WO1996012740A1 - SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY - Google Patents

SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY Download PDF

Info

Publication number
WO1996012740A1
WO1996012740A1 PCT/US1995/013841 US9513841W WO9612740A1 WO 1996012740 A1 WO1996012740 A1 WO 1996012740A1 US 9513841 W US9513841 W US 9513841W WO 9612740 A1 WO9612740 A1 WO 9612740A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
seq
gly
leu
val
Prior art date
Application number
PCT/US1995/013841
Other languages
French (fr)
Inventor
Chang Yi Wang
Alan M. Walfield
Original Assignee
United Biomedical, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by United Biomedical, Inc. filed Critical United Biomedical, Inc.
Priority to EP95938912A priority Critical patent/EP0787150A1/en
Priority to AU40120/95A priority patent/AU4012095A/en
Publication of WO1996012740A1 publication Critical patent/WO1996012740A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6018Lipids, e.g. in lipopeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/64Medicinal preparations containing antigens or antibodies characterised by the architecture of the carrier-antigen complex, e.g. repetition of carrier-antigen units
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18411Morbillivirus, e.g. Measles virus, canine distemper
    • C12N2760/18422New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates to the use of a composition of a synthetic peptide immunogen comprising a target antigenic site and a helper T cell epitope covalently linked in a linear tandem form. More particularly, the present invention relates to the use of such a composition to elicit the production in healthy mammals, including humans, of high titer antibodies specific to sites on the e heavy chain of B cell-expressed membrane-bound human IgE, i.e., sites on the extracellular segment of the anchor domain of membrane-bound human e- chain and to the use of such a composition as a vaccine to provide an immunotherapy for the treatment of allergy.
  • Immunotherapy for the prevention of IgE-mediated allergic responses such as asthma and hay fever has involved desensitization or hyposensitization by administering a gradually increasing amount of an allergen to a patient to reduce the effects of subsequent exposure to that allergen 1) .
  • Limitations to such an allergen-based immunotherapy include difficulties in identifying the allergen involved and the adverse reactions frequently caused by the use of the identified allergen 0 '.
  • Other treatments for the relief of allergies employ therapeutic compounds to block the cascade of cellular events that is responsible for allergic reactions. These compounds include anti-histamines, decongestants, ⁇ 2 agonists, and corticosteroids.
  • Anti-histamines, decongestants, and ⁇ 2 agonists act on events downstream of IgE in the allergic cascade, making them palliative remedies which address allergic symptoms rather than preventative treatments which must act on events closer to the initiation of IgE- mediated allergic reactions. These palliative remedies provide relief that is short term and partial, frequently accompanied by adverse side effects. For example, anti- histamines may cause restlessness or sedation, and ⁇ 2 agonists are sometimes associated with increased morbidity in asthmatic patients.
  • corticosteroids are powerful immunosuppressants and are highly efficacious for the treatment of allergic symptoms. However, they produce adverse hormonal activities and may cause an undesirably broad immunosuppression.
  • a means of suppression selectively targeted to IgE This may be accomplished by suppressing IgE synthesis directly or indirectly. Indirect suppression can be accomplished by desensitization or by inhibition of IL-4 and other T cell-produced mediators of IgE synthesis 05 .
  • Direct suppression as suggested by Chang et al. (4) , can be accomplished by specifically targeting IgE-producing B cells with selective antibodies.
  • Chang et al. 4 and others (9) have studied human e-chains and corresponding antibodies, as well as the genes and mRNAs by which the e-chains are encoded. They have elucidated the molecular basis for the expression of two types of IgE: the secreted and membrane-bound forms by B cells committed to IgE synthesis.
  • the membrane-bound form of IgE may be distinguished from the secreted form by an additional membrane anchoring domain that extends from the C-terminus of the heavy chains and is contiguous with the CH4 constant domain of IgE.
  • the membrane-bound form is distinctive to the surface of B cells committed to IgE synthesis.
  • IgE-secreting cells By targeting such cells with antibodies specific for the exposed extracellular portion of that anchor domain, such cells may be eliminated or inactivated.
  • the mechanisms for elimination of IgE-secreting cells by such antibodies can be through antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytolysis (6,7) .
  • ADCC antibody-dependent cellular cytotoxicity
  • the reduction of circulating IgE and IgE-expressing cells by anti-IgE antibodies has been demonstrated in vivo in mice (10) , and by the inhibition in vivo of passive cutaneous anaphylaxis in a rat model. It has also been demonstrated in human IgE- secreting cell lines where anti-IgE was shown to lead to reductions in cell growth, decreased IgE accumulation and cytolysis in complement-mediated and ADCC-mediated cytolysis assays (11) .
  • mlgE membrane-bound IgE
  • Another objective is to design optimal peptide immunogens, with specific amino acid sequences taken from the human mlgE heavy chain membrane anchoring domain (SEQ ID NOS:l,2) and attached to peptides containing promiscuous human helper T cell epitopes in specified orientations which, when introduced into mammals, including humans, will stimulate production of efficacious antibodies to the sites on human mlgE anchor domain.
  • These antibodies may result in the reduction in IgE- producing B lymphocytes and thereby attenuate allergen- induced IgE production, constrain mast cell activation by IgE-allergen complexes, reduce the consequent release of chemical mediators such as histamines responsible for allergic symptoms and depress IgE- ediated passive cutaneous anaphylaxis (PCA) .
  • PCA passive cutaneous anaphylaxis
  • Another objective is to develop an effective mlgE e-chain peptide-based vaccine, employing compositions containing such linear peptide immunogens, so as to provide immunotherapy for the treatment of allergic reactions.
  • a series of linearly arranged synthetic peptides which contain either of two peptide sequences corresponding to sites on the exposed portion of the membrane anchoring domain of human mlgE (SEQ ID NOS:l,2) or their immunogenic analogs thereof together with a portion of a helper T-cell epitope (Th epitope) are made by solid phase synthesis.
  • Compositions of the invention are used to immunize healthy mammals, e.g. rats and humans for the production of high titer antisera that is specific for the mlgE anchor membrane sites (SEQ ID N0S:1,2) and free of irrelevant antibodies.
  • vaccines containing the synthetic peptide compositions as the key immunogen may also be prepared with an immunologically effective amount of linear synthetic peptide in the presence of a proper adjuvant and/or delivery vehicle. It is expected that such vaccine compositions will elicit a more focused anti-IgE peptide response than those of the peptide-carrier protein conjugates currently used by Chang et al. (4,6*8) , thus providing better immunotherapy for the treatment of allergy.
  • This invention is directed to the use of a novel group of peptide-based immunogens for the generation of high titer antibodies to mlgE anchor membrane sites (SEQ ID NOS:l,2) on human IgE e heavy chain in healthy mammals, including humans, ultimately for the treatment of IgE- mediated allergic diseases.
  • Table I shows the arrangement and the amino acid sequence of the Membrane Anchor Domain of the e heavy chain of human membrane-bound IgE (mlgE) , as deduced from the nucleic acid sequence of the predominant species of mRNA that codes for membrane-bound e chain* 7,8 '.
  • the regions on the e chain sequence of the peptides used as the target immunogens of the invention are underlined: a single underline for SEQ ID NO:l, and a double underline for SEQ ID NO:2.
  • the allergen/antibody binding crosslinks the bivalent surface-bound IgE and induces conformational changes in the distal Fc region of IgE, the region of IgE in direct contact with a high affinity Fc receptor on the mast/basophil cell surface as well as receptor site(s) on the cell surface.
  • the conformational changes activate the cell-IgE-allergen complex with the resultant release from the cell of chemical mediators, including histamine, inducing allergic symptoms and the further secretion of IgE.
  • the secretory IgE which mediates the allergic reaction is produced by terminally differentiated B cells in response to allergen.
  • B cells committed to IgE synthesis also display membrane-bound IgE (mlgE) on their surface.
  • mlgE membrane-bound IgE
  • the mlgE molecules are allergen receptors and are believed to play regulatory roles in the maturation of the B cells, and in activation of the B cells by allergen-specific T cells.
  • the mlgE is distinguishable from the secreted IgE by a membrane- anchoring segment which extends from the C-terminus of the heavy chains which serves to attach the mlgE to the cell membrane.
  • amino acid sequences for two immunogenic sites on the extracellular portion of the anchor domain were deduced (SEQ ID NOS:l,2). These sites are present on alternative forms of mlgE that result from different mRNA splicing events. Both sites are present on the predominant species of membrane-bound e-chain, in the orientation shown in Table I. The presence and specificity of these sites as well as their accessibility to antibodies were confirmed with specific antibodies' 4,8 '.
  • anti-IgE antibodies directed against such specific mlgE sites can lead to the reduction in the numbers of IgE-producing B cells and a concomitant reduction in circulating IgE, possibly through the lytic removal of the IgE-expressing cells 00 - 11 '.
  • it is desirable to target anti-IgE antibodies to the membrane-anchor domain because the domain is a surface marker specific to IgE- expressing cells. This target site is not available on secreted IgE.
  • anti-mlgE cannot bind and crosslink IgE bound to receptors on mast cells and basophils, and cannot by itself induce histamine release.
  • the removal of IgE-expressing cells in hosts suffering from allergic reactions may result in the down-regulation of IgE production and have a therapeutic outcome.
  • Such interventions employed in the treatment of allergy through the use of specific anti-IgE antibodies can be achieved either passively, through prophylactic treatment with specific "site-directed" antibodies to IgE, or, more preferably, actively, by providing the host with a vaccine comprised of site-directed peptide immunogens, to elicit the production by the host of its own site-directed anti-IgE antibodies. It is believed that active immunization will provide a more effective and longer lasting form of protection.
  • the sites on the extracellular segment of the membrane anchor domain (SEQ ID NOS:l,2), arranged on membrane-bound IgE as shown in Table I, have been confirmed as immunogenic sites that are acessible to antibodies through the cross-reactivities of the surface of IgE-secreting cells to antibodies that had been generated by immunizing animals with mlgE anchor membrane peptides coupled to a carrier protein, keyhole limpet hemocyanin (KLH) (4,8) .
  • KLH keyhole limpet hemocyanin
  • the peptide immunogens of the current invention while being substantially incapable of mediating non- cytolytic histamine release, are capable of eliciting antibodies with serological cross-reactivity with the target amino acid sequences of the extracellular region of the human mlgE membrane anchor domain (SEQ ID NOS:l,2) .
  • the initial dose e.g. 0.2-2.5 mg; preferably 1 mg, of immunogen is to be administered by injection, preferably intramuscularly, followed by repeat (booster) doses. Dosage will depend on the age, weight and general health of the patient as is well known in the vaccine and therapeutic arts.
  • immunogen relates to a synthetic peptide which is capable of inducing antibodies against the mlgE membrane anchor domain (SEQ ID NOS:l,2), leading to suppression of IgE-mediated basophil and mast cell degranulation.
  • the immunogen of the present invention includes linear synthetic peptides which contain promiscuous helper T cell epitopes (Th epitopes) .
  • the Th peptides are covalently attached to the mlgE membrane anchor domain peptide (SEQ ID NOS:l,2), with a spacer, so as to be adjacent to either the N- or C-terminus of the membrane anchor peptides, in order to evoke efficient antibody responses.
  • the immunogen may also comprise an immune stimulatory amino acid sequence corresponding to a domain of an invasin protein from the bacteria Yersinia spp 05 '. The invasin domain is attached through a spacer to a Th peptide.
  • the immunogen of the present invention minimizes the generation of irrelevant antibodies to elicit a more focused immune response to the "target sequences", the desired reactivity to mlgE membrane anchor sites (SEQ ID N0S:1,2), without producing undesirable side effects which may complicate the immunotherapy process for the treatment of allergy.
  • the desired target sequence is short, such as the 26 and 17 amino acid mlgE peptides (SEQ ID NOS:l,2) of the present invention
  • T cell-dependent antigens i.e. the presence of a T helper epitope is required to render such antigens immunogenic.
  • A is an amino acid, ⁇ -NH 2 , a fatty acid or a derivative thereof, or an invasin domain;
  • B is an amino acid
  • Th is a helper T cell epitope or an immune enhancing analog or segment thereof;
  • mlgE peptide is:
  • n is from 1 to about 10
  • m is from 1 to about 4
  • o is from 0 to about 10.
  • the peptide immunogen of the present invention comprises from about 20 to about 100 amino acid residues, preferably from about 20 to about 50 amino acid residues and more preferably from about 20 to about 35 amino acid residues.
  • A is an amino acid
  • it can be any non- naturally occurring or any naturally occurring amino acid.
  • Non-naturally occurring amino acids include, but are not limited to, ⁇ -alanine, ornithine, norleucine, norvaline, hydroxyproline, thyroxine, ⁇ -amino butyric acid, homoserine, citrulline and the like.
  • Naturally-occurring amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine.
  • m is greater than one, and two or more of the A groups are amino acids, then each amino acid may be independently the same or different.
  • A is a fatty acid, such as stearic acid, palmitic acid or a fatty acid derivative, such as tripalmitoyl cysteine (Pam 3 Cys) , it acts as an adjuvant by enhancing the immune stimulating properties of the Th epitope 06 '.
  • the fatty acid moiety or its derivative is preferably located at the amino terminus of the mlgE peptide.
  • the peptide im uogen comprises 2 or 3 additional A moieties which are amino acids.
  • the fatty acid is selected from the group with a hydrocarbon chain of 8 to 24 carbon atoms. The hydrocarbon chain can be saturated or unsaturated.
  • A When A is an invasin domain, it is an immune stimulatory epitope from the invasin protein of a Yersinia species. This immune stimulatory property results from the capability of this invasin domain to interact with the ⁇ l integrin molecules present on T cells, particularly activated immune or memory T cells.
  • the specific sequence for an invasin domain found to interact with the ⁇ l integrins has been decribed by Brett et al ⁇ 15) , incorporated herein by reference.
  • the invasin domain (Inv) for linkage to a promiscuous Th epitope has the sequence:
  • Thr-Tyr-Gln-Phe (SEQ ID NO:3) or is an immune stimulatory ar.alog thereof from the corresponding region in another Yersinia species invasin protein.
  • Such analogs thus may contain substitutions, deletions or insertions of amino acid residues to accommodate strain to strain variation, provided that the analogs retain immune stimulatory properties.
  • n is 1 and A is ⁇ -NH 2 . In another embodiment, n is 4 and A is ⁇ -NH 2 , an invasin domain (Inv) , glycine and glycine, in that order.
  • Inv invasin domain
  • B is a spacer and is an amino acid which can be naturally occurring or the non-naturally occurring amino acids as described above.
  • Each B is independently the same or different.
  • the amino acids of B can form a flexible hinge, or spacer, to enhance the immune response to the Th epitope and mlgE peptides and their immunogenic analogs thereof.
  • Examples of sequences encoding flexible hinges are found in the immunoglobulin heavy chain hinge region. Flexible hinge sequences are often proline rich.
  • One particularly useful flexible hinge is provided by the sequence Pro-Pro-Xaa-Pro-Xaa-Pro (SEQ ID N0:4) , where Xaa is any amino acid, and preferably aspartic acid.
  • Immunogenicity can also be improved through the addition of spacer residues (e.g. Gly-Gly) between the promiscuous Th epitope and the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof.
  • spacer residues e.g. Gly-Gly
  • the spacer glycine residues can disrupt any artifactual secondary structures created by the joining of the Th epitope with the the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof and thereby eliminate interference between the T and/or B cell responses.
  • the conformational separation between the helper cell and the antibody eliciting domains thus permits more efficient interactions between the presented immunogen and the appropriate Th and B cells.
  • Th is a sequence of amino acids (natural or non- natural amino acids) that comprises a Th epitope.
  • a Th epitope can consist of a continuous or discontinuous epitope. Hence not every amino acid of Th is necessarily part of the epitope. Accordingly, Th epitopes, including analogs and segments of Th epitopes, are capable of enhancing or stimulating an immune response to the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunological analogs thereof. Th epitopes that are immunodominant and promiscuous are highly and broadly reactive in animal and human populations with widely divergent MHC types 07 ' 19 '.
