WO2010002251A1 - Vaccine against amyloid folding intermediate - Google Patents

Vaccine against amyloid folding intermediate Download PDF

Info

Publication number
WO2010002251A1
WO2010002251A1 PCT/NL2009/050387 NL2009050387W WO2010002251A1 WO 2010002251 A1 WO2010002251 A1 WO 2010002251A1 NL 2009050387 W NL2009050387 W NL 2009050387W WO 2010002251 A1 WO2010002251 A1 WO 2010002251A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
disease
alzheimer
vgsnk
vgsnkg
Prior art date
Application number
PCT/NL2009/050387
Other languages
French (fr)
Inventor
Peter Hoogerhout
Gerarda Petronella Johanna Maria Van Den Dobbelsteen
Original Assignee
De Staat Der Nederlanden, Vert. Door De Minister Van Vws
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 MX2011000134A priority Critical patent/MX2011000134A/en
Priority to ES09773784T priority patent/ES2397641T3/en
Priority to CA2729414A priority patent/CA2729414C/en
Priority to CN2009801306774A priority patent/CN102112488A/en
Priority to AU2009266552A priority patent/AU2009266552B2/en
Priority to EP09773784A priority patent/EP2297196B1/en
Priority to RU2011103448/10A priority patent/RU2546234C2/en
Priority to BRPI0914925A priority patent/BRPI0914925A2/en
Application filed by De Staat Der Nederlanden, Vert. Door De Minister Van Vws filed Critical De Staat Der Nederlanden, Vert. Door De Minister Van Vws
Priority to NZ590324A priority patent/NZ590324A/en
Priority to JP2011516186A priority patent/JP5722770B2/en
Priority to KR1020117002526A priority patent/KR101694788B1/en
Priority to US13/002,174 priority patent/US9045555B2/en
Publication of WO2010002251A1 publication Critical patent/WO2010002251A1/en
Priority to IL210364A priority patent/IL210364A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1716Amyloid plaque core protein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers

Definitions

  • the invention relates to an improved vaccine which can be used to treat the Alzheimer's disease.
  • Alzheimer's disease is a conformational neurodegenerative disorder (Sadowski & Wisniewski 2004, Blennow et al. 2006, Editorials Nature Med. 2006).
  • a characteristic of the disease is formation of plaques in the brain or in brain blood vessels. These plaques originate from a neuronal membrane -bound protein, the amyloid precursor protein.
  • An ⁇ -helical fragment of 38-43 (typically 42) amino acid residues is cleaved enzymatically from the protein thus forming a peptide called "soluble A ⁇ ⁇ probably first adopts an extended conformation and is present in all body fluids. If soluble A ⁇ reaches a high concentration, it will undergo conformational changes and form aggregates.
  • Fibrillar A ⁇ has a cross-beta spine structure (Sawaya et al. 2007) and is eventually deposited in the brain to form the neurodegenerative plaques.
  • a disadvantage of the "pre-aggregated" A ⁇ l-42 suspension is that physical properties of this material are ill-defined.
  • a far more serious problem was induction of meningoencephalitis as a vaccine-related side effect in 6% of the patients during a phase II clinical trial (Check 2002, Gilman et al. 2005).
  • This side effect is caused by a cellular inflammatory reaction, attributed to a ThI cellular response to epitopes located in the central and C-terminal part of A ⁇ 1-42 (McLaurin et al. 2002, Gelinas et al 2004). It has been demonstrated that beneficial antibodies induced by A ⁇ l-42 are directed against the N-terminus (McLaurin et al. 2002, (Lee et al.
  • the present invention provides an improved vaccine, which does not have all the drawbacks of existing vaccines: less to no toxicity and still able to induce an effective antibody response for immunization.
  • the vaccine proposed in the present invention is a new analogue of the ⁇ -amyloid peptide.
  • a peptide comprising the following sequence XiX 2 X 3 VGSN-Z, X 2 X 3 VGSNK-Z or X 3 VGSNKG-Z, wherein Xi is A or G, X 2 is E, G, Q or K, X 3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX 2 X 3 VGSN-Z, X 2 X 3 VGSNK-Z or X 3 VGSNKG-Z.
  • the peptides of the invention are modified peptides which shall be understood herein as peptides that are not a naturally occurring A ⁇ 1-42.
  • Z may also be defined as an agent stabilizing the conformation Of XiX 2 X 3 VGSN (SEQ ID NO:1), X 2 X 3 VGSNK (SEQ ID NO:2) or X 3 VGSNKG (SEQ ID NO:3) as likely adopted in A ⁇ 1-42, preferably as adopted in A ⁇ 1-42.
  • the peptide sequences XiX 2 X 3 VGSN, X 2 X 3 VGSNK and X 3 VGSNKG as identified above correspond respectively to amino acids 22-28 and 23-29 of A ⁇ 1-42.
  • a preferred peptide sequence is X 2 X 3 VGSNK which corresponds to amino acids 22-28 of A ⁇ 1-42.
  • X 1 , X 2 and X 3 as indicated herein come from the presence of several known mutations within the human population in the sequence of A ⁇ 1-42:
  • X 2 is amino acid 22 and is predominantly E in the population.
  • the Arctic (E22G), Dutch (E22Q), and Italian (E22K) mutations are also known.
  • E22 ⁇ another mutation has been identified (E22 ⁇ ) (Tomiyama et al. 2008).
  • X3 is amino acid 23 and is predominantly D.
  • D23N the Iowa mutation has already been identified (D23N). Therefore, it is obvious for the skilled person that if any other mutations would later be identified in a specific portion of A ⁇ 1-42 as identified herein; i.e. amino acid 21-27, 22-28 or 23-29, the sequence of the peptide of the invention may possibly be adapted to take into account of this later identified mutation.
  • a peptide of the invention is preferably tested as set out in example 2: ELISA.
  • the use of peptide-BSA conjugates as coating antigens in the ELISA allows determination of the anti -peptide titre, whereas coating of oligomeric and fibrillar A ⁇ 1-42 allows detection of specific cross-reactivity.
  • any other peptide sequence derived from A ⁇ 1-42 and incorporated in a peptide according to the invention and which is able to adopt the conformation Of XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG as likely adopted in A ⁇ 1-42 is also encompassed by the present invention.
  • the peptide of the invention consists of the formula
  • XiX 2 X 3 VGSN-Z X 2 X 3 VGSNK-Z or X 3 VGSNKG-Z
  • Xi is A or G
  • X 2 is E, G, Q or K
  • X 3 is D or N
  • Z is an agent stabilizing the bend present within the peptide sequence XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG.
  • a peptide of the invention it is critical that the bend present within XiX 2 X 3 VGSN, X 2 X 3 VGSNK or XiX 2 X 3 VGSN is stabilised since we aim at designing a peptide which mimics a conformational epitope present in folded A ⁇ 1-42 as expressed in monomer, soluble oligomer, fibrils, or neurodegenerative plaques. Any way of achieving this stabilisation is encompassed by the present invention.
  • the skilled person after having synthesized such a peptide of the invention may test its conformation by a method known in the art, for example by NMR as referred to in Example 1.
  • one first way of achieving this stabilisation is to cyclise a peptide of the invention. Therefore, a preferred peptide of the invention is a cyclic peptide.
  • the skilled person knows how to cyclise a peptide.
  • the actual cyclization reaction can be performed between any successive positions, including Z, in the sequence.
  • the actual cyclization reaction can be performed on a precursor sequence not yet containing Z, but yielding Z as a result of the cyclization.
  • Cyclisation can be carried out by linking, preferably by covalently linking, the N- terminal amino acid of the peptide sequence, preferably X 1 , X 2 or X 3 in respectively XiX 2 X 3 VGSN-Z, X 2 X 3 VGSNK-Z or X 3 VGSNKG-Z, to Z.
