergen in a composition comprising Ara h 1 and/or Ara h 2, said method comprising administering to said subject an effective amount of an immunomodulatory
composition according to any one of claims .1 to 1.4 .tor a time and under conditions Sufficient to remove or reduce the presence or function in said subject of T celts directed to said Ara h 1 and/or. Ara h 2 o other allergen.
16. Use of an immunomodulatory composition according to any one of claims 1 to 14 in the manufacture of a medicament for the treatment of a condition in a mammal, which condition is characterised by an aberrant, unwanted or otherwise inappropriate immune response to Ara h 1. and/or Ara h 2.
.17. The method or use according to any one of claims .15 or 16 wherein condition is hypersensitivit to peanuts or tree nuts which contain Ara h 1 and Ara h 2 or Ara h Hike or Ara h 2-like molecules.
18. The method or use according to claim 1.7 wherein said nuts are hazelnuts, almond or brazil nuts.
19. The method or use according to an any one of claims 15 to 18 wherein said method desensitises or induces immunological tolerance to Ara h 1 and/or Ara h 2 or other allergen of said composition.
20. The method or use according to claim 19 wherein said desensitizatioii or tolerance is achieved by inducing T cell anergy or apoptosis.
21. The method or use according to claim 19 wherein said desensirisation or tolerance is achieved by inducing Ara h 1 or Ara h 2-specific Treg cells.
22. The method or use according to any one of claims 15 to 21 wherein said composition i administered intradermally or transdermally,
23. The method or use according to any one of claims 15 to 22 wherein said subject is a human.
PCT/AU2014/050249 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof WO2015042664A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
JP2016517506A JP7007534B2 (en) 2013-09-25 2014-09-25 New immunotherapeutic composition and its use
CA2925480A CA2925480A1 (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof
BR112016006813A BR112016006813A2 (en) 2013-09-25 2014-09-25 NEW IMMUNOTHERAPEUTIC COMPOSITION AND ITS USES
RU2016115732A RU2689552C2 (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and use thereof
MX2016003363A MX2016003363A (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof.
EP14848639.2A EP3049098A4 (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof
CN201480052398.1A CN105992589A (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof
AU2014328483A AU2014328483B2 (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof
US15/024,666 US11266737B2 (en) 2013-09-25 2014-09-25 Immunotherapeutic composition and uses thereof
KR1020167010829A KR20160075532A (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof
ZA2016/02043A ZA201602043B (en) 2013-09-25 2016-03-29 Novel immunotherapeutic composition and uses thereof
AU2020200883A AU2020200883B2 (en) 2013-09-25 2020-02-07 Novel immunotherapeutic composition and uses thereof
US17/672,571 US20220280637A1 (en) 2013-09-25 2022-02-15 Novel immunotherapeutic composition and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2013903686A AU2013903686A0 (en) 2013-09-25 Novel immunotherapeutic composition and uses thereof
AU2013903686 2013-09-25

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/024,666 A-371-Of-International US11266737B2 (en) 2013-09-25 2014-09-25 Immunotherapeutic composition and uses thereof
US17/672,571 Division US20220280637A1 (en) 2013-09-25 2022-02-15 Novel immunotherapeutic composition and uses thereof

Publications (1)

Publication Number Publication Date
WO2015042664A1 true WO2015042664A1 (en) 2015-04-02

Family

ID=52741648

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2014/050249 WO2015042664A1 (en) 2013-09-25 2014-09-25 Novel immunotherapeutic composition and uses thereof

Country Status (12)

Country Link
US (2) US11266737B2 (en)
EP (1) EP3049098A4 (en)
JP (1) JP7007534B2 (en)
KR (1) KR20160075532A (en)
CN (1) CN105992589A (en)
AU (2) AU2014328483B2 (en)
BR (1) BR112016006813A2 (en)
CA (1) CA2925480A1 (en)
MX (1) MX2016003363A (en)
RU (1) RU2689552C2 (en)
WO (1) WO2015042664A1 (en)
ZA (1) ZA201602043B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11980658B2 (en) 2012-10-30 2024-05-14 Aravax Pty Ltd Immunotherapeutic molecules and uses thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2914286T3 (en) 2012-10-30 2022-01-31 Aravax Pty Ltd Novel immunotherapeutic molecules and uses thereof
WO2020186190A2 (en) * 2019-03-13 2020-09-17 Psomagen, Inc. Epitope-based approach for allergy treatments and inhibitors for crohn's disease

