WO2006121656A2 - Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease - Google Patents

Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease Download PDF

Info

Publication number
WO2006121656A2
WO2006121656A2 PCT/US2006/016481 US2006016481W WO2006121656A2 WO 2006121656 A2 WO2006121656 A2 WO 2006121656A2 US 2006016481 W US2006016481 W US 2006016481W WO 2006121656 A2 WO2006121656 A2 WO 2006121656A2
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
ompc
immunogenic fragment
conjugate
adjuvant
Prior art date
Application number
PCT/US2006/016481
Other languages
French (fr)
Other versions
WO2006121656A3 (en
Inventor
Victor M. Garsky
Joseph G Joyce
Paul M. Keller
Gene Kinney
Xiaoping Liang
John W. Shiver
Original Assignee
Merck & Co., 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
Priority to US11/919,897 priority Critical patent/US7850973B2/en
Priority to BRPI0610093-7A priority patent/BRPI0610093A2/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to CN2006800153890A priority patent/CN101171031B/en
Priority to MX2007013825A priority patent/MX2007013825A/en
Priority to NZ562434A priority patent/NZ562434A/en
Priority to JP2008510087A priority patent/JP2008540417A/en
Priority to EP06751930A priority patent/EP1879613B1/en
Priority to AT06751930T priority patent/ATE535252T1/en
Priority to AU2006246382A priority patent/AU2006246382A1/en
Priority to CA002607868A priority patent/CA2607868A1/en
Publication of WO2006121656A2 publication Critical patent/WO2006121656A2/en
Publication of WO2006121656A3 publication Critical patent/WO2006121656A3/en
Priority to ZA2007/08635A priority patent/ZA200708635B/en
Priority to IL186793A priority patent/IL186793A0/en
Priority to NO20076239A priority patent/NO20076239L/en
Priority to US12/907,493 priority patent/US20110052611A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0007Nervous system antigens; Prions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6068Other bacterial proteins, e.g. OMP
    • 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
    • A61K2039/6087Polysaccharides; Lipopolysaccharides [LPS]
    • 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
    • A61K2039/6093Synthetic polymers, e.g. polyethyleneglycol [PEG], Polymers or copolymers of (D) glutamate and (D) lysine
    • 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