  • the C-terminal amino acid of the peptide sequences XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG is non-engaged in the cyclisation.
  • cyclisation is performed in solid phase.
  • D23 side-chain linked to the solid phase
  • D27 side-chain linked to the solid phase
  • K28 side-chain linked to the solid phase
  • cyclisation is performed between amino acids in the contraloop Z, such as e.g. from D to G (becomes N-G) or from G to K*, if Z is YNGK
  • cyclise in solution e.g. from G25 to S26 or from G to K*, if Z is YNGK. Cyclisation is thought to be important to stabilise the bend present within XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG.
  • Another preferred way of cyclisation of a peptide is to add a cysteine at the N- and C-termini of the peptide sequence, or by adding a cysteine at the N-terminus of the peptide sequence and another one to Z.
  • the presence of two cysteines will allow to carry out a disulfide cyclisation, as is well-known to the skilled person.
  • a second way of achieving this stabilisation is to use Z.
  • Z is an agent stabilizing the bend present within XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG in a peptide of the invention.
  • Z stabilizes the bend present within XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG to ensure the peptide will likely adopt the conformation of folded A ⁇ 1-42.
  • Z stabilizes the bend present within XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG to ensure these peptides will adopt the conformation of folded A ⁇ 1-42.
  • Z may be any agent known to the skilled person as stabilising a bend, a turn or loop.
  • Z may be defined as a "contra-turn" agent with a high probability to form a ⁇ -turn (Hutchinson et al., 1998; Woolfson et al., 1993).
  • Z may be an amino acid, an oligopeptide, a peptide, a polypeptide, a protein, an antigen, a mono- or oligosaccharide, and/or a steroid.
  • Z is a peptide fragment of 8, 7, 6, 5 or 4 amino acid, in increasing preference with decreasing length.
  • the peptide fragment of 4-8 amino acids is a "contra-turn" agent with a high propensity for a ⁇ -turn conformation.
  • a preferred peptide fragment is a tetrapeptide selected from the group consisting of YNGK, TCGV, CGNT, LCGT, LKGT, GAIK, GAIC, AIIK, and AIIC. More preferably, the tetrapeptide is selected from the group consisting of YNGK, TCGV, CGNT, LCGT and LKGT.
  • proteins that may be used for Z are HSA, IgG' s and other serum proteins.
  • antigens are (bacterial) toxins and virus-like particles.
  • Z may also be a steroid scaffold such as described in e.g. Bode et al. (2007, J.Pept.Sci., 13:702-708) Suitable steroid scaffold for use as Z include e.g.
  • bile acids and derivatives thereof such as e.g. cholic acid, deoxycholic acid and methyl 7- ⁇ -acetoxy-3 ⁇ -amino-12 «-amino-5/?- cholan-24-oate.
  • the peptide sequences are connected to the C-3 and C-12 positions of the steroid scaffold, e.g. as described by Bode et al. (2007, supra).
  • Z may be linked to the peptide sequence before cyclisation and optionally cyclised together with the rest of the peptide sequence.
  • Z is preferably a relatively short molecule like an oligopeptide: an amino acid, a dipeptide, a tripeptide, a tetrapeptide, a pentapeptide.
  • the total number of amino acids is preferably ten or eleven. Even more preferably, this number is eleven.
  • Z may comprise or consist of an amino acid present in the corresponding A ⁇ 1-42 sequence to align with the corresponding X 1 X 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG sequence.
  • Z may be linked to the cyclised peptide sequence.
  • Z may be a relatively bigger molecule than in the previous embodiment: a polypeptide or a protein for example.
  • Z is linked to the peptide sequence and is subsequently cyclised with the rest of the peptide sequence.
  • Z is formed as a result of the cyclization reaction.
  • Z being the tetrapeptide as defined above, such as e.g. YNGK.
  • the tetrapeptide comprises at least one of a cysteine and a lysine to allow selective conjugation of the peptide to a carrier molecule as described below.
  • the lysine preferably is a modified lysine such as N 1 ⁇ -(S- acetylmercaptoacety ⁇ lysine (Lys-SAMA).
  • a modified lysine such as N 1 ⁇ -(S- acetylmercaptoacety ⁇ lysine (Lys-SAMA).
  • the presence of at least one of a cysteine and a Lys-SAMA residue in the tetrapeptide allows for selective conjugation of the peptide of the invention to a sulfhydryl-reactive carrier such as a carrier protein.
  • a peptide consisting of the formula X 2 X 3 VGSNK-Z wherein X 2 is E, G, Q or K, X 3 is D or N and Z is an agent stabilizing the bend present within X 2 X 3 VGSNK.
  • Z is YNGK, wherein even more preferably, K in YNGK is a modified lysine (Lys-SAMA) to allow selective conjugation of the peptide.
  • a peptide comprising the following sequence X 3 VGSNKG-Z, wherein X 3 is D or N and Z is an agent stabilizing the bend present within X 3 VGSNKG.
  • Z is YNGK wherein even more preferably, K in YNGK is a modified lysine (Lys-SAMA) to allow selective conjugation of the peptide.
  • a peptide comprising the sequence X 2 X 3 VGSNKGAI-Z wherein X 2 is E, X 3 is D, and Z is a modified lysine (Lys-SAMA) and a peptide comprising the sequence
  • VGSNKG-Z wherein Z is is a modified lysine (Lys-SAMA) were both found to generate antibody responses to the immunizing peptides themselves, however the thus generated antibodies failed to cross react with oligomer or fibrillar A ⁇ 1-42.
  • a peptide of the invention may be present as a single peptide or incorporated into a fusion molecule, such as a fusion protein.
  • a peptide may further be modified by deletion or substitution of one or more amino acids, by extension at the N- and/or C- terminus with additional amino acids or functional groups, which may improve bio- availability, targeting to T-cells, or comprise or release immune modulating substances that provide adjuvant or (co)stimulatory functions.
  • the impact of these modifications is preferably tested on the conformation of the synthetised peptide. This may be done by NMR for example.
  • the conformation of XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG as likely adopted in A ⁇ 1-42, preferably as adopted in A ⁇ 1-42 has not been modified.
  • the optional additional amino acids at the N- and/or C-terminus are preferably not present in the corresponding positions in the amino acid sequence of the protein it derives from, i.e. the A ⁇ 1-42 amino acid sequence.
  • this peptide in order to improve the immunogenicity of a peptide of the invention, this peptide, preferably a cyclic peptide as described above is conjugated to an immunogenic carrier molecule, preferably selectively through linkage of Z and the immunogenic carrier molecule.
  • an immunogenic carrier molecule preferably is a carrier that when conjugated to a peptide of the invention induces an immune response to the peptide of the invention upon administration to a subject such as a mammal.
  • the immunogenic carrier may also have adjuvant-activity as later defined herein.
  • Numerous of immunogenic carrier molecules are known to the skilled person (Hermanson, G. T., 1996, Bioconjugate techniques. Academic Press, San Diego; Drijfhout and Hoogerhout, 2000).
  • Suitable immunogenic carrier molecules include e.g. bacterial toxins or toxoids such as exotoxins and variants thereof with reduced toxicity.
  • Preferred immunogenic carrier molecules include diphtheria toxoid CRM 197 , a serum albumin (e.g. human serum albumin) and tetanus toxoid (Beuvery et al, 1986; Claesson et al, 2005).
  • composition comprising a peptide as defined herein.
  • a composition may be a pharmaceutical composition or a medicament.