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005121166A1 (en) * 2004-06-10 2005-12-22 Monash University Novel immunointeractive molecules and uses thereof
US20100291145A1 (en) * 1999-09-14 2010-11-18 Antigen Express, Inc. Ii-KEY/ANTIGENIC EPITOPE HYBRID PEPTIDE VACCINES

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5558869A (en) 1992-12-30 1996-09-24 University Of Arkansas Major peanut allergen ara h II
AU729836B2 (en) 1995-12-29 2001-02-08 University Of Arkansas, The Peanut allergens and methods
US20030202980A1 (en) 1995-12-29 2003-10-30 Caplan Michael J. Methods and reagents for decreasing clinical reaction to allergy
US6835824B1 (en) * 1995-12-29 2004-12-28 University Of Arkansas Peanut allergens and methods
JP2002500198A (en) 1998-01-09 2002-01-08 サーカシア リミテッド Methods and compositions for desensitization
DE69921773D1 (en) 1998-01-16 2004-12-16 Univ Johns Hopkins ORAL ADMINISTRATION OF NUCLEIC ACID VACCINE THROUGH PARTICLE COMPLEXES
CA2319437C (en) 1998-01-31 2009-06-16 University Of Arkansas Methods and reagents for decreasing allergic reactions
AU3085299A (en) 1998-03-12 1999-09-27 Board Of Trustees Of The University Of Arkansas, The Tertiary structure of peanut allergen ara h 1
AU3717600A (en) 1999-03-02 2000-09-21 Board Of Trustees Of The University Of Arkansas, The Methods and reagents for decreasing clinical reaction to allergy
AU3865400A (en) 1999-03-03 2000-09-21 Mount Sinai School Of Medicine Of The City University Of New York, The Animal model of allergies
WO2000054803A2 (en) 1999-03-16 2000-09-21 Panacea Pharmaceuticals, Llc Immunostimulatory nucleic acids and antigens
US20030235594A1 (en) 1999-09-14 2003-12-25 Antigen Express, Inc. Ii-Key/antigenic epitope hybrid peptide vaccines
AU2065801A (en) 1999-12-06 2001-06-12 Panacea Pharmaceuticals, Llc Passive desensitization
US20020147140A1 (en) 2000-01-31 2002-10-10 Rosen Craig A. Nucleic acids, proteins, and antibodies
CZ20031653A3 (en) 2000-11-16 2004-03-17 Alk-Abelló A/S Mutant allergens
RU2285042C2 (en) 2000-11-16 2006-10-10 Альк-Абелло А/С New mutant allergens
AU2002306850A1 (en) 2001-03-16 2002-10-03 Panacea Pharmaceuticals Methods and reagents for decreasing clinical reaction to allergy
GB0110432D0 (en) 2001-04-27 2001-06-20 Plant Bioscience Ltd Lantibiotic production
MXPA03010037A (en) 2001-05-01 2004-06-30 Univ California Fusion molecules and methods for treatment of immune diseases.
EP1453539B1 (en) 2001-12-05 2008-11-19 Circassia Limited Immunotherapeutic methods and systems
AU2003294220A1 (en) * 2002-09-17 2004-04-23 Antigen Express, Inc. Ii-KEY/ANTIGENIC EPITOPE HYBRID PEPTIDE VACCINES
US7179645B2 (en) 2002-09-24 2007-02-20 Antigen Express, Inc. Ii-Key/antigenic epitope hybrid peptide vaccines
US7923209B2 (en) * 2003-03-14 2011-04-12 Anergis, S.A. Allergen peptide fragments and use thereof
US7566456B2 (en) 2005-06-23 2009-07-28 Haiming Chen Allergen vaccine proteins for the treatment and prevention of allergic diseases
FI20075063A0 (en) * 2007-02-01 2007-02-01 Vactech Oy Prevention of allergic sensitization
GB0710529D0 (en) 2007-06-01 2007-07-11 Circassia Ltd Vaccine
ATE482721T1 (en) 2007-08-15 2010-10-15 Circassia Ltd PEPTIDES FOR DESENSITIZATION TO ALLERGENS
GB2455108A (en) 2007-11-28 2009-06-03 Circassia Ltd T-Cell dependent method for detecting non-allergic or intrinsic disorders
EP2140880B1 (en) 2008-07-04 2012-11-14 HAL Allergy Holding B.V. Modification of allergens
ATE536880T1 (en) 2008-08-15 2011-12-15 Circassia Ltd VACCINE CONTAINING AMB A 1 PEPTIDES FOR THE TREATMENT OF RAPE ALLERGY
US8821887B2 (en) 2008-08-15 2014-09-02 Circassia Limited T-cell antigen peptide from allergen for stimulation of IL-10 production
GB0821806D0 (en) 2008-11-28 2009-01-07 Circassia Ltd Compositions with reduced dimer formation
US8802375B2 (en) 2009-09-14 2014-08-12 Icahn School Of Medicine At Mount Sinai Methods for characterizing antibody binding affinity and epitope diversity in food allergy
US20130004528A1 (en) 2010-02-26 2013-01-03 Benaroya Research Institute Direct analysis of antigen-specific immune response
US9517257B2 (en) 2010-08-10 2016-12-13 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
PL2914286T3 (en) 2012-10-30 2022-01-31 Aravax Pty Ltd Novel immunotherapeutic molecules and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100291145A1 (en) * 1999-09-14 2010-11-18 Antigen Express, Inc. Ii-KEY/ANTIGENIC EPITOPE HYBRID PEPTIDE VACCINES
WO2005121166A1 (en) * 2004-06-10 2005-12-22 Monash University Novel immunointeractive molecules and uses thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
PRICKETT, S.R. ET AL.: "Ara h 1 CD 4+ T cell epitope-based peptides: candidates for a peanut allergy therapeutic", CLINICAL & EXPERIMENTAL ALLERGY, vol. 43, no. ISSUE, June 2013 (2013-06-01), pages 684 - 697, XP055338039 *
PRICKETT, S.R. ET AL.: "Ara h 2 peptides containing dominant CD 4+ T- cell epitopes: Candidates for a peanut allergy therapeutic", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 127, no. 3, March 2011 (2011-03-01), pages 608 - 615 .E5, XP028196570 *
See also references of EP3049098A4 *
SRIVASTAVA, K.D.: "Immunotherapy With Modified Peanut Allergens in a Murine Model of Peanut Allergy", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 109, no. ISSUE, January 2002 (2002-01-01), pages S287, XP005395989 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11980658B2 (en) 2012-10-30 2024-05-14 Aravax Pty Ltd Immunotherapeutic molecules and uses thereof