Definitions

  • the present invention relates to compositions and methods for the prevention and treatment of amyloidogenic diseases and, in particular, Alzheimer's disease.
  • Alzheimer's disease is characterized by progressive memory impairment and cognitive decline. Its hallmark pathological lesions are amyloid deposits (senile plaques), neurofibrillary tangles and neuronal loss in specific brain regions.
  • the amyloid deposits are composed of amyloid beta peptides (A ⁇ ) of 40 to 43 amino acid residues, which are the proteolytic products of the amyloid precursor protein (APP).
  • Neurofibrillary tangles are the intracellular filamentous aggregates of hyperphosphorylated tau proteins (Selkoe, Science, 275: 630-631, 1997).
  • AD Alzheimer's disease
  • a ⁇ is believed to play a pivotal role in the disease process, also know as the amyloid cascade hypothesis (Golde, Brain Pathol., 15: 84-87, 1995).
  • a ⁇ is prone to form various forms of aggregates, ranging from small oligomers to large, elongated profibrile structures.
  • These aggregates are neurotoxic and are responsible for the synaptic pathology associated with the memory loss and cognition decline in the early stage of the disease (Klein et al, Neurobiol. Aging, 25: 569-580, 2004).
  • Betabloc showed regional clearance of senile plaques (Nicoll et al., Nature Med., 9: 448-452, 2003; Ferrer et al, Brain Pathol.. 14: 11-20, 2004; Masliah et al, Neurology, 64: 129-131, 2005).
  • This data collectively indicates that vaccines that effectively elicit antibody responses to A ⁇ antigens are efficacious against the pathological senile plaques found in AD.
  • the mechanism of vaccine or antibody efficacy remains to be defined.
  • peripheral sink refers to a change in the equilibrium of brain and plasma A ⁇ stores resulting in a net efflux of central A ⁇ to the periphery (see, for example, Deane et al., J. Neurosci., 25: 11495-11503, 2005; DeMattos et al, Pro. Natl. Acad. Sci. USA. 98: 8931-8932, 2001).
  • mice expressing the wild-type human A ⁇ sequence plus the Swedish mutation responded to active immunization with both increases in plasma A ⁇ and subsequent decreases in central A ⁇ (Cribbs et al., 7th International Conference on AD/PD, Sorrento, Italy, 2005). Accordingly, it is expected that any active vaccine immunogen capable of generating an immune response that results in the elevation of plasma A ⁇ levels will be useful for the treatment of Alzheimer's disease and related disorders characterized by elevated brain A ⁇ levels.
  • Applicants herein have surprisingly found that an antigen which eliminated T-cell epitopes, to avoid a self T-cell response, is immunogenic and elevates plasma A ⁇ levels. This represents a potential means to produce a safe and effective AD vaccine. Applicants herein provide such an antigen and a formulation for use as an AD vaccine.
  • the invention provides a pharmaceutical composition comprising an immunogenic fragment of A ⁇ , lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to A ⁇ .
  • this composition comprises linear 8 amino acid peptides (8- mers) of A ⁇ .
  • this composition comprises multivalent linear 8-mers interspersed with at least one spacer or a multivalent branched multiple antigenic peptide (MAP).
  • MAP multivalent branched multiple antigenic peptide
  • the pharmaceutical composition can be used as a vaccine for AD and related amyloid diseases.
  • the pharmaceutical composition is an A ⁇ plasma elevating agent comprising an immunogenic fragment of A ⁇ , lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to A ⁇ that elevate plasma A ⁇ levels.
  • the pharmaceutical composition can be used as a vaccine for AD and related amyloid diseases characterized by elevated brain A ⁇ levels.
  • the pharmaceutical composition is linked to a carrier molecule to form a conjugate, wherein the carrier helps to elicit an immune response comprising antibodies to the A ⁇ fragment.
  • the carrier is the outer membrane protein complex of Neisseria meningitides (OMPC).
  • the pharmaceutical composition is administered with a pharmaceutically acceptable adjuvant.
  • the adjuvant is an
  • the invention provides methods for preventing or treating a disease associated with amyloid deposits of A ⁇ in the brain of a patient.
  • diseases include Alzheimer's disease, Down's syndrome, cognitive impairment or other forms of senile dementia.
  • the method comprises administering an immunogenic fragment of A ⁇ , lacking a T-cell epitope, selected from the group consisting of linear 8 amino acid peptides (8-mers), a multivalent linear peptides interspersed with at least one spacer and a multivalent branched multiple antigenic peptide (MAP).
  • the immunogenic fragment comprises a multivalent linear peptide with a polyethylene glycol (PEG) spacer.
  • the immunogenic fragment comprises a multivalent branched MAP, A ⁇ (3-10)/(21-28) conjugate, Construct No. 12, Figure 6A, conjugated to OMPC.
  • Such methods entail the administration of an effective dose of an immunogenic fragment of A ⁇ , lacking a T-cell epitope, to patients in need of such treatment that will induce an immune response in the form of antibodies to A ⁇ . Said antibody response is capable of elevating plasma A ⁇ levels.
  • the immunogenic fragment to be administered is linked to a carrier molecule.
  • the immunogenic fragment is administered with an adjuvant.
  • Figure 1 represents synthetic 8-amino acid peptides (8-mers) (SEQ ID NOS: 2-36) derived from A ⁇ (1-42) (SEQ ID NO: 1) from which peptides were selected to conduct a linear peptide scan to identify the epitopes of A ⁇ .
  • Figure 2 represents the 8-mers selected for conjugation to KLH ( Figure 2A) and OMPC ( Figure 2B).
  • Figure 3 represents the immunogenicity of selected A ⁇ conjugates, described in Figure 2, after the first (PDl), second (PD2) and third dose (PD3).
  • Figure 4 represents the cross-reactivity of sera extracted from a guinea pig previously immunized with an A ⁇ (3-1O)-KLH conjugate (SEQ ID NO: 40) on human AD brain tissue.
  • Figure 4A shows immunoreactivity of the anti-A ⁇ monoclonal antibody 6F3D (which recognizes amino acids 8-17 of A ⁇ ). The staining pattern reveals extensive amyloid pathology in this human brain.
  • Figure 4B demonstrates a lack of immunoreactivity of this same brain to the pre-immune sera from the immunized guinea pig prior to immunization.
  • Figure 4C shows the immunoreactivity of the sera from an immunized guinea pig following immunization
  • Figure 5 shows representative multivalent linear 8-mer peptides, which were selected based on the immunogenicity of the separate 8-mers in guinea pig studies (Example 3). These conjugates were synthesized as described and conjugated to OMPC (Example IJ and 1.K).
  • Figure 6 shows representative multivalent branched MAP conjugates, which were selected based on the immunogenicity of the separate 8-mers in guinea pig studies (Example 3).
  • Figure 6A shows representative divalent MAPs and
  • Figure 6B shows representative bromoacetyl-cysteine MAPs. These conjugates were synthesized as described and conjugated to OMPC (Example 2).
  • Figure 7 represents the anti-A ⁇ 4o titer from sera collected from rhesus monkeys following 1 (PDl) or 2 (PD2) injections with an A ⁇ (1-18) peptide conjugated to OMPC formulated in Merck alum alone or Merck alum plus MX (ISCOMATRIX , CSL, Ltd., Parkville, Australia) as an adjuvant.
  • Figure 8 represents the increase in plasma A ⁇ levels following administration of a A ⁇ conjugate.
  • Figure 8A shows a greater than three-fold elevation following administration of a MAP construct comprising A ⁇ (3-10)/(21-28) (Construct No. 12, Figure 6A) conjugated to OMPC versus the monomeric constructs, A ⁇ (3-10) (SEQ ID NO: 69) and A ⁇ (21-28) (SEQ ID NO: 73) (o , Construct No. 12, Figure 6A; •, A ⁇ (3-10) (SEQ ID NO 69), A, A ⁇ (21-28) (SEQ ID NO: 73).
  • Figure 8B shows that plasma A ⁇ levels are independent of titer levels ( ⁇ , Construct No. 12, Figure 6A; •, A ⁇ (3-10) (SEQ ID NO 69), ⁇ , A ⁇ (21-28) (SEQ ID NO: 73).
  • 8-mer refers to an eight amino acid peptide which corresponds to a fragment of A ⁇ , an analog of a natural A ⁇ peptide or a peptide mimetic.
  • One or more 8-mers may be combined with at least one spacer to form a multivalent linear peptide or to form a multivalent branched MAP.
  • a ⁇ conjugate means an 8-mer or immunogenic fragment of A ⁇ that is chemically or biologically linked to a carrier, such as keyhole limpet hemocyanin or the outer membrane protein complex of Nesseria rneningitidies (OMPC).
  • a carrier such as keyhole limpet hemocyanin or the outer membrane protein complex of Nesseria rneningitidies (OMPC).
  • a ⁇ peptide means any of the A ⁇ peptides described herein, including, but not limited to, linear 8-mers, multivalent linear peptides with at least one spacer and multivalent branched multiple antigenic peptides (MAPs).
  • MAPs multivalent branched multiple antigenic peptides
  • epitope refers to a site on an antigen to which B and/or T cells respond.
  • B- cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • T-cell epitopes consist of peptides which are capable of forming complexes with host MHC molecules.
  • T-cell epitopes for a human MHC class I molecules which are responsible for induction of CD8+ T-cell responses, generally comprise 9 to 11 amino acid residues
  • epitopes for human MHC class II molecules which are responsible for CD4+ T-cell responses, typically comprise 12 or more amino acid residues
  • B cells are capable of recognizing peptides as small as 4 amino acids in length. It is the T-cell epitope/MHC complexes that are recognized by T-cell receptors leading to T cell activation.
  • T-cell response refers to an 8-mer or an A ⁇ fragment that is capable of inducing an immune response in the form of antibodies to A ⁇ , but which response does not include a T-cell response to the self antigen, A ⁇ .
  • immunological or “immune” or “immunogenic” response refers to the development of a humoral (antibody mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) response directed against an antigen in a vertebrate individual.
  • a humoral antibody mediated
  • a cellular mediated by antigen-specific T cells or their secretion products
  • Such a response can be an active response induced by administration of an immunogen or a passive response induced by administration of an antibody.
  • multivalent peptide refers to peptides having more than one antigenic determinant.
  • pharmaceutical composition means a chemical or biological composition suitable for administration to a mammalian individual. As used herein, it refers to a composition comprising 8-mers, immunogenic fragments of A ⁇ and A ⁇ conjugates described herein to be administered optionally with or without an adjuvant.
  • Applicants herein used the differing nature of T and B cell epitopes to design the peptides used for the invention.
  • the vaccine constructs were designed, by restricting the linear peptide size to eight amino acids and, if necessary, removing any potential C-terminal T-cell epitope anchor residues.
  • one aspect of the present invention was the identification of A ⁇ fragments that are immunogenic, but lack a T-cell epitope, for use as an AD vaccine.
  • Those skilled in the art would appreciate that previous teachings in the field did not predict, for example, that an 8-mer would produce an immunogenic response and did not distinguish the usefulness of fragments from different regions of the A ⁇ peptide. See, for example, U.S. Pat. Nos. 6,808,712 and 6,787,144.
  • An additional aspect of the invention herein includes the identification of A ⁇ plasma elevating agents comprising an immunogenic fragment of A ⁇ , lacking a T-cell epitope, that induce an immune response in the form of antibodies to A ⁇ and that elevate plasma A ⁇ levels.
  • Such agents can be used as an AD vaccine and for related amyloid diseases characterized by elevated brain A ⁇ levels.
  • Prior to Applicants' invention it was not known or predictable which immunogenic fragments of A ⁇ would result in elevated plasma A ⁇ levels.
  • the A ⁇ plasma elevating agents described herein act to induce an immune response in the form of antibodies to A ⁇ that, according to the peripheral sink theory of A ⁇ clearance, produce elevated levels of plasma A ⁇ that leads to subsequent decreases in brain A ⁇ .
  • the invention provides compositions and methods for prophylactic and therapeutic treatment of disease characterized by accumulation of amyloid deposits.
  • Amyloid deposits comprise a peptide aggregated to an insoluble mass. The nature of the peptide varies in different disease but in most cases, the aggregate' has a ⁇ -pleated sheet structure and stains with Congo Red dye.
  • Diseases characterized by amyloid deposits include Alzheimer's disease (AD), both late and early onset. In both diseases, the amyloid deposit comprises a peptide termed amyloid beta (A ⁇ ), which accumulates in the brain of affected individuals.
  • AD Alzheimer's disease
  • a ⁇ amyloid beta
  • the term "amyloid disease” also refers to disease characterized by elevated brain A ⁇ levels.
  • Therapeutic agents for use in the present invention induce an immune response in the form of antibodies to A ⁇ .
  • Induction of an immune response can be active as when an immunogen is administered to induce antibodies or T cells reactive with A ⁇ in an individual or passive, as when an antibody is administered that itself binds to A ⁇ in the individual.
  • the therapeutic agent to be used in preventing or treating amyloid diseases, such as AD include peptide fragments of A ⁇ , which can be any of the naturally occurring forms (i.e. A ⁇ 39, A ⁇ 40, A ⁇ 42, A ⁇ 42, or A ⁇ 43). These sequences are known in the art, see, for example, Hardy et al., TINS 20: 155-158, 1997.
  • the therapeutic agent is, in a preferred embodiment, an immunogenic fragment, lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to A ⁇ .
  • the immunogenic fragment of A ⁇ can be in the form of an 8-mer, a multivalent linear A ⁇ conjugate having at least one PEG spacer or a multivalent branched MAP A ⁇ conjugate.
  • the therapeutic agent can be administered in the form of a pharmaceutical composition.
  • the therapeutic agent is an A ⁇ plasma elevating agent capable of inducing an immune response in the form of antibodies to A ⁇ and that elevate plasma A ⁇ levels in an individual.
  • Such agents can comprise a naturally occurring peptide fragment or may include one or more substitutions, additions or deletions, and may include synthetic or non-naturally occurring amino acids. Fragments and constructs can be screened for prophylactic and therapeutic efficacy in the assays described in the examples herein.
  • the therapeutic agents comprise a peptide fragment of A ⁇
  • such agents may also include peptides and other compounds that do not necessarily have a significant amino acid sequence similarity with A ⁇ , but that nevertheless can serve as mimetics of A ⁇ and induce a similar immune response.
  • peptides and proteins forming ⁇ -pleated sheets can be screened for suitability for the invention herein.
  • combinatorial libraries and other compounds can be screened for suitability for the invention herein.
  • Such identified therapeutic agents can be linked either chemically or biologically to a carrier to facilitate their use as an immunogen.
  • a carrier include serum albumins, keyhole limpet hemocyanin (KLH), immunoglobulin molecules, ovalbumin, tetanus toxoid protein, or a toxoid from other pathogenic bacteria, such as diphtheria, E. coli, cholera, or H. pylori, or an attenuated toxin derivative.
  • the carrier is the outer membrane protein complex of Neisseria meningitides (OMPC).
  • the invention herein also contemplates the use of such therapeutic agents in a pharmaceutical composition comprising an 8-mer or immunogenic fragment of A ⁇ , which may be linked to a carrier, to be administered optionally with an adjuvant.
  • Suitable adjuvants include aluminum salts (alum), a lipid, such as 3 De-O-acylated monophosphoryl lipid A (MPL) or a saponin-based adjuvant.
  • the adjuvant is an aluminum adjuvant (Merck alum adjuvant, MAA) or a saponin-based adjuvant (ISCOMATRK * , CSL Ltd, Parkville, Australia.
  • Effective doses of the compositions of the invention herein for the prophylactic or therapeutic treatment of AD and other amyloid diseases will vary depending upon many factors including, but not limited to, means of administration, target site, physiological state of the patient, other medications administered and whether treatment is a therapeutic, i.e. after on-set of disease symptoms, or prophylactic, i.e. to prevent the on-set of disease symptoms.
  • the patient is human and the therapeutic agent is to be administered by injection.
  • the amount of immunogen or therapeutic agent to be employed will also depend on whether an adjuvant is to be administered either concomitantly or sequentially, with higher doses being employed in the absence of an adjuvant.
  • an immunogen or therapeutic agent to be administered will vary, but amounts ranging from 0.5-50 ⁇ g of peptide (based on the A ⁇ peptide content) per injection are considered for human use. Those skilled in the art would know how to formulate compositions comprising antigens of the type described herein.
  • the administration regimen would consist of a primary immunization followed by booster injections at set intervals.
  • the intervals between the primary immunization and the booster immunization, the intervals between the booster injections, and the number of booster immunizations will depend on the antibody titers and duration elicited by the vaccine. It will also depend on the functional efficacy of the antibody responses, namely, levels of antibody titers required to prevent AD development or exerting therapeutic effects in AD patients.
  • a typical regimen will consist of an initial set of injections at 1, 2 and 6 months. Another regimen will consist of initial injections at 1 and 2 months. For either regimen, booster injections will be given either every six months or yearly, depending on the antibody titers and durations.
  • An administration regimen can also be on an as-needed basis as determined by the monitoring of immune responses in the patient.
  • peptides were modified by the addition of triple lysine (KKK) (SEQ ID NOS: 52, 53, 54, 56, 59, 60, 62, 64 and 65) or glutamine (EEE) (SEQ ID NOS: 50, 51 and 61) residues or the use of a polyethelyene glycol (PEG) (SEQ E) NOS: 55 and 63) spacer.
  • KKK triple lysine
  • EEEE glutamine
  • peptides spanning the sequences of A ⁇ corresponding to residues (11-18) and (13-20) were made in multiple forms, the first with a 6-aminohexanoic acid (Aha) spacer plus a functional group for chemical cross-linking at N-terminus and the other form with Aha and the functional group at C- terminus.
  • Aha 6-aminohexanoic acid
  • the immunogenic fragments may be 8-mer peptides (eight amino acid residues) derived from the naturally occurring, i.e. wild type, or synthetic A ⁇ (SEQ ID NO:1) or any mutation or variation thereof.
  • Such mutation or variant can be produced by synthetic or recombinant means known to those of ordinary skill in the art.
  • One example of such a variant is the EV substrate (EVEFRHDSGYEVHHQKLVFFAEDVGSNKGA ⁇ GLMVGGVVIA) (SEQ ID NO: 66) a peptide corresponding to A ⁇ (1-42) in which positions 1 and 2 of wild type A ⁇ have been varied.
  • a ⁇ conjugates for use in formulating a vaccine was based on the immunogenicity of the 8-mers.
  • the 29 peptides (Figure 2A) were conjugated to EXH to form an A ⁇ conjugate and tested in guinea pigs ( Figure 3).
  • a ⁇ (1-18)-KLH (SEQ ID NO: 37) was included in this analysis.
  • Guinea pigs were immunized as described in Example 3.B with conjugated immunogens formulated in alum plus 50 ⁇ g of ISCOMATRIX® (CSL, Ltd., Parkville, Australia).
  • ISCOMATRIX® CSL, Ltd., Parkville, Australia.
  • guinea pigs were immunized three times at four week intervals.
  • blood samples were collected and tested by ELISA for antibody titers against A ⁇ 40 peptide. These titers are shown in Figure 3 as post- dose 1 (PDl), post-dose 2 (PD2) and post-dose 3 (PD3), respectively.
  • regions corresponding to A ⁇ amino acids 1-8, 3- 10, 7-14, 11-18, 13-20, 15-22, 19-26, 21-28, 23-30, 27-34 and 29-36 were highly immunogenic (>1: 10000) including: regions 1-8, 15-22, 21-28, 23-30 and 29-36.
  • regions 1-8, 15-22, 21-28, 23-30 and 29-36 are highly immunogenic, while other regions (e.g., 5-12, 25-32, 31-38 and 35-42) are non-immunogenic (titers ⁇ 1:300).
  • the results also demonstrate that while the A ⁇ conjugates were capable of eliciting an A ⁇ 4o peptide-specific antibody response, not all fragments of A ⁇ were equally immunogenic.