  • a peptide or a peptide composition further comprises a pharmaceutical excipient and/or a pharmaceutically acceptable carrier and/or an immune modulator. Any known inert pharmaceutically acceptable carrier and/or excipient may be added to a composition. Formulation of medicaments, and the use of pharmaceutically acceptable excipients are known and customary in the art and for instance described in Remington; The Science and Practice of Pharmacy, 21 nd Edition 2005, University of Sciences in Philadelphia.
  • a pharmaceutical composition may further comprise pharmaceutically acceptable stabilizing agents, osmotic agents, buffering agents, dispersing agents, and the like.
  • the preferred form of the pharmaceutical composition depends on the intended mode of administration and therapeutic application.
  • the pharmaceutical carrier can be any compatible, non-toxic substance suitable to deliver the active ingredients, i.e. a peptide to a patient.
  • Pharmaceutically acceptable carriers for intranasal delivery are exemplified by water, buffered saline solutions, glycerin, polysorbate 20, cremophor EL, and an aqueous mixture of caprylic/capric glyceride, and may be buffered to provide a neutral pH environment.
  • compositions of the invention are preferably administered by bolus injection.
  • a typical pharmaceutical composition for intramuscular injection would be made up to contain, for example, 1 - 10 ml of phosphate buffered saline and 1 to 100 ⁇ g, preferably 15-45 ⁇ g of a modified conjugated peptide.
  • the active ingredient can be administered in liquid dosage forms, such as elixirs, syrups, and suspensions.
  • Liquid dosage forms for oral administration can contain colouring and flavouring to increase patient acceptance.
  • any known immune modulator in particular modulators leading to a balanced Th2/Thl response, like aluminium phosphate or aluminium hydroxide, may be added to a composition.
  • the immune modulator is an adjuvant.
  • the composition comprises a peptide as earlier defined herein and at least one adjuvant.
  • An adjuvant is herein defined to include any substance or compound that, when used in combination with a peptide, to immunise a mammal, preferably a human, stimulates the immune system, thereby provoking, enhancing or facilitating the immune response against the peptide, preferably without generating a specific immune response to the adjuvant itself.
  • Preferred adjuvants enhance the immune response against a given antigen by at least a factor of 1.5, 2, 2.5, 5, 10 or 20, as compared to the immune response generated against the peptide under the same conditions but in the absence of the adjuvant.
  • Tests for determining the statistical average enhancement of the immune response against a given peptide as produced by an adjuvant in a group of animals or humans over a corresponding control group are available in the art.
  • An adjuvant as used herein will usually be a compound that is foreign to a mammal, thereby excluding immunostimulatory compounds that are endogenous to mammals, such as e.g. interleukins, interferons and other hormones.
  • a composition of the present invention may contain at least one adjuvant.
  • An adjuvant used in the present invention will be selected so that the effect of the peptide is not inhibited.
  • Adjuvants used in the present invention are those which are physiologically acceptable to humans, these include, but are not limited to aluminium hydroxide, aluminium phosphate, oil/surfactant based emulsion adjuvants such as Montanide TM in which different surfactants (especially mannityl oleate) are combined with a mineral oil, squalene-containing emulsions such as MF59TM, monophosphoryl lipid A, or Neisseriae mutant lipopolysaccharide (as described in PCT/NL98/0063).
  • a medicament may be administered as a single administration.
  • the administration of a peptide as earlier herein defined and/or an adjuvant may be repeated if needed and/or distinct peptides and/or distinct adjuvants may be sequentially administered.
  • Peptide, composition and medicament of the invention are preferably formulated to be suitable for intravenous or subcutaneous, or intramuscular administration, although other administration routes can be envisaged, such as mucosal administration or intradermal and/or intracutaneous administration, e.g. by injection.
  • a peptide as described herein is for use as a medicament. More preferably, this medicament is a vaccine against the Alzheimer's disease. Even more preferably, the medicament is for preventing, delaying and/or treating the Alzheimer's disease.
  • a vaccine as defined herein may be used for the prophylactic protection against the Alzheimer's disease or for the treatment of this disease.
  • an organism or an individual or a subject may be an animal or a human being.
  • the organism is a human being.
  • an organism treated is suspected to have a high risk of developing the Alzheimer's disease due for example to potential genetic predisposition, and/or to the age of the subject and/or to the lifestyle of a subject (for example nutritional habit and/or to the absence of physical activity) and/or to any other known parameter indicating this subject has an increased risk of developing the Alzheimer's disease.
  • prevention shall be understood to include complete prevention, prophylaxis, as well as lowering the individual's risk of falling ill with said disease or condition and delaying the onset of the disease or condition.
  • prevention thus also comprises the treatment of persons suspected to be at risk falling ill with said disease or condition.
  • the term shall also be understood to include alleviation of symptoms already developed.
  • delay means administration of a peptide to an organism, i.e. a patient being in a pre-stage of the condition to be treated in which patients a pre-form of the corresponding condition is diagnosed by methods known in the art.
  • treatment or “treating” is understood the management and care of a patient for the purpose of combating the disease, condition, or disorder.
  • “treating the Alzheimer's disease and/or delaying its progression” preferably means that a therapeutically effective amount of a peptide is added. It refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, i.e. treat the Alzheimer's disease and/or delay its progression.
  • the amount of a peptide may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the pharmacological agent to elicit a desired response in the individual.
  • a therapeutic effect (leading to treating the Alzheimer's disease and/or delaying its progression) is preferably one that results in at least one of:
  • a reduction in the load of beta plaques present in the brain of a treated patient preferably means that the amount of a peptide added will be able to prevent de novo formation of beta plaques to at least some extent and/or that existing plaques will be to at least some extent inhibited in their ability to expand.
  • the deposition of beta plaques will not increase in a treated patient in terms of beta plaque load as identified by using an imaging technique such as PET scan (Henriksen, Yousef ⁇ et al., 2008) and/or magnetic resonance imaging (MRI) (O'Brien, 2007).
  • the quantity of beta plaque load is proportional to the tracer uptake.
  • the load of beta plaques will decrease of at least 2%, 5%, 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more. Even more preferably, no beta plaque will be detected.
  • the visualisation of beta plaque should be done approximately at least one day, at least one week, at least one month after vaccination or more. (Meyer- Luehmann, Spires-Jones et al., 2008). If need be, one may decide to vaccinate several times and to regularly monitor the load of beta plaques.
  • a reduction of the amount of soluble A ⁇ oligomers or pref ⁇ brils present in the brain of a treated patient preferably means that the amount of a peptide added will be able to prevent de novo formation of soluble A ⁇ oligomers or pref ⁇ brils to at least some extent and/or that existing soluble A ⁇ oligomers or pre fibrils will be to at least some extent inhibited in their ability to expand.
  • the amount of soluble A ⁇ oligomers or pref ⁇ brils will not increase in a treated patient in terms of a surface as identified by an imaging technique as defined above.
  • the amount of soluble A ⁇ oligomers or pref ⁇ brils will decrease of at least 2%, 5%, 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more. More preferably, the amount of soluble A ⁇ oligomers or pref ⁇ brils will not be detectable using the same method.
  • a soluble A ⁇ oligomer, pref ⁇ bril or protofibril is an indication for an assembly of 2-24 A ⁇ monomers (Haas and Selkoe 2007).
  • a reduction of the severity of a symptom associated with Alzheimer's disease preferably means an improvement of cognition as measured with a psychological test to assess the improvement of cognition in patients suffering from the Alzheimer's disease.
  • the use of a peptide or of a composition as defined herein for the manufacture of a medicament against the Alzheimer's disease is a vaccine. More preferably, the vaccine is for preventing, delaying and/or treating the Alzheimer's disease.
  • a method of preventing, delaying and/or treating the Alzheimer's disease by administering a peptide or a composition as defined herein to a patient in a need thereof.