  • the Th domain of the subject peptides has from about 10 to about 50 amino acids and preferably from about 10 to about 30 amino acids. When multiple Th epitopes are present (i.e. m ⁇ 2), then each Th epitope is independently the same or different.
  • Th epitope analogs include substitutions, additions, deletions and insertions of from one to about 10 amino acid residues in the Th epitope.
  • Th segments are contiguous portions of a Th epitope that are sufficient to enhance or stimulate an immune response to the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunological analogs thereof.
  • Th epitopes of the present invention include hepatitis B surface and core antigen helper T cell epitopes (HB,Th and HB c Th) , pertussis toxin helper T cell epitopes (PT Th) , tetanus toxin helper T cell epitopes (TT Th) , measles virus F protein helper T cell epitopes (MV F Th) , Chla ydia trachomatis major outer membrane protein helper T ceil epitopes (CT Th) , diphtheria toxin helper T cell epitopes (DT Th) , Plasmodi ⁇ m falciparu circumsporozoite helper T cell epitopes (PF Th) , Schistosoma mansoni triose phosphate isomerase helper T cell epitopes (SM Th) , Escherichia coli TraT helper T cell epitopes (TraT Th) and immune-enhancing analogs and segments of any of these
  • Ser-Leu-Asp SEQ ID NO: 5
  • PT 1A Th Tyr-Met-Ser-Gly-Leu-Ala-Val-Arg-Val-His-Val-Ser- Lys-Glu-Glu (SEQ ID NO: 9)
  • MV 'pF2 Th: Gly-Ile-Leu-Glu-Ser-Arg-Gly-Ile-Lys-Ala-Arg-Ile- Thr-His-Val-Asp-Thr-Glu-Ser-Tyr (SEQ ID NO: 14) TT 4 Th: Trp-Val-Arg-Asp-Ile-Ile-Asp-Asp-Phe-Thr-Asn-Glu-
  • CT Th Ala-Leu-Asn-Ile-Trp-Asp-Arg-Phe-Asp-Val-Phe-Cys-
  • TraT 3 Th Ser-Thr-Glu-Thr-Gly-Asn-Gln-His-His-Tyr-Gln-Thr- Arg-Val-Val-Ser-Asn-Ala-Asn-Lys (SEQ ID NO:24)
  • the linear synthetic peptides of this invention as described by the formulas (A) n - (Th) m - (B) 0 - (mlgE peptide) or (mlgE peptide) - (B) 0 - (Th) m - (A) n , have the Th epitope covalently attached through spacer B to the N terminus of either of the the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof.
  • the Th epitope is HB S Th, PT, Th, PT 2 Th, TT, Th, TT 3 Th, or MV F , Th.
  • the sequence of the mlgE membrane anchor peptide comprises:
  • Immunogenic peptide analogs of the mlgE anchor peptides may further comprise substitutions, additions, deletions, or insertions of from one to about four amino acid residues provided that the peptide analogs are capable of eliciting immune responses crossreactive with the mlgE membrane anchor peptides (SEQ ID NOS:l,2).
  • the substitutions, additions, and insertions can be accomplished with natural or non-natural amino acids as defined herein.
  • preferred peptide immunogens of this invention are the linear synthetic peptides containing the mlgE membrane anchor peptides (SEQ ID NOS:l,2) or immunological analogs thereof and Th.
  • the more preferred peptide immunogens are those linear constructs containing the mlgE membrane anchor peptides (SEQ ID NOS:l,2) or immunogenic analogs thereof; a spacer (e.g Gly-Gly) ; a Th epitope selected from the group consisting of HB S Th, PT, Th, PT 2 Th, TT, Th, TT 3 Th, and MV ⁇ Th (SEQ ID NOS:5,6,11,7,10,12, respectively); and, optionally, an Inv domain (SEQ ID NO:3) or analog thereof.
  • peptide immunogens of this invention can be made by chemical synthesis methods which are well known to the ordinarily skilled artisan. See, for example, Grant, ed. Synthetic Peptides 00 '. Hence, peptides can be synthesized using the automated Merrifield techniques of solid phase synthesis with the ⁇ -NH 2 protected by either t- Boc or F-moc chemistry using side chain protected amino acids on, for example, an Applied Biosystems Peptide Synthesizer Model 430A or 431.
  • A is a fatty acid, it can be easily added to the N-terminal amino group of the synthesized peptide by the well known dicyclohexyl-carbodiimide coupling method.
  • Pam 3 Cys, lipoamino acid S-[2,3- Bis(palmitoyloxy) - (2R) -propyl-N-palmitoyl- (R) -cysteine may also be synthesized by chemical methods.
  • Pam 3 Cys can be coupled to, for example, the N terminus of the mlgE peptide by solid-phase synthesis using Pam 3 Cys-OH in the final coupling step to link the lipoamino acid to a resin- bound mlgE peptide chain.
  • the solid-phase peptide can be elongated with additional serine and lysine residues at the N-terminus.
  • the resin After complete assembly of the desired immunogen, the resin is treated according to standard procedures to cleave the peptide from the resin and deblock the functional groups on the amino acid side chains.
  • the free peptide is purified by HPLC and characterized biochemically, for example, by amino acid analysis or by sequencing. Purification and characterization methods for peptides are well known to one of ordinary skill in the art.
  • a cysteine can be added to the C terminus of a Th-containing peptideand the thiol group of cysteine may be used to form a covalent bond to an electrophilic group such as an N° chloroacetyl-modified or a maleimide-derivatized cu- or e-NH 2 group of a lysine residue attached to the N-terminus of an mlgE membrane anchor peptide (SEQ ID N0S:1 or 2) or immunogenic analogs thereof.
  • an electrophilic group such as an N° chloroacetyl-modified or a maleimide-derivatized cu- or e-NH 2 group of a lysine residue attached to the N-terminus of an mlgE membrane anchor peptide (SEQ ID N0S:1 or 2) or immunogenic analogs thereof.
  • the subject immunogen may also be polymerized. Polymerization can be accomplished for example by reaction between glutaraldehyde and the -NH 2 groups of the lysine residues using routine methodology.
  • the linear "A-Th-spacer- (mlgE peptide) " or " (mlgE peptide) -spacer- (Th) m - (A) n " immunogen can be polymerized or co-polymerized by utilization of an additional cysteine added to the N-terminus of the linear "A-Th-spacer- (mlgE peptide) or "(mlgE peptide) -spacer- (Th) m - (A) n " immunogen.
  • the thiol group of the N-terminal cysteine can be used for the formation of a "thioether" bond with haloacetyl- modified amino acid or a maleimide-derivatized or- or e-NH 2 group of a lysine residue that is attached to the N- terminus of a branched poly-lysyl core molecule (e.g., ⁇ 2 ⁇ , K ⁇ K ⁇ or K-g ⁇ i ⁇ K ) .
  • a branched poly-lysyl core molecule e.g., ⁇ 2 ⁇ , K ⁇ K ⁇ or K-g ⁇ i ⁇ K
  • the longer linear peptide immunogens can be synthesized by well known recombinant DNA techniques. Any standard manual on DNA technology provides detailed protocols to produce the peptides of the invention.
  • a gene encoding a peptide of this invention the amino acid sequence is reverse translated into a nucleic acid sequence, and preferably using optimized codon usage for the organism in which the gene will be expressed.
  • a synthetic gene is made, typically by synthesizing overlapping oligonucleotides which encode the peptide and any regulatory elements, if necessary.
  • the synthetic gene is inserted in a suitable cloning vector and recombinants are obtained and characterized.
  • the peptide is then expressed under suitable conditions appropriate for the selected expression system and host.
  • the peptide is purified and characterized by standard methods.
  • the efficacy of the immunogen of the present invention can be established by injecting an animal, for example, rats, followed by monitoring the humoral immune response to the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof, as detailed in the Examples.
  • Another aspect of this invention provides a vaccine composition
  • a vaccine composition comprising an immunologically effective amount of one or more of the immunogens of this invention in a pharmaceutically acceptable delivery system.
  • Such vaccine compositions are used for prevention of atopic allergic reactions including allergic rhinitis, those of food allergies, asthma, anaphylaxis, and other IgE-mediated hypersensitive reactions such as virally- induced asthma.
  • the subject peptides can be formulated as a vaccine composition using adjuvants, pharmaceutically-acceptable carriers or other ingredients routinely provided in vaccine compositions.
  • Such formulations are readily determined by one of ordinary skill in the art and include formulations for immediate release and/or for sustained release, and for induction of systemic immunity and/or induction of localized mucosal immunity, which may be accomplished by, for example, immunogen entrapment by microparticles.
  • the present vaccines can be administered by any convenient route including subcutaneous, oral, intramuscular, or other parenteral or enteral route. Similarly the vaccines can be administered as a single dose or multiple doses. Immunization schedules are readily determined by the ordinarily skilled artisan.
  • Adjuvants or emulsifiers that can be used in this invention include alum, incomplete Freund's adjuvant, liposyn, saponin, squalene, L121, emulsigen and ISA 720 as well as the other efficacious adjuvants and emulsifiers.
  • the vaccine composition of the instant invention contain an effective amount of one or more of the immunogens of the present invention and a pharmaceutically acceptable carrier.
  • a composition in a suitable dosage unit form generally contains about 0.5 ⁇ g to about 1 mg of the immunogen per kg body weight. When delivered in multiple doses, it may be conveniently divided into an appropriate amount per dosage unit form.
  • Vaccines which contain cocktails of the subject immunogens with two or more of the Th epitopes may enhance immunoefficacy in a broader population and thus provide an improved immune response to the mlgE membrane anchor peptide (SEQ ID NOS:l,2).
  • a cocktail of Peptide Nos. 1-4 of Example 1 and 7-10 of Example 4 is useful.
  • Other immune stimulatory synthetic peptide-based mlgE anchor peptide immunogens are arrived at through modification into lipopeptides, such as Pam 3 Cys, so as to provide a built-in adjuvant for a potent vaccine.
  • the immune response to synthetic mlgE anchor peptide- containing immunogens can be improved by delivery through entrapment in or on biodegradable microparticles of the type described by O'Hagan et al (2i) .
  • the immunogens can be encapsulated with or without an adjuvant, including covalently attached Pam 3 Cys, and such microparticles can carry an immune stimulatory adjuvant such as Freund's Incomplete Adjuvant or alum.
  • the microparticles function to potentiate immune responses to an immunogen, including localized mucosal immunity which may be especially applicable to mucosally localized allergic reactions, and to provide time-controlled release for sustained or periodic responses, for oral administration, and for topical administration 01,22 '.
  • peptide immunogens and compositions are provided in the following examples to illustrate the invention.
  • the peptide immunogen of the invention can be useful for the amelioration of IgE-mediated allergic disorders.
  • Immunogens 1-4 (Table III) were synthesized by solid phase synthesis using F-moc chemistry on an Applied Biosystems Peptide Synthesizer Model 430A or 431 according to manufacturer's instructions. After complete assembly of the peptide, the resin was treated according to standard procedures to cleave the peptide from the resin and deprotect the functional groups on amino acid side chains. The free peptide was then purified by HPLC and characterized biochemically for amino acid content and sequence.
  • Peptide immunogens Nos. 1-4 from the amino terminus to the carboxyl terminus, are symbolized as A-Th-B- (mlgE peptide) or (mlgE peptide) -B- Th-A, where "A” is NH 2 - , "B” is a Gly-Gly spacer, "Th” is the measles virus Fl helper T cell epitope MV F , Th (SEQ ID NO:12) , and "(mlgE peptide)” is either mlgE peptide of SEQ ID NO:l or mlgE peptide of SEQ ID NO:2.
  • Adjuvant Freund's Complete/Incomplete
  • Assay ELISAs for anti-peptide activity. Blood is collected and processed into serum, and stored prior to titering by ELISA with the target peptides (SEQ ID NOS:1,2) .
  • Anti-peptide antibody activities are determined by ELISA (enzyme-linked immunosorbent assay) using 96-well flat bottom microtiter plates which are coated with the corresponding target mlgE peptide epitope as the immunosorbent, either Peptide 5 for mlgE anchor peptide site described by SEQ ID NO:l or Peptide No. 6, described by SEQ ID N0:2. Aliquots (100 ⁇ L) of a peptide immunogen solution at a concentration of 5 ⁇ g/mL are incubated for 1 hour at 37°C. The plates are blocked by another incubation at 37°C for 1 hour with a 3% gelatin/PBS solution. The blocked plates are then dried and used for the assay.
  • ELISA enzyme-linked immunosorbent assay
  • the plates are washed six times with 0.05% PBS/Tween ® buffer.
  • 100 ⁇ L of horseradish peroxidase labelled goat-anti-rat antibody is added at a dilution of 1:1,000 in conjugate dilution buffer (Phosphate buffer containing 0.5M NaCl, and normal goat serum) .
  • the plates are incubated for 1 hour at 37°C before being washed as above. Aliquots (100 ⁇ L) of o-phenylenediamine substrate solution are then added.
  • the color is allowed to develop for 5-15 minutes before the enzymatic color reaction is stopped by the addition of 50 ⁇ L 2N H 2 S0 4 .
  • the A 492nm of the contents of each well is read in a plate reader.
  • the peptide immunogen compositions of this invention evoke antibodies that target IgE-secreting human B cells and inhibit IgE production. Unlike most antibodies to IgE, the mlgE peptide-elicited antibodies do not bind to cells bearing only the secretory form of IgE bound to receptors and are therefore incapable, by themselves, of triggering the release of the chemical mediators of allergic response from IgE-sensitized mast cells and basophils. These biological activities are of relevance to immunotherapy for allergy and can be observed in the rat antisera to Peptide immunogens Nos. 1-4 by assaying for the following functions:
  • a human IgE- producing B cell line for example the myeloma cell line SKO-007 (ATCC, Rockville MD) or an EBV-transformed B cell line 8866, is incubated with serial dilutions of the rat anti-mlgE and antibody binding is detected using FITC- labeled ant-rat IgG and quantitated by fluorescence flow cytometry* 4 '.
  • the extent of binding by the rat anti-mlgE antibodies to normal human basophils, prepared from peripheral blood and loaded or sensitized with secreted IgE, is evaluated in a similar fashion (5) .
  • IgE-secretin ⁇ cells Reduction in the I ⁇ E accumulation of IgE-secretin ⁇ cells.
  • IgE accumulates in the culture medium of myeloma cell line SKO-007 and in like IgE-secreting cell lines.
  • Peptide immunogens Nos. 1-4 results in a decrease of IgE secreted into the medium
  • the cells are treated with a range of antibody concentrations, and IgE levels in the medium are monitored over time by IgE-specific ELISA 01 '. Efficacious antibodies result in a dose-related reduction in accumulated IgE.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • Th-spacer- (mlgE peptide) and (mlgE peptide) -spacer-Th peptides wherein there are more than one Th epitope for recognition by diverse MHC types can serve to broaden immune responsiveness in a genetically diverse human population.
  • Promiscuous Th epitopes useful for this purpose are selected from Table II.
  • the Th epitopes of Table II that are useful for such peptide cocktails include but are not limited to the MV F , Th peptide (SEQ ID NO:12) used in Peptide Nos. 1-4 (SEQ ID NOS:25-28) and the HB S Th peptide (SEQ ID NO:5) .
  • Peptides containing either of the two anchor membrane mlgE peptide sequences (SEQ ID NOS:l or 2) and the HB, Th peptide are described in Table IV as Peptide immunogens Nos. 7-10 (SEQ ID NOS: 29-32).
  • Peptide immunogens Nos. 7-10 are synthesized as described in Example 1 and combined with each other and with Peptide immunogens Nos. 1-4, in equal molar ratios, to formulate into a peptide cocktail.
  • a composition of the invention formulated as a cocktail is evaluated for immunopotency in rats by the protocol described below.
  • Alum Alluminum hydroxide
  • CFA/IFA groups receive CFA week 0, IFA weeks 2 and 4.
  • Alum groups receive Alum formulations for all 3 doses
  • This study is designed to demonstrate improved immunogenicity for this embodiment of the peptide invention, and to demonstrate efficacy for a composition of the invention formulated with a pharmaceutically acceptable adjuvant, Alum.
  • Th peptide sequences from measles virus F and hepatitis B surface antigen are promiscuous for multiple human HLA DR antigens, so as to provide maximum immunogenicity in a genetically diverse human population.
  • these Th peptides are derived from children's vaccines, childhood vaccinations are a potential source of Th memory in an immunized human population.
  • children's vaccines have the potential to afford enhanced immunopotency to anti- allergy vaccines comprised of mixtures of such Th peptides.
  • compositions of the invention formulated as a mixture of such linear "Th-Spacer- (mlgE Peptide) " and " (mlgE Peptide) -spacer-Th” peptide constructs, in a widely acceptable adjuvant, Alum.
  • Efficacy and safety of the clinical composition are evaluated by comparisons of serological tests, skin test reaction, by recording patient usage of hay fever medication, by physical examination for allergic symptoms and adverse reactions, and by interviews to obtain subjective patient assessments of the product.
  • Serological evaluations include the aforementioned ELISAs for antipeptide titers, and a standard automated spectrofluorimetric assay to determine reduction in histamine levels 03 ' as well as to ascertain that the products do not trigger histamine release.
  • the skin test is an intradermal test in which a standardized solution of allergens is injected into the upper layers of the skin. Reactions to the allergens are quantitated in the skin test by determining the area of the typical "wheal and flare" produced in response to the allergens.
  • the expected results include significant improvement in allergic symptoms at the endpoint of the study, and no evidence of histamine release. This experiment demonstrates the clinical efficacy and safety of a pharmaceutically acceptable composition of the invention.
  • ADDRESSEE Maria C.H. Lin
  • B STREET: 345 Park Avenue
  • Lys Lys Lys Leu Arg Arg Leu Leu Tyr Met lie Tyr Met Ser 1 5 10
  • Lys Lys Gin Tyr lie Lys Ala Asn Ser Lys Phe lie Gly 1 5 10 lie Thr Glu Leu 15
  • Trp Val Arg Asp lie lie Asp Asp Phe Thr Asn Glu Ser 1 5 10