  • FIG. 4 the immunogenic response produced by the A ⁇ (3-1O)-KLH (SEQ ID NO: 40) conjugate produced an antibody response that was directed against human AD brain tissue.
  • Figure 4A demonstrates immunoreactivity of the monoclonal anti-A ⁇ antibody 6F3D (Vector Laboratories). As shown, this brain has extensive A ⁇ deposits in a manner expected to be typical for human AD.
  • Figure 4B demonstrates a lack of immunoreactivity of sera from a pre-immunized guinea pig.
  • Figure 4C shows positive immunoreactivity of sera from this same guinea pig following two injections of the A ⁇ (3-1O)-KLH (SEQ ID NO: 40) conjugate.
  • the invention described herein includes multivalent peptide conjugates such as those shown in Figures 5, 6A and 6B.
  • Multivalent branched MAP-OMPC conjugates ( Figures 6A and 6B) were generated by using a lysine-based scaffold, whereas multivalent linear 8-mer-OMPC conjugates ( Figure 5) were prepared using a PEG linker.
  • a PEG linker compared to conventional amino acid linkers that can also be used herein, offers the advantage of lower immunogenicity and greater peptide solubility.
  • the immunogenic fragment is a multivalent MAP conjugated to OMPC.
  • ISCOMATRK® (CSL, Ltd., Parkville, Australia). Blood samples were collected and used to determine the antibody titers against A ⁇ . Interim analysis of this ongoing study demonstrated that at post-dose 1 (PDl) the monkeys receiving 5 ⁇ g vaccine in alum failed to develop any detectable titers, while those receiving 30 ⁇ g vaccine in alum developed low A ⁇ 4o specific titers. All monkeys that received the alum plus ISCOMATRK® formulation developed significant antibody titers.
  • the peptides intended for conjugation to maleimide derivatized carrier proteins were synthesized with a cysteine residue at the carboxy terminus.
  • the spacer, Aha (6-aminohexanoic acid) was incorporated between the primary peptide sequence and the carboxy terminal cysteine as a structural element for minimizing steric accessibility to carrier protein during conjugation.
  • solubilizing residues represented by EEE, KKK or PEG were introduced at the C-terminus in sequences 14,15,16 17,18,19,20,23,24,25,26,27,28,29.
  • the PEG unit was introduced as, O-(N ⁇ Fmoc-2- aminoethyl)-O'-(2-carboxyethyl)-undecaethylenegIycol [FmOC-NHCH 2 CH 2 O(CH 2 CH 2 O)I 0 CH 2 CH 2 OCH 2 CH 2 CO 2 H].
  • the A ⁇ peptides were prepared by solid-phase synthesis on an automated peptide synthesizer using Fmoc chemistry protocols as supplied by the manufacturer (Applied Biosystems, Foster City, CA). Following assembly the resin bound peptide was deprotected and cleaved from the resin using a cocktail of 94.5% trifluoroacetic acid, 2.5% 1,2- ethanedithiol, 1% triisopropylsilane and 2.5% H 2 O. Following a two hour treatment the reaction was filtered, concentrated and the resulting oil triturated with ethyl ether. The solid product was filtered, dissolved in 50% acetic acid/H 2 O and freeze-dried.
  • the A ⁇ peptides (8-mers), 2 mg, containing a C- terminal cysteine was suspended in 1 ml of commercial maleimide conjugation buffer (83 mM sodium phosphate, 0.1 M EDTA, 0.9 M NaCl, 0.02% sodium azide, pH 7.2 (Pierce Biotechnology, Rockford, IL).
  • a 2 mg sample of commercial maleimide-activated KLH (Pierce Biotechnology, Rockford, IL) was added to the peptide and allowed to react at 25 0 C for four hours.
  • the conjugate was separated from unreacted peptide and reagents by exhaustive dialysis versus PBS buffer using 100,000 Da dialysis tubing.
  • the amount of peptide incorporated into the conjugate was estimated by amino acid analysis following a 70 hour acid hydrolysis. Peptide concentrations were determined to be between 0.24 and 0.03 mg/ml.
  • Bromoacetylated peptides were prepared by standard t-Boc solid-phase synthesis, using a double coupling protocol for the introduction of amino acids on the Applied Biosystems model 430A automated synthesizer. Starting with p-methylbenzhydrylamine resin the carboxy terminal amino acid t- B oc-Ly s (Fmoc)-OH was introduced followed by the subsequent amino acids in the sequence. Aha was introduced as a spacer to all of these sequences and a PEG unit in sequences 35 and 37 to aid in aqueous solubility.
  • the PEG unit was introduced as O-(N-Boc-2-aminoethyl)-O'-(N-diglycolyl-2-aminoethyl) hexaethyleneglycol [BOC-NHCH 2 CH 2 O(CH 2 CH 2 O) 6 CH 2 CH 2 NHCOCH 2 OCH 2 CO 2 H].
  • the amino terminous was capped by the coupling of acetic acid.
  • the Fmoc protecting group on the epsilon amino group of the carboxy terminal lysine was removed by treatment with piperidine. Subsequently the N amino group was reacted with Bromoacetic anhydride in methylene chloride as the solvent for 30 minutes.
  • t-Boc protected amino acids were assembled on the ABI automated synthesizer C, Lys(Fmoc), Aha, Y, G, S, D, H, R, F, E followed by coupling with acetic acid.
  • the N amino Fmoc protecting group of lysine was removed and the synthesis continued with the introduction of the following t-Boc protected amino acids: Aha, H, H, V, E, Y, G, S, D followed by coupling with acetic acid.
  • the resin bound peptide was isolated, purified and characterized as in Example l.C. Note: Instead of 10% anisole as in Example l.C, a 1:1 mixture of p-cresol: p-thiocresol was used as a scavenger during HF cleavage.
  • Divalent MAPs Construct Nos. 10, 13 and 14, Figure 6A, were prepared as described in Example 6.F.
  • the PEG unit was introduced as 0-(N-Boc-2-aminoethyl)-0'-(N-diglycolyl-2-aminoethyl) hexaethyleneglycol (t-Boc-NHCH 2 CH 2 O(CH 2 CH 2 O) 6 CH 2 CH 2 NHCOCH 2 OCH 2 CO 2 H).
  • Fmoc-Lys (t-Boc)-OH was coupled to MBHA resin. Following removal of the t-Boc protecting group on the N ⁇ amino group of lysine the sequence was extended with the introduction of the following t-Boc protected amino acids: Aha, Y,G, S, D, H, R, F, E, followed by coupling of acetic acid. The Na Fmoc protecting group on lysine was removed by manual treatment with piperidine.
  • This example presents the chemical conjugation of peptides derived from human A ⁇ 42 to purified Outer Membrane Protein Complex (OMPC) of Neisseria meningitidis, type B.
  • OMPC Outer Membrane Protein Complex
  • the chemical nature of the coupling is reaction between haloacetyl-derivatized peptide and thiol-derivatized protein of the membrane complex.
  • Amyloid peptides were synthesized as described above with a bromoacetyl functionality on the N-terminus for divalent linear epitope peptides or on the C-terminus or attached through the epsilon amino group of a lysine residue for monovalent linear and branched MAP forms.
  • the BrAc group was separated from the mature peptide by a spacer consisting of 6-aminohexanoic acid (Aha). Refer to sequences described above. Conjugation will be described for the representative peptide, A ⁇ (3-10). All manipulation of OMPC-containing solutions was performed in a laminar flow environment following standard aseptic techniques.
  • OMPC Purified, sterile OMPC, obtainable from a process such as that described in Fu, U.S. Pat. No. 5,494,808 used for the production of PedvaxHIB® and pneumococcal conjugate vaccines, was thiolated on a portion of its surface-accessible lysine residues using the reagent N- acetylhomocysteinethiolactone (NAHT, Aldrich, St. Louis, MO). OMPC in water, 117mg, was pelleted by centrifugation at 289,000 x g for 60 minutes at 4°C and the supernatant was discarded.
  • NAHT N- acetylhomocysteinethiolactone
  • N2-sparged activation buffer (0.11 M sodium borate, pH 11) was added to the centrifuge tube and the pellet was dislodged with a glass stir rod. The suspension was transferred to a glass Dounce homogenizer and resuspended with 30 strokes. The centrifuge tube was washed and the wash dounced with 30 strokes. Re-suspended pellet and wash were combined in a clean vessel to give a OMPC concentration of 10 mg/mL. Solid DTT and EDTA were dissolved in N2-sparged activation buffer and charged to the reaction vessel at a ratio of 0.106 mg DTT/mg OMPC and 0.57 mg EDTA/mg OMPC.
  • NAHT was dissolved in N2-sparged water and charged to the reaction at the ratio of 0.89 mg NAHT/mg OMPC. Reaction proceeded for three hours at ambient temperature, protected from light in a N2 hood.
  • OMPC was pelleted as described above and re-suspended at 6 mg/mL by Dounce homogenization in N2-sparged conjugation buffer (25 mM sodium borate, pH 8.5, 0.15 M NaCl) to wash the pellet.
  • N2-sparged conjugation buffer 25 mM sodium borate, pH 8.5, 0.15 M NaCl
  • the OMPC was pelleted as above and re-suspended at 10 mg/mL by Dounce homogenization in N2-sparged conjugation buffer. An aliquot was removed for free thiol determination by Ellman assay and the bulk product was stored on ice in dark until use. Measured thiol content was between 0.2 to 0.3 ⁇ mol/mL.
  • conjugate and control were transferred to 100,000 Da molecular weight cut-off dialysis units and dialyzed exhaustively against at least five changes of conjugation buffer.
  • samples were transferred to 15 ml polypropylene centrifuge tubes and centrifuged at 2,280 x g for five minutes at 4°C to remove any aggregated material. Aliquots were removed for analysis and the bulk was stored at 4°C.
  • Total protein was determined by the modified Lowry assay and samples of conjugate and control were analyzed by quantitative amino acid analysis (AAA).
  • AAA quantitative amino acid analysis
  • Peptide to OMPC molar ratios were determined from quantitation of the unique residue S-carboxymethylhomocysteine which was released upon acid hydrolysis of the nascent peptide-OMPC bond.
  • the OMPC-specific concentration was determined from hydrolysis-stable residues which were absent from the peptide sequence and thus unique to OMPC protein. Assuming 1 mol of peptide for every mol SCMHC, the ratio of SCMHC/OMPC was thus equivalent to the peptide/OMPC content.
  • the mass loading of peptide could be calculated from this ratio using the peptide molecular weight and an average OMPC mass ' of 40,000,000 Da.
  • the covalent nature of the conjugation was qualitatively confirmed by SDS-PAGE analysis using 4-20% Tris-glycine gels (Invitrogen, Carlsbad, CA) where an upward shift in mobility was observed for the Coomassie-stained conjugate bands relative to control.
  • This example describes the formulation and administration of the A ⁇ conjugates capable of inducing an immune response in the form of antibodies to A ⁇ .
  • a second adjuvant such as the saponin-based adjuvant, ISCOMATRDC (CSL Ltd.,
  • conjugates are diluted one times saline at a designated peptide concentration and mixed with two times alum (Merck, Product No. 39943), which corresponds to 900 ⁇ g/mL Merck alum prepared in sterile saline (150 mM sterile sodium chloride solution).
  • target concentration in the vaccine is 450 ⁇ g/mL Merck alum or one time Merck alum.
  • Target peptide (antigen) concentrations for animal studies were as follows: for mice - 12.1 ⁇ g/mL (Dose 0.1 mL); for monkeys - 10 ⁇ g/mL or 60 ⁇ g/mL (Dose 0.5 mL) and for guinea pigs - 12.5 ⁇ g/mL (Dose 0.4 mL). The mix is incubated for two hours at room temperature. To obtain the injection dose, the alum-absorbed conjugates are diluted with one time alum to reach the target peptide concentration. Where a second ® adjuvant is needed, i.e. ISCOMATRIX, the target concentration was 10 ⁇ g/ML for mice studies, 0, 100 or 200 ⁇ g/mL for monkey studies and 125 ⁇ g/mL for guinea pigs.
  • ISCOMATRIX is dialyzed into sterile saline solution at 2-8° C. Sterile saline solution is
  • ISCOMATRIX changed 2-3 times during dialysis.
  • ISCOMATRIX is filter sterilized using a syringe filter (0.22 uM Millex-GV syringe filter, Millipore, Billerica, MA).
  • concentration of sterile, dialyzed ISCOMATRIX R is determined by RP-HPLC.
  • ISCOMATRIX is stored sterile at 2-8°C until use. 2.
  • a ⁇ peptide-OMPC conjugate stocks are diluted into sterile IX saline solution.
  • the diluted AD peptide-OMPC conjugate stocks are then added to 2X Merck alum in IX sterile saline solution and mixed for one hour on a rotating wheel at room temperature.
  • the mixture is allowed to settle on the bench top for 15 minutes at room temperature and is then centrifuged at 1500 rpm for ten minutes.
  • the supernatant is decanted off gently (UV analysis of supernatant is performed to determine % A ⁇ peptide-OMPC conjugate bound to alum) and the pellet is resuspended in sterile IX saline.
  • the mixture is aliquoted into sterile 3 mL tubing glass vials and then stored at 2-8 0 C until final formulation with ISCOMATRIX®.
  • OMPC/alum in saline is determined by static light scattering to confirm binding and monitor particle stability.
  • the sterile, dialyzed ISCOMATRIX R in IX saline is added to A ⁇ peptide-OMPC/alum in sterile 150 mM NaCl while vortexing. Vials are stoppered, capped and crimped to completely seal.
  • Vaccine is stored at 2-8 0 C prior to injection. Prior to injection, each vaccine is vortexed for 3-5 minutes.
  • Two animals per group were immunized with the A ⁇ peptide - E-LH conjugates shown in Figure 2A intramuscularly with 400 ⁇ l of a conjugate vaccine (8 ⁇ g by peptide content or 50 ⁇ g by total conjugate) in the presence of 40 ⁇ g of ISCOMATRIX.®
  • the immunizations were performed three times in four-week intervals. Serum samples were collected before first immunization (pre-bleeds) and three weeks after each immunization and stored at 4°C prior to antibody titer determinations.
  • the antibody titers were determined by ELISA according to the protocol that follows using A ⁇ 40 as the target antigen.
  • the ELISA based analysis is as follows: Ninety six-well plates were coated with 50 ⁇ l per well of A ⁇ at a concentration of 4 ⁇ g/ml in 50 mM bicarbonate buffer, pH 9.6, at 4°C overnight. Plates were washed with phosphate buffered saline (PBS) and blocked with 3% skim milk in PBS containing 0.05% Tween-20 (milk-PBST). Testing samples were diluted in a 4-fold series in PBST. One hundred ⁇ l of a diluted sample was added to each well, and the plates were incubated at 24°C for two hours and then washed six times with PBST.
  • PBS phosphate buffered saline
  • milk-PBST 0.05% Tween-20
  • regions corresponding to A ⁇ amino acids 1-8, 3-10, 7-14, 11-18, 13- 20, 15-22, 19-26, 21-28, 23-30, 27-34 and 29-36 were highly immunogenic (>1: 10000) including: regions 1-8, 15-22, 21-28, 23-30 and 29-36.
  • the results demonstrate that 8-mer conjugates are capable of eliciting an A ⁇ 4o specific antibody response.
  • fragments of A ⁇ were equally immunogenic.
  • regions of A ⁇ e.g., 5-12, 25-32, 31-38 and 35-42 are non-immunogenic (titers ⁇ 1:300).
  • ISCOMATRJX® can provide an improved immune response.
  • Applicants used an A ⁇ (1-18) peptide conjugated to OMPC as the model antigen.
  • Rhesus monkeys (Macaca mulatto) were maintained in accordance with the institutional animal care protocols of Merck Research Laboratories and New Iberia Research Center (The University of Louisiana at Lafayette, New Iberia, LA).
  • a ⁇ (1-18)-OMPC conjugates Five groups of monkeys, three per group, were given the following A ⁇ (1-18)-OMPC conjugates: (1) 5 ⁇ g conjugate (based on peptide content) in alum, (2) 5 ⁇ g conjugate in alum + 100 ⁇ g ISCOMATRK®, (3) 5 ⁇ g conjugate in alum + 50 mg ISCOMATRK®, (4), 30 ⁇ g conjugate in alum, (2) 30 ⁇ g conjugate in alum + 100 ⁇ g ISCOMATRK®.
  • the immunizations were carried out by intramuscular injections in 0.5 ml aliquots at weeks 0, 8 and 24 using tuberculin syringes (Becton- Dickinson, Franklin Lakes, NJ). Serum samples were collected at four week intervals starting from week 0 (pre-bleed) and the tested for antibody titers against A ⁇ 40 by ELISA, performed as described in the preceding example.
  • Sections were blocked by a ten minute exposure to 10% goat serum followed by a five minute wash with PBS. Sections were then incubated with diluted guinea pig sera at 4° C overnight or for one hour at room temperature. Following a five minute PBS wash, sections were incubated for ten minutes with diluted biotinylated goat anti- guinea pig IgG or biotinylated horse anti-mouse antibody (1 drop in 5 ml PBS). Sections were washed for five minutes in PBS and subsequently incubated with ABC solution (Vectorstain ABC kit; Vector Laboratories, Inc.) for thirty minutes. Sections were washed with PBS for five minutes.
  • ABC solution Vectorstain ABC kit; Vector Laboratories, Inc.
  • Sections were then stained with DAB (DakoCytomation) for five minutes and washed with dd H2O. Sections were then counterstained in hematoxylin for thirty seconds and dehydrated in graded EtOH and Xylenes (70% EtOH for five minutes, 80% EtOH for five minutes, 100% EtOH for five minutes and xylenes for five minutes). Sections were then cover-slipped and evaluated by light microscopy.
  • the immunogenic response produced by the A ⁇ (3-1O)-KLH conjugate produced an antibody response that was directed against human AD brain tissue. As shown in Figure 4, this human brain section has extensive A ⁇ deposition in a manner typical to that expected for human AD.
  • Enzyme-Linked ImmunoSpot Assay
  • Immunogen fragments possessing T-cell epitopes are identified by the presence of a dark spot on the surface of a white membrane; each spot indicates the presence of a T- cell that has secreted interferon gamma (IFN- ⁇ ) in response to the antigen (i.e. immunogenic fragment).
  • IFN- ⁇ interferon gamma
  • PBMCs peripheral blood monocytes
  • PBMCs were centrifuged and re-suspended in RlO. PBMCs were counted on a Z2 Coulter counter (Beckman Coulter, Fullerton, CA).
  • Each well of the aspirated plate received either 0.4 ⁇ g of A ⁇ 1-40, A ⁇ 1-20, PHA (phytohemagglutinin, Cat No. L-8902, Sigma, St. Louis, MO, positive control), or diluted DMSO (Sigma, negative control); 400000 PBMCs were then added to each well. Plates were incubated for 18- 24 hours at 37 0 C in a humid CO 2 incubator. Plates were washed in PBS with 5% FBS and 0.005%
  • Applicants have established that spot forming cells per million PBMCs (SFC/ 10 6 PBMCs) must exceed 55 and must exceed 4-fold the negative control to be defined as a positive result; failing to meet both these criteria defines a negative result.
  • Rhesus macaques were vaccinated with either a MAP construct comprising A ⁇ (3-10)/(21-28) (Construct No. 12, Figure 6A) conjugated to OMPC or with both of two monomeric constructs, A ⁇ (3-10) (SEQ ID NO: 69) and A ⁇ (21-28) (SEQ ID NO: 73) conjugated to OMPC.
  • the rhesus primates received vaccinations every four weeks with bleeds collected and analyzed four weeks following each injection. Anti-A ⁇ 4o titers and total A ⁇ .40 levels were determined.
  • Plasma A ⁇ i.40 levels were determined in these immunized animals using a 6E10/G210 ELISA.
  • This assay measures A ⁇ i.40 using a sandwich ELISA comprising an N-terminal capture antibody 6E10 (A ⁇ 1-8) (Signet Laboratories, Dedham, MA) and a C-terminal A ⁇ 4o neo-epitope antibody (G210) (The Genetics Company, Inc., Zurich, Switzerland) conjugated with alkaline phosphatase.
  • the antibody, 6E10 was coated onto plates at a concentration of 5ug/ml. Diluted plasma samples (1:3) were used at 50 ⁇ l/well in triplicates.
  • a ⁇ i_4o standards were prepared from 400 pM - 3pM in 6E10 immuno-depleted rhesus plasma matrix. This assay has a signal-to-noise ratio of about 5-20.
  • the CDP-star alkaline phosphatase substrate was obtained from Applied Biosystems, Foster City, CA.
  • the identification of immunogens, either 8-mers or MAPs, lacking a T-cell epitope, that elevate plasma A ⁇ following immunization provides a method for selecting said 8-mers or MAPs for use in a vaccine construct.
  • those skilled in the art are now able to characterize said vaccine constructs both quantitatively (i.e., immunogenicity) and qualitatively (i.e., nature of the polyclonal antibody response - ability to elevate plasma AB levels).
  • the invention herein is not limited to 8-amino acid A ⁇ fragments, but is inclusive of any antigen capable of producing a polyclonal antibody response in the host organism that is reactive to A ⁇ .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides compositions and methods for the treatment of diseases associated with amyloid deposits of Aβ in the brain of a patient, such as Alzheimer’s disease. Such methods entail administering an immunogenic fragment of Aβ, lacking a T-cell epitope, capable of inducing a beneficial immune response in the form of antibodies to Aβ. In another aspect, the immunogenic fragment of Aβ is capable of elevating plasma Aβ levels. The immunogenic fragments comprise linear or multivalent peptides of Aβ. Pharmaceutical compositions comprise the immunogenic fragment chemically linked to a carrier molecule which may be administered with an adjuvant.