  • the invention is not limited to any ways of generating a peptide as long as the generated peptide comprises, consists or overlaps with any of the given modified sequences as identified herein and had the required conformation as earlier defined herein.
  • a method for producing a modified cyclic peptide as defined herein comprising the following steps: a) synthesising a cyclic peptide comprising the sequence X 1 X 2 X 3 VGSN-Z,
  • X 2 X 3 VGSNK-Z or X 3 VGSNKG-Z wherein Xi is A or G, X 2 is E, G, Q or K, X 3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX 2 X 3 VGSN, X 2 X 3 VGSNK or X 3 VGSNKG; and b) optionally conjugating an immunogenic carrier molecule to the cyclic peptide obtained in b), preferably through linkage of Z to the immunogenic carrier molecule.
  • an antibody directed against a modified (cyclic) peptide of the invention as defined herein.
  • the skilled person knows how to produce such an antibody in an animal. Methods for generating antibodies or antibody- fragments that specifically bind to a given polypeptide are described in e.g. Harlow and Lane (1988, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY) and WO 91/19818; WO 91/18989; WO 92/01047; WO 92/06204; WO 92/18619; and US 6,420,113 and references cited therein.
  • the term "specific binding,” as used herein, includes both low and high affinity specific binding.
  • Specific binding can be exhibited, e.g., by a low affinity antibody or antibody-fragment having a Kd of at least about 10 "4 M. Specific binding also can be exhibited by a high affinity antibody or antibody-fragment, for example, an antibody or antibody-fragment having a Kd of at least about of 10 "7 M, at least about 10 "8 M, at least about 10 "9 M, at least about 10 "10 M, or can have a Kd of at least about 10 "11 M or 10 "12 M or greater.
  • a method for diagnosing a neurodegenerative disease or condition comprising determining the presence or absence of a beta amyloid plaque (i.e. neurodegenerative plaque) in the brain of a patient using an antibody as defined herein.
  • a beta amyloid plaque i.e. neurodegenerative plaque
  • the presence of a beta amyloid plaque in the subject's brain (or a sample thereof) is indicative for the subject being at risk of developing a neurodegenerative disease or condition such as Alzheimer's disease or indicative for the diagnosis of the neurodegenerative disease or condition, such as Alzheimer's disease.
  • such method is used for prognosing or diagnosing Alzheimer's disease in the brain of a patient.
  • diagnosis means either a predictive risk assessment of a subject for developing later the Alzheimer's disease or preferably an assessment of the development of the Alzheimer's disease in a subject.
  • a subject may be an animal or a human being.
  • a subject is a human being.
  • the method is carried out in vitro or ex vzVo in a sample obtained from a subject.
  • the sample preferably comprises brain tissue isolated from a subject. More preferably, the tissue is brain blood vessel.
  • a detection of the presence of a beta amyloid plaque is revealed by the binding of an antibody of the invention to a brain sample as assayed by ELISA as explained in the example.
  • the diagnosis method may be sequentially applied to a subject to monitor the development of the disease.
  • the verb "to comprise” and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded.
  • the verb "to consist” may be replaced by "to consist essentially of meaning that a peptide or a composition as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention.
  • reference to an element by the indefinite article “a” or “an” does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements.
  • the indefinite article “a” or “an” thus usually means “at least one”.
  • FIG. 1 Model of an A ⁇ l-42 fibril.
  • a and B represent a dimeric cross- ⁇ unit and C the assembled fibril. [Credit: Olofsson et al. 2006 J. Biol. Chem. 281, 477-483].
  • Figure 2 Sequences of amyloid-derived cyclic peptides (K* is a modified lysine residue for conjugation purposes).
  • Figure 3 IgG-antibody titres of pooled mice sera against homologous peptide-BSA conjugates. The titre is the 10 log of the reciprocal serum dilution at 50% of the maximum optical density at 450 nm in ELISA.
  • Figure 4 OD450nm of pooled mice sera as a function of dilution with coating oligomeric or fibrillar A ⁇ 1-42.
  • Figure 5 IgG antibody titers of individual mice sera of groups 8 and 9 against oligomeric or fibrillar A ⁇ 1-42 The titre is the reciprocal serum dilution at 50% of the maximum optical density at 450 nm in ELISA.
  • Figure 6 Immunohistochemical staining of human brain section donor 99-30 (Braak 6) with a mouse serum (1 :300) immunized with cyclo[A ⁇ (22-28)-YNGK]/tetanus toxoid conjugate (a) and the control monoclonal 6E10 (1 :15.000) in (b). Similar pattern of plaques were recognized by the mouse sera as positive control. (Microscope: Leica DMRE fitted with a DC300 camera)
  • a ⁇ l-42 fibrils has been resolved by NMR spectroscopy (Olofsson et al. 2006). Hydrogen/deuterium exchange experiments with fibrils showed that the regions GIu -GIy and Lys -Ala in the A ⁇ sequence are shielded from solvent, whereas the N-terminus Asp'-Tyr 10 and the two-residue fragment Ser 26 -Asn 27 are solvent-accessible.
  • the NMR data are consistent: Asp'-Tyr 10 and the two-residue fragment Ser 26 -Asn 27 are solvent-accessible.
  • the NMR data are consistent with a model of the fibril as depicted in Figure 1C. The predicted structure is a twisted cross- ⁇ spine.
  • Figure IA and IB show a section, the dimeric cross- ⁇ unit.
  • each monomer contains two antiparallel ⁇ sheets which are connected by a turn composed of
  • Example 2 specificity of some of the modified cyclised peptides for a conformational epitope of folded A ⁇ 1-42
  • the ⁇ -(2,4-dimethoxybenzyl) ester of N ⁇ -fluorenylmethoxycarbonyl-L-aspartic acid was coupled through its side-chain to a polymer for the synthesis of peptide amides (for later conversion of the starting Asp into Asn).
  • the resin-bound linear peptide was converted to cyclo[GK*EDVGSNKYD(resin)].
  • cyclo[GK*EDVGSNKYD] ⁇ cyclo [EDVGSNKYNGK*] the peptide from group 9 (Table 1), was obtained.
  • the peptides from groups 4-8 were prepared similarly.
  • the peptide from group 10, cyclo [AEDVGSNYNGK*] was prepared from the side-chain-protected resin-bound linear precursor YNGK* AED VGSD(resin).
  • the peptides were purified by reverse- phase high performance liquid chromatography and characterized by ion-spray mass spectrometry (MH + f 0U nd/caicuiated, see table 1).
  • the purified peptides were coupled to either bromoacetylated tetanus toxoid or maleimidyl-modified bovine serum albumin (BSA) (modifying reagent: NHS-PEO 2 -Maleimide, Pierce) (Drijfhout JW et al., 2000).
  • BSA bovine serum albumin
  • Lyophilized A ⁇ 1-42 (Anaspec) was dissolved in trifluoroacetic acid at a concentration of 1.0 mM, left to stand at room temperature for 1 h and dried under a stream of nitrogen and, thereafter, in a vacuum (1 mm Hg) for 15 min. The peptide was then redissolved in hexafluoroisopropanol at a concentration of 1.0 mM and, after 1 h of incubation at room temperature, dried as described above (Zeng et al., 2001). The peptide was stored at -20 0 C for 18-20 h.
  • oligomeric or fibrillar A ⁇ (l-42) Disaggregated of A ⁇ 1-42 was dissolved in dimethyl sulfoxide at a concentration of 5.0 mM, diluted 50-fold with either phosphate buffered saline (PBS), pH 7.2, or 10 mM hydrochloric acid.
  • PBS phosphate buffered saline
  • pH 7.2 pH 7.2
  • 10 mM hydrochloric acid phosphate buffered saline
  • the solution in PBS was incubated at 4 0 C for 24 h (to give oligomers), whereas the solution in 10 mM HCl was incubated at 37 0 C for 24 h (to give fibrils) (Stine et al).