Abstract

The present invention relates to a method for eliciting the production in healthy mammals, including humans, of high titer antibodies specific for sites on the extracellular segment of the anchor domain of the membrane-bound ε heavy chain of B cell-expressed humain IgE by the use of a composition comprising a synthetic peptide immunogen containing extracellular membrane anchor sites, to reduce IgE-secreting B leukocytes and allergen-induced IgE production. It also relates to the use of optimally designed, carrier protein free, IgE ε-chain related immunogens as key components in a synthetic vaccine to provide an immunotherapy for the treatment of allergy. The subject peptides contain immune stimulator sequences, including a tandemly linked helper T cell epitope, to aid in stimulating the immune response towards the mIgE membrane anchor domain.

Description

o
SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY
FIELD OF THE INVENTION The present invention relates to the use of a composition of a synthetic peptide immunogen comprising a target antigenic site and a helper T cell epitope covalently linked in a linear tandem form. More particularly, the present invention relates to the use of such a composition to elicit the production in healthy mammals, including humans, of high titer antibodies specific to sites on the e heavy chain of B cell-expressed membrane-bound human IgE, i.e., sites on the extracellular segment of the anchor domain of membrane-bound human e- chain and to the use of such a composition as a vaccine to provide an immunotherapy for the treatment of allergy.
BACKGROUND OF THE INVENTION Immunotherapy for the prevention of IgE-mediated allergic responses such as asthma and hay fever, as known and practiced since early in this century, has involved desensitization or hyposensitization by administering a gradually increasing amount of an allergen to a patient to reduce the effects of subsequent exposure to that allergen1). Limitations to such an allergen-based immunotherapy include difficulties in identifying the allergen involved and the adverse reactions frequently caused by the use of the identified allergen0'. Other treatments for the relief of allergies employ therapeutic compounds to block the cascade of cellular events that is responsible for allergic reactions. These compounds include anti-histamines, decongestants, β2agonists, and corticosteroids. Anti-histamines, decongestants, and β2 agonists act on events downstream of IgE in the allergic cascade, making them palliative remedies which address allergic symptoms rather than preventative treatments which must act on events closer to the initiation of IgE- mediated allergic reactions. These palliative remedies provide relief that is short term and partial, frequently accompanied by adverse side effects. For example, anti- histamines may cause restlessness or sedation, and β2 agonists are sometimes associated with increased morbidity in asthmatic patients.
In contrast to anti-histamines, decongestants and β2 agonists, corticosteroids are powerful immunosuppressants and are highly efficacious for the treatment of allergic symptoms. However, they produce adverse hormonal activities and may cause an undesirably broad immunosuppression. To avoid the shortcomings of these therapeutic compounds, it would be desirable to prevent allergic responses at the level of IgE, with a means of suppression selectively targeted to IgE. This may be accomplished by suppressing IgE synthesis directly or indirectly. Indirect suppression can be accomplished by desensitization or by inhibition of IL-4 and other T cell-produced mediators of IgE synthesis05. Direct suppression, as suggested by Chang et al.(4), can be accomplished by specifically targeting IgE-producing B cells with selective antibodies.
Chang et al.4") and others(9) have studied human e-chains and corresponding antibodies, as well as the genes and mRNAs by which the e-chains are encoded. They have elucidated the molecular basis for the expression of two types of IgE: the secreted and membrane-bound forms by B cells committed to IgE synthesis. The membrane-bound form of IgE (mlgE) may be distinguished from the secreted form by an additional membrane anchoring domain that extends from the C-terminus of the heavy chains and is contiguous with the CH4 constant domain of IgE. The membrane-bound form is distinctive to the surface of B cells committed to IgE synthesis. By targeting such cells with antibodies specific for the exposed extracellular portion of that anchor domain, such cells may be eliminated or inactivated. The mechanisms for elimination of IgE-secreting cells by such antibodies can be through antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytolysis(6,7). The reduction of circulating IgE and IgE-expressing cells by anti-IgE antibodies has been demonstrated in vivo in mice(10), and by the inhibition in vivo of passive cutaneous anaphylaxis in a rat model. It has also been demonstrated in human IgE- secreting cell lines where anti-IgE was shown to lead to reductions in cell growth, decreased IgE accumulation and cytolysis in complement-mediated and ADCC-mediated cytolysis assays(11). By determining the nucleotide sequences of pertinent segments in human genomic DNA and in mRNA from human mlgE-expressing B cells, amino acid sequences for the extracellular portion of the anchor domain were predicted. The presence and specificity of these sites as well as their accessibility to antibodies were confirmed with specific antibodies(4,). These polyclonal and monoclonal antibodies were anti-peptide antibodies derived from immunizations with e chain-related "peptide-carrier protein conjugate"(4,8). The carrier protein was Keyhole Limpet Hemocyanin (KLH) known to be useful for its capability of stimulating antibody responses to a target peptide. This approach can be used to demonstrate the utility of the mlgE-specific peptide immunogen for the immunotherapy of allergic disease either by passive immunization with monoclonal antibodies, or by active immunization*6'7*.
The feasibility of using such a peptide-KLH vaccine to provide immunotherapy to patients with IgE- mediated sensitivities has been reported by Stanworth et al. using a different e-chain peptide(12,13). A rabbit anti- peptide serum, selected from bleeds obtained from multiple immunizations which produced better-than-average anti- peptide titer, reduced induced histamine release from rat peritoneal mast cells in a titer-dependent fashion. This inhibitory activity was further confirmed by in vivo tests in a rat passive cutaneous anaphylaxis (PCA) model. The effect of this rabbit anti-peptide serum on anaphylaxis was assessed by measurement of the area of blueing and by an estimate of the color intensity when given to rats which had been previously sensitized by multiple allergen application prior to anaphylactic challenge with allergen. These results were preliminary indications for the feasibility of using peptide-based vaccines for the treatment of allergy. However, this peptide-conjugate strategy has met with considerable difficulties. For example, it has been found that the resultant clones and antisera raised by such conjugates contain more antibodies directed at the epitopes on the protein carrier, KLH, than to the target-peptide1). Other major deficiencies of protein carrier conjugate vaccines include: less-than- optimal immune stimulatory capability, manufacturing difficulties stemming from the poorly defined composition of the carrier protein, and the non-uniformity of the conjugation reaction.
Those skilled in the art usually use a conjugated protein carrier for synthetic peptide immunogens because small peptides are poor immunogens. In order to be rendered immunogenic, these peptides are usually conjugated to large carrier proteins chemically or by gene fusion. These processes, however, can produce unpredictable conformational changes in peptides. Moreover, the immune response is frequently misdirected to the immunodominant carrier. Consequently, the development of a potent vaccine that provides long-lasting protection from allergies awaits further immunogen design. Careful explorations into the design of "optimal immunogens, " including full validation by the results of extensive experimental trials, still remain to be accomplished. LIST OF REFERENCES
1. Noon. "Prophylactic inoculation against hay fever." Lancet. i:1572-1573 (1911) .
2. World Health Organization and International Union of Immunological Societies Working Group Report: Current status of allergen immunotherapy. Lancet, i:259-261 (1989) .
3. Pene et al . Proc. Natl. Acad. Sci. USA.. 85:6880-6884 (1988) .
4. Davis, Gosset, and Chang. Bio/Technol .. 9:53-56 (1991) .
5. Sun, Liou, Sun, Gosset, et al . J. Immunol . , 146:199-205 (1991) .
6. Chang. Antigenic epitopes of IgE present on B cell but not basophil surface. U.S. patent no. 5,091,313 (1992) .
7. Chang. Newly identified human epsilon immunoglobulin peptides and related products. U.S. patent no. 5,274,075 (1993) .
8. Peng, Davis, Sun, et al . J. Immunol .. 148:129- 136 (1992) .
9. Saxon, Kurbe-Leamer, et al. J. Immunol .. 147:4000-4006 (1991) .
10. Haba and Nisonoff . Proc. Natl. Acad. Sci. USA., 87:3363-3367 (1990) .
11. Davis, Gosset, Pinkston, Liou, et al . Springer Semin. Immunopath. , 15:51-73 (1993) .
12. Stanworth, Jones, Lewin, and Nayyar. Lancet, 336:1279-1281 (1990) .
13. Stanworth, Lewin, Nayyar, and Jones. Immunoreactive peptides and antibodies and their use in anti-allergy treatment. EPO 403 312 Al (1990) .
14. Stanworth, and Burt. Mol . Immunol ■ , 23:1231-1235 (1986) . 15. Brett, et al . Eur. J. Immunol . , 23:1608-1614
(1993) .
16. Weismuller, et al . Int. J. Peptide Res.. 40:255-260 (1992) .
17. Celis, et al . J. Immunol .. 140:1808-1815 (1988) .
18. Demotz, et al . J. Immunol.. 142:394-402 (1989)
19. Chong ,et al. Infect. Immun.. 60:4640-4647
(1992) .
20. Grant, ed. Synthetic Peptides: A User's Guide. W.H.Freeman & Co., New York, NY, (1992) pp. 382
21. O'Hagan, et al . Vaccine. 9:768-771 (1991) .
22 . Eldridge , et al . Molec . Immunol . . 28 : 287 -294
( 1991 ) .
OBJECTS OF THE INVENTION
It is an objective of the present invention to employ a group of membrane-bound IgE (mlgE) e-chain peptide immunogens chemically synthesized linearly in tandem with T helper epitope peptides, which when introduced to mammals, including humans, can elicit high titer antibodies to exposed peptide sites of the membrane anchoring domain of human mlgE.
Another objective is to design optimal peptide immunogens, with specific amino acid sequences taken from the human mlgE heavy chain membrane anchoring domain (SEQ ID NOS:l,2) and attached to peptides containing promiscuous human helper T cell epitopes in specified orientations which, when introduced into mammals, including humans, will stimulate production of efficacious antibodies to the sites on human mlgE anchor domain. These antibodies may result in the reduction in IgE- producing B lymphocytes and thereby attenuate allergen- induced IgE production, constrain mast cell activation by IgE-allergen complexes, reduce the consequent release of chemical mediators such as histamines responsible for allergic symptoms and depress IgE- ediated passive cutaneous anaphylaxis (PCA) . The end result is expected to be the reduction of allergic symptoms.
Another objective is to develop an effective mlgE e-chain peptide-based vaccine, employing compositions containing such linear peptide immunogens, so as to provide immunotherapy for the treatment of allergic reactions.
SUMMARY OF THE INVENTION According to the present invention, a series of linearly arranged synthetic peptides which contain either of two peptide sequences corresponding to sites on the exposed portion of the membrane anchoring domain of human mlgE (SEQ ID NOS:l,2) or their immunogenic analogs thereof together with a portion of a helper T-cell epitope (Th epitope) are made by solid phase synthesis. Compositions of the invention are used to immunize healthy mammals, e.g. rats and humans for the production of high titer antisera that is specific for the mlgE anchor membrane sites (SEQ ID N0S:1,2) and free of irrelevant antibodies.
According to the present invention, vaccines containing the synthetic peptide compositions as the key immunogen may also be prepared with an immunologically effective amount of linear synthetic peptide in the presence of a proper adjuvant and/or delivery vehicle. It is expected that such vaccine compositions will elicit a more focused anti-IgE peptide response than those of the peptide-carrier protein conjugates currently used by Chang et al.(4,6*8), thus providing better immunotherapy for the treatment of allergy.
DETAILED DESCRIPTION OF THE INVENTION
This invention is directed to the use of a novel group of peptide-based immunogens for the generation of high titer antibodies to mlgE anchor membrane sites (SEQ ID NOS:l,2) on human IgE e heavy chain in healthy mammals, including humans, ultimately for the treatment of IgE- mediated allergic diseases.
Table I shows the arrangement and the amino acid sequence of the Membrane Anchor Domain of the e heavy chain of human membrane-bound IgE (mlgE) , as deduced from the nucleic acid sequence of the predominant species of mRNA that codes for membrane-bound e chain*7,8'. The regions on the e chain sequence of the peptides used as the target immunogens of the invention are underlined: a single underline for SEQ ID NO:l, and a double underline for SEQ ID NO:2. TABLE I
CH4 Domain | Membrane Anchor Domain/extracellular segment O. ValAsnPro | GlvLeuAlaGlvGlvSerAlaGlnSerGlnArgAlaProAspArgVal
LeuCvsHisSerGlvGlnGlnGlnGlvLeuProArgAlaAlaGlvGlvSerValProHisPro
/extracellular ArgCvsHisCvsGlvAlaGlvArgAlaAspTrDProGlvProProGluLeuAspValCvsVal
segment 1 /transmembrane anchor segment
GluGluAlaGluGlvGluAlaProTrpTrijTroThrGlvLeuCvsIlePheAlaAlaLeuPhe
/cytoplasmic anchor segment ul^uSerValSerTyrSerAlaAlal^uThrLeuLeuMetValGlnArgPheLeuSerAla
ThrArgGlnGlyArgProGlnTlirSerLeiAspTyrThrAsnValLeuGlnProHisAla
It is generally accepted by people skilled in the art that allergic symptoms, the immediate result of IgE-dependent hypersensitivities, are caused by the chemical mediators released by mast cells and basophils. This release is triggered when secretory IgE ligated to receptors on a mast cell or basophil is bound by the allergen for which the receptor-bound IgE is specific. The triggering event is actuated after the allergen binds to the the Fab' portion of the surface-bound IgE in an antigen-antibody type interaction. The allergen/antibody binding crosslinks the bivalent surface-bound IgE and induces conformational changes in the distal Fc region of IgE, the region of IgE in direct contact with a high affinity Fc receptor on the mast/basophil cell surface as well as receptor site(s) on the cell surface. By a mechanism as yet not precisely known, the conformational changes activate the cell-IgE-allergen complex with the resultant release from the cell of chemical mediators, including histamine, inducing allergic symptoms and the further secretion of IgE. The secretory IgE which mediates the allergic reaction is produced by terminally differentiated B cells in response to allergen.