Description

TITLE OF THE INVENTION
PEPTIDE CONJUGATE COMPOSITIONS AND METHODS FOR THE PREVENTION AND
TREATMENT OF ALZHEIMER'S DISEASE
FIELD OF THE INVENTION
The present invention relates to compositions and methods for the prevention and treatment of amyloidogenic diseases and, in particular, Alzheimer's disease.
BACKGROUND OF THE INVENTION Alzheimer's disease (AD) is characterized by progressive memory impairment and cognitive decline. Its hallmark pathological lesions are amyloid deposits (senile plaques), neurofibrillary tangles and neuronal loss in specific brain regions. The amyloid deposits are composed of amyloid beta peptides (Aβ) of 40 to 43 amino acid residues, which are the proteolytic products of the amyloid precursor protein (APP). Neurofibrillary tangles are the intracellular filamentous aggregates of hyperphosphorylated tau proteins (Selkoe, Science, 275: 630-631, 1997).
The pathogenesis of AD has not been fully understood, but it is expected to be a multi- factored event. Accumulation and aggregation of Aβ in brain tissue is believed to play a pivotal role in the disease process, also know as the amyloid cascade hypothesis (Golde, Brain Pathol., 15: 84-87, 1995). According to this hypothesis, Aβ, particularly Aβ42, is prone to form various forms of aggregates, ranging from small oligomers to large, elongated profibrile structures. These aggregates are neurotoxic and are responsible for the synaptic pathology associated with the memory loss and cognition decline in the early stage of the disease (Klein et al, Neurobiol. Aging, 25: 569-580, 2004). A recent publication suggests that reduction of Aβ in a triple transgenic mouse model also prevents intracellular tau deposition (Oddo et al, Proc. Neuron, 43:321-332, 2004). This finding suggests that the extracellular amyloid deposition may be causative for subsequent neurofibrillary tangle formation, which may in turn lead to neuronal loss.
Immunization of APP transgenic mice with Aβ antigen can reduce the brain Aβ deposits and mitigate disease progression. This was first reported by Shenk et al., Nature, 400: 173-177, 1999, and has now been corroborated by a large number of studies involving different transgenic animal models, various active vaccines as well as passive immunization with Aβ specific monoclonal antibodies (Bard et al., Nature Med, 6: 916-919, 2000; Janus et al, Nature, 408: 979-982, 2000; Morgan et al, Nature, 408: 982-985, 2000; DeMattos et al, Proc. Natl. Acad. Sci., 98: 8850-8855, 2001; Bacskai et al, J. Neurosci., 22: 7873-7878, 2002; Wilcock et al, J. Neurosci., 23: 3745-3751, 2003). Consistent with these animal data, three published evaluations of postmortem human brain tissues from patients who had previously received active immunization with a pre-aggregated Aβ (1-42) immunogen (AN1792,
Betabloc) showed regional clearance of senile plaques (Nicoll et al., Nature Med., 9: 448-452, 2003; Ferrer et al, Brain Pathol.. 14: 11-20, 2004; Masliah et al, Neurology, 64: 129-131, 2005). This data collectively indicates that vaccines that effectively elicit antibody responses to Aβ antigens are efficacious against the pathological senile plaques found in AD. However, the mechanism of vaccine or antibody efficacy remains to be defined.
The most advanced immunotherapy-based AD program in the public domain had been an active immunization Phase II vaccine trial using AN1792 (Betabloc), a vaccine composed of pre- aggregated Aβ (1-42) co-administered with the adjuvant, QS-21™ (Antigenics, New York, NY). In January 2002, this study was terminated when four patients showed symptoms consistent with meningoencephalitis (Senior, Lancet Neurol., 1: 3, 2002). Ultimately, 18 of 298 treated patients developed signs of menigoencephalitis (Orgogozo et ah, Neurology, 61: 46-54, 2003). There was no correlation between encephalitis and antibody titer and it has been reported that the likely causative mechanism for this effect was activation of T-cells to the self-immunogen, particularly the mid- and carboxy-terminal portion of the Aβ42 peptide (Monsonego et ah, J. Clin. Invest., 112: 415-422, 2003).
In support of this conclusion, postmortem examination of brain tissues from two vaccine recipients that developed encephalitis revealed substantial meningeal infiltration of CD4+ T cells in one patient (Nicoll et al. , Nature Med.. 9: 448-452, 2003) and CD4+, CD8+, CD3+, CD5+, CD7+ T cells in the other (Ferrer et al., Brain Pathol., 14: 11-20, 2004).
Current evidence suggests that increases in plasma Aβ levels following passive or active immunization reflect the initiation of a peripheral sink as a precursor to subsequent decreases in brain Aβ. The peripheral sink refers to a change in the equilibrium of brain and plasma Aβ stores resulting in a net efflux of central Aβ to the periphery (see, for example, Deane et al., J. Neurosci., 25: 11495-11503, 2005; DeMattos et al, Pro. Natl. Acad. Sci. USA. 98: 8931-8932, 2001). Other studies suggest that this increase in plasma Aβ observed following anti-Aβ immunotherapy is necessary for subsequent decreases in central Aβ to be realized (Cribbs et al., 7th International Conference on AD/PD, Sorrento, Italy, 2005). Thus, when two amino acids within Aβ are substituted (for example, such as occurs with the Dutch and Iowa mutations) the peptide is no longer able to cross from central to peripheral compartments (Davis et al., Neurobiol. Aging, in press, available on line 18 August 2005). When mice expressing this mutant form of Aβ and the Swedish mutation were immunized, no elevations in plasma Aβ were found and no subsequent lowering of brain Aβ was noted. By contrast, mice expressing the wild-type human Aβ sequence plus the Swedish mutation responded to active immunization with both increases in plasma Aβ and subsequent decreases in central Aβ (Cribbs et al., 7th International Conference on AD/PD, Sorrento, Italy, 2005). Accordingly, it is expected that any active vaccine immunogen capable of generating an immune response that results in the elevation of plasma Aβ levels will be useful for the treatment of Alzheimer's disease and related disorders characterized by elevated brain Aβ levels.
Applicants herein have surprisingly found that an antigen which eliminated T-cell epitopes, to avoid a self T-cell response, is immunogenic and elevates plasma Aβ levels. This represents a potential means to produce a safe and effective AD vaccine. Applicants herein provide such an antigen and a formulation for use as an AD vaccine. SUMMARY OF THE INVENTION
In one embodiment, the invention provides a pharmaceutical composition comprising an immunogenic fragment of Aβ, lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to Aβ. In one aspect, this composition comprises linear 8 amino acid peptides (8- mers) of Aβ. In still another aspect, this composition comprises multivalent linear 8-mers interspersed with at least one spacer or a multivalent branched multiple antigenic peptide (MAP). The pharmaceutical composition can be used as a vaccine for AD and related amyloid diseases.
In another embodiment of the invention, the pharmaceutical composition is an Aβ plasma elevating agent comprising an immunogenic fragment of Aβ, lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to Aβ that elevate plasma Aβ levels. The pharmaceutical composition can be used as a vaccine for AD and related amyloid diseases characterized by elevated brain Aβ levels.
In still another embodiment of the invention, the pharmaceutical composition is linked to a carrier molecule to form a conjugate, wherein the carrier helps to elicit an immune response comprising antibodies to the Aβ fragment. In a preferred embodiment of the invention, the carrier is the outer membrane protein complex of Neisseria meningitides (OMPC).
In a further embodiment of the invention, the pharmaceutical composition is administered with a pharmaceutically acceptable adjuvant. In a preferred embodiment the adjuvant is an
® aluminum adjuvant (Merck alum adjuvant, MAA) or a saponin-based adjuvant (ISCOMATRIX , CSL
Ltd., Parkville, Australia).
In still another embodiment, the invention provides methods for preventing or treating a disease associated with amyloid deposits of Aβ in the brain of a patient. Such diseases include Alzheimer's disease, Down's syndrome, cognitive impairment or other forms of senile dementia. The method comprises administering an immunogenic fragment of Aβ, lacking a T-cell epitope, selected from the group consisting of linear 8 amino acid peptides (8-mers), a multivalent linear peptides interspersed with at least one spacer and a multivalent branched multiple antigenic peptide (MAP). In a preferred embodiment the immunogenic fragment comprises a multivalent linear peptide with a polyethylene glycol (PEG) spacer. In a more preferred embodiment the immunogenic fragment comprises a multivalent branched MAP, Aβ (3-10)/(21-28) conjugate, Construct No. 12, Figure 6A, conjugated to OMPC.
Such methods entail the administration of an effective dose of an immunogenic fragment of Aβ, lacking a T-cell epitope, to patients in need of such treatment that will induce an immune response in the form of antibodies to Aβ. Said antibody response is capable of elevating plasma Aβ levels. In another aspect of this embodiment, the immunogenic fragment to be administered is linked to a carrier molecule. In yet another aspect of this embodiment, the immunogenic fragment is administered with an adjuvant. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 represents synthetic 8-amino acid peptides (8-mers) (SEQ ID NOS: 2-36) derived from Aβ (1-42) (SEQ ID NO: 1) from which peptides were selected to conduct a linear peptide scan to identify the epitopes of Aβ.
Figure 2 represents the 8-mers selected for conjugation to KLH (Figure 2A) and OMPC (Figure 2B).
Figure 3 represents the immunogenicity of selected Aβ conjugates, described in Figure 2, after the first (PDl), second (PD2) and third dose (PD3).
Figure 4 represents the cross-reactivity of sera extracted from a guinea pig previously immunized with an Aβ (3-1O)-KLH conjugate (SEQ ID NO: 40) on human AD brain tissue. Figure 4A shows immunoreactivity of the anti-Aβ monoclonal antibody 6F3D (which recognizes amino acids 8-17 of Aβ). The staining pattern reveals extensive amyloid pathology in this human brain. Figure 4B demonstrates a lack of immunoreactivity of this same brain to the pre-immune sera from the immunized guinea pig prior to immunization. Figure 4C shows the immunoreactivity of the sera from an immunized guinea pig following immunization
Figure 5 shows representative multivalent linear 8-mer peptides, which were selected based on the immunogenicity of the separate 8-mers in guinea pig studies (Example 3). These conjugates were synthesized as described and conjugated to OMPC (Example IJ and 1.K).
Figure 6 shows representative multivalent branched MAP conjugates, which were selected based on the immunogenicity of the separate 8-mers in guinea pig studies (Example 3). Figure 6A shows representative divalent MAPs and Figure 6B shows representative bromoacetyl-cysteine MAPs. These conjugates were synthesized as described and conjugated to OMPC (Example 2). Figure 7 represents the anti-Aβ4o titer from sera collected from rhesus monkeys following 1 (PDl) or 2 (PD2) injections with an Aβ (1-18) peptide conjugated to OMPC formulated in Merck alum alone or Merck alum plus MX (ISCOMATRIX , CSL, Ltd., Parkville, Australia) as an adjuvant.
Figure 8 represents the increase in plasma Aβ levels following administration of a Aβ conjugate. Figure 8A shows a greater than three-fold elevation following administration of a MAP construct comprising Aβ (3-10)/(21-28) (Construct No. 12, Figure 6A) conjugated to OMPC versus the monomeric constructs, Aβ (3-10) (SEQ ID NO: 69) and Aβ (21-28) (SEQ ID NO: 73) (o , Construct No. 12, Figure 6A; •, Aβ (3-10) (SEQ ID NO 69), A, Aβ (21-28) (SEQ ID NO: 73). Figure 8B shows that plasma Aβ levels are independent of titer levels (□ , Construct No. 12, Figure 6A; •, Aβ (3-10) (SEQ ID NO 69), ▲ , Aβ (21-28) (SEQ ID NO: 73). DEFINITIONS
The term "8-mer" refers to an eight amino acid peptide which corresponds to a fragment of Aβ, an analog of a natural Aβ peptide or a peptide mimetic. One or more 8-mers may be combined with at least one spacer to form a multivalent linear peptide or to form a multivalent branched MAP.
The term "Aβ conjugate" means an 8-mer or immunogenic fragment of Aβ that is chemically or biologically linked to a carrier, such as keyhole limpet hemocyanin or the outer membrane protein complex of Nesseria rneningitidies (OMPC).
The term "Aβ peptide" means any of the Aβ peptides described herein, including, but not limited to, linear 8-mers, multivalent linear peptides with at least one spacer and multivalent branched multiple antigenic peptides (MAPs).
The term "epitope" refers to a site on an antigen to which B and/or T cells respond. B- cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. T-cell epitopes consist of peptides which are capable of forming complexes with host MHC molecules. T-cell epitopes for a human MHC class I molecules, which are responsible for induction of CD8+ T-cell responses, generally comprise 9 to 11 amino acid residues, while epitopes for human MHC class II molecules, which are responsible for CD4+ T-cell responses, typically comprise 12 or more amino acid residues (Bjorkman et al. Nature 329:506-512, 1987; Madden et al. Cell 75:693-708; Batalia and Collins; Engelhard Annu Rev Immunol., 12: 181-207-622. 1995; Madden, Annu Rev Immunol., 13:587-622. 1995). Unlike T cells, B cells are capable of recognizing peptides as small as 4 amino acids in length. It is the T-cell epitope/MHC complexes that are recognized by T-cell receptors leading to T cell activation. The term "immunogenic fragment of Aβ" or "immunogenic fragment of Aβ, lacking a
T-cell response," as used herein refers to an 8-mer or an Aβ fragment that is capable of inducing an immune response in the form of antibodies to Aβ, but which response does not include a T-cell response to the self antigen, Aβ.
The term "immunological" or "immune" or "immunogenic" response refers to the development of a humoral (antibody mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) response directed against an antigen in a vertebrate individual. Such a response can be an active response induced by administration of an immunogen or a passive response induced by administration of an antibody.
The term "multivalent peptide" refers to peptides having more than one antigenic determinant. The term "pharmaceutical composition" means a chemical or biological composition suitable for administration to a mammalian individual. As used herein, it refers to a composition comprising 8-mers, immunogenic fragments of Aβ and Aβ conjugates described herein to be administered optionally with or without an adjuvant.
DETAILED DESCRIPTION OF THE INVENTION
As previously described, preclinical studies suggest that active immunization resulting in an anti-Aβ polyclonal antibody response provides efficacy against the pathological and cognitive symptoms associated with AD in transgenic mice that overexpress the amyloid precursor protein (Bard et al., Nature Med.. 6: 916-919, 2000; Janus et al, Nature, 408: 979-982, 2000; Morgan et al., Nature, 408: 982-985, 2000; DeMattos et al, Proe. Natl. Acad. Sci.. 98: 8850-8855, 2001; Bacskai etal, J. Neurosci.. 22: 7873-7878, 2002; Wilcoc, et al, J. Neurosci.. 23: 3745-3751, 2003). These preclinical studies are supported by a single clinical trial where an aggregate form of Aβ42 was used as an active immunogen.
Preliminary evidence from this study suggests that pathological (Nicoll et al, Nature Med., 9: 448-452, 2003; Ferrer et al, Brain Pathol.. 14: 11-20, 2004; Masliah et al, Neurology, 64:129-131, 2005) and cognitive improvements (Gilman et al, Neurology, 64 (9): 1553-1562, 2005) were found following treatment. While these findings are encouraging and consistent with preclinical studies, the treatment proved unsafe and was terminated following the appearance of meningoencephalitis in approximately 6% of the treated patients (Orgogozo et al, Neurology, 61: 46-54, 2003). Thus, there exists a need for active immunization procedures capable of an efficacious immune response and devoid of adverse safety issues.
Progress in understanding the nature of the adverse events in this preliminary clinical trial has been made. Several investigators have now reported the presence of CD4+ and CD8+ positive meningeal infiltrates on post-mortem evaluation (Nicoll, et al, Nature Med., 9: 448-452, 2003; Ferrer et al., Brain Pathol., 14: 11-20, 2004) suggestive of a T-cell response directed at the self-peptide Aβ42- However, while those skilled in the art would recognize the need to avoid a self-directed T-cell response while maintaining an appreciable antibody response (B-cell mediated), the means to produce an agent having this property is not known. This difficulty is compounded by a lack of predictive animal models or other preclinical assays with predictive validity for these activities.
To this end, Applicants herein used the differing nature of T and B cell epitopes to design the peptides used for the invention. The vaccine constructs were designed, by restricting the linear peptide size to eight amino acids and, if necessary, removing any potential C-terminal T-cell epitope anchor residues.
Accordingly, one aspect of the present invention was the identification of Aβ fragments that are immunogenic, but lack a T-cell epitope, for use as an AD vaccine. Prior to the present application, it was not definitively known which amino acid fragments of the Aβ peptide would produce an immunogenic response that would also be deficient in a T-cell epitope. Those skilled in the art would appreciate that previous teachings in the field did not predict, for example, that an 8-mer would produce an immunogenic response and did not distinguish the usefulness of fragments from different regions of the Aβ peptide. See, for example, U.S. Pat. Nos. 6,808,712 and 6,787,144.
An additional aspect of the invention herein includes the identification of Aβ plasma elevating agents comprising an immunogenic fragment of Aβ, lacking a T-cell epitope, that induce an immune response in the form of antibodies to Aβ and that elevate plasma Aβ levels. Such agents can be used as an AD vaccine and for related amyloid diseases characterized by elevated brain Aβ levels. Prior to Applicants' invention, it was not known or predictable which immunogenic fragments of Aβ would result in elevated plasma Aβ levels. Without wishing to be bound by any theory, it is believed that the Aβ plasma elevating agents described herein act to induce an immune response in the form of antibodies to Aβ that, according to the peripheral sink theory of Aβ clearance, produce elevated levels of plasma Aβ that leads to subsequent decreases in brain Aβ. Moreover, while individual 8-mers or immunogenic fragments of Aβ may be capable of inducing an immune response such that plasma Aβ levels are elevated, Applicants found that a multivalent branched MAP, Aβ (3-10)/(21-28) (Construct No. 12, Figure 6A), conjugated to OMPC, was particularly effective in elevating plasma Aβ levels relative to those of its constituent monomeric constructs, Aβ (3-10) (SEQ ID NO: 69) or Aβ (21-28) (SEQ ID NO: 73).
Amyloid Diseases
The invention provides compositions and methods for prophylactic and therapeutic treatment of disease characterized by accumulation of amyloid deposits. Amyloid deposits comprise a peptide aggregated to an insoluble mass. The nature of the peptide varies in different disease but in most cases, the aggregate' has a β-pleated sheet structure and stains with Congo Red dye. Diseases characterized by amyloid deposits include Alzheimer's disease (AD), both late and early onset. In both diseases, the amyloid deposit comprises a peptide termed amyloid beta (Aβ), which accumulates in the brain of affected individuals. Thus, the term "amyloid disease" also refers to disease characterized by elevated brain Aβ levels.
Therapeutic Agents
Therapeutic agents for use in the present invention induce an immune response in the form of antibodies to Aβ. Induction of an immune response can be active as when an immunogen is administered to induce antibodies or T cells reactive with Aβ in an individual or passive, as when an antibody is administered that itself binds to Aβ in the individual.
The therapeutic agent to be used in preventing or treating amyloid diseases, such as AD, include peptide fragments of Aβ, which can be any of the naturally occurring forms (i.e. Aβ39, Aβ40, Aβ42, Aβ42, or Aβ43). These sequences are known in the art, see, for example, Hardy et al., TINS 20: 155-158, 1997. As used herein, the therapeutic agent is, in a preferred embodiment, an immunogenic fragment, lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to Aβ. The immunogenic fragment of Aβ can be in the form of an 8-mer, a multivalent linear Aβ conjugate having at least one PEG spacer or a multivalent branched MAP Aβ conjugate. The therapeutic agent can be administered in the form of a pharmaceutical composition. In an another embodiment, the therapeutic agent is an Aβ plasma elevating agent capable of inducing an immune response in the form of antibodies to Aβ and that elevate plasma Aβ levels in an individual. Such agents can comprise a naturally occurring peptide fragment or may include one or more substitutions, additions or deletions, and may include synthetic or non-naturally occurring amino acids. Fragments and constructs can be screened for prophylactic and therapeutic efficacy in the assays described in the examples herein.
While in a preferred embodiment the therapeutic agents comprise a peptide fragment of Aβ, such agents may also include peptides and other compounds that do not necessarily have a significant amino acid sequence similarity with Aβ, but that nevertheless can serve as mimetics of Aβ and induce a similar immune response. For example, peptides and proteins forming β-pleated sheets can be screened for suitability for the invention herein. Similarly, combinatorial libraries and other compounds can be screened for suitability for the invention herein.
Such identified therapeutic agents can be linked either chemically or biologically to a carrier to facilitate their use as an immunogen. Such carriers include serum albumins, keyhole limpet hemocyanin (KLH), immunoglobulin molecules, ovalbumin, tetanus toxoid protein, or a toxoid from other pathogenic bacteria, such as diphtheria, E. coli, cholera, or H. pylori, or an attenuated toxin derivative. In a preferred embodiment of the invention the carrier is the outer membrane protein complex of Neisseria meningitides (OMPC).
The invention herein also contemplates the use of such therapeutic agents in a pharmaceutical composition comprising an 8-mer or immunogenic fragment of Aβ, which may be linked to a carrier, to be administered optionally with an adjuvant. Suitable adjuvants include aluminum salts (alum), a lipid, such as 3 De-O-acylated monophosphoryl lipid A (MPL) or a saponin-based adjuvant. In a preferred embodiment the adjuvant is an aluminum adjuvant (Merck alum adjuvant, MAA) or a saponin-based adjuvant (ISCOMATRK * , CSL Ltd, Parkville, Australia.