  • mice Groups of eight female Balb/c mice of 6-8 weeks of age were immunized subcutaneously on days 0 and 28 with either 25 ⁇ ig A ⁇ 1-42 in PBS without adjuvant or with 50 ⁇ g peptide -TTd conjugate and 75 ⁇ g A1PO4 in PBS. Small serum samples were collected on day 0. The mice were bled on day 42.
  • ELISA Microtitre plates (Greiner 655092) were coated with A ⁇ l-42 or peptide-BSA conjugates.
  • Freshly prepared A ⁇ 1-42 oligomers or fibrils were diluted to a final concentration of 2.5 ⁇ M (11.3 ⁇ g/ml) in 0.04 M sodium carbonate/bicarbonate buffer, pH 9.7.
  • Peptide-BSA conjugates in phosphate buffered saline, pH 7.2 (PBS) had a total protein concentration of 0.5 ⁇ g/ml. Aliquots (100 ⁇ l) of these solutions were transferred into wells of the plates. The plates were incubated for 90 min at 37 0 C. The plates were further processed as described earlier (Westdijk, Van den Ijssel et al, 1997).
  • mice sera (l :300;first antibody) in 3% NDS + 0.4% TritonXIOO in 0.05 M PB was added and incubated O/N at RT in a box with wet tissues. Sections were washed with 0.05M PB; at least 30 min with one or more exchanges. Then sections were incubated with Donkey-anti-Mouse -Cy 3 1 : 1400 in 0.05 M PB for 2 hours. Sections were washed with 0.05M PB; at least 30 min with one or more exchanges. Sections were sealed in Vectashield with Dapi (Vector). Mouse Monoclonal 6E10 to beta amyloid 1-17 (Abeam, Cambridge, UK) was used as positive control (1 :15,000). Reference list
  • Prototype Alzheimer's disease vaccine using the immunodominant B cell epitope from ⁇ -amyloid and promiscuous T cell epitope pan HLA DR-binding peptide. J. Immunol. 174, 1580-1586.
  • Amyloid-beta immunotherapy for the prevention and treatment of Alzheimer's disease lessons from mice, monkeys, and humans. Rejuvenation Res. 9, 77-84. Lemere,C.A., Maier,M., Peng,Y., Jiang,L., SeabrookJ.J., 2007. Novel Abeta immunogens: is shorter better? Curr.Alzheimer Res. 4, 427-436.
  • Short amyloid ⁇ (A ⁇ ) immunogens reduce cerebral A ⁇ load and learning deficits in an Alzheimer's disease mouse model in the absence of an A ⁇ -specif ⁇ c cellular immune response. J. Neurosci. 26, 4717-4728.
  • Therapeutically effective antibodies against amyloid-beta peptide target amyloid-beta residues 4-10 and inhibit cytotoxicity and f ⁇ brillogenesis. Nature Med. 1263-1269.
  • Alzheimer amyloid- ⁇ -(l-42) fibrils as determined by solution NMR spectroscopy. J.
  • Crystal structure of an anti -meningococcal subtype P 1.4 PorA antibody provides basis for peptide -vaccine design. J. MoI. Biol. 351, 1070-1080. Sadowski M and Wisniewski T, 2004. Vaccines for conformational disorders. Expert

Abstract

The invention relates to an improved vaccine to treat Alzheimer's disease.

Description

Vaccine against amyloid folding intermediate
Field of the invention
The invention relates to an improved vaccine which can be used to treat the Alzheimer's disease.
Background of the invention
It is estimated by the World Health Organization that 18 million people suffer from Alzheimer's disease worldwide (Vas et al. 2001). In the Netherlands, approximately 250,000 people have the Alzheimer's disease. The problem is expanding with increasing average age of the population. Care for a patient in a nursing home is estimated to cost €30,000-60,000 per year (McDonnell et al. 2001). Vaccination would be cost-effective.
Alzheimer's disease is a conformational neurodegenerative disorder (Sadowski & Wisniewski 2004, Blennow et al. 2006, Editorials Nature Med. 2006). A characteristic of the disease is formation of plaques in the brain or in brain blood vessels. These plaques originate from a neuronal membrane -bound protein, the amyloid precursor protein. An α-helical fragment of 38-43 (typically 42) amino acid residues is cleaved enzymatically from the protein thus forming a peptide called "soluble Aβ β probably first adopts an extended conformation and is present in all body fluids. If soluble Aβ reaches a high concentration, it will undergo conformational changes and form aggregates. A plethora of aggregates has been found in vitro or in vivo, including multiple monomer conformers, different types of oligomers, Aβ-derived diffusable ligands, protofibrils, fibrils, and spheroids (adopted from Klein et al. 2004). Fibrillar Aβ has a cross-beta spine structure (Sawaya et al. 2007) and is eventually deposited in the brain to form the neurodegenerative plaques.
Immunization of transgenic mice (Schenk et al. 1999) and human patients in a phase I clinical trial (Hock et al. 2002) with a suspension of "pre-aggregated" Aβl-42 seemed to be beneficial. Antibodies in human immune sera recognized plaques, Aβ deposits and β-amyloid in brain blood vessels. The antibodies did not recognize the amyloid precursor protein or soluble Aβ.
A disadvantage of the "pre-aggregated" Aβl-42 suspension is that physical properties of this material are ill-defined. However, a far more serious problem was induction of meningoencephalitis as a vaccine-related side effect in 6% of the patients during a phase II clinical trial (Check 2002, Gilman et al. 2005). This side effect is caused by a cellular inflammatory reaction, attributed to a ThI cellular response to epitopes located in the central and C-terminal part of Aβ 1-42 (McLaurin et al. 2002, Gelinas et al 2004). It has been demonstrated that beneficial antibodies induced by Aβl-42 are directed against the N-terminus (McLaurin et al. 2002, (Lee et al. 2005). It was therefore proposed to use C-terminally truncated Aβ peptides as immunogens (Sigurdsson et al. 2004, Lemere et al. 2006, Gevorkian et al. 2004, Lemere et al. 2007). Such short peptides are poorly immunogenic. In order to increase the immunogenicity, multiple copies of the peptide should be coupled to non-immunogenic carriers (with the aim of inducing IgM) or to carriers providing heterologous T cell epitopes (Agadjanyan et al. 2005, Ghochikyan et al. 2006, Maier et al. 2006, (Movsesyan et al. 2008)). In neither of these conjugates the peptide is expected to adopt the conformation of residues 4-10 as exposed by the β amyloid oligomers or pre-fibrils. Thus, antibodies induced with truncated peptide conjugates are expected to be weakly specific for the oligomers or pre-fibrils.
Therefore, there is still a need for an efficient medicament, preferably a vaccine against the Alzheimer's disease. The present invention provides an improved vaccine, which does not have all the drawbacks of existing vaccines: less to no toxicity and still able to induce an effective antibody response for immunization. The vaccine proposed in the present invention is a new analogue of the β-amyloid peptide.