Besides secreting the circulatory IgE which becomes bound to mast cells and basophils, B cells committed to IgE synthesis also display membrane-bound IgE (mlgE) on their surface. The mlgE molecules are allergen receptors and are believed to play regulatory roles in the maturation of the B cells, and in activation of the B cells by allergen-specific T cells. The mlgE is distinguishable from the secreted IgE by a membrane- anchoring segment which extends from the C-terminus of the heavy chains which serves to attach the mlgE to the cell membrane. By determining the nucleotide sequences of pertinent segments in human genomic DNA and in mRNA from human mlgE-expressing B cells, amino acid sequences for two immunogenic sites on the extracellular portion of the anchor domain were deduced (SEQ ID NOS:l,2). These sites are present on alternative forms of mlgE that result from different mRNA splicing events. Both sites are present on the predominant species of membrane-bound e-chain, in the orientation shown in Table I. The presence and specificity of these sites as well as their accessibility to antibodies were confirmed with specific antibodies'4,8'. The presence of anti-IgE antibodies directed against such specific mlgE sites, through either active or passive immunization, can lead to the reduction in the numbers of IgE-producing B cells and a concomitant reduction in circulating IgE, possibly through the lytic removal of the IgE-expressing cells00-11'. Moreover, it is desirable to target anti-IgE antibodies to the membrane-anchor domain because the domain is a surface marker specific to IgE- expressing cells. This target site is not available on secreted IgE. Thus, such anti-mlgE cannot bind and crosslink IgE bound to receptors on mast cells and basophils, and cannot by itself induce histamine release. The removal of IgE-expressing cells in hosts suffering from allergic reactions may result in the down-regulation of IgE production and have a therapeutic outcome.
Such interventions employed in the treatment of allergy through the use of specific anti-IgE antibodies, i.e. a kind of immunotherapy, can be achieved either passively, through prophylactic treatment with specific "site-directed" antibodies to IgE, or, more preferably, actively, by providing the host with a vaccine comprised of site-directed peptide immunogens, to elicit the production by the host of its own site-directed anti-IgE antibodies. It is believed that active immunization will provide a more effective and longer lasting form of protection.
The sites on the extracellular segment of the membrane anchor domain (SEQ ID NOS:l,2), arranged on membrane-bound IgE as shown in Table I, have been confirmed as immunogenic sites that are acessible to antibodies through the cross-reactivities of the surface of IgE-secreting cells to antibodies that had been generated by immunizing animals with mlgE anchor membrane peptides coupled to a carrier protein, keyhole limpet hemocyanin (KLH)(4,8).
A major deficiency of these prototype "mlgE peptide" vaccines are their weak immunogenicities, an inherent problem associated with almost all self-antigens. In the present invention, specific examples are provided for the linkage of synthetic immunostimulatory elements to the mlgE peptides (SEQ ID NOS:l,2) in specified orientations such that potent antibodies directed to these sites on mlgE can be broadly generated in a genetically diverse host population. In turn, these antibodies may lead to reduction of IgE-expressing cells, reduced levels of circulating IgE, and diminished IgE-mediated responses, thus resulting in an effective treatment for the prevention of IgE-mediated allergic diseases.
The peptide immunogens of the current invention, while being substantially incapable of mediating non- cytolytic histamine release, are capable of eliciting antibodies with serological cross-reactivity with the target amino acid sequences of the extracellular region of the human mlgE membrane anchor domain (SEQ ID NOS:l,2) .
The initial dose, e.g. 0.2-2.5 mg; preferably 1 mg, of immunogen is to be administered by injection, preferably intramuscularly, followed by repeat (booster) doses. Dosage will depend on the age, weight and general health of the patient as is well known in the vaccine and therapeutic arts.
While there is no particular limitation to species of mammals provided for the preparation of antibodies, it is generally preferred to use mice, rabbits, guinea pigs, pigs, goats, rats or sheep, etc. as the hosts. For active immunization, the term "immunogen" referred to herein relates to a synthetic peptide which is capable of inducing antibodies against the mlgE membrane anchor domain (SEQ ID NOS:l,2), leading to suppression of IgE-mediated basophil and mast cell degranulation. The immunogen of the present invention includes linear synthetic peptides which contain promiscuous helper T cell epitopes (Th epitopes) . The Th peptides are covalently attached to the mlgE membrane anchor domain peptide (SEQ ID NOS:l,2), with a spacer, so as to be adjacent to either the N- or C-terminus of the membrane anchor peptides, in order to evoke efficient antibody responses. The immunogen may also comprise an immune stimulatory amino acid sequence corresponding to a domain of an invasin protein from the bacteria Yersinia spp05'. The invasin domain is attached through a spacer to a Th peptide.
The immunogen of the present invention minimizes the generation of irrelevant antibodies to elicit a more focused immune response to the "target sequences", the desired reactivity to mlgE membrane anchor sites (SEQ ID N0S:1,2), without producing undesirable side effects which may complicate the immunotherapy process for the treatment of allergy. However, when the desired target sequence is short, such as the 26 and 17 amino acid mlgE peptides (SEQ ID NOS:l,2) of the present invention, one faces other challenges because such short peptide antigens are usually T cell-dependent antigens, i.e. the presence of a T helper epitope is required to render such antigens immunogenic. Much effort had to be expended in the design of linear synthetic immunogens containing the short mlgE anchor membrane peptides (SEQ ID N0S:1,2) to provide for functional T-cell epitopes.
The peptides of this invention are represented by the formulas
(A)n-(Th)m-(B)0-(mIgE peptide) wherein: A is an amino acid, α-NH2, a fatty acid or a derivative thereof, or an invasin domain;
B is an amino acid;
Th is a helper T cell epitope or an immune enhancing analog or segment thereof; mlgE peptide is:
Gly-Leu-Ala-Gly-Gly-Ser-Ala-Gln-Ser-Gln-Arg-Ala-
Pro-Asp-Arg-Val-Leu-Cys-His-Ser-Gly-Gln-Gln-Gln-
Gly-Leu (SEQ ID NO:l); or
Pro-Glu-Leu-Asp-Val-Cys-Val-Glu-Glu-Ala-Glu-Gly-
Glu-Ala-Pro-Trp-Thr (SEQ ID NO:2); or an immunogenic analog thereof; n is from 1 to about 10; m is from 1 to about 4; and o is from 0 to about 10.
The peptide immunogen of the present invention comprises from about 20 to about 100 amino acid residues, preferably from about 20 to about 50 amino acid residues and more preferably from about 20 to about 35 amino acid residues.
When A is an amino acid, it can be any non- naturally occurring or any naturally occurring amino acid. Non-naturally occurring amino acids include, but are not limited to, β-alanine, ornithine, norleucine, norvaline, hydroxyproline, thyroxine, γ-amino butyric acid, homoserine, citrulline and the like. Naturally-occurring amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine. Moreover, when m is greater than one, and two or more of the A groups are amino acids, then each amino acid may be independently the same or different. When A is a fatty acid, such as stearic acid, palmitic acid or a fatty acid derivative, such as tripalmitoyl cysteine (Pam3Cys) , it acts as an adjuvant by enhancing the immune stimulating properties of the Th epitope06'. The fatty acid moiety or its derivative is preferably located at the amino terminus of the mlgE peptide. When A is a fatty acid or its derivative, the peptide im uogen comprises 2 or 3 additional A moieties which are amino acids. As used herein, the fatty acid is selected from the group with a hydrocarbon chain of 8 to 24 carbon atoms. The hydrocarbon chain can be saturated or unsaturated.
When A is an invasin domain, it is an immune stimulatory epitope from the invasin protein of a Yersinia species. This immune stimulatory property results from the capability of this invasin domain to interact with the βl integrin molecules present on T cells, particularly activated immune or memory T cells. The specific sequence for an invasin domain found to interact with the βl integrins has been decribed by Brett et al <15), incorporated herein by reference. In a preferred embodiment, the invasin domain (Inv) for linkage to a promiscuous Th epitope has the sequence:
Thr-Ala-Lys-Ser-Lys-Lys-Phe-Pro-Ser-Tyr-Thr-Ala-
Thr-Tyr-Gln-Phe (SEQ ID NO:3) or is an immune stimulatory ar.alog thereof from the corresponding region in another Yersinia species invasin protein. Such analogs thus may contain substitutions, deletions or insertions of amino acid residues to accommodate strain to strain variation, provided that the analogs retain immune stimulatory properties.
In one embodiment, n is 1 and A is α-NH2. In another embodiment, n is 4 and A is α-NH2, an invasin domain (Inv) , glycine and glycine, in that order.
B is a spacer and is an amino acid which can be naturally occurring or the non-naturally occurring amino acids as described above. Each B is independently the same or different. In addition, the amino acids of B can form a flexible hinge, or spacer, to enhance the immune response to the Th epitope and mlgE peptides and their immunogenic analogs thereof. Examples of sequences encoding flexible hinges are found in the immunoglobulin heavy chain hinge region. Flexible hinge sequences are often proline rich. One particularly useful flexible hinge is provided by the sequence Pro-Pro-Xaa-Pro-Xaa-Pro (SEQ ID N0:4) , where Xaa is any amino acid, and preferably aspartic acid.
Immunogenicity can also be improved through the addition of spacer residues (e.g. Gly-Gly) between the promiscuous Th epitope and the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof. In addition to physically separating the Th epitope from the B cell epitope, i.e., the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunological analogs thereof, the spacer glycine residues can disrupt any artifactual secondary structures created by the joining of the Th epitope with the the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof and thereby eliminate interference between the T and/or B cell responses. The conformational separation between the helper cell and the antibody eliciting domains thus permits more efficient interactions between the presented immunogen and the appropriate Th and B cells.
Th is a sequence of amino acids (natural or non- natural amino acids) that comprises a Th epitope. A Th epitope can consist of a continuous or discontinuous epitope. Hence not every amino acid of Th is necessarily part of the epitope. Accordingly, Th epitopes, including analogs and segments of Th epitopes, are capable of enhancing or stimulating an immune response to the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunological analogs thereof. Th epitopes that are immunodominant and promiscuous are highly and broadly reactive in animal and human populations with widely divergent MHC types07'19'. The Th domain of the subject peptides has from about 10 to about 50 amino acids and preferably from about 10 to about 30 amino acids. When multiple Th epitopes are present (i.e. m ≥ 2), then each Th epitope is independently the same or different.
Th epitope analogs include substitutions, additions, deletions and insertions of from one to about 10 amino acid residues in the Th epitope. Th segments are contiguous portions of a Th epitope that are sufficient to enhance or stimulate an immune response to the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunological analogs thereof.
Th epitopes of the present invention include hepatitis B surface and core antigen helper T cell epitopes (HB,Th and HBcTh) , pertussis toxin helper T cell epitopes (PT Th) , tetanus toxin helper T cell epitopes (TT Th) , measles virus F protein helper T cell epitopes (MVF Th) , Chla ydia trachomatis major outer membrane protein helper T ceil epitopes (CT Th) , diphtheria toxin helper T cell epitopes (DT Th) , Plasmodiυm falciparu circumsporozoite helper T cell epitopes (PF Th) , Schistosoma mansoni triose phosphate isomerase helper T cell epitopes (SM Th) , Escherichia coli TraT helper T cell epitopes (TraT Th) and immune-enhancing analogs and segments of any of these Th epitopes. Examples of Th epitope sequences are provided below as Table II: TABLE II HB, Th: Phe-Phe-Leu-Leu-Thr-Arg-Ile-Leu-Thr-Ile-Pro-Gln-
Ser-Leu-Asp (SEQ ID NO: 5)
PT, Th: Lys-Lys-Leu-Arg-Arg-Leu-Leu-Tyr-Met-Ile-Tyr-Met-
Ser-Gly-Leu-Ala-Val-Arg-Val-His-Val-Ser-Lys-Glu- Glu-Gln-Tyr-Tyr-Asp-Tyr (SEQ ID NO: 6)
TT, Th: Lys-Lys-Gln-Tyr-Ile-Lys-Ala-Asn-Ser-Lys-Phe-Ile-
Gly-Ile-Thr-Glu-Leu (SEQ ID NO: 7)
TT2 Th: Lys-Lys-Phe-Asn-Asn-Phe-Thr-Val-Ser-Phe-Trp-Leu-
Arg-Val-Pro-Lys-Val-Ser-Ala-Ser-His-Leu
(SEQ ID NO: 8)
PT1A Th: Tyr-Met-Ser-Gly-Leu-Ala-Val-Arg-Val-His-Val-Ser- Lys-Glu-Glu (SEQ ID NO: 9)
TT3 Th: Tyr-Asp-Pro-Asn-Tyr-Leu-Arg-Thr-Asp-Ser-Asp-Lys-
Asp-Arg-Phe-Leu-Gln-Thr-Met-Val-Lys-Leu-Phe-Asn- Arg-Ile-Lys (SEQ ID NO: 10)
PT2 Th: Gly-Ala-Tyr-Ala-Arg-Cys-Pro-Asn-Gly-Thr-Arg-Ala-
Leu-Thr-Val-Ala-Glu-Leu-Arg-Gly-Asn-Ala-Glu-Leu
(SEQ ID NO: 11)
MV, Th; Leu-Ser-Glu-Ile-Lys-Gly-Val-Ile-Val-His-Arg-Leu- Glu-Gly-Val (SEQ ID NO: 12)
HBC Th: Val-Ser-Phe-Gly-Val-Trp-Ile-Arg-Thr-Pro-Pro-Ala- Tyr-Arg-Pro-Pro-Asn-Ala-Pro-Ile-Leu
(SEQ ID NO: 13)
MV 'pF2, Th: Gly-Ile-Leu-Glu-Ser-Arg-Gly-Ile-Lys-Ala-Arg-Ile- Thr-His-Val-Asp-Thr-Glu-Ser-Tyr (SEQ ID NO: 14) TT4 Th: Trp-Val-Arg-Asp-Ile-Ile-Asp-Asp-Phe-Thr-Asn-Glu-
Ser-Ser-Gln-Lys-Thr (SEQ ID NO: 15)
TT5 Th: Asp-Val-Ser-Thr-Ile-Val-Pro-Tyr-Ile-Gly-Pro-Ala- Leu-Asn-Ile-Val (SEQ ID NO: 16)
CT Th: Ala-Leu-Asn-Ile-Trp-Asp-Arg-Phe-Asp-Val-Phe-Cys-
Thr-Leu-Gly-Ala-Thr-Thr-Gly-Tyr-Leu-Lys-Gly-Asn- Ser (SEQ ID NO: 17)
DT, Th: Asp-Ser-Glu-Thr-Ala-Asp-Asn-Leu-Glu-Lys-Thr-Val- Ala-Ala-Leu-Ser-Ile-Leu-Pro-Gly-Ile-Gly-Cys
(SEQ ID NO:18)
DT2 Th: Glu-Glu-Ile-Val-Ala-Gln-Ser-Ile-Ala-Leu-Ser-Ser-
Leu-Met-Val-Ala-Gln-Ala-Ile-Pro-Leu-Val-Gly-Glu- Leu-Val-Asp-Ile-Gly-Phe-Ala-Ala-Thr-Asn-Phe-Val- Glu-Ser-Cys (SEQ ID NO: 19)
PF Th: Asp-Ile-Glu-Lys-Lys-Ile-Ala-Lys-Met-Glu-Lys-Ala-
Ser-Ser-Val-Phe-Asn-Val-Val-Asn-Ser
(SEQ ID NO: 20)
SM Th: Lys-Trp-Phe-Lys-Thr-Asn-Ala-Pro-Asn-Gly-Val-Asp-
Glu-Lys-Ile-Arg-Ile (SEQ ID NO:21)
TraT, Th: Gly-Leu-Gln-Gly-Lys-His-Ala-Asp-Ala-Val-Lys-Ala- Lys-Gly (SEQ ID NO: 22)
TraT2 Th: Gly-Leu-Ala-Ala-Gly-Leu-Val-Gly-Met-Ala-Ala-Asp- Ala-Met-Val-Glu-Asp-Val-Asn (SEQ ID NO: 23)
TraT3 Th: Ser-Thr-Glu-Thr-Gly-Asn-Gln-His-His-Tyr-Gln-Thr- Arg-Val-Val-Ser-Asn-Ala-Asn-Lys (SEQ ID NO:24) The linear synthetic peptides of this invention, as described by the formulas (A)n- (Th)m- (B)0- (mlgE peptide) or (mlgE peptide) - (B)0- (Th)m- (A)n, have the Th epitope covalently attached through spacer B to the N terminus of either of the the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof. In a preferred embodiment the Th epitope is HBS Th, PT, Th, PT2 Th, TT, Th, TT3 Th, or MVF, Th.