Treatment Regimes
Effective doses of the compositions of the invention herein for the prophylactic or therapeutic treatment of AD and other amyloid diseases will vary depending upon many factors including, but not limited to, means of administration, target site, physiological state of the patient, other medications administered and whether treatment is a therapeutic, i.e. after on-set of disease symptoms, or prophylactic, i.e. to prevent the on-set of disease symptoms. In a preferred embodiment the patient is human and the therapeutic agent is to be administered by injection. The amount of immunogen or therapeutic agent to be employed will also depend on whether an adjuvant is to be administered either concomitantly or sequentially, with higher doses being employed in the absence of an adjuvant.
The amount of an immunogen or therapeutic agent to be administered will vary, but amounts ranging from 0.5-50 μg of peptide (based on the Aβ peptide content) per injection are considered for human use. Those skilled in the art would know how to formulate compositions comprising antigens of the type described herein.
The administration regimen would consist of a primary immunization followed by booster injections at set intervals. The intervals between the primary immunization and the booster immunization, the intervals between the booster injections, and the number of booster immunizations will depend on the antibody titers and duration elicited by the vaccine. It will also depend on the functional efficacy of the antibody responses, namely, levels of antibody titers required to prevent AD development or exerting therapeutic effects in AD patients. A typical regimen will consist of an initial set of injections at 1, 2 and 6 months. Another regimen will consist of initial injections at 1 and 2 months. For either regimen, booster injections will be given either every six months or yearly, depending on the antibody titers and durations. An administration regimen can also be on an as-needed basis as determined by the monitoring of immune responses in the patient.
Identification of AD vaccine epitopes. In order to determine which 8-amino acid fragments ("8-mers") of the Aβ peptide were sufficient to produce an immunogenic response, Applicants systematically scanned the entire length of Aβ42 with small (8 amino acids) overlapping synthetic peptides derived from the naturally occurring Aβ sequence (SEQ ID NO. 1) as shown in Figure 1 (SEQ ID NOS: 2-37). Twenty nine overlapping eight amino acid peptides, spanning the entire length of Aβ42, were synthesized (Figure 2A) for use as antigens. To improve solubility, several of the peptides were modified by the addition of triple lysine (KKK) (SEQ ID NOS: 52, 53, 54, 56, 59, 60, 62, 64 and 65) or glutamine (EEE) (SEQ ID NOS: 50, 51 and 61) residues or the use of a polyethelyene glycol (PEG) (SEQ E) NOS: 55 and 63) spacer. For this reason, peptides spanning the sequences of Aβ corresponding to residues (11-18) and (13-20) were made in multiple forms, the first with a 6-aminohexanoic acid (Aha) spacer plus a functional group for chemical cross-linking at N-terminus and the other form with Aha and the functional group at C- terminus. As a control, Applicants included a longer peptide, Aβ (1-18).
As used herein, the immunogenic fragments may be 8-mer peptides (eight amino acid residues) derived from the naturally occurring, i.e. wild type, or synthetic Aβ (SEQ ID NO:1) or any mutation or variation thereof. Such mutation or variant can be produced by synthetic or recombinant means known to those of ordinary skill in the art. One example of such a variant is the EV substrate (EVEFRHDSGYEVHHQKLVFFAEDVGSNKGAΠGLMVGGVVIA) (SEQ ID NO: 66) a peptide corresponding to Aβ (1-42) in which positions 1 and 2 of wild type Aβ have been varied. Aβ conjugates for use in vaccine formulation
Selection of Aβ conjugates for use in formulating a vaccine was based on the immunogenicity of the 8-mers. In order to determine the immunogenicity of the 8-mer in a species with a sequence identical to the human Aβ sequence, the 29 peptides (Figure 2A) were conjugated to EXH to form an Aβ conjugate and tested in guinea pigs (Figure 3). As a control immunogen, Aβ (1-18)-KLH (SEQ ID NO: 37) was included in this analysis.
Guinea pigs were immunized as described in Example 3.B with conjugated immunogens formulated in alum plus 50 μg of ISCOMATRIX® (CSL, Ltd., Parkville, Australia). In order to distinguish immunogenic from non-immunogenic Aβ42 fragments, guinea pigs were immunized three times at four week intervals. Three weeks after each immunization, blood samples were collected and tested by ELISA for antibody titers against Aβ40 peptide. These titers are shown in Figure 3 as post- dose 1 (PDl), post-dose 2 (PD2) and post-dose 3 (PD3), respectively.
Following the first injection (PDl) some peptide regions elicited appreciable antibody titers as did the 18-mer control. In particular, Aβ conjugates corresponding to Aβ amino acids 1-8, 2-9, 3-10, 17-24, 21-28, and 33-40 all produced titers in excess of 1:800. After the second injection (PD2), 15 of the Aβ conjugates elicited antibody titers in excess of 1: 1000. Analysis at post-dose 3 (PD3) further confirmed that certain regions of Aβ are more immunogenic relative to others. Eleven regions demonstrated titers greater than 1:6000. These included regions corresponding to Aβ amino acids 1-8, 3- 10, 7-14, 11-18, 13-20, 15-22, 19-26, 21-28, 23-30, 27-34 and 29-36. Of these regions, five regions were highly immunogenic (>1: 10000) including: regions 1-8, 15-22, 21-28, 23-30 and 29-36. This data suggests that certain 8-amino acid regions of Aβ are highly immunogenic, while other regions (e.g., 5-12, 25-32, 31-38 and 35-42) are non-immunogenic (titers < 1:300). The results also demonstrate that while the Aβ conjugates were capable of eliciting an Aβ4o peptide-specific antibody response, not all fragments of Aβ were equally immunogenic.
Immunoreactivity of Aβ peptide-KLH conjugates
In order to demonstrate that the immune sera generated from the guinea pigs following immunization with the Aβ peptide-KLH conjugates is relevant to human AD, a study was performed to evaluate the immunoreactivity of polyclonal sera from a guinea pig immunized with an Aβ (3-1O)-KLH (SEQ ID NO: 40) conjugate. The serum sample collected four weeks following the second injection of Aβ (3-1O)-KLH (SEQ ID NO: 40) conjugate from a guinea pig was tested for reactivity with human AD brain tissues by immunohistochemistry (Example 4).
As depicted in Figure 4 the immunogenic response produced by the Aβ (3-1O)-KLH (SEQ ID NO: 40) conjugate produced an antibody response that was directed against human AD brain tissue. Figure 4A demonstrates immunoreactivity of the monoclonal anti-Aβ antibody 6F3D (Vector Laboratories). As shown, this brain has extensive Aβ deposits in a manner expected to be typical for human AD. Figure 4B demonstrates a lack of immunoreactivity of sera from a pre-immunized guinea pig. Figure 4C shows positive immunoreactivity of sera from this same guinea pig following two injections of the Aβ (3-1O)-KLH (SEQ ID NO: 40) conjugate. Collectively, this data demonstrates that the immunogenicity found by ELISA contains a significant antibody response directed against human Aβ found in this AD tissue. These results confirm and extend the unexpected finding of the differential immunogenicity imparted by particular fragments of Aβ to further demonstrate that this response is directed in a manner consistent with a therapeutic application.
Generation of OMPC conjugates and multiple antigenic conjugates
On the basis of immunogenicity in guinea pigs, the relative location of the peptide fragment within the Aβ42 amino acid sequence, the solubility of the Aβ fragments and the feasibility of using OMPC as a carrier protein, Applicants selected seven 8-mers (Figure 2B) for OMPC conjugation. These peptide fragments correspond to the following amino acid regions of Aβ: 1-8, 2-9, 3-10, 7-14, 17- 24, 21-28 and 33-40.
The invention described herein includes multivalent peptide conjugates such as those shown in Figures 5, 6A and 6B. Multivalent branched MAP-OMPC conjugates (Figures 6A and 6B) were generated by using a lysine-based scaffold, whereas multivalent linear 8-mer-OMPC conjugates (Figure 5) were prepared using a PEG linker. Those skilled in the art will appreciate that a PEG linker, compared to conventional amino acid linkers that can also be used herein, offers the advantage of lower immunogenicity and greater peptide solubility. In a preferred embodiment of the invention, the immunogenic fragment is a multivalent MAP conjugated to OMPC. It should be understood by those skilled in the art that such a conjugation is not a 1: 1 ratio of peptide to carrier. Rather, a plurality of peptides is attached in a spherical manner to each OMPC molecule. It will be further appreciated by those skilled in the art that the use of multivalent linear constructs and MAPs will enhance solubility, formulation stability, immunogenicity and the diversity of the polyclonal response.
Immunogenicity of OMPC conjugate vaccines
In an effort to evaluate the immunogenicity of an Aβ peptide - OMPC conjugate and to further evaluate the benefit of an adjuvant with this vaccine construct, Applicants initiated a study in rhesus monkeys. Rhesus monkeys were vaccinated with an Aβ (l-18)-0MPC conjugate (dose based on the Aβ peptide content), which was formulated either in Merck alum adjuvant (MAA) or MAA and
ISCOMATRK® (CSL, Ltd., Parkville, Australia). Blood samples were collected and used to determine the antibody titers against Aβ^. Interim analysis of this ongoing study demonstrated that at post-dose 1 (PDl) the monkeys receiving 5 μg vaccine in alum failed to develop any detectable titers, while those receiving 30 μg vaccine in alum developed low Aβ4o specific titers. All monkeys that received the alum plus ISCOMATRK® formulation developed significant antibody titers. At post-dose 2 (PD2) both doses of the Aβ conjugate in alum alone produced similar titer levels, whereas the cohorts receiving the alum plus ISCOMATRIX® developed 10-fold higher antibody titers relative to the alum alone cohorts. The results of this study confirmed that the Aβ-OMPC conjugate is immunogenic in non-human primates. The data further demonstrated that the efficacy of such a conjugate vaccine is significantly enhanced by a saponin-based adjuvant such as ISCOMATRJX®.
EXAMPLES
EXAMPLE 1
Preparation of Aβ Conjugates This example describes the preparation of Aβ peptide fragments subsequently used for the Aβ conjugates to induce an immune response in the form of antibodies to Aβ.
A. Preparation of Aβ (8-mers) peptides (SEQ ID NOS.: 37-65; Figure 2A)
The peptides intended for conjugation to maleimide derivatized carrier proteins were synthesized with a cysteine residue at the carboxy terminus. The spacer, Aha (6-aminohexanoic acid) was incorporated between the primary peptide sequence and the carboxy terminal cysteine as a structural element for minimizing steric accessibility to carrier protein during conjugation. Additionally, solubilizing residues represented by EEE, KKK or PEG were introduced at the C-terminus in sequences 14,15,16 17,18,19,20,23,24,25,26,27,28,29. The PEG unit was introduced as, O-(N~Fmoc-2- aminoethyl)-O'-(2-carboxyethyl)-undecaethylenegIycol [FmOC-NHCH2CH2O(CH2CH2O)I0 CH2CH2OCH2CH2CO2H].
Starting with Rink Amide MBHA resin the Aβ peptides were prepared by solid-phase synthesis on an automated peptide synthesizer using Fmoc chemistry protocols as supplied by the manufacturer (Applied Biosystems, Foster City, CA). Following assembly the resin bound peptide was deprotected and cleaved from the resin using a cocktail of 94.5% trifluoroacetic acid, 2.5% 1,2- ethanedithiol, 1% triisopropylsilane and 2.5% H2O. Following a two hour treatment the reaction was filtered, concentrated and the resulting oil triturated with ethyl ether. The solid product was filtered, dissolved in 50% acetic acid/H2O and freeze-dried. Purification of the semi-pure product was achieved by RPHPLC using a 0.1% TFA/H2O/acetonitrile gradient on a C-18 support. Fractions were evaluated by analytical HPLC. Pure fractions (>98%) were pooled and freeze-dried. Identity was confirmed by amino acid analysis and mass spectral analysis.
B. Preparation of Aβ peptide-KLH conjugates (SEQ ID NOS.: 37-65; Figure 2A)
For preparing the KLH conjugates, the Aβ peptides (8-mers), 2 mg, containing a C- terminal cysteine was suspended in 1 ml of commercial maleimide conjugation buffer (83 mM sodium phosphate, 0.1 M EDTA, 0.9 M NaCl, 0.02% sodium azide, pH 7.2 (Pierce Biotechnology, Rockford, IL). A 2 mg sample of commercial maleimide-activated KLH (Pierce Biotechnology, Rockford, IL) was added to the peptide and allowed to react at 25 0C for four hours. The conjugate was separated from unreacted peptide and reagents by exhaustive dialysis versus PBS buffer using 100,000 Da dialysis tubing. The amount of peptide incorporated into the conjugate was estimated by amino acid analysis following a 70 hour acid hydrolysis. Peptide concentrations were determined to be between 0.24 and 0.03 mg/ml.
C. Synthesis of bromoacetylated Aβ peptides (SEQ ID NOS.: 67-77; Figure 2B)
Bromoacetylated peptides were prepared by standard t-Boc solid-phase synthesis, using a double coupling protocol for the introduction of amino acids on the Applied Biosystems model 430A automated synthesizer. Starting with p-methylbenzhydrylamine resin the carboxy terminal amino acid t- B oc-Ly s (Fmoc)-OH was introduced followed by the subsequent amino acids in the sequence. Aha was introduced as a spacer to all of these sequences and a PEG unit in sequences 35 and 37 to aid in aqueous solubility. The PEG unit was introduced as O-(N-Boc-2-aminoethyl)-O'-(N-diglycolyl-2-aminoethyl) hexaethyleneglycol [BOC-NHCH2CH2O(CH2CH2O)6CH2CH2NHCOCH2OCH2CO2H]. The amino terminous was capped by the coupling of acetic acid. After assembly of the primary sequence the Fmoc protecting group on the epsilon amino group of the carboxy terminal lysine was removed by treatment with piperidine. Subsequently the N amino group was reacted with Bromoacetic anhydride in methylene chloride as the solvent for 30 minutes. Deprotection and removal of the peptide from the resin support were achieved by treatment with liquid hydrofluoric acid and 10% anisole as a scavenger. The peptides were purified by preparative HPLC on reverse phase C- 18 silica columns using a 0.1% TFA/acetonitrile gradient. Identity and homogeneity of the peptides were confirmed by analytical HPLC and mass spectral analysis.
D. Synthesis of bromoacetylated divalent MAP, Construct No. 8, Figure 6A The synthesis of bromoacetylated branched multiple antigenic peptides (MAPs) is similar to that described in Example l.C. Following coupling of the carboxyterminal Fmoc-Lys(ivDde)- OH [ivDde = 1, (4,4-Dimethyl-2, 6-dioxo-cyclohexylidene)-3-methyl-butyl] to MBHA resin the α-amino Fmoc protecting group was removed using piperidine and the synthesis continued with the introduction of t-Boc-Lys(Fmoc)-OH. After deprotection of the t-Boc group the sequence was extended with the following t-Boc protected amino acids: Alia, Y, G, S, D, H, R, F, E and the amino terminous capped by coupling acetic acid on the ABI synthesizer. The side chain lysine Fmoc protecting group was removed with piperidine and the Nε arm of lysine extended on the ABI synthesizer with the introduction of the following protected amino acids: Aha, H, H, V, E, Y, G, S, D and the amino terminous capped by coupling acetic acid. Removal of the ivDde protecting group was by treatment with 5% hydrazine in dimethylformamide for 5 minutes providing the unblocked N amino group on the carboxy terminal lysine which was further elaborated with bromoacetic anhydride as described in Example l.C. Cleavage of the peptide from the resin, its subsequent purification and characterization are as described in Example l.C.
E. Synthesis of bromoacetylated MAPs, Construct Nos. 11 and 12, Figure 6A MAP Constructs Nos. 11 and 12 were prepared as described in Example l.D.
F. Synthesis of cysteine multivalent MAP, Construct No. 9, Figure 6A
Starting with MBHA resin the following t-Boc protected amino acids were assembled on the ABI automated synthesizer C, Lys(Fmoc), Aha, Y, G, S, D, H, R, F, E followed by coupling with acetic acid. The N amino Fmoc protecting group of lysine was removed and the synthesis continued with the introduction of the following t-Boc protected amino acids: Aha, H, H, V, E, Y, G, S, D followed by coupling with acetic acid. The resin bound peptide was isolated, purified and characterized as in Example l.C. Note: Instead of 10% anisole as in Example l.C, a 1:1 mixture of p-cresol: p-thiocresol was used as a scavenger during HF cleavage.
G. Synthesis of cysteine divalent MAPs, Construct Nos. 10, 13 and 14, Figure 6A
Divalent MAPs, Construct Nos. 10, 13 and 14, Figure 6A, were prepared as described in Example 6.F. The PEG unit was introduced as 0-(N-Boc-2-aminoethyl)-0'-(N-diglycolyl-2-aminoethyl) hexaethyleneglycol (t-Boc-NHCH2CH2O(CH2CH2O)6 CH2CH2NHCOCH2OCH2CO2H).
H. Synthesis of bromoacetylated multivalent MAP, Construct No. 16, Figure 6B
Using the ABI automated synthesizer Fmoc-Lys (t-Boc)-OH was coupled to MBHA resin. Following removal of the t-Boc protecting group on the Nε amino group of lysine the sequence was extended with the introduction of the following t-Boc protected amino acids: Aha, Y,G, S, D, H, R, F, E, followed by coupling of acetic acid. The Na Fmoc protecting group on lysine was removed by manual treatment with piperidine. The sequence was further elaborated (on ABI synthesizer) with the introduction of Fmoc-Lys (t-Boc)-OH followed by the following t-Boc protected amino acids: Aha, H, H, V, E, Y, G, S, D and coupling of acetic acid. The lysine Fmoc N amino protecting group was removed as previously described and the synthesis continued with the introduction of Fmoc-Lys(t-Boc)- OH followed by the t-Boc protected amino acids: Aha, K, N, S, G, V, D, E, A and acetic acid coupling. The N Fmoc protecting on lysine was removed and the synthesis continued with the introduction Fmoc- Lys(t-Boc)-OH followed by the following t-Boc protected amino acids: Aha, V, V, G, G, V, M, L, G and- acetic acid coupling. Following removal of the Na Fmoc protecting group of lysine the resin bound peptide was reacted with bromoacetic anhydride as in Example l.C. Isolation and characterization of the final product was as in Example l.C.
I. Synthesis of multivalent MAPs, Construct Nos. 15 and 17, Figure 6B The synthesis of MAP Aβ conjugates, Construct Nos. 15 and 17, Figure 6B, are as described in Example l.F and I.H.
J. Synthesis of bromoacetylated multivalent linear peptide, Construct No. 1, Figure 5 Starting with MBHA resin the primary sequence was synthesized using t-Boc chemistry on the ABI automated synthesizer as described in Example 6.A. The interspaced PEG units were manually introduced as the Fmoc-l-amino-4, 7, 10-trioxa 13-tridecanamine succinic acid [Fmoc- NHCH2CH2CH2O(CH2 CH2O)2 CH2CH2CH2 NHCOCH2CH2CO2H] using BOP reagent as the coupling agent. Piperidine was used for deprotection of the Fmoc group. Bromoacetylation of the amino terminus was as described in Example l.C. Isolation and characterization of the desired product was as in Example l.C.
K. Synthesis of multivalent linear Aβ peptides, Construct Nos. 2, 5, 6 and 7, Figure 5
The synthesis of multivalent linear Aβ peptides, Construct Nos. 2, 5, 6 and 7 are as described in Example IJ.
EXAMPLE 2 Chemical conjugation of Aβ peptides to OMPC
This example presents the chemical conjugation of peptides derived from human Aβ42 to purified Outer Membrane Protein Complex (OMPC) of Neisseria meningitidis, type B. The chemical nature of the coupling is reaction between haloacetyl-derivatized peptide and thiol-derivatized protein of the membrane complex. Amyloid peptides were synthesized as described above with a bromoacetyl functionality on the N-terminus for divalent linear epitope peptides or on the C-terminus or attached through the epsilon amino group of a lysine residue for monovalent linear and branched MAP forms. The BrAc group was separated from the mature peptide by a spacer consisting of 6-aminohexanoic acid (Aha). Refer to sequences described above. Conjugation will be described for the representative peptide, Aβ (3-10). All manipulation of OMPC-containing solutions was performed in a laminar flow environment following standard aseptic techniques.
A. Thiolation of OMPC
Purified, sterile OMPC, obtainable from a process such as that described in Fu, U.S. Pat. No. 5,494,808 used for the production of PedvaxHIB® and pneumococcal conjugate vaccines, was thiolated on a portion of its surface-accessible lysine residues using the reagent N- acetylhomocysteinethiolactone (NAHT, Aldrich, St. Louis, MO). OMPC in water, 117mg, was pelleted by centrifugation at 289,000 x g for 60 minutes at 4°C and the supernatant was discarded. N2-sparged activation buffer (0.11 M sodium borate, pH 11) was added to the centrifuge tube and the pellet was dislodged with a glass stir rod. The suspension was transferred to a glass Dounce homogenizer and resuspended with 30 strokes. The centrifuge tube was washed and the wash dounced with 30 strokes. Re-suspended pellet and wash were combined in a clean vessel to give a OMPC concentration of 10 mg/mL. Solid DTT and EDTA were dissolved in N2-sparged activation buffer and charged to the reaction vessel at a ratio of 0.106 mg DTT/mg OMPC and 0.57 mg EDTA/mg OMPC. After gentle mixing, NAHT was dissolved in N2-sparged water and charged to the reaction at the ratio of 0.89 mg NAHT/mg OMPC. Reaction proceeded for three hours at ambient temperature, protected from light in a N2 hood. At completion, OMPC was pelleted as described above and re-suspended at 6 mg/mL by Dounce homogenization in N2-sparged conjugation buffer (25 mM sodium borate, pH 8.5, 0.15 M NaCl) to wash the pellet. For final re-suspension, the OMPC was pelleted as above and re-suspended at 10 mg/mL by Dounce homogenization in N2-sparged conjugation buffer. An aliquot was removed for free thiol determination by Ellman assay and the bulk product was stored on ice in dark until use. Measured thiol content was between 0.2 to 0.3 μmol/mL.
B. Conjugation of Aβ peptide to OMPC
Functional BrAc content of peptide was assumed to be 1:1 on a molar basis. Sufficient peptide was weighed to give a 1.6 molar excess of BrAc over total thiol. The targeted total OMPC protein for each conjugation was 15 mg. Peptides were re-suspended in N2-sρarged conjugation buffer at 2.6 mg/mL and slowly added to thiolated OMPC solution. The reactions were protected from light and incubated at ambient temperature for about 22 hours. Residual free OMPC thiol groups were quenched with a 5-fold molar excess of N-ethylmaleimide for 18 hours at ambient temperature. A thiolated OMPC-only control was carried through the conjugation protocol in parallel. Upon completion of quenching, conjugate and control were transferred to 100,000 Da molecular weight cut-off dialysis units and dialyzed exhaustively against at least five changes of conjugation buffer. Upon completion of dialysis, samples were transferred to 15 ml polypropylene centrifuge tubes and centrifuged at 2,280 x g for five minutes at 4°C to remove any aggregated material. Aliquots were removed for analysis and the bulk was stored at 4°C.