Description of the invention Peptides of the invention In a first aspect of the invention, there is provided a peptide comprising the following sequence XiX2X3VGSN-Z, X2X3VGSNK-Z or X3VGSNKG-Z, wherein Xi is A or G, X2 is E, G, Q or K, X3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX2X3VGSN-Z, X2X3VGSNK-Z or X3VGSNKG-Z. The peptides of the invention are modified peptides which shall be understood herein as peptides that are not a naturally occurring Aβ 1-42. Z may also be defined as an agent stabilizing the conformation Of XiX2X3VGSN (SEQ ID NO:1), X2X3VGSNK (SEQ ID NO:2) or X3VGSNKG (SEQ ID NO:3) as likely adopted in Aβ 1-42, preferably as adopted in Aβ 1-42. The peptide sequences XiX2X3VGSN, X2X3 VGSNK and X3 VGSNKG as identified above correspond respectively to amino acids 22-28 and 23-29 of Aβ 1-42. A preferred peptide sequence is X2X3VGSNK which corresponds to amino acids 22-28 of Aβ 1-42. The different possible identities for X1, X2 and X3 as indicated herein come from the presence of several known mutations within the human population in the sequence of Aβ 1-42: X2 is amino acid 22 and is predominantly E in the population. However, the Arctic (E22G), Dutch (E22Q), and Italian (E22K) mutations are also known. Recently, another mutation has been identified (E22Δ) (Tomiyama et al. 2008). X3 is amino acid 23 and is predominantly D. However, the Iowa mutation has already been identified (D23N). Therefore, it is obvious for the skilled person that if any other mutations would later be identified in a specific portion of Aβ 1-42 as identified herein; i.e. amino acid 21-27, 22-28 or 23-29, the sequence of the peptide of the invention may possibly be adapted to take into account of this later identified mutation.
Several overlapping peptide sequences were tested (see the example). As far as we are aware, two of the tested peptide sequences (aa 22-28 or 23-29) were found able to induce an antibody response in mice, which antibody was specifically able to recognize a conformational epitope of Aβ 1-42 as expressed in monomer, soluble oligomer (Haass and Selkoe 2007, Lambert et al, 2007, and Wash and Selkoe, 2007) fibrils, or neurodegenerative plaques. It seems that the recognition of oligomeric Ab is even more crucial than the recognition of fibril or plaque, since oligomeric Ab is more toxic to neurons. Clearance of soluble oligomers rapidly improves cognition while plaques are still present. The functionality of a peptide of the invention is preferably tested as set out in example 2: ELISA. The use of peptide-BSA conjugates as coating antigens in the ELISA allows determination of the anti -peptide titre, whereas coating of oligomeric and fibrillar Aβ 1-42 allows detection of specific cross-reactivity. The skilled person will understand that any other peptide sequence derived from Aβ 1-42 and incorporated in a peptide according to the invention and which is able to adopt the conformation Of XiX2X3VGSN, X2X3VGSNK or X3VGSNKG as likely adopted in Aβ 1-42 is also encompassed by the present invention. In an embodiment, the peptide of the invention consists of the formula
XiX2X3VGSN-Z, X2X3VGSNK-Z or X3VGSNKG-Z, wherein Xi is A or G, X2 is E, G, Q or K, X3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX2X3VGSN, X2X3VGSNK or X3VGSNKG. In a peptide of the invention, it is critical that the bend present within XiX2X3VGSN, X2X3VGSNK or XiX2X3VGSN is stabilised since we aim at designing a peptide which mimics a conformational epitope present in folded Aβ 1-42 as expressed in monomer, soluble oligomer, fibrils, or neurodegenerative plaques. Any way of achieving this stabilisation is encompassed by the present invention. The skilled person after having synthesized such a peptide of the invention may test its conformation by a method known in the art, for example by NMR as referred to in Example 1.
In one preferred embodiment, one first way of achieving this stabilisation is to cyclise a peptide of the invention. Therefore, a preferred peptide of the invention is a cyclic peptide. The skilled person knows how to cyclise a peptide. The actual cyclization reaction can be performed between any successive positions, including Z, in the sequence. In addition, the actual cyclization reaction can be performed on a precursor sequence not yet containing Z, but yielding Z as a result of the cyclization. Cyclisation can be carried out by linking, preferably by covalently linking, the N- terminal amino acid of the peptide sequence, preferably X1, X2 or X3 in respectively XiX2X3VGSN-Z, X2X3VGSNK-Z or X3VGSNKG-Z, to Z. In this way, the C-terminal amino acid of the peptide sequences XiX2X3VGSN, X2X3VGSNK or X3VGSNKG is non-engaged in the cyclisation. Conveniently, cyclisation is performed in solid phase. For example, D23, side-chain linked to the solid phase, can be cyclised to E22, which on a carbonic acid linker yields cyclo-E22-D23. In another option D27, side-chain linked to the solid phase, can be cyclised to K28, which on amide linker yields cyclo- N27-K28. Preferably cyclisation is performed between amino acids in the contraloop Z, such as e.g. from D to G (becomes N-G) or from G to K*, if Z is YNGK It is also possible to cyclise in solution, e.g. from G25 to S26 or from G to K*, if Z is YNGK. Cyclisation is thought to be important to stabilise the bend present within XiX2X3VGSN, X2X3VGSNK or X3VGSNKG.
Another preferred way of cyclisation of a peptide is to add a cysteine at the N- and C-termini of the peptide sequence, or by adding a cysteine at the N-terminus of the peptide sequence and another one to Z. The presence of two cysteines will allow to carry out a disulfide cyclisation, as is well-known to the skilled person. In another preferred embodiment, a second way of achieving this stabilisation is to use Z. As earlier indicated herein, Z is an agent stabilizing the bend present within XiX2X3VGSN, X2X3VGSNK or X3VGSNKG in a peptide of the invention. In a preferred embodiment, Z stabilizes the bend present within XiX2X3VGSN, X2X3VGSNK or X3VGSNKG to ensure the peptide will likely adopt the conformation of folded Aβ 1-42. In a more preferred embodiment, Z stabilizes the bend present within XiX2X3VGSN, X2X3VGSNK or X3VGSNKG to ensure these peptides will adopt the conformation of folded Aβ 1-42. From studies, it is believed that there is a bend in conformation of folded Aβ 1-42, which bend is predicted to be present in a position corresponding to the position between the S and the N in XiX2X3VGSN, X2X3VGSNK or X3VGSNKG.
Z may be any agent known to the skilled person as stabilising a bend, a turn or loop. Z may be defined as a "contra-turn" agent with a high probability to form a β-turn (Hutchinson et al., 1998; Woolfson et al., 1993). Z may be an amino acid, an oligopeptide, a peptide, a polypeptide, a protein, an antigen, a mono- or oligosaccharide, and/or a steroid. In a preferred embodiment, Z is a peptide fragment of 8, 7, 6, 5 or 4 amino acid, in increasing preference with decreasing length. Preferably the peptide fragment of 4-8 amino acids is a "contra-turn" agent with a high propensity for a β-turn conformation. A preferred peptide fragment is a tetrapeptide selected from the group consisting of YNGK, TCGV, CGNT, LCGT, LKGT, GAIK, GAIC, AIIK, and AIIC. More preferably, the tetrapeptide is selected from the group consisting of YNGK, TCGV, CGNT, LCGT and LKGT. Most preferably, the tetrapeptide is selected from the group consisting of YNGK, TcGV, CGNT, LcGT and LkGT, wherein c = D- Cys and wherein k = D-Lys (see e.g. Oomen et al. 2003; and Oomen et al. 2005). Examples of proteins that may be used for Z are HSA, IgG' s and other serum proteins. Examples of antigens are (bacterial) toxins and virus-like particles. Z may also be a steroid scaffold such as described in e.g. Bode et al. (2007, J.Pept.Sci., 13:702-708) Suitable steroid scaffold for use as Z include e.g. bile acids and derivatives thereof such as e.g. cholic acid, deoxycholic acid and methyl 7-α-acetoxy-3α-amino-12«-amino-5/?- cholan-24-oate. Preferably in the peptides of the invention, the peptide sequences are connected to the C-3 and C-12 positions of the steroid scaffold, e.g. as described by Bode et al. (2007, supra). Z may be linked to the peptide sequence before cyclisation and optionally cyclised together with the rest of the peptide sequence. In this embodiment, Z is preferably a relatively short molecule like an oligopeptide: an amino acid, a dipeptide, a tripeptide, a tetrapeptide, a pentapeptide. In this preferred embodiment, the total number of amino acids (from the peptide sequence originating from Aβ 1-42 and from Z) is preferably ten or eleven. Even more preferably, this number is eleven. Z may comprise or consist of an amino acid present in the corresponding Aβ 1-42 sequence to align with the corresponding X1X2X3VGSN, X2X3VGSNK or X3VGSNKG sequence.