The sequence of the mlgE membrane anchor peptide comprises:
Gly-Leu-Ala-Gly-Gly-Ser-Ala-Gln-Ser-Gln- Arg-Ala-Pro-Asp-Arg-Val-Leu-Cys-His-Ser- Gly-Gln-Gln-Gln-Gly-Leu (SEQ ID NO:l); or Pro-Glu-Leu-Asp-Val-Cys-Val-Glu-Glu-Ala- Glu-Gly-Glu-Ala-Pro-Trp-Thr (SEQ ID NO:2). Immunogenic peptide analogs of the mlgE anchor peptides (SEQ ID NOS:l,2) according to the invention, may further comprise substitutions, additions, deletions, or insertions of from one to about four amino acid residues provided that the peptide analogs are capable of eliciting immune responses crossreactive with the mlgE membrane anchor peptides (SEQ ID NOS:l,2). The substitutions, additions, and insertions can be accomplished with natural or non-natural amino acids as defined herein.
Accordingly, preferred peptide immunogens of this invention are the linear synthetic peptides containing the mlgE membrane anchor peptides (SEQ ID NOS:l,2) or immunological analogs thereof and Th. The more preferred peptide immunogens are those linear constructs containing the mlgE membrane anchor peptides (SEQ ID NOS:l,2) or immunogenic analogs thereof; a spacer (e.g Gly-Gly) ; a Th epitope selected from the group consisting of HBS Th, PT, Th, PT2 Th, TT, Th, TT3 Th, and MVπ Th (SEQ ID NOS:5,6,11,7,10,12, respectively); and, optionally, an Inv domain (SEQ ID NO:3) or analog thereof. The peptide immunogens of this invention can be made by chemical synthesis methods which are well known to the ordinarily skilled artisan. See, for example, Grant, ed. Synthetic Peptides00'. Hence, peptides can be synthesized using the automated Merrifield techniques of solid phase synthesis with the α-NH2protected by either t- Boc or F-moc chemistry using side chain protected amino acids on, for example, an Applied Biosystems Peptide Synthesizer Model 430A or 431.
When A is a fatty acid, it can be easily added to the N-terminal amino group of the synthesized peptide by the well known dicyclohexyl-carbodiimide coupling method.
Pam3Cys, lipoamino acid S-[2,3- Bis(palmitoyloxy) - (2R) -propyl-N-palmitoyl- (R) -cysteine may also be synthesized by chemical methods. Pam3Cys can be coupled to, for example, the N terminus of the mlgE peptide by solid-phase synthesis using Pam3Cys-OH in the final coupling step to link the lipoamino acid to a resin- bound mlgE peptide chain. To improve the solubility of the final coupled lipopeptide product, the solid-phase peptide can be elongated with additional serine and lysine residues at the N-terminus.
After complete assembly of the desired immunogen, the resin is treated according to standard procedures to cleave the peptide from the resin and deblock the functional groups on the amino acid side chains. The free peptide is purified by HPLC and characterized biochemically, for example, by amino acid analysis or by sequencing. Purification and characterization methods for peptides are well known to one of ordinary skill in the art.
Other chemical means to generate the linear peptide constructs of the invention containing mlgE and Th sites include the ligation of the haloacetylated and the cysteinylated peptide through the formation of a "thioether" linkage. For example, a cysteine can be added to the C terminus of a Th-containing peptideand the thiol group of cysteine may be used to form a covalent bond to an electrophilic group such as an N° chloroacetyl-modified or a maleimide-derivatized cu- or e-NH2 group of a lysine residue attached to the N-terminus of an mlgE membrane anchor peptide (SEQ ID N0S:1 or 2) or immunogenic analogs thereof. In this manner, a construct with Th- (mlgE peptide) or its reverse, (mlgE peptide) -Th, may be obtained.
The subject immunogen may also be polymerized. Polymerization can be accomplished for example by reaction between glutaraldehyde and the -NH2 groups of the lysine residues using routine methodology. By another method, the linear "A-Th-spacer- (mlgE peptide) " or " (mlgE peptide) -spacer- (Th)m- (A)n" immunogen can be polymerized or co-polymerized by utilization of an additional cysteine added to the N-terminus of the linear "A-Th-spacer- (mlgE peptide) or "(mlgE peptide) -spacer- (Th)m- (A)n" immunogen. The thiol group of the N-terminal cysteine can be used for the formation of a "thioether" bond with haloacetyl- modified amino acid or a maleimide-derivatized or- or e-NH2 group of a lysine residue that is attached to the N- terminus of a branched poly-lysyl core molecule (e.g., κ2κ, K^K^ or K-g ^i^K ) .
Alternatively, the longer linear peptide immunogens can be synthesized by well known recombinant DNA techniques. Any standard manual on DNA technology provides detailed protocols to produce the peptides of the invention. To construct a gene encoding a peptide of this invention, the amino acid sequence is reverse translated into a nucleic acid sequence, and preferably using optimized codon usage for the organism in which the gene will be expressed. Next, a synthetic gene is made, typically by synthesizing overlapping oligonucleotides which encode the peptide and any regulatory elements, if necessary. The synthetic gene is inserted in a suitable cloning vector and recombinants are obtained and characterized. The peptide is then expressed under suitable conditions appropriate for the selected expression system and host. The peptide is purified and characterized by standard methods.
The efficacy of the immunogen of the present invention can be established by injecting an animal, for example, rats, followed by monitoring the humoral immune response to the mlgE membrane anchor peptides (SEQ ID NOS:l,2) and immunogenic analogs thereof, as detailed in the Examples.
Another aspect of this invention provides a vaccine composition comprising an immunologically effective amount of one or more of the immunogens of this invention in a pharmaceutically acceptable delivery system. Such vaccine compositions are used for prevention of atopic allergic reactions including allergic rhinitis, those of food allergies, asthma, anaphylaxis, and other IgE-mediated hypersensitive reactions such as virally- induced asthma.
Accordingly, the subject peptides can be formulated as a vaccine composition using adjuvants, pharmaceutically-acceptable carriers or other ingredients routinely provided in vaccine compositions. Such formulations are readily determined by one of ordinary skill in the art and include formulations for immediate release and/or for sustained release, and for induction of systemic immunity and/or induction of localized mucosal immunity, which may be accomplished by, for example, immunogen entrapment by microparticles. The present vaccines can be administered by any convenient route including subcutaneous, oral, intramuscular, or other parenteral or enteral route. Similarly the vaccines can be administered as a single dose or multiple doses. Immunization schedules are readily determined by the ordinarily skilled artisan. Adjuvants or emulsifiers that can be used in this invention include alum, incomplete Freund's adjuvant, liposyn, saponin, squalene, L121, emulsigen and ISA 720 as well as the other efficacious adjuvants and emulsifiers.
The vaccine composition of the instant invention contain an effective amount of one or more of the immunogens of the present invention and a pharmaceutically acceptable carrier. Such a composition in a suitable dosage unit form generally contains about 0.5 μg to about 1 mg of the immunogen per kg body weight. When delivered in multiple doses, it may be conveniently divided into an appropriate amount per dosage unit form.
Vaccines which contain cocktails of the subject immunogens with two or more of the Th epitopes may enhance immunoefficacy in a broader population and thus provide an improved immune response to the mlgE membrane anchor peptide (SEQ ID NOS:l,2). For example, a cocktail of Peptide Nos. 1-4 of Example 1 and 7-10 of Example 4 is useful. Other immune stimulatory synthetic peptide-based mlgE anchor peptide immunogens are arrived at through modification into lipopeptides, such as Pam3Cys, so as to provide a built-in adjuvant for a potent vaccine. The immune response to synthetic mlgE anchor peptide- containing immunogens can be improved by delivery through entrapment in or on biodegradable microparticles of the type described by O'Hagan et al(2i) . The immunogens can be encapsulated with or without an adjuvant, including covalently attached Pam3Cys, and such microparticles can carry an immune stimulatory adjuvant such as Freund's Incomplete Adjuvant or alum. The microparticles function to potentiate immune responses to an immunogen, including localized mucosal immunity which may be especially applicable to mucosally localized allergic reactions, and to provide time-controlled release for sustained or periodic responses, for oral administration, and for topical administration01,22'.
Specific peptide immunogens and compositions are provided in the following examples to illustrate the invention. The peptide immunogen of the invention can be useful for the amelioration of IgE-mediated allergic disorders.
EXAMPLE 1
SYNTHESIS OF PEPTIDE IMMUNOGENS NOS 1-4 "Th-SPACER- (mlσE PEPTIDE)"
AND " (mlσE PEPTIDE) -SPACER-Th"
Immunogens 1-4 (Table III) were synthesized by solid phase synthesis using F-moc chemistry on an Applied Biosystems Peptide Synthesizer Model 430A or 431 according to manufacturer's instructions. After complete assembly of the peptide, the resin was treated according to standard procedures to cleave the peptide from the resin and deprotect the functional groups on amino acid side chains. The free peptide was then purified by HPLC and characterized biochemically for amino acid content and sequence.
The structures of Peptide immunogens Nos. 1-4, from the amino terminus to the carboxyl terminus, are symbolized as A-Th-B- (mlgE peptide) or (mlgE peptide) -B- Th-A, where "A" is NH2- , "B" is a Gly-Gly spacer, "Th" is the measles virus Fl helper T cell epitope MVF, Th (SEQ ID NO:12) , and "(mlgE peptide)" is either mlgE peptide of SEQ ID NO:l or mlgE peptide of SEQ ID NO:2. Thus, Peptide No. 1 may be more explicitly represented as "MVF1 Th-GG- (mlgE l) » and Peptide No. 2 as "(mlgE 2)-GG-MVF, Th", for examples. The actual sequences for Peptides 1-4 (SEQ ID NOS:25-28) are shown in Table 2. TABLE 2 Amino Acid Sequences of Peptide Nos. 1-4
Peptide Sequence
1. MVF,Th-GG-mIgEl Leu-Ser-Glu-Ile-Lys-Gly-Val-Ile-Val- His-Arg-Leu-Glu-Gly-Val-Gly-Gly-Leu- Ala-Gly-Gly-Ser-Ala-Gln-Ser-Gln-Arg- Ala-Pro-Asp-Arg-Val-Leu-Cys-His-Ser- Gly-Gln-Gln-Gln-Gly-Leu
(SEQ ID No:25)
2. mIgEl-GG-MVF1Th Leu-Ala-Gly-Gly-Ser-Ala-Gln-Ser-Gln- Arg-Ala-Pro-Asp-Arg-Val-Leu-Cys-His- Ser-Gly-Gln-Gln-Gln-Gly-Leu-Gly-Gly- Leu-Ser-Glu-Ile-Lys-Gly-Val-Ile-Val- His-Arg-Leu-Glu-Gly-Val
(SEQ ID No: 26)
3. MVF,Th-GG-mIgE2S Leu-Ser-Glu-Ile-Lys-Gly-Val-Ile-Val- His-Arg-Leu-Glu-Gly-Val-Gly-Gly-Pro- Glu-Leu-Asp-Val-Cys-Val-Glu-Glu-Ala- Glu-Gly-Glu-Ala-Pro-Trp-Thr
(SEQ ID No:27)
4. mIgE2-GG-MVFITh Pro-Glu-Leu-Asp-Val-Cys-Val-Glu-Glu- Ala-Glu-Gly-Glu-Ala-Pro-Trp-Thr-Gly- Gly-Leu-Ser-Glu-Ile-Lys-Gly-Val-Ile- Val-His-Arg-Leu-Glu-Gly-Val
(SEQ ID Nθ:28)
EXAMPLE 2
IMMUNIZATION OF RATS WITH "Th-SPACER- (mlσE PEPTIDE) "
AND "(mlσE PEPTIDE) -SPACER-Th" LINEAR CONSTRUCTS, PEPTIDE
NOS. 1-4. AND EVALUATION OF IMMUNOGENICITY BY ELISA
Efficacy of Peptide immunogens Nos. 1-4 is evaluated on groups of five rats by the experimental immunization protocol outlined below and by serological assay for determination of immunogenicity. Experimental Design:
Immunogen: Peptide Nos. 1-4 (1 per group)
Dose: 100 μg per immunization
Route: intramuscular
Adjuvant: Freund's Complete/Incomplete
Dose Schedule: week 0 (FCA) , 3 and 6 weeks
( IFA)
Bleed Schedule : weeks 0 , 3 , 6 , 8 , 10
Species : Sprague-Dawley rats
Group size : 5
Assay: ELISAs for anti-peptide activity. Blood is collected and processed into serum, and stored prior to titering by ELISA with the target peptides (SEQ ID NOS:1,2) .
Anti-peptide antibody activities are determined by ELISA (enzyme-linked immunosorbent assay) using 96-well flat bottom microtiter plates which are coated with the corresponding target mlgE peptide epitope as the immunosorbent, either Peptide 5 for mlgE anchor peptide site described by SEQ ID NO:l or Peptide No. 6, described by SEQ ID N0:2. Aliquots (100 μL) of a peptide immunogen solution at a concentration of 5 μg/mL are incubated for 1 hour at 37°C. The plates are blocked by another incubation at 37°C for 1 hour with a 3% gelatin/PBS solution. The blocked plates are then dried and used for the assay. Aliquots (100 μL) of the test rat sera, starting with a 1:10 dilution in a sample dilution buffer and ten-fold serial dilutions thereafter, are added to the peptide coated plates. The plates are incubated for 1 hour at 37°C.
The plates are washed six times with 0.05% PBS/Tween® buffer. 100 μL of horseradish peroxidase labelled goat-anti-rat antibody is added at a dilution of 1:1,000 in conjugate dilution buffer (Phosphate buffer containing 0.5M NaCl, and normal goat serum) . The plates are incubated for 1 hour at 37°C before being washed as above. Aliquots (100 μL) of o-phenylenediamine substrate solution are then added. The color is allowed to develop for 5-15 minutes before the enzymatic color reaction is stopped by the addition of 50 μL 2N H2S04. The A492nm of the contents of each well is read in a plate reader.
All sera are assayed by anti-peptide ELISA and those samples which give A492nm values of ≥ 0.2 at a 1:100 dilution are recorded as seropositive. Normal rat serum is used as a negative control. Results are also compared to positive control serum from rats immunized with KLH conjugates of either Peptide No. 5 or 6 described above, to demonstrate improved immunogenicity for peptide of the invention.
EXAMPLE 3
EVALUATION OF RAT ANTI-SERA TO PEPTIDE NOS. 1-4 FOR FUNCTIONAL EFFICACY ON HUMAN IσE-SECRETING CELL LINES
The peptide immunogen compositions of this invention evoke antibodies that target IgE-secreting human B cells and inhibit IgE production. Unlike most antibodies to IgE, the mlgE peptide-elicited antibodies do not bind to cells bearing only the secretory form of IgE bound to receptors and are therefore incapable, by themselves, of triggering the release of the chemical mediators of allergic response from IgE-sensitized mast cells and basophils. These biological activities are of relevance to immunotherapy for allergy and can be observed in the rat antisera to Peptide immunogens Nos. 1-4 by assaying for the following functions:
1. Antibody binding to IσE-secretinσ B cells and not bindinσ to IσE-sensitized basophils. A human IgE- producing B cell line, for example the myeloma cell line SKO-007 (ATCC, Rockville MD) or an EBV-transformed B cell line 8866, is incubated with serial dilutions of the rat anti-mlgE and antibody binding is detected using FITC- labeled ant-rat IgG and quantitated by fluorescence flow cytometry*4'. The extent of binding by the rat anti-mlgE antibodies to normal human basophils, prepared from peripheral blood and loaded or sensitized with secreted IgE, is evaluated in a similar fashion(5).