C. Analysis of Aβ peptide-OMPC conjugates
Total protein was determined by the modified Lowry assay and samples of conjugate and control were analyzed by quantitative amino acid analysis (AAA). Peptide to OMPC molar ratios were determined from quantitation of the unique residue S-carboxymethylhomocysteine which was released upon acid hydrolysis of the nascent peptide-OMPC bond. The OMPC-specific concentration was determined from hydrolysis-stable residues which were absent from the peptide sequence and thus unique to OMPC protein. Assuming 1 mol of peptide for every mol SCMHC, the ratio of SCMHC/OMPC was thus equivalent to the peptide/OMPC content. The mass loading of peptide could be calculated from this ratio using the peptide molecular weight and an average OMPC mass' of 40,000,000 Da. The covalent nature of the conjugation was qualitatively confirmed by SDS-PAGE analysis using 4-20% Tris-glycine gels (Invitrogen, Carlsbad, CA) where an upward shift in mobility was observed for the Coomassie-stained conjugate bands relative to control.
The calculated molar loading ratios (mol peptide/mol OMPC) for all conjugated BrAc peptides were:
Figure imgf000018_0001
EXAMPLE 3 Immunogenicity of Aβ conjugates
This example describes the formulation and administration of the Aβ conjugates capable of inducing an immune response in the form of antibodies to Aβ.
A. Formulation of vaccine conjugates The Aβ peptide-KLH conjugate vaccines were formulated in ISCOMATRTX® (CSL
Ltd., Parkville, Australia). AU Aβ peptide-OMPC conjugate vaccines were formulated in alum, either
® with or without a second adjuvant, such as the saponin-based adjuvant, ISCOMATRDC (CSL Ltd.,
Parkville, Australia). All the sample manipulations were performed under sterile conditions.
For the alum formulations, conjugates are diluted one times saline at a designated peptide concentration and mixed with two times alum (Merck, Product No. 39943), which corresponds to 900 μg/mL Merck alum prepared in sterile saline (150 mM sterile sodium chloride solution). Thus, target concentration in the vaccine is 450 μg/mL Merck alum or one time Merck alum. Target peptide (antigen) concentrations for animal studies were as follows: for mice - 12.1 μg/mL (Dose 0.1 mL); for monkeys - 10 μg/mL or 60 μg/mL (Dose 0.5 mL) and for guinea pigs - 12.5 μg/mL (Dose 0.4 mL). The mix is incubated for two hours at room temperature. To obtain the injection dose, the alum-absorbed conjugates are diluted with one time alum to reach the target peptide concentration. Where a second ® adjuvant is needed, i.e. ISCOMATRIX, the target concentration was 10 μg/ML for mice studies, 0, 100 or 200 μg/mL for monkey studies and 125 μg/mL for guinea pigs.
1. ISCOMATRIX preparation
® Using a cassette membrane (Slide-A-Lyzer Dialysis Cassett,10K MWCO, Pierce,
® Rockford, IL), ISCOMATRIX is dialyzed into sterile saline solution at 2-8° C. Sterile saline solution is
® changed 2-3 times during dialysis. After completion of dialysis, ISCOMATRIX is filter sterilized using a syringe filter (0.22 uM Millex-GV syringe filter, Millipore, Billerica, MA). The concentration of sterile, dialyzed ISCOMATRIX R is determined by RP-HPLC. ISCOMATRIX is stored sterile at 2-8°C until use. 2. Aβ peptide-OMPC conjugateand Merck alum preparation
Aβ peptide-OMPC conjugate stocks are diluted into sterile IX saline solution. The diluted AD peptide-OMPC conjugate stocks are then added to 2X Merck alum in IX sterile saline solution and mixed for one hour on a rotating wheel at room temperature. The mixture is allowed to settle on the bench top for 15 minutes at room temperature and is then centrifuged at 1500 rpm for ten minutes. The supernatant is decanted off gently (UV analysis of supernatant is performed to determine % Aβ peptide-OMPC conjugate bound to alum) and the pellet is resuspended in sterile IX saline. The mixture is aliquoted into sterile 3 mL tubing glass vials and then stored at 2-80C until final formulation with ISCOMATRIX®.
® 3. Formulation of Aβ peptide-OMPC/alum and ISCOMATRIX vaccine
® Prior to final formulation with ISCOMATRIX, the particle size of the Aβ peptide-
OMPC/alum in saline is determined by static light scattering to confirm binding and monitor particle stability. The sterile, dialyzed ISCOMATRIX R in IX saline is added to Aβ peptide-OMPC/alum in sterile 150 mM NaCl while vortexing. Vials are stoppered, capped and crimped to completely seal. Vaccine is stored at 2-80C prior to injection. Prior to injection, each vaccine is vortexed for 3-5 minutes.
B. Immunogenicity of conjugate vaccines in guinea pigs
Six to ten week old female guinea pigs were obtained from Harlan Inc., Indianapolis, IA and maintained in the animal facilities of Merck research Laboratories in accordance with institutional guidelines. All animal experiments were approved by Merck Research Laboratories Institutional Animal Care and Use Committee (IACUC). Antigens were prepared in phosphate-buffered saline and formulated in the designated adjuvant.
Two animals per group were immunized with the Aβ peptide - E-LH conjugates shown in Figure 2A intramuscularly with 400 μl of a conjugate vaccine (8 μg by peptide content or 50 μg by total conjugate) in the presence of 40 μg of ISCOMATRIX.® The immunizations were performed three times in four-week intervals. Serum samples were collected before first immunization (pre-bleeds) and three weeks after each immunization and stored at 4°C prior to antibody titer determinations. The antibody titers were determined by ELISA according to the protocol that follows using Aβ40 as the target antigen. The ELISA based analysis is as follows: Ninety six-well plates were coated with 50 μl per well of Aβ at a concentration of 4 μg/ml in 50 mM bicarbonate buffer, pH 9.6, at 4°C overnight. Plates were washed with phosphate buffered saline (PBS) and blocked with 3% skim milk in PBS containing 0.05% Tween-20 (milk-PBST). Testing samples were diluted in a 4-fold series in PBST. One hundred μl of a diluted sample was added to each well, and the plates were incubated at 24°C for two hours and then washed six times with PBST. Fifty μl of HRP-conjugated secondary antibodies at 1:5000 dilution in milk-PBST was added per well and the plates were incubated at 24°C for one hour. The plates were washed three times and 100 μl of 1 mg/ml o-phenylenediamine dihydrochloride in 100 mM sodium citrate, pH 4.5 was added per well. After 30 minutes incubation at 240C, the reaction was stopped by adding 100 μl of IN H2SO4 per well, and the plates were read at 490 nm using an ELISA plate reader. The antibody titer was defined as the reciprocal of the highest dilution that gave an OD490 nm value above the mean plus two standard deviations of the conjugate control wells.
The results of this analysis, shown in Figure 3, demonstrated that following the first injection (PDl) some peptide regions elicited appreciable antibody titers as did the 18-mer control. In particular, Aβ peptide fragments corresponding to Aβ amino acids 1-8, 2-9, 3-10, 17-24, 21-28, and 33- 40 all produced titers in excess of 1:800. After the second injection (PD2), 15 of the 8-mer conjugates elicited antibody titers in excess of 1: 1000. Analysis at post-dose 3 (PD3) further confirmed that certain regions of the Aβ peptide were more immunogenic relative to others. Eleven regions demonstrated titers greater than 1:6000. These included regions corresponding to Aβ amino acids 1-8, 3-10, 7-14, 11-18, 13- 20, 15-22, 19-26, 21-28, 23-30, 27-34 and 29-36. Of these regions, five regions were highly immunogenic (>1: 10000) including: regions 1-8, 15-22, 21-28, 23-30 and 29-36. The results demonstrate that 8-mer conjugates are capable of eliciting an Aβ4o specific antibody response. Unexpectedly, and contrary to previous teachings, not all fragments of Aβ were equally immunogenic. In fact, these data suggest that certain 8-mers are highly immunogenic, while other regions of Aβ (e.g., 5-12, 25-32, 31-38 and 35-42) are non-immunogenic (titers < 1:300).
C. Immunogenicity of conjugate vaccines in rhesus monkeys
A study was conducted in non-human primates, i.e. rhesus monkeys, comparable to that done with guinea pigs to determine whether Aβ peptide-OMPC conjugates and an alum and ISCOMATRIX® adjuvant provided an immune response. Rhesus monkeys (Macaca mulatto) were maintained in accordance with the institutional animal care protocols of Merck Research Laboratories and New Iberia Research Center (The University of Louisiana at Lafayette, New Iberia, LA).
Applicants used Aβ peptides conjugated to OMPC as the model antigens, including, the
8-mers shown in Figure 2B: Aβ (1-8) (SEQ. ID NO: 67), Aβ (3-10) (SEQ. ID NO: 69), Aβ (7-14) (SEQ ID NO: 70), Aβ (17-24) (SEQ ID NO. 72), Aβ (21-28) (SEQ ID NO: 73) and Aβ (33-40) (SEQ ID NO.
74); the divalent linear peptides shown in Figure 5: Aβ (3-10) (7-14) (Construct No. 1), Aβ (3-10) (21-
28) (Construct No. 2), Aβ (l-8)(21-28) (Construct No. 5); and the multivalent branched MAPs shown in Figure 6A: Aβ (3-10)(7-14) (Construct No. 8), Aβ (l-8)(21-28) (Construct No. 11), Aβ (3-10) (21-28) (Construct No. 12).
Rhesus macaques (N=3) were immunized with 5 μg of each of the vaccine formulated in Merck alum adjuvant (MAA) plus 100 ug of ISCOMATRK® every four weeks. Serum samples were collected four weeks following each injection and determined for Aβ specific antibody responses by ELISA. Consistent with the results from the guinea pig studies, all conjugates were found to be immunogenic in monkeys. Aβ specific antibody titers were detectable after single injections and further boosted after the subsequent injections. Generally for the conjugates tested, the peak titers were reached after the second or third immunization where geometric mean liters ranged from 25,000 to 500,000. These results confirm the finding that the Aβ conjugates described herein are capable of eliciting an Aβ specific antibody response.
D. Adjuvant effect on immunogenicity of conjugate vaccines in rhesus monkeys
An additional study was conducted in non-human primates, i.e. rhesus monkeys, to determine whether an Aβ peptide-OMPC conjugate and a saponin-based adjuvant, such as
ISCOMATRJX®, can provide an improved immune response. Applicants used an Aβ (1-18) peptide conjugated to OMPC as the model antigen. Rhesus monkeys (Macaca mulatto) were maintained in accordance with the institutional animal care protocols of Merck Research Laboratories and New Iberia Research Center (The University of Louisiana at Lafayette, New Iberia, LA). Five groups of monkeys, three per group, were given the following Aβ (1-18)-OMPC conjugates: (1) 5 μg conjugate (based on peptide content) in alum, (2) 5 μg conjugate in alum + 100 μg ISCOMATRK®, (3) 5 μg conjugate in alum + 50 mg ISCOMATRK®, (4), 30 μg conjugate in alum, (2) 30 μg conjugate in alum + 100 μg ISCOMATRK®. The immunizations were carried out by intramuscular injections in 0.5 ml aliquots at weeks 0, 8 and 24 using tuberculin syringes (Becton- Dickinson, Franklin Lakes, NJ). Serum samples were collected at four week intervals starting from week 0 (pre-bleed) and the tested for antibody titers against Aβ40 by ELISA, performed as described in the preceding example.
Interium analysis of this ongoing study demonstrated that at PDl the monkeys receiving 5 meg conjugate vaccine in alum failed to develop any detectable titers, while those receiving 30 μg conjugate vaccine in alum developed low Aβ4o specific titers. All monkeys that received the alum plus ISCOMATRK® formulation developed significant antibody titers. At PD2, both doses of immunogen in alum alone produced similar titer levels, whereas the cohorts receiving the alum plus ISCOMATRK® developed 10-fold higher antibody titers relative to the alum alone condition. The results of this study confirmed that this Aβ peptide-OMPC conjugate is immunogenic in non-human primates. The data further demonstrate that the efficacy of such a conjugate vaccine is significantly enhanced by a saponin- based adjuvant such as ISCOMATRK®. EXAMPLE 4 Immunoreactivity of guinea pig polyclonal sera
In order to demonstrate that the immune sera generated from the guinea pigs above (Example 3.B) following immunization with 8-mer KLH conjugates is relevant to human AD, a study was performed to evaluate the immunoreactivity of polyclonal sera from a guinea pig immunized with an Aβ (3-1O)-KLH immunogen. Four weeks following a second injection of this construct blood was collected from a representative guinea pig according to the following methodology.
Reactivity of the polyclonal sera was evaluated on human AD brain sections (BioChain Institute, Inc., Hayward, CA). Human brain sections were prepared by incubating at 6O0C for thirty minutes followed by two five minute xylene washes at room temperature. Sections were subsequently immersed in 100% EtOH twice for five minute each followed by a five minute immersion in ddH2θ. Sections were immersed for three minutes in 99% formic acid followed by a brief wash in ddH2θ and a five minute immersion in phosphate buffered solution (PBS). Sections were then incubated with a peroxidase blocker for ten minutes followed by a five minute PBS wash. Sections were blocked by a ten minute exposure to 10% goat serum followed by a five minute wash with PBS. Sections were then incubated with diluted guinea pig sera at 4° C overnight or for one hour at room temperature. Following a five minute PBS wash, sections were incubated for ten minutes with diluted biotinylated goat anti- guinea pig IgG or biotinylated horse anti-mouse antibody (1 drop in 5 ml PBS). Sections were washed for five minutes in PBS and subsequently incubated with ABC solution (Vectorstain ABC kit; Vector Laboratories, Inc.) for thirty minutes. Sections were washed with PBS for five minutes. Sections were then stained with DAB (DakoCytomation) for five minutes and washed with dd H2O. Sections were then counterstained in hematoxylin for thirty seconds and dehydrated in graded EtOH and Xylenes (70% EtOH for five minutes, 80% EtOH for five minutes, 100% EtOH for five minutes and xylenes for five minutes). Sections were then cover-slipped and evaluated by light microscopy. The immunogenic response produced by the Aβ (3-1O)-KLH conjugate produced an antibody response that was directed against human AD brain tissue. As shown in Figure 4, this human brain section has extensive Aβ deposition in a manner typical to that expected for human AD. While pre-immunized guinea pig sera demonstrates a lack of immunoreactivity when exposed to this tissue, positive immunoreactivity of sera from this same guinea pig is noted following two injections of the Aβ (3-10)=-KLH construct. These data demonstrate that the immunogenicity found by ELISA, and illustrated in Figure 3, contains a significant antibody response directed against human Aβ found in this AD tissue. Thus, the results extend the unexpected finding of differential immunogenicity by some Aβ fragments to further demonstrate that this response is directed in a manner consistent with therapeutic application. EXAMPLE 5 Identification of immunogenic fragments lacking T-cell epitopes
To identify immunogenic fragments lacking a T-cell epitope for use in the invention herein, the following Enzyme-Linked ImmunoSpot (ELISpot) assay can be used as a method to assess T- cell responses to a particular antigen. Immunogen fragments possessing T-cell epitopes are identified by the presence of a dark spot on the surface of a white membrane; each spot indicates the presence of a T- cell that has secreted interferon gamma (IFN-γ) in response to the antigen (i.e. immunogenic fragment). Those skilled in the art of vaccines and immunology are familiar with this assay, see, for example, Larsson et ah, AIDS 3: 767-777, 1999, and Mwau et al, AIDS Research and Human Retroviruses 18: 611-618, 2002. A recent review can be found in A.E. Kalyuzhny, Methods MoI Biol. 302: 15-31, 2005.
Applicants used peripheral blood monocytes (PBMCs) from rhesus macaques (New Iberia Research Center, The University of Louisiana at Lafayette, New Iberia, LA) for response to the peptides Aβl-40 (American Peptide Co., Sunnyvale, CA) (amino acid sequence DAEFRHDSGYEVHHQKLVEFAEDVG SNKGAnGLMVGGVV) (SEQ ID NO: 78) and Aβl-20 (Synpep, Dublin, CA) (amino acid sequence DAEFRHDSG YEVHHQKLVFF) (SEQ ID NO: 79).
Purified monoclonal mouse anti-monkey IFN-γ (clone MD-I, Cat No. CT 525,
U-CyTech biosciences, Utrecht, The Netherlands) was diluted in phosphate buffered saline (PBS) with 1% penicillin and streptomycin sulfate (GIBCO® Penicillin-Streptomycin, Cat. No. 15140-122, Invitrogen.Carlsbad, CA), then added to 96-well HTS IP sterile plates (Cat. No. MSIPS4W10, Millipore, Billerica, MA), and incubated for greater than twelve hours at 40C. Plates were washed and RlO [RPMI medium 1640 (GIBCO Cat. No. 11875-093), 10% Fetal bovine serum (HyClone SH30070.03, Logan,
® ®
UT), 0.1 % 50 mM 2-Mercaptoethanol (GIBCO Cat. No. 21985-023), 1 % IM HEPES Buffer (GIBCO
® 15630-080), 1 % 20OmM L-glutamine (GIBCO Cat. No. 25030-081), 1 % 10OmM MEM sodium pyruvate solution (GIBCO R Cat. No. 11360-070), 1% penicillin-streptomycin solution (GIBCO R Cat. No. 15140-122)] was added before incubation for at least two hours at 370C. PBMCs were centrifuged and re-suspended in RlO. PBMCs were counted on a Z2 Coulter counter (Beckman Coulter, Fullerton, CA). Each well of the aspirated plate received either 0.4μg of Aβ 1-40, Aβ 1-20, PHA (phytohemagglutinin, Cat No. L-8902, Sigma, St. Louis, MO, positive control), or diluted DMSO (Sigma, negative control); 400000 PBMCs were then added to each well. Plates were incubated for 18- 24 hours at 370C in a humid CO2 incubator. Plates were washed in PBS with 5% FBS and 0.005%
Tween; biotin-conjugated anti-monkey IFN-γ polyclonal antibodies (U-Cy Tech biosciences, Utrecht, The Netherlands) were diluted in the same media and added to each plate; plates were then incubated at 40C for 18-24 hours. Streptavidin-AP (Cat. No.l3043E, BD PharMingen, Franklin Lakes, NJ) was diluted in the same media and added to washed plates; plates were incubated at room temperature and in the dark for two hours. Filtered 1-Step NBT/BCIP Substrate (Pierce, Rockford, IL, Cat. No. 34042) was added and a further incubation at room temperature in the dark for ten minutes was performed. After washing, plates were allowed to dry before being imaged with a CCD camera and the spots within each well were automatically counted by computer.
Applicants have established that spot forming cells per million PBMCs (SFC/ 106 PBMCs) must exceed 55 and must exceed 4-fold the negative control to be defined as a positive result; failing to meet both these criteria defines a negative result. Rhesus macaques were vaccinated with either a MAP construct comprising Aβ (3-10)/(21-28) (Construct No. 12, Figure 6A) conjugated to OMPC or with both of two monomeric constructs, Aβ (3-10) (SEQ ID NO: 69) and Aβ (21-28) (SEQ ID NO: 73) conjugated to OMPC. Each macaque was assayed during the vaccination regimen at monthly intervals for three or four months; the highest signal ever recorded against either Aβ 1-40 or Aβ 1-20 is only 18 SFC/106 PBMCs, significantly below the 55 SFC/106 PBMCs criterion. Thus, all resulted in a negative score, providing strong evidence that the vaccines did not elicit T-cell responses and, as such, did not include a T-cell epitope.
EXAMPLE 6
Elevation of plasma Aβ
Rhesus macaque non-human primates (N=3) were immunized with 5 μg of the MAP Aβ (3-10)/(21-28) conjugate (Construct No. 12, Figure 6A) or its monomeric constituent conjugate, Aβ (3-
10) (SEQ ID NO: 69) and Aβ (21-28) (SEQ ID NO: 73) linked to OMPC as the carrier and formulated in
® MAA plus 100 μg ISCOMATREX . The rhesus primates received vaccinations every four weeks with bleeds collected and analyzed four weeks following each injection. Anti-Aβ4o titers and total Aβχ.40 levels were determined.
Plasma Aβi.40 levels were determined in these immunized animals using a 6E10/G210 ELISA. This assay measures Aβi.40 using a sandwich ELISA comprising an N-terminal capture antibody 6E10 (Aβ 1-8) (Signet Laboratories, Dedham, MA) and a C-terminal Aβ4o neo-epitope antibody (G210) (The Genetics Company, Inc., Zurich, Switzerland) conjugated with alkaline phosphatase. The antibody, 6E10, was coated onto plates at a concentration of 5ug/ml. Diluted plasma samples (1:3) were used at 50μl/well in triplicates. Aβi_4o standards were prepared from 400 pM - 3pM in 6E10 immuno-depleted rhesus plasma matrix. This assay has a signal-to-noise ratio of about 5-20. The CDP-star alkaline phosphatase substrate was obtained from Applied Biosystems, Foster City, CA.
® SuperBlock, a pre-formulated blocking buffer, was obtained from Pierce Biotechnology, Rockford, IL
(Cat#37515). Counts from individual samples, run in triplicate, were converted to actual analyte concentrations using a third order spline fit to the standards. QC samples were run to evaluate plate to plate variability of the signal. As depicted in Figure 8A, the results of these analyses demonstrated a greater than 3-fold increase in plasma Aβ4Q at PD3 following immunization with the MAP Aβ (3-10)/(21-28) construct (Construct No.12, Figure 6A). This increase in plasma Aβ4Q was not observed in animals immunized with the monomeric Aβ conjugate/OMPC vaccine constructs. Specifically, immunization using either Aβ (3-10) (SEQ ID NO: 69) or Aβ (21-28) (SEQ ID NO: 73) produced a lack of, or appreciably lower, response on this measure. It was notable that these differences were independent of titer levels as depicted in Figure 8B. Collectively, these data demonstrate that some constructs have an advantage relative to other immunogenic constructs with respect to their ability to elevate plasma Aβ levels. Those skilled in the art would appreciate that this selectivity of immunogenic fragments, i.e. the ability to elevate plasma Aβ levels, has not been shown prior to the invention herein and was not predictable from the prior art. As such, the identification of immunogens, either 8-mers or MAPs, lacking a T-cell epitope, that elevate plasma Aβ following immunization, provides a method for selecting said 8-mers or MAPs for use in a vaccine construct. As a result of the invention herein, those skilled in the art are now able to characterize said vaccine constructs both quantitatively (i.e., immunogenicity) and qualitatively (i.e., nature of the polyclonal antibody response - ability to elevate plasma AB levels). It will be further appreciated by those skilled in the art that the invention herein is not limited to 8-amino acid Aβ fragments, but is inclusive of any antigen capable of producing a polyclonal antibody response in the host organism that is reactive to Aβ.