Alternatively, Z may be linked to the cyclised peptide sequence. In this embodiment, Z may be a relatively bigger molecule than in the previous embodiment: a polypeptide or a protein for example.
Best results were obtained when both ways are combined (cyclisation and the presence of Z) for stabilizing the peptide. Even more preferably, Z is linked to the peptide sequence and is subsequently cyclised with the rest of the peptide sequence. Alternatively, Z is formed as a result of the cyclization reaction. Within this preferred embodiment, best results were obtained with Z being the tetrapeptide as defined above, such as e.g. YNGK. More preferably, the tetrapeptide comprises at least one of a cysteine and a lysine to allow selective conjugation of the peptide to a carrier molecule as described below. The lysine preferably is a modified lysine such as N1^-(S- acetylmercaptoacety^lysine (Lys-SAMA). The presence of at least one of a cysteine and a Lys-SAMA residue in the tetrapeptide allows for selective conjugation of the peptide of the invention to a sulfhydryl-reactive carrier such as a carrier protein.
In a most preferred embodiment, there is provided a peptide consisting of the formula X2X3VGSNK-Z wherein X2 is E, G, Q or K, X3 is D or N and Z is an agent stabilizing the bend present within X2X3VGSNK. Preferably, Z is YNGK, wherein even more preferably, K in YNGK is a modified lysine (Lys-SAMA) to allow selective conjugation of the peptide.
In another most preferred embodiment, there is provided a peptide comprising the following sequence X3VGSNKG-Z, wherein X3 is D or N and Z is an agent stabilizing the bend present within X3VGSNKG. Preferably, Z is YNGK wherein even more preferably, K in YNGK is a modified lysine (Lys-SAMA) to allow selective conjugation of the peptide. A peptide comprising the sequence X2X3 VGSNKGAI-Z wherein X2 is E, X3 is D, and Z is a modified lysine (Lys-SAMA) and a peptide comprising the sequence
VGSNKG-Z wherein Z is is a modified lysine (Lys-SAMA) were both found to generate antibody responses to the immunizing peptides themselves, however the thus generated antibodies failed to cross react with oligomer or fibrillar Aβ 1-42.
A peptide of the invention may be present as a single peptide or incorporated into a fusion molecule, such as a fusion protein. A peptide may further be modified by deletion or substitution of one or more amino acids, by extension at the N- and/or C- terminus with additional amino acids or functional groups, which may improve bio- availability, targeting to T-cells, or comprise or release immune modulating substances that provide adjuvant or (co)stimulatory functions. The impact of these modifications is preferably tested on the conformation of the synthetised peptide. This may be done by NMR for example. It is important that in a thus obtained peptide, the conformation of XiX2X3VGSN, X2X3VGSNK or X3VGSNKG as likely adopted in Aβ 1-42, preferably as adopted in Aβ 1-42 has not been modified. The optional additional amino acids at the N- and/or C-terminus are preferably not present in the corresponding positions in the amino acid sequence of the protein it derives from, i.e. the Aβ 1-42 amino acid sequence. Therefore, in an even more preferred embodiment, in order to improve the immunogenicity of a peptide of the invention, this peptide, preferably a cyclic peptide as described above is conjugated to an immunogenic carrier molecule, preferably selectively through linkage of Z and the immunogenic carrier molecule. Such a peptide is called a conjugated peptide. Therefore, in a preferred embodiment, a peptide of the invention is a conjugated peptide, more preferably a conjugated cyclic peptide. An immunogenic carrier molecule preferably is a carrier that when conjugated to a peptide of the invention induces an immune response to the peptide of the invention upon administration to a subject such as a mammal. The immunogenic carrier may also have adjuvant-activity as later defined herein. Numerous of immunogenic carrier molecules are known to the skilled person (Hermanson, G. T., 1996, Bioconjugate techniques. Academic Press, San Diego; Drijfhout and Hoogerhout, 2000). Suitable immunogenic carrier molecules include e.g. bacterial toxins or toxoids such as exotoxins and variants thereof with reduced toxicity. Preferred immunogenic carrier molecules include diphtheria toxoid CRM197, a serum albumin (e.g. human serum albumin) and tetanus toxoid (Beuvery et al, 1986; Claesson et al, 2005). Composition
In a further aspect, there is provided a composition comprising a peptide as defined herein. Such a composition may be a pharmaceutical composition or a medicament.
In a further preferred embodiment, a peptide or a peptide composition further comprises a pharmaceutical excipient and/or a pharmaceutically acceptable carrier and/or an immune modulator. Any known inert pharmaceutically acceptable carrier and/or excipient may be added to a composition. Formulation of medicaments, and the use of pharmaceutically acceptable excipients are known and customary in the art and for instance described in Remington; The Science and Practice of Pharmacy, 21nd Edition 2005, University of Sciences in Philadelphia.
A pharmaceutical composition may further comprise pharmaceutically acceptable stabilizing agents, osmotic agents, buffering agents, dispersing agents, and the like. The preferred form of the pharmaceutical composition depends on the intended mode of administration and therapeutic application. The pharmaceutical carrier can be any compatible, non-toxic substance suitable to deliver the active ingredients, i.e. a peptide to a patient. Pharmaceutically acceptable carriers for intranasal delivery are exemplified by water, buffered saline solutions, glycerin, polysorbate 20, cremophor EL, and an aqueous mixture of caprylic/capric glyceride, and may be buffered to provide a neutral pH environment. Pharmaceutically acceptable carriers for parenteral delivery are exemplified by sterile buffered 0.9% NaCl or 5% glucose optionally supplemented with a 20% albumin. Preparations for parental administration must be sterile. The parental route for administration of the active ingredients is in accord with known methods, e.g. injection or infusion by subcutaneous, intravenous, intraperitoneal, intramuscular, intraarterial or intralesional routes. A composition of the invention is preferably administered by bolus injection. A typical pharmaceutical composition for intramuscular injection would be made up to contain, for example, 1 - 10 ml of phosphate buffered saline and 1 to 100 μg, preferably 15-45 μg of a modified conjugated peptide. For oral administration, the active ingredient can be administered in liquid dosage forms, such as elixirs, syrups, and suspensions. Liquid dosage forms for oral administration can contain colouring and flavouring to increase patient acceptance. Methods for preparing parenterally, orally or intranasally administrable compositions are well known in the art and described in more detail in various sources, including, for example, Remington's Pharmaceutical Science (15th ed., Mack
Publishing, Easton, PA, 1980) (incorporated by reference in its entirety for all purposes).
Any known immune modulator, in particular modulators leading to a balanced Th2/Thl response, like aluminium phosphate or aluminium hydroxide, may be added to a composition. Preferably, the immune modulator is an adjuvant. More preferably, the composition comprises a peptide as earlier defined herein and at least one adjuvant. An adjuvant is herein defined to include any substance or compound that, when used in combination with a peptide, to immunise a mammal, preferably a human, stimulates the immune system, thereby provoking, enhancing or facilitating the immune response against the peptide, preferably without generating a specific immune response to the adjuvant itself. Preferred adjuvants enhance the immune response against a given antigen by at least a factor of 1.5, 2, 2.5, 5, 10 or 20, as compared to the immune response generated against the peptide under the same conditions but in the absence of the adjuvant. Tests for determining the statistical average enhancement of the immune response against a given peptide as produced by an adjuvant in a group of animals or humans over a corresponding control group are available in the art. An adjuvant as used herein will usually be a compound that is foreign to a mammal, thereby excluding immunostimulatory compounds that are endogenous to mammals, such as e.g. interleukins, interferons and other hormones.