2. Reduction in the IσE accumulation of IgE-secretinσ cells. IgE accumulates in the culture medium of myeloma cell line SKO-007 and in like IgE-secreting cell lines. To determine whether treatment of IgE-secreting cells with the rat anti-mlgE sera elicited by Peptide immunogens Nos. 1-4 results in a decrease of IgE secreted into the medium, the cells are treated with a range of antibody concentrations, and IgE levels in the medium are monitored over time by IgE-specific ELISA01'. Efficacious antibodies result in a dose-related reduction in accumulated IgE.
3. Lvsis of IσE-secretinσ cells by antibody dependent cell-mediated cytotoxicity (ADCC) . The capability of a range of concentrations of the rat anti-mlgE sera to induce cytolysis of SKO-007 cells, or the like, by ADCC activity is evaluated for percent lysis by an assay employing effector cells from several donors01'.
4. Inability to induce histamine release from basophils. Rat anti-mlgE sera is used at several concentrations to show a dose-related ability to induce histamine release from IgE-sensitized isolated peripheral blood basophils. Histamine-release is quantitated by fluorometric assay(5). An inability to induce histamine release is evidence for the efficacy and safety of the peptide immunogen of the present invention. EXAMPLE 4
COCKTAIL OF PEPTIDE IMMUNOGENS TO FURTHER BROADEN THE RESPONSIVE POPULATION
A cocktail of immunogenic Th-spacer- (mlgE peptide) and (mlgE peptide) -spacer-Th peptides wherein there are more than one Th epitope for recognition by diverse MHC types can serve to broaden immune responsiveness in a genetically diverse human population. Promiscuous Th epitopes useful for this purpose are selected from Table II. The Th epitopes of Table II that are useful for such peptide cocktails include but are not limited to the MVF, Th peptide (SEQ ID NO:12) used in Peptide Nos. 1-4 (SEQ ID NOS:25-28) and the HBS Th peptide (SEQ ID NO:5) . Peptides containing either of the two anchor membrane mlgE peptide sequences (SEQ ID NOS:l or 2) and the HB, Th peptide are described in Table IV as Peptide immunogens Nos. 7-10 (SEQ ID NOS: 29-32).
Peptide immunogens Nos. 7-10 are synthesized as described in Example 1 and combined with each other and with Peptide immunogens Nos. 1-4, in equal molar ratios, to formulate into a peptide cocktail. A composition of the invention formulated as a cocktail is evaluated for immunopotency in rats by the protocol described below. Experimental Design:
Immunogens: (1) Cocktail : Peptide Nos. 1-4 and 7-10 (2) Positive Control:
Individual KLH conjugates of Peptide Nos. 5 and 6 combined 1:1 (One immunogen per group of rats) Dose: Molar equivalents of each synthetic peptide immunogen or mlgE peptide equivalent, to equal 100 μg total or 12.5 μg of each peptide per immunization Route: intramuscular Adjuvants: (1) Freund's Complete/Incomplete
(2) 0.4% Alum (Aluminum hydroxide) (One of either adjuvant per immunogen per group) .
Dose Schedule week 0, 2 and 4 weeks CFA/IFA groups receive CFA week 0, IFA weeks 2 and 4. Alum groups receive Alum formulations for all 3 doses)
Bleed Schedule weeks 0, 3, 6 and 8 Species: Sprague-Dawley rats
Group size: 5, 4 groups Assay: 2 ELISAs for anti-peptide activity, solid-phase substrates are Peptide Nos. 5 and 6 (SEQ ID NOS:1, 2) Blood is collected, processed into serum and stored prior for determination of seroconversion by the two anti-peptide ELISAs as described in Example 2.
This study is designed to demonstrate improved immunogenicity for this embodiment of the peptide invention, and to demonstrate efficacy for a composition of the invention formulated with a pharmaceutically acceptable adjuvant, Alum.
TABLE IV Amino Acid Sequences of Peptide Nos. 7-10
Peptide Sequence h-GG-mlgEl Phe-Phe -Leu -Leu-Thr- Arg - I le-Leu- Thr- Ile-Pro-Gln-Ser-Leu-Asp-Gly- Gly-Leu-Ala-Gly-Gly-Ser-Ala-Gln- Ser-Gln-Arg-Ala-Pro-Asp-Arg-Val- Leu-Cys-His-Ser-Gly-Gln-Gln-Gln- Gly-Leu (SEQ ID No : 29)
l-GG-HBsTh Leu-Ala-Gly-Gly-Ser-Ala-Gln-Ser- Gln-Arg-Ala-Pro-Asp-Arg-Val -Leu- Cys-His-Ser-Gly-Gln-Gln-Gln-Gly- Leu -Gly-Gly- Phe-Phe -Leu -Leu-Thr - Arg- I le-Leu-Thr- lie -Pro-Gin- Ser- Leu-Asp (SEQ ID No : 30)
h-GG-mIgE2 Phe -Phe -Leu -Leu-Thr- Arg- I le-Leu- Thr- lie -Pro- Gin- Ser-Leu-Asp-Gly- Gly-Pro-Glu-Leu-Asp-Val -Cys-Val - Glu-Glu-Ala-Glu-Gly-Glu-Ala-Pro- Trp-Thr (SEQ ID No : 31)
E2 -GG-HBsTh Pro-Glu-Leu-Asp-Val -Cys-Val -Glu- Glu-Ala-Glu-Gly-Glu-Ala-Pro-Trp- Thr-Gly-Gly-Phe-Phe-Leu-Leu-Thr- Arg- Ile-Leu-Thr- Ile-Pro-Gln-Ser- Leu-Asp (SEQ ID No : 32 ) EXAMPLE 5
CLINICAL TRIAL TO DEMONSTRATE THERAPEUTIC EFFICACY OF COCKTAIL
Individual constructs carrying Th peptide sequences from measles virus F and hepatitis B surface antigen are promiscuous for multiple human HLA DR antigens, so as to provide maximum immunogenicity in a genetically diverse human population. Moreover, because these Th peptides are derived from children's vaccines, childhood vaccinations are a potential source of Th memory in an immunized human population. Thus, children's vaccines have the potential to afford enhanced immunopotency to anti- allergy vaccines comprised of mixtures of such Th peptides. The clinical protocol below is designed to demonstrate efficacy for compositions of the invention formulated as a mixture of such linear "Th-Spacer- (mlgE Peptide) " and " (mlgE Peptide) -spacer-Th" peptide constructs, in a widely acceptable adjuvant, Alum.
Experimental Design: Subjects: Hay fever patients
Season & Duration: Hay fever seasons, 8 weeks
Groups: 4 groups, 1 group/im unogen/dose N=15 per group,
12 receive immunogen, 3 receive placebo
Immunogen: Cocktail 1; Peptide Nos. 1-4, 7-10 Adjuvant: 0.2% Alum
Dose: Molar equivalents of each synthetic peptide to equal 500 μg total or 62.5 μg of each peptide per dose
Route: intramuscular Dose Schedule: week 0, and 4 weeks Evaluation schedule: weeks 0, 4, and 6 Blood is collected, processed into serum, and stored prior to titering by ELISA as described in Example 2.
Efficacy and safety of the clinical composition are evaluated by comparisons of serological tests, skin test reaction, by recording patient usage of hay fever medication, by physical examination for allergic symptoms and adverse reactions, and by interviews to obtain subjective patient assessments of the product. Serological evaluations include the aforementioned ELISAs for antipeptide titers, and a standard automated spectrofluorimetric assay to determine reduction in histamine levels03' as well as to ascertain that the products do not trigger histamine release. The skin test is an intradermal test in which a standardized solution of allergens is injected into the upper layers of the skin. Reactions to the allergens are quantitated in the skin test by determining the area of the typical "wheal and flare" produced in response to the allergens. The expected results include significant improvement in allergic symptoms at the endpoint of the study, and no evidence of histamine release. This experiment demonstrates the clinical efficacy and safety of a pharmaceutically acceptable composition of the invention.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: United Biomedical Inc.;
Walfield, Alan M. ; Wang, Chang Yi (ii) TITLE OF INVENTION: Synthetic IgE Membrane Anchor Peptide Immunogens for the Treatment of Allergy
(iii) NUMBER OF SEQUENCES: 32
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Maria C.H. Lin (B) STREET: 345 Park Avenue
(C) CITY: New York
(D) STATE: NY
(E) COUNTRY: USA
(F) ZIP: 10154
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: WordPerfect 5.1
(vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 08/328,519
(B) FILING DATE: 25-OCT-1994 (C) CLASSIFICATION:
(vii) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: TO BE ASSIGNED
(B) FILING DATE: 25-OCT-1995
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION: (A) NAME: Lin, Maria C.H.
(B) REGISTRATION NUMBER: 29,323
(C) REFERENCE/DOCKET NUMBER: 1151-4117
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 212-758-4800
(B) TELEFAX: 212-751-6849
(C) TELEX: 421792
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 26 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:
Gly Leu Ala Gly Gly Ser Ala Gin Ser Gin Arg Ala Pro 1 5 10
Asp Arg Val Leu Cys His Ser Gly Gin Gin Gin Gly Leu 15 20 25
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
10 (A) LENGTH: 17 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
15
Pro Glu Leu Asp Val Cys Val Glu Glu Ala Glu Gly Glu 1 5 10
Ala Pro Trp Thr 15
20 (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
25 (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
Thr Ala Lys Ser Lys Lys Phe Pro Ser Tyr Thr Ala Thr 1 5 10
Tyr Gin Phe 30 15
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 6 amino acids -- (B) TYPE: amino acid
JD (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4
Pro Pro Xaa Pro Xaa Pro
1 5
(2) INFORMATION FOR SEQ ID NO:5 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:
Phe Phe Leu Leu Thr Arg lie Leu Thr lie Pro Gin 1 5 10
Ser Leu Asp 15
(2) INFORMATION FOR SEQ ID NO:6 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6
Lys Lys Leu Arg Arg Leu Leu Tyr Met lie Tyr Met Ser 1 5 10
Gly Leu Ala Val Arg Val His Val Ser Lys Glu Glu Gin 15 20 25
Tyr Tyr Asp Tyr 30
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 17 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear o
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
Lys Lys Gin Tyr lie Lys Ala Asn Ser Lys Phe lie Gly 1 5 10 lie Thr Glu Leu 15
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 22 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:
Lys Lys Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg 1 5 10
Val Pro Lys Val Ser Ala Ser His Leu 15 20
(2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9:
Tyr Met Ser Gly Leu Ala Val Arg Val His Val Ser Lys 1 5 10
Glu Glu 15
(2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:
Tyr Asp Pro Asn Tyr Leu Arg Thr Asp Ser Asp Lys Asp 1 5 10
Arg Phe Leu Gin Thr Met Val Lys Leu Phe Asn Arg lie 15 20 25
Lys
(2) INFORMATION FOR SEQ ID NO:11:
10
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
15
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:
Gly Ala Tyr Ala Arg Cys Pro Asn Gly Thr Arg Ala Leu 1 5 10
Thr Val Ala Glu Leu Arg Gly Asn Ala Glu Leu 15 20
20
(2) INFORMATION FOR SEQ ID NO:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 amino acids
(B) TYPE: amino acid
25 (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:
Leu Ser Glu lie Lys Gly Val lie Val His Arg Leu Glu 1 5 10
30
Gly Val 15
(2) INFORMATION FOR SEQ ID NO:13:
^ (i) SEQUENCE CHARACTERISTICS:
" (A) LENGTH: 21 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
Val Ser Phe Gly Val Trp lie Arg Thr Pro Pro Ala Tyr 1 5 10
Arg Pro Pro Asn Ala Pro lie Leu 15 20
(2) INFORMATION FOR SEQ ID NO:14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:
Gly lie Leu Glu Ser Arg Gly lie Lys Ala Arg lie Thr 1 5 10
His Val Asp Thr Glu Ser Tyr 15 20
(2) INFORMATION FOR SEQ ID NO: 15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 17 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15:
Trp Val Arg Asp lie lie Asp Asp Phe Thr Asn Glu Ser 1 5 10
Ser Gin Lys Thr 15
(2) INFORMATION FOR SEQ ID NO: 16: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 16 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:
Asp Val Ser Thr lie Val Pro Tyr lie Gly Pro Ala Leu 1 5 10
Asn He Val 15
(2) INFORMATION FOR SEQ ID NO:17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 25 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
Ala Leu Asn He Trp Asp Arg Phe Asp Val Phe Cys Thr
1 5 10 Leu Gly Ala Thr Thr Gly Tyr Leu Lys Gly Asn Ser 15 20 25
(2) INFORMATION FOR SEQ ID NO: 18:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 23 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:18: Asp Ser Glu Thr Ala Asp Asn Leu Glu Lys Thr Val Ala 1 5 10
Ala Leu Ser He Leu Pro Gly He Gly Cys 15 20
(2) INFORMATION FOR SEQ ID NO: 19 (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 39 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:19:
Glu Glu He Val Ala Gin Ser He Ala Leu Ser Ser Leu 1 5 10
Met Val Ala Gin Ala He Pro Leu Val Gly Glu Leu Val 15 20 25
Asp He Gly Phe Ala Ala Thr Asn Phe Val Glu Ser Cys 30 35
(2) INFORMATION FOR SEQ ID NO:20: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20:
Asp He Glu Lys Lys He Ala Lys Met Glu Lys Ala Ser 1 5 10
Ser Val Phe Asn Val Val Asn Ser 15 20
(2) INFORMATION FOR SEQ ID NO:21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 17 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21:
Lys Trp Phe Lys Thr Asn Ala Pro Asn Gly Val Asp Glu 1 5 10
Lys He Arg He 15 (2) INFORMATION FOR SEQ ID NO:22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:
Gly Leu Gin Gly Lys His Ala Asp Ala Val Lys Ala Lys
1 5 10
Gly
(2) INFORMATION FOR SEQ ID NO:23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23:
Gly Leu Ala Ala Gly Leu Val Gly Met Ala Ala Asp Ala i 5 10
Met Val Glu Asp Val Asn
12
(2) INFORMATION FOR SEQ ID NO:24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:
Ser Thr Glu Thr Gly Asn Gin His His Tyr Gin Thr Arg 1 5 10
Val Val Ser Asn Ala Asn Lys 15 20 (2) INFORMATION FOR SEQ ID NO:25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:
Leu Ser Glu He Lys Gly Val He Val His Arg Leu Glu
1 5 10
Gly Val Gly Gly Leu Ala Gly Gly Ser Ala Gin Ser Gin 15 20 25
Arg Ala Pro Asp Arg Val Leu Cys His Ser Gly Gin Gin 30 35
Gin Gly Leu 40
(2) INFORMATION FOR SEQ ID NO:26
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 43 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:26:
Gly Leu Ala Gly Gly Ser Ala Gin Ser Gin Arg Ala Pro
1 5 10
Asp Arg Val Leu Cys His Ser Gly Gin Gin Gin Gly Leu 15 20 25
Gly Gly Leu Ser Glu He Lys Gly Val He Val His Arg 30 35
Leu Glu Gly Val 40
(2) INFORMATION FOR SEQ ID NO:27:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 34 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:27:
Leu Ser Glu He Lys Gly Val He Val His Arg Leu Glu 1 5 10
Gly Val Gly Gly Pro Glu Leu Asp Val Cys Val Glu Glu 15 20 25
Ala Glu Gly Glu Ala Pro Trp Thr 30
(2) INFORMATION FOR SEQ ID NO:28:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:28:
Pro Gly Leu Asp Val Cys Val Glu Glu Ala Glu Gly Glu 1 5 10
Ala Pro Trp Thr Gly Gly Leu Ser Glu He Lys Gly Val 15 20 25
He Val His Arg Leu Glu Gly Val 30
[ 2 ] INFORMATION FOR SEQ ID NO:29:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:29:
Phe Phe Leu Leu Thr Arg He Leu Thr He Pro Gin Ser 1 5 10
Leu Asp Gly Gly Leu Ala Gly Gly Ser Ala Gin Ser Gin 15 20 25 Arg Ala Pro Asp Arg Val Leu Cys His Ser Gly Gin Gin 30 35
Gin Gly Leu 40
(2) INFORMATION FOR SEQ ID NO:30:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 43 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:30:
Gly Leu Ala Gly Gly Ser Ala Gin Ser Gin Arg Ala Pro 1 5 10 Asp Arg Val Leu Cys His Ser Gly Gin Gin Gin Gly Leu 15 20 25
Gly Gly Phe Phe Leu Leu Thr Arg He Leu Thr He Pro 30 35
Gin Ser Leu Asp 40
(2) INFORMATION FOR SEQ ID NO: 31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:31
Phe Phe Leu Leu Thr Arg He Leu Thr He Pro Gin Ser
1 5 10
Leu Asp Gly Gly Pro Glu Leu Asp Val Cys Val Glu Glu
15 20 25
Ala Glu Gly Glu Ala Pro Trp Thr 30 (2) INFORMATION FOR SEQ ID NO:32:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:32:
Pro Glu Leu Asp Val Cys Val Glu Glu Ala Glu Gly Glu 1 5 10
Ala Pro Trp Thr Gly Gly Phe Phe Leu Leu Thr Arg He 15 20 25
Leu Thr He Pro Gin Ser Leu Asp 30