Also Published As

Publication number Publication date
RU2016115732A (en) 2017-10-30
AU2020200883B2 (en) 2022-05-12
JP2016533341A (en) 2016-10-27
CN105992589A (en) 2016-10-05
BR112016006813A2 (en) 2017-09-19
AU2014328483B2 (en) 2019-11-14
AU2014328483A1 (en) 2016-04-21
JP7007534B2 (en) 2022-01-24
EP3049098A1 (en) 2016-08-03
ZA201602043B (en) 2022-08-31
US20160375130A1 (en) 2016-12-29
EP3049098A4 (en) 2017-08-16
MX2016003363A (en) 2016-09-23
AU2020200883A1 (en) 2020-02-27
US20220280637A1 (en) 2022-09-08
US11266737B2 (en) 2022-03-08
RU2016115732A3 (en) 2018-06-19
RU2689552C2 (en) 2019-05-28
KR20160075532A (en) 2016-06-29
CA2925480A1 (en) 2015-04-02

Similar Documents

Publication Publication Date Title
CA2744787C (en) Compositions and methods for treatment of celiac disease
US20220280637A1 (en) Novel immunotherapeutic composition and uses thereof
AU2019240574B2 (en) Novel immunotherapeutic molecules and uses thereof
KR20120120965A (en) Peptides for vaccine against birch allergy
EA020792B1 (en) Grass peptides for vaccine
AU2013204429B2 (en) Compositions and methods for treatment of celiac disease
CA3182111A1 (en) Immunogenic peptides with extended oxidoreductase motifs
AU2012292060B2 (en) Hypoallergenic variants of Mal d 1, the major allergen from Malus domestica
US11980658B2 (en) Immunotherapeutic molecules and uses thereof
AU2003213861B2 (en) Immunotherapeutic and immunoprophylactic reagents
AU2005252268B2 (en) Novel immunointeractive molecules and uses thereof
AU780049B2 (en) Immunointeractive molecules and their uses in treating subjects suffering from HEV B 5 allergies
EP2855515A1 (en) Alternaria peptides

Legal Events

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

Ref document number: 14848639

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/003363

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2925480

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016517506

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15024666

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016006813

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2014328483

Country of ref document: AU

Date of ref document: 20140925

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2014848639

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014848639

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016115732

Country of ref document: RU

Kind code of ref document: A

Ref document number: 20167010829

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016006813

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160328