Claims

WHAT IS CLAMED:
1. A pharmaceutical composition comprising an immunogenic fragment of Aβ, lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to said Aβ fragment.
2. An immunogenic fragment of claim 1 wherein said immunogenic fragment is capable of elevating plasma Aβ levels.
3. A composition of claim 2 wherein said immunogenic fragment is selected from the group consisting of a linear 8 amino acid peptide (8-mer) of Aβ, a multivalent linear peptide interspersed with at least one spacer and a multivalent branched multiple antigen peptide (MAP).
4. A pharmaceutical composition of claim 3 further comprising a carrier molecule linked to said composition to form a conjugate, wherein the carrier promotes an immune response comprising antibodies to the Aβ fragment.
5. A pharmaceutical composition of claim 4 wherein the carrier molecule is selected from the group consisting of serum albumins, keyhole limpet hemocyanin, immunoglobulin molecules and Neisseria meningitidies outer membrane protein complex (OMPC).
6. A pharmaceutical composition of claim 5 wherein the carrier molecule is OMPC.
7. A pharmaceutical composition of claim 4 further comprising a pharmaceutically acceptable adjuvant.
8. A pharmaceutical composition of claim 7 wherein the pharmaceutically acceptable adjuvant is selected from the group of an aluminum hydroxide and a saponin-based adjuvant.
9. A pharmaceutical composition of claim 9 wherein the pharmaceutically acceptable adjuvant is a saponin-based adjuvant.
10. A method for preventing or treating a disease associated with amyloid deposits of Aβ in the brain of a patient, comprising administering an effective dose of an immunogenic fragment of Aβ, lacking a T-cell epitope, capable of inducing an immune response in the form of antibodies to said Aβ fragment.
11. A method of claim 10 wherein the immunogenic fragment is capable of elevating plasma Aβ levels.
12. A method of claim 11 wherein the immunogenic fragment is selected from the group consisting of a linear 8 amino acid peptide (8-mer) of Aβ, a multivalent linear peptide interspersed with at least one spacer and a multivalent branched multiple antigen peptide (MAP).
13. A method of claim 12 wherein the immunogenic fragment is linked to a carrier molecule to form a conjugate and where said carrier promotes an immune response comprising antibodies to the Aβ fragment.
14. A method of claim 13 wherein said carrier molecule is selected from the group consisting of serum albumins, keyhole limpet hemocyanin, immunoglobulin molecules and Neisseria meningitidies outer membrane protein complex (OMPC).
15. A method of claim 14 wherein the carrier molecule is OMPC.
16. A method of claim 13 wherein the conjugate is administered with a pharmaceutically acceptable adjuvant.
17. A method of claim 14 wherein the pharmaceutically acceptable adjuvant is selected from the group of an aluminum hydroxide and a saponin-based adjuvant.
18. A method of claim 179 wherein the pharmaceutically acceptable adjuvant is a saponin-based adjuvant.
PCT/US2006/016481 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease WO2006121656A2 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
AU2006246382A AU2006246382A1 (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of Alzheimer's disease
AT06751930T ATE535252T1 (en) 2005-05-05 2006-05-01 PEPTIDE CONJUGATE COMPOSITIONS AND METHODS FOR PREVENTING AND TREATING ALZHEIMER'S DISEASE
CN2006800153890A CN101171031B (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
BRPI0610093-7A BRPI0610093A2 (en) 2005-05-05 2006-05-01 pharmaceutical composition, and method for preventing or treating a disease associated with alpha-beta amyloid deposits in a patient's brain
NZ562434A NZ562434A (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
JP2008510087A JP2008540417A (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for prevention and treatment of Alzheimer's disease
CA002607868A CA2607868A1 (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
US11/919,897 US7850973B2 (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
MX2007013825A MX2007013825A (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease.
EP06751930A EP1879613B1 (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
ZA2007/08635A ZA200708635B (en) 2005-05-05 2007-10-10 Peptide conjugate compositions and methods for the prevention and treatment of alzheimers disease
IL186793A IL186793A0 (en) 2005-05-05 2007-10-18 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
NO20076239A NO20076239L (en) 2005-05-05 2007-12-04 Peptide conjugate preparations and methods for the prevention and treatment of Alzheimer's disease
US12/907,493 US20110052611A1 (en) 2005-05-05 2010-11-08 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US67788605P 2005-05-05 2005-05-05
US60/677,886 2005-05-05