Claims

o We claim:
1. A peptide immunogen selected from the group consisting of:
(A)n-(Th)m-(B)0-(mIgE peptide) or (mlgE peptide)- (B)0-(Th)m- (A)n wherein: A is an amino acid, -NH2, a fatty acid or a derivative thereof, or an invasin domain having immmune stimulatory property; B is an amino acid;
Th is a helper T cell epitope, or an immune enhancing analog or segment thereof; mlgE peptide is SEQ ID N0:1, SEQ ID NO:2, or an immunogenic analog thereof; n is from 1 to about 10; m is from 1 to about 4; and o is from 0 to about 10.
2. The peptide immunogen of Claim 1 wherein said Th has an amino acid sequence selected from the group of sequences selected from SEQ ID NOS:5-24 or an immunogenic analog or segment thereof.
3. The peptide immunogen of Claim 1 having an amino acid sequence selected from the group consisting of: SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31 and SEQ ID NO:32.
4. The peptide immunogen of Claim 2 further comprising a fatty acid.
5. The peptide immunogen of Claim 2 further comprising a fatty acid derivative. o
6. The peptide immunogen of Claim 5 wherein the fatty acid derivative is Pam3Cys.
7. The peptide immunogen of Claim 3 further comprising a fatty acid.
8. The peptide immunogen of Claim 3 further comprising a fatty acid derivative.
9. The peptide immunogen of Claim 8 wherein the fatty acid derivative is Pam3Cys.
10. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 1 and a pharmaceutically acceptable delivery system.
11. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 2 and a pharmaceutically acceptable delivery system.
12. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 3 and a pharmaceutically acceptable delivery system.
13. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 4 and a pharmaceutically acceptable delivery system.
14. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 5 and a pharmaceutically acceptable delivery system.
15. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 6 and a pharmaceutically acceptable delivery system. o
16. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 7 and a pharmaceutically acceptable delivery system.
17. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 8 and a pharmaceutically acceptable delivery system.
18. A vaccine composition comprising an effective amount of a peptide immunogen of Claim 9 and a pharmaceutically acceptable delivery system.
19. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 1.
20. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 2.
21. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 3.
22. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 4.
23. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 5.
24. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 6. o
25. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 7.
26. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 8.
27. A method of treating allergic reactions by administering to a patient an effective amount of a composition according to Claim 9.
PCT/US1995/013841 1994-10-25 1995-10-25 SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY WO1996012740A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP95938912A EP0787150A1 (en) 1994-10-25 1995-10-25 SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY
AU40120/95A AU4012095A (en) 1994-10-25 1995-10-25 Synthetic ige membrane anchor peptide immunogens for the treatment of allergy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32851994A 1994-10-25 1994-10-25
US08/328,519 1994-10-25

Publications (1)

Publication Number Publication Date
WO1996012740A1 true WO1996012740A1 (en) 1996-05-02

Family

ID=23281314

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/013841 WO1996012740A1 (en) 1994-10-25 1995-10-25 SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY

Country Status (5)

Country Link
EP (1) EP0787150A1 (en)
CN (1) CN1167491A (en)
AU (1) AU4012095A (en)
CA (1) CA2203165A1 (en)
WO (1) WO1996012740A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6610297B1 (en) 1996-03-01 2003-08-26 Novartis Ag Peptide immunogens for vaccination against and treatment of allergy
US6913749B2 (en) 1998-11-02 2005-07-05 Resistentia Pharmaceuticals Ab Immunogenic polypeptides for inducing anti-self IgE responses
US20060052592A1 (en) * 2002-06-20 2006-03-09 Levinson Arnold I Vaccines for suppressing ige-mediated allergic disease and methods for using the same
US7265208B2 (en) 2001-05-01 2007-09-04 The Regents Of The University Of California Fusion molecules and treatment of IgE-mediated allergic diseases
EP2021027A2 (en) * 2006-05-03 2009-02-11 Guthrie Foundation For Education&Research Immunoglobulin associated cell-surface determinants in the treatment of b-cell disorders
WO2007041171A3 (en) * 2005-09-29 2009-04-09 Medimmune Inc Method of identifying membrane lg specific antibodies and use thereof for targeting immunoglobulin-producing precursor cells
US7604955B2 (en) 2001-08-13 2009-10-20 Swey-Shen Alex Chen Immunoglobulin E vaccines and methods of use thereof
WO2011012240A2 (en) 2009-07-25 2011-02-03 Emc Microcollections Gmbh Lipopeptides for therapy and prophylaxis of allergic illnesses
US20120149881A1 (en) * 2007-03-22 2012-06-14 Genentech, Inc. Apoptotic anti-ige antibodies
US8460664B2 (en) 2009-02-25 2013-06-11 Academia Sinica Anti-CεmX antibodies capable of binding to human mIgE on B lymphocytes
US20150118256A1 (en) * 2010-06-07 2015-04-30 Pfizer Vaccines Llc IGE CH3 Peptide Vaccine
US20150203550A1 (en) * 2012-06-18 2015-07-23 Nippon Zenyaku Kogyo Co., Ltd. IgE PEPTIDE VACCINE
WO2017005851A1 (en) 2015-07-07 2017-01-12 Affiris Ag Vaccines for the treatment and prevention of ige mediated diseases
US9587034B2 (en) 2012-04-20 2017-03-07 Academia Sinica Anti-mIgE antibodies that bind to the junction between CH4 and CεmX domains
WO2019133024A1 (en) * 2017-12-31 2019-07-04 United Biomedical, Inc. Peptide immunogens and formulations thereof targeting membrane-bound ige for treatment of ige mediated allergic diseases
US11439682B2 (en) 2017-10-31 2022-09-13 Oneness Biotech Co., Ltd. Treating IgE-mediated allergic diseases

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102004035337A1 (en) * 2004-07-21 2006-03-16 Merck Patent Gmbh Variations of the group 1 allergens from Poaceae with reduced allergenicity and preserved T cell reactivity
BRPI0920124A2 (en) * 2008-10-15 2015-12-22 Alk Abelló As pharmaceutical product to increase allergy vaccine dose

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4722840A (en) * 1984-09-12 1988-02-02 Chiron Corporation Hybrid particle immunogens
US5231026A (en) * 1987-12-31 1993-07-27 Tanox Biosystems, Inc. DNA encoding murine-human chimeric antibodies specific for antigenic epitopes of IgE present on the extracellular segment of the membrane domain of membrane-bound IgE
US5260416A (en) * 1987-12-31 1993-11-09 Tanox Biosystems, Inc. Antigenic epitopes present on membrane-bound but not secreted IgE
US5274075A (en) * 1987-12-31 1993-12-28 Tanox Biosystems, Inc. Newly identified human epsilon immunoglobulin peptides and related products
US5281699A (en) * 1990-06-01 1994-01-25 Tanox Biosystems, Inc. Treating B cell lymphoma or leukemia by targeting specific epitopes on B cell bound immunoglobulins
US5342924A (en) * 1987-12-31 1994-08-30 Tanox Biosystems, Inc. Extracellular segments of human ε immunoglobulin anchoring peptides and antibodies specific therefor

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4722840A (en) * 1984-09-12 1988-02-02 Chiron Corporation Hybrid particle immunogens
US5231026A (en) * 1987-12-31 1993-07-27 Tanox Biosystems, Inc. DNA encoding murine-human chimeric antibodies specific for antigenic epitopes of IgE present on the extracellular segment of the membrane domain of membrane-bound IgE
US5260416A (en) * 1987-12-31 1993-11-09 Tanox Biosystems, Inc. Antigenic epitopes present on membrane-bound but not secreted IgE
US5274075A (en) * 1987-12-31 1993-12-28 Tanox Biosystems, Inc. Newly identified human epsilon immunoglobulin peptides and related products
US5342924A (en) * 1987-12-31 1994-08-30 Tanox Biosystems, Inc. Extracellular segments of human ε immunoglobulin anchoring peptides and antibodies specific therefor
US5281699A (en) * 1990-06-01 1994-01-25 Tanox Biosystems, Inc. Treating B cell lymphoma or leukemia by targeting specific epitopes on B cell bound immunoglobulins

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
EUROPEAN J. IMMUNOLOGY, Volume 17, issued 1987, LECLERC et al., "A Synthetic Vaccine Constructed by Copolymerization of Band T Cell Determinants", pages 269-273. *
MOLECULAR IMMUNOLOGY, Volume 23, Number 1, issued 1986, STANWORTH et al., "Anti -e- Chain Antibodies as Probes in the Study of Mast-Cell Triggering", pages 1231-1235. *
THE JOURNAL OF IMMUNOLOGY, Volume 148, Number 1, issued 01 January 1992, PENG et al., "A New Isoform of Human Membrane Bound IgE", pages 129-136. *
THE LANCET, Volume 336, issued 1990, STANWORTH et al., "Allergy Treatment with a Peptide Vaccine", pages 1279-1281. *

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6610297B1 (en) 1996-03-01 2003-08-26 Novartis Ag Peptide immunogens for vaccination against and treatment of allergy
US6913749B2 (en) 1998-11-02 2005-07-05 Resistentia Pharmaceuticals Ab Immunogenic polypeptides for inducing anti-self IgE responses
US7459158B2 (en) 1998-11-02 2008-12-02 Resistentia Pharmaceuticals Ab Immunogenic polypeptides for inducing anti-self IgE responses
US7879334B1 (en) 2001-05-01 2011-02-01 The Regents Of The University Of California Fusion molecules and treatment of IgE-mediated allergic diseases
US7265208B2 (en) 2001-05-01 2007-09-04 The Regents Of The University Of California Fusion molecules and treatment of IgE-mediated allergic diseases
US7534440B2 (en) 2001-05-01 2009-05-19 The Regents Of The University Of California Fusion molecules and methods for treatment of immune diseases
US7879324B2 (en) 2001-05-01 2011-02-01 The Regents Of The University Of California Fusion molecules and methods for treatment of immune diseases
US7604955B2 (en) 2001-08-13 2009-10-20 Swey-Shen Alex Chen Immunoglobulin E vaccines and methods of use thereof
US20060052592A1 (en) * 2002-06-20 2006-03-09 Levinson Arnold I Vaccines for suppressing ige-mediated allergic disease and methods for using the same
US20140127244A1 (en) * 2002-06-20 2014-05-08 Arnold I. Levinson Vaccines for suppressing ige-mediated allergic disease and methods for using the same
US9408897B2 (en) 2002-06-20 2016-08-09 The Trustees Of The University Of Pennsylvania Vaccines for suppressing IgE-mediated allergic disease and methods for using the same
AU2006297304B2 (en) * 2005-09-29 2012-05-17 Medimmune, Llc Method of identifying membrane LG specific antibodies and use thereof for targeting immunoglobulin-producing precursor cells
WO2007041171A3 (en) * 2005-09-29 2009-04-09 Medimmune Inc Method of identifying membrane lg specific antibodies and use thereof for targeting immunoglobulin-producing precursor cells
US8137670B2 (en) 2005-09-29 2012-03-20 Medimmune, Llc Method of identifying membrane IgE specific antibodies and use thereof for targeting IgE producing precursor cells
US8404236B2 (en) 2005-09-29 2013-03-26 Medimmune, Llc Method of identifying membrane Ig specific antibodies and use thereof for targeting immunoglobulin-producing precursor cells
EP2021027A4 (en) * 2006-05-03 2010-01-27 Guthrie Foundation For Educati Immunoglobulin associated cell-surface determinants in the treatment of b-cell disorders
EP2021027A2 (en) * 2006-05-03 2009-02-11 Guthrie Foundation For Education&Research Immunoglobulin associated cell-surface determinants in the treatment of b-cell disorders
US8632775B2 (en) * 2007-03-22 2014-01-21 Genentech, Inc. Apoptotic anti-IgE antibodies
US20120149881A1 (en) * 2007-03-22 2012-06-14 Genentech, Inc. Apoptotic anti-ige antibodies
US8586040B2 (en) 2007-03-22 2013-11-19 Genentech, Inc. Methods of using apoptotic anti-IGE antibodies
US8618274B2 (en) 2007-03-22 2013-12-31 Genentech, Inc. Nucleic acid encoding apoptotic anti-IgE antibodies
US8460664B2 (en) 2009-02-25 2013-06-11 Academia Sinica Anti-CεmX antibodies capable of binding to human mIgE on B lymphocytes
US8741294B2 (en) 2009-02-25 2014-06-03 Academia Sinica Anti-CεmX antibodies capable of binding to human mIgE on B lymphocytes
US20140220042A1 (en) * 2009-02-25 2014-08-07 Academia Sinica Anti-c (epsilon)mx antibodies capable of binding to human mige on b lymphocytes
US8974794B2 (en) 2009-02-25 2015-03-10 Academia Sinica C(epsilon)mX peptides for inducing immune responses to human mIgE on B lymphocytes
WO2011012240A2 (en) 2009-07-25 2011-02-03 Emc Microcollections Gmbh Lipopeptides for therapy and prophylaxis of allergic illnesses
DE102009034779A1 (en) 2009-07-25 2011-02-03 Emc Microcollections Gmbh Synthetic analogues of bacterial lipopeptides and their application for the therapy and prophylaxis of allergic diseases
US20150118256A1 (en) * 2010-06-07 2015-04-30 Pfizer Vaccines Llc IGE CH3 Peptide Vaccine
US9249233B2 (en) * 2010-06-07 2016-02-02 Pfizer Vaccines Llc IgE CH3 peptide vaccine
US9587034B2 (en) 2012-04-20 2017-03-07 Academia Sinica Anti-mIgE antibodies that bind to the junction between CH4 and CεmX domains
US9657071B2 (en) * 2012-06-18 2017-05-23 Nippon Zenyaku Kogyo Co., Ltd. IgE peptide vaccine
US20150203550A1 (en) * 2012-06-18 2015-07-23 Nippon Zenyaku Kogyo Co., Ltd. IgE PEPTIDE VACCINE
WO2017005851A1 (en) 2015-07-07 2017-01-12 Affiris Ag Vaccines for the treatment and prevention of ige mediated diseases
US11439682B2 (en) 2017-10-31 2022-09-13 Oneness Biotech Co., Ltd. Treating IgE-mediated allergic diseases
WO2019133024A1 (en) * 2017-12-31 2019-07-04 United Biomedical, Inc. Peptide immunogens and formulations thereof targeting membrane-bound ige for treatment of ige mediated allergic diseases
JP2021515744A (en) * 2017-12-31 2021-06-24 ユナイテッド・バイオメディカル・インコーポレーテッドUnited Biomedical Incorporated Peptide immunogens targeting membrane-bound IgE and their preparations for the treatment of IgE-mediated allergic diseases
RU2769983C2 (en) * 2017-12-31 2022-04-12 Юнайтед Байомедикал, Инк. Peptide immunogens and compositions containing said immunogens targeting membrane-bound ige for treating ige-mediated allergic diseases
TWI804553B (en) * 2017-12-31 2023-06-11 美商聯合生物醫學公司 Peptide immunogens and formulations thereof targeting membrane-bound ige for treatment of ige mediated allergic diseases
US20230270848A1 (en) * 2017-12-31 2023-08-31 United Biomedical, Inc. PEPTIDE IMMUNOGENS AND FORMULATIONS THEREOF TARGETING MEMBRANE-BOUND IgE FOR TREATMENT OF IgE MEDIATED ALLERGIC DISEASES

Also Published As

Publication number Publication date
AU4012095A (en) 1996-05-15
CA2203165A1 (en) 1996-05-02
CN1167491A (en) 1997-12-10
EP0787150A1 (en) 1997-08-06

Similar Documents

Publication Publication Date Title
AU757919B2 (en) Peptide composition as immunogen for the treatment of allergy
WO1996012740A1 (en) SYNTHETIC IgE MEMBRANE ANCHOR PEPTIDE IMMUNOGENS FOR THE TREATMENT OF ALLERGY
CA2329772C (en) Artificial t helper cell epitopes as immune stimulators for synthetic peptide immunogens
AU2195395A (en) Synthetic peptide based immunogens for the treatment of allergy
CA1329766C (en) T cell epitopes of the hepatitis b virus nucleocapsid protein
AU687805B2 (en) Immunogenic LHRH peptide constructs and synthetic universal immune stimulators for vaccines
JP2003529319A (en) Methods of eliciting broadly neutralizing antibodies targeting HIV-1 gp41
GB2251186A (en) Polypeptide for use in treatment of autoimmune disease
WO2002051860A2 (en) Synthetic peptide composition as immunogens for prevention of urinary tract infection
IE911527A1 (en) Improved vaccine compositions
JP2004520817A (en) vaccine
JPH02502637A (en) Synthetic anti-worm vaccine
IE911614A1 (en) Epitopes of the pre-s region of hepatitis b virus surface¹antigen
MXPA00011938A (en) Peptide composition as immunogen for the treatment of allergy
Robinson The immune responses of mice to rubella virus synthetic peptides
MXPA00011939A (en) Artificial t helper cell epitopes as immune stimulators for synthetic peptideimmunogens
MXPA00011752A (en) Synthetic somatostatin immunogen for growth promotion in farm animals

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 95196496.8

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA CN JP US

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2203165

Country of ref document: CA

Ref document number: 2203165

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1995938912

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995938912

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995938912

Country of ref document: EP