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/907,493 Division US20110052611A1 (en) 2005-05-05 2010-11-08 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease

Publications (2)

Publication Number Publication Date
WO2006121656A2 true WO2006121656A2 (en) 2006-11-16
WO2006121656A3 WO2006121656A3 (en) 2007-01-04

Family

ID=37019042

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/016481 WO2006121656A2 (en) 2005-05-05 2006-05-01 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease

Country Status (16)

Country Link
US (2) US7850973B2 (en)
EP (2) EP2269633A1 (en)
JP (1) JP2008540417A (en)
KR (1) KR20080005260A (en)
CN (1) CN101171031B (en)
AT (1) ATE535252T1 (en)
AU (1) AU2006246382A1 (en)
BR (1) BRPI0610093A2 (en)
CA (1) CA2607868A1 (en)
IL (1) IL186793A0 (en)
MX (1) MX2007013825A (en)
NO (1) NO20076239L (en)
NZ (1) NZ562434A (en)
RU (1) RU2406529C2 (en)
WO (1) WO2006121656A2 (en)
ZA (1) ZA200708635B (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007064917A2 (en) * 2005-11-30 2007-06-07 Abbott Laboratories Methods of preparation of recombinant forms of human beta-amyloid protein and uses of these proteins
WO2008150949A1 (en) * 2007-05-30 2008-12-11 Abbott Laboratories HUMANIZED ANTIBODIES TO Aß(20-42) GLOBULOMER AND USES THEREOF
WO2008156621A1 (en) 2007-06-12 2008-12-24 Ac Immune S.A. Monoclonal anti beta amyloid antibody
EP2020602A1 (en) * 2007-08-02 2009-02-04 Araclon Biotech High sensitivty immunoassays and kits for the determination of peptides and proteins of biological interest
WO2009054537A1 (en) 2007-10-25 2009-04-30 Kagoshima University PEPTIDE VACCINE USING MIMIC MOLECULE OF AMYLOID β-PEPTIDE
WO2009047002A3 (en) * 2007-10-12 2009-08-06 Fond I R C C S Istituto Neurol Products and their use for the diagnosis, prevention, and/or care of human and/or animal pathologies characterised by the anomalous deposition of b-amyloid and/or amyloid-like substance in human and/or animal organs and tissues, and screening method for determining the risk of such pathologies
WO2010002251A1 (en) * 2008-07-01 2010-01-07 De Staat Der Nederlanden, Vert. Door De Minister Van Vws Vaccine against amyloid folding intermediate
WO2010005858A1 (en) * 2008-07-08 2010-01-14 Merck & Co., Inc. Vaccine for the treatment of alzheimer's disease
EP2149380A1 (en) * 2008-07-29 2010-02-03 Medivet Pharma, S.L. Veterinary immunotherapy compositions for the Aged Related Cognitive Dysfunction.
WO2010136487A1 (en) * 2009-05-26 2010-12-02 Araclon Biotech S.L. Albumin-amyloid peptide conjugates and uses thereof
WO2011106732A1 (en) 2010-02-25 2011-09-01 Wyeth Llc Pet monitoring of ab-directed immunotherapy
WO2012072611A1 (en) * 2010-11-29 2012-06-07 Philipps-Universität Marburg SYNTHETIC LIGANDS FOR HUMAN ANTI-Aβ ANTIBODIES
WO2013020723A1 (en) 2011-08-05 2013-02-14 Glaxosmithkline Biologicals S.A. Compositions and uses thereof for the treatment or prevention of alzheimer's disease
US8535673B2 (en) 1997-12-02 2013-09-17 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US20140039155A1 (en) * 2008-10-06 2014-02-06 Commonwealth Scientific And Industrial Research Organisation Amyloid-beta peptide crystal structure
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
EP2952524A1 (en) 2007-10-17 2015-12-09 Janssen Sciences Ireland UC Immunotherapy regimes dependent on apoe status
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
RU2679059C1 (en) * 2018-04-17 2019-02-05 Александр Олегович Морозов Peptide and method of treatment of alzheimer's disease
RU2679080C1 (en) * 2018-04-17 2019-02-05 Александр Олегович Морозов Peptide and method of treatment of alzheimer's disease
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
PT2238166E (en) 2007-10-05 2014-02-11 Genentech Inc Use of anti-amyloid beta antibody in ocular diseases
EP2440234A4 (en) * 2009-06-10 2013-11-06 Univ New York Immunological targeting of pathological tau proteins
RU2607368C2 (en) 2010-07-30 2017-01-10 Ац Иммуне С.А. Safe and functional humanized antibodies
US20120328605A1 (en) * 2010-10-27 2012-12-27 Daniel Larocque Compositions and uses
WO2013059322A2 (en) * 2011-10-17 2013-04-25 Lawrence Steinman Amyloid beta peptide as a therapy for inflammation
WO2015157820A1 (en) * 2014-04-15 2015-10-22 Griffith University Group a streptococcus vaccine
EP3166688A4 (en) 2014-07-08 2017-12-20 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
WO2017027685A2 (en) 2015-08-13 2017-02-16 New York University Antibody-based molecules specific for the truncated asp421 epitope of tau and their uses in the diagnosis and treatment of tauopathy
ES2571055B1 (en) * 2016-02-15 2016-12-28 Araclon Biotech, S.L. Amyloid conjugate and its uses and procedures
EP3444344A4 (en) * 2016-04-14 2020-02-19 TAO Health Life Pharma Co., Ltd. Amylospheroid (aspd)-like structure and pharmaceutical composition
KR101898623B1 (en) 2016-11-10 2018-09-13 주식회사 브레인숲 Paper toy brock
MX2020004338A (en) 2017-10-25 2020-10-14 Janssen Pharmaceuticals Inc Compositions of phosphorylated tau peptides and uses thereof.
CN116063382A (en) * 2022-11-17 2023-05-05 山东大学 Polypeptide material with third-order nonlinear optical performance and preparation method and application thereof

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5494808A (en) 1994-09-15 1996-02-27 Merck & Co., Inc. Defined medium OMPC fermentation process
US7964192B1 (en) * 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US6905686B1 (en) * 1997-12-02 2005-06-14 Neuralab Limited Active immunization for treatment of alzheimer's disease
JP3854481B2 (en) * 2000-11-17 2006-12-06 三菱重工業株式会社 Wet flue gas desulfurization apparatus and wet flue gas desulfurization method
EP1397380B1 (en) * 2001-06-20 2006-12-27 Ramot at Tel Aviv University Ltd. Multiple antigenic peptide displaying multiple copies of an epitope of a plaque-forming polypeptide and methods of using same
MY144532A (en) * 2001-08-20 2011-09-30 Lundbeck & Co As H Novel method for down-regulation of amyloid
EP1594969B1 (en) * 2003-02-01 2015-05-20 Janssen Sciences Ireland UC Active immunization to generate antibodies to soluble a-beta
WO2005058940A2 (en) * 2003-12-17 2005-06-30 Wyeth Immunogenic peptide carrier conjugates and methods of producing same

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BATALIA; COLLINS, ENGELHARD ANNU REV IMMUNOL., vol. 12, 1995, pages 181 - 207,622
BJORKMAN ET AL., NATURE, vol. 329, 1987, pages 506 - 512
MADDEN ET AL., CELL, vol. 75, pages 693 - 708
MADDEN, ANNU REV IMMUNOL., vol. 13, 1995, pages 587 - 622

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8535673B2 (en) 1997-12-02 2013-09-17 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8642044B2 (en) 1997-12-02 2014-02-04 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US8263558B2 (en) 2005-11-30 2012-09-11 Abbott Laboratories Methods of preparation of recombinant forms of human beta-amyloid protein and uses of these proteins
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
WO2007064917A3 (en) * 2005-11-30 2008-03-13 Abbott Lab Methods of preparation of recombinant forms of human beta-amyloid protein and uses of these proteins
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
WO2007064917A2 (en) * 2005-11-30 2007-06-07 Abbott Laboratories Methods of preparation of recombinant forms of human beta-amyloid protein and uses of these proteins
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
WO2008150949A1 (en) * 2007-05-30 2008-12-11 Abbott Laboratories HUMANIZED ANTIBODIES TO Aß(20-42) GLOBULOMER AND USES THEREOF
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
WO2008156621A1 (en) 2007-06-12 2008-12-24 Ac Immune S.A. Monoclonal anti beta amyloid antibody
EP2527366A1 (en) * 2007-06-12 2012-11-28 AC Immune S.A. Monoclonal anti beta amyloid antibody
AU2008267037B2 (en) * 2007-06-12 2014-08-07 Ac Immune S.A. Monoclonal anti beta amyloid antibody
US9585956B2 (en) 2007-06-12 2017-03-07 Ac Immune S.A. Polynucleotides encoding anti-amyloid beta monoclonal antibodies
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
EP2020602A1 (en) * 2007-08-02 2009-02-04 Araclon Biotech High sensitivty immunoassays and kits for the determination of peptides and proteins of biological interest
KR101461538B1 (en) * 2007-08-02 2014-11-14 아라클론 바이오테크, 에스.엘. Highly sensitive immunoassays and kits for the determination of peptides and proteins of biological interest
WO2009015696A1 (en) * 2007-08-02 2009-02-05 Araclon Biotech High sensitivty immunoassays and kits for the determination of peptides and proteins of biological interest
WO2009047002A3 (en) * 2007-10-12 2009-08-06 Fond I R C C S Istituto Neurol Products and their use for the diagnosis, prevention, and/or care of human and/or animal pathologies characterised by the anomalous deposition of b-amyloid and/or amyloid-like substance in human and/or animal organs and tissues, and screening method for determining the risk of such pathologies
EP2952524A1 (en) 2007-10-17 2015-12-09 Janssen Sciences Ireland UC Immunotherapy regimes dependent on apoe status
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
WO2009054537A1 (en) 2007-10-25 2009-04-30 Kagoshima University PEPTIDE VACCINE USING MIMIC MOLECULE OF AMYLOID β-PEPTIDE
WO2010002251A1 (en) * 2008-07-01 2010-01-07 De Staat Der Nederlanden, Vert. Door De Minister Van Vws Vaccine against amyloid folding intermediate
US9045555B2 (en) 2008-07-01 2015-06-02 De Staat Der Nederlanden, Vert. Door De Minister Van Vws Vaccine against amyloid folding intermediate
JP2011526885A (en) * 2008-07-01 2011-10-20 デ スタート デル ネダーランデン、フェルト.ドール デ ミニスター ファン フォルクスヘーゾンドハイド、フェルジイン アン スポルト Vaccine against amyloid folding intermediate
WO2010005858A1 (en) * 2008-07-08 2010-01-14 Merck & Co., Inc. Vaccine for the treatment of alzheimer's disease
EP2149380A1 (en) * 2008-07-29 2010-02-03 Medivet Pharma, S.L. Veterinary immunotherapy compositions for the Aged Related Cognitive Dysfunction.
WO2010012749A1 (en) * 2008-07-29 2010-02-04 Medivet Pharma, S.L. Immunotherapy compositions for the treatment and prevention of amyloidosis
US20140039155A1 (en) * 2008-10-06 2014-02-06 Commonwealth Scientific And Industrial Research Organisation Amyloid-beta peptide crystal structure
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
EP2258398A1 (en) * 2009-05-26 2010-12-08 Araclón Biotech, S. L. Albumin-amyloid peptide conjugates and uses thereof
CN102458478A (en) * 2009-05-26 2012-05-16 阿拉克隆生物技术商业有限公司 Albumin-amyloid peptide conjugates and uses thereof
CN102458478B (en) * 2009-05-26 2014-03-19 阿拉克隆生物技术商业有限公司 Albumin-amyloid peptide conjugates and uses thereof
WO2010136487A1 (en) * 2009-05-26 2010-12-02 Araclon Biotech S.L. Albumin-amyloid peptide conjugates and uses thereof
WO2011106732A1 (en) 2010-02-25 2011-09-01 Wyeth Llc Pet monitoring of ab-directed immunotherapy
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2012072611A1 (en) * 2010-11-29 2012-06-07 Philipps-Universität Marburg SYNTHETIC LIGANDS FOR HUMAN ANTI-Aβ ANTIBODIES
WO2013020723A1 (en) 2011-08-05 2013-02-14 Glaxosmithkline Biologicals S.A. Compositions and uses thereof for the treatment or prevention of alzheimer's disease
WO2013020724A1 (en) 2011-08-05 2013-02-14 Glaxosmithkline Biologicals S.A. Composition comprising a tlr agonist and an antibody specific for an antigen and uses thereof as vaccine
WO2013020722A3 (en) * 2011-08-05 2013-04-04 Glaxosmithkline Biologicals S.A. Composition comprising small peptide as amyloid beta mimotope and adjuvant for use as vaccine against alzheimer's disease
RU2679059C1 (en) * 2018-04-17 2019-02-05 Александр Олегович Морозов Peptide and method of treatment of alzheimer's disease
RU2679080C1 (en) * 2018-04-17 2019-02-05 Александр Олегович Морозов Peptide and method of treatment of alzheimer's disease
WO2019203687A1 (en) * 2018-04-17 2019-10-24 Александр Олегович МОРОЗОВ Peptide and method of treating alzheimer's disease
WO2019203686A1 (en) * 2018-04-17 2019-10-24 Александр Олегович МОРОЗОВ Peptide and method of treating alzheimer's disease

Also Published As

Publication number Publication date
EP1879613B1 (en) 2011-11-30
CA2607868A1 (en) 2006-11-16
ZA200708635B (en) 2008-10-29
EP2269633A1 (en) 2011-01-05
US20090098155A1 (en) 2009-04-16
ATE535252T1 (en) 2011-12-15
EP1879613A2 (en) 2008-01-23
MX2007013825A (en) 2008-01-18
US20110052611A1 (en) 2011-03-03
RU2406529C2 (en) 2010-12-20
NO20076239L (en) 2008-01-30
IL186793A0 (en) 2008-02-09
KR20080005260A (en) 2008-01-10
BRPI0610093A2 (en) 2008-12-09
CN101171031A (en) 2008-04-30
CN101171031B (en) 2011-09-28
AU2006246382A1 (en) 2006-11-16
RU2007145052A (en) 2009-06-10
JP2008540417A (en) 2008-11-20
WO2006121656A3 (en) 2007-01-04
NZ562434A (en) 2010-04-30
US7850973B2 (en) 2010-12-14

Similar Documents

Publication Publication Date Title
US7850973B2 (en) Peptide conjugate compositions and methods for the prevention and treatment of alzheimer&#39;s disease
AU2017203425B2 (en) Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer&#39;s type
RU2390350C2 (en) Active immunisation for creating soluble a-beta antibodies
US9535076B2 (en) Methods and compositions for eliciting an amyloid-selective immune response
JP5559789B2 (en) Immunotherapeutic composition for the treatment of Alzheimer&#39;s disease
JP2006516535A (en) Immunogens and corresponding antibodies specific for high molecular weight aggregation intermediates common to amyloids formed from proteins of different sequences
US20110002949A1 (en) Vaccine for the treatment of alzheimer&#39;s disease
US20220023401A1 (en) Peptide vaccine for prevention and immunotherapy of dementia of the alzheimer&#39;s type
RU2811687C2 (en) Peptide vaccine for prevention and immunotherapy of alzheimer-type dementia
JP2023519104A (en) Peptide immunogens targeting IAPP for preventing and treating disorders associated with aggregated islet amyloid polypeptide (IAPP)
JP2011201902A (en) Active immunization to generate antibody to soluble a-beta

Legal Events

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

Ref document number: 200680015389.0

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 562434

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 186793

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2006246382

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/013825

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2008510087

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2006751930

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2607868

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 11919897

Country of ref document: US

Ref document number: 1020077025682

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006246382

Country of ref document: AU

Date of ref document: 20060501

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 9383/DELNP/2007

Country of ref document: IN

Ref document number: 2007145052

Country of ref document: RU

ENP Entry into the national phase

Ref document number: PI0610093

Country of ref document: BR