A composition of the present invention may contain at least one adjuvant. An adjuvant used in the present invention will be selected so that the effect of the peptide is not inhibited. Adjuvants used in the present invention are those which are physiologically acceptable to humans, these include, but are not limited to aluminium hydroxide, aluminium phosphate, oil/surfactant based emulsion adjuvants such as Montanide ™ in which different surfactants (especially mannityl oleate) are combined with a mineral oil, squalene-containing emulsions such as MF59™, monophosphoryl lipid A, or Neisseriae mutant lipopolysaccharide (as described in PCT/NL98/0063).
A medicament may be administered as a single administration. Alternatively, the administration of a peptide as earlier herein defined and/or an adjuvant may be repeated if needed and/or distinct peptides and/or distinct adjuvants may be sequentially administered. Peptide, composition and medicament of the invention are preferably formulated to be suitable for intravenous or subcutaneous, or intramuscular administration, although other administration routes can be envisaged, such as mucosal administration or intradermal and/or intracutaneous administration, e.g. by injection. Accordingly in a preferred embodiment, a peptide as described herein is for use as a medicament. More preferably, this medicament is a vaccine against the Alzheimer's disease. Even more preferably, the medicament is for preventing, delaying and/or treating the Alzheimer's disease. A vaccine as defined herein may be used for the prophylactic protection against the Alzheimer's disease or for the treatment of this disease.
In the context of the invention, an organism or an individual or a subject may be an animal or a human being. Preferably, the organism is a human being. Preferably, an organism treated is suspected to have a high risk of developing the Alzheimer's disease due for example to potential genetic predisposition, and/or to the age of the subject and/or to the lifestyle of a subject (for example nutritional habit and/or to the absence of physical activity) and/or to any other known parameter indicating this subject has an increased risk of developing the Alzheimer's disease.
The term "prevention" shall be understood to include complete prevention, prophylaxis, as well as lowering the individual's risk of falling ill with said disease or condition and delaying the onset of the disease or condition. The term "prevention" thus also comprises the treatment of persons suspected to be at risk falling ill with said disease or condition. The term shall also be understood to include alleviation of symptoms already developed.
The term "delaying" used herein means administration of a peptide to an organism, i.e. a patient being in a pre-stage of the condition to be treated in which patients a pre-form of the corresponding condition is diagnosed by methods known in the art.
The term "treatment" or "treating" "is understood the management and care of a patient for the purpose of combating the disease, condition, or disorder. Within the context of the invention, "treating the Alzheimer's disease and/or delaying its progression" preferably means that a therapeutically effective amount of a peptide is added. It refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, i.e. treat the Alzheimer's disease and/or delay its progression.
The amount of a peptide may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the pharmacological agent to elicit a desired response in the individual.
A therapeutic effect (leading to treating the Alzheimer's disease and/or delaying its progression) is preferably one that results in at least one of:
• a reduction of the load of beta plaques present in the brain;
• a reduction of the amount of soluble Aβ oligomers or prefϊbrils present in the brain; and,
• a reduction of the severity of a symptom associated with the Alzheimer' s disease. A reduction in the load of beta plaques present in the brain of a treated patient preferably means that the amount of a peptide added will be able to prevent de novo formation of beta plaques to at least some extent and/or that existing plaques will be to at least some extent inhibited in their ability to expand. Preferably, in this context, the deposition of beta plaques will not increase in a treated patient in terms of beta plaque load as identified by using an imaging technique such as PET scan (Henriksen, Yousefϊ et al., 2008) and/or magnetic resonance imaging (MRI) (O'Brien, 2007). In a PET scan, the quantity of beta plaque load is proportional to the tracer uptake. Preferably, the load of beta plaques will decrease of at least 2%, 5%, 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more. Even more preferably, no beta plaque will be detected. The skilled person knows that the visualisation of beta plaque should be done approximately at least one day, at least one week, at least one month after vaccination or more. (Meyer- Luehmann, Spires-Jones et al., 2008). If need be, one may decide to vaccinate several times and to regularly monitor the load of beta plaques.
A reduction of the amount of soluble Aβ oligomers or prefϊbrils present in the brain of a treated patient preferably means that the amount of a peptide added will be able to prevent de novo formation of soluble Aβ oligomers or prefϊbrils to at least some extent and/or that existing soluble Aβ oligomers or pre fibrils will be to at least some extent inhibited in their ability to expand. Preferably, in this context, the amount of soluble Aβ oligomers or prefϊbrils will not increase in a treated patient in terms of a surface as identified by an imaging technique as defined above. Preferably, the amount of soluble Aβ oligomers or prefϊbrils will decrease of at least 2%, 5%, 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more. More preferably, the amount of soluble Aβ oligomers or prefϊbrils will not be detectable using the same method. In the context of the invention, a soluble Aβ oligomer, prefϊbril or protofibril is an indication for an assembly of 2-24 Aβ monomers (Haas and Selkoe 2007).
Within the context of the invention, "a reduction of the severity of a symptom associated with Alzheimer's disease" preferably means an improvement of cognition as measured with a psychological test to assess the improvement of cognition in patients suffering from the Alzheimer's disease.
Use
Accordingly, in a further aspect, there is provided the use of a peptide or of a composition as defined herein for the manufacture of a medicament against the Alzheimer's disease. Preferably, the medicament is a vaccine. More preferably, the vaccine is for preventing, delaying and/or treating the Alzheimer's disease.
Accordingly, in another further aspect, there is provided a method of preventing, delaying and/or treating the Alzheimer's disease by administering a peptide or a composition as defined herein to a patient in a need thereof.
Method of svnthesising a peptide
The art currently knows many ways of generating a peptide of the invention. The invention is not limited to any ways of generating a peptide as long as the generated peptide comprises, consists or overlaps with any of the given modified sequences as identified herein and had the required conformation as earlier defined herein.
Accordingly, in a further aspect there is provided a method for producing a modified cyclic peptide as defined herein , said method comprising the following steps: a) synthesising a cyclic peptide comprising the sequence X1X2X3 VGSN-Z,
X2X3VGSNK-Z or X3VGSNKG-Z, wherein Xi is A or G, X2 is E, G, Q or K, X3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX2X3VGSN, X2X3VGSNK or X3VGSNKG; and b) optionally conjugating an immunogenic carrier molecule to the cyclic peptide obtained in b), preferably through linkage of Z to the immunogenic carrier molecule.
Each step of this method is known to the skilled person and has been extensively described in the example.
Antibody
In a further aspect, there is provided an antibody directed against a modified (cyclic) peptide of the invention as defined herein. The skilled person knows how to produce such an antibody in an animal. Methods for generating antibodies or antibody- fragments that specifically bind to a given polypeptide are described in e.g. Harlow and Lane (1988, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY) and WO 91/19818; WO 91/18989; WO 92/01047; WO 92/06204; WO 92/18619; and US 6,420,113 and references cited therein. The term "specific binding," as used herein, includes both low and high affinity specific binding. Specific binding can be exhibited, e.g., by a low affinity antibody or antibody-fragment having a Kd of at least about 10"4 M. Specific binding also can be exhibited by a high affinity antibody or antibody-fragment, for example, an antibody or antibody-fragment having a Kd of at least about of 10"7 M, at least about 10"8 M, at least about 10"9 M, at least about 10"10 M, or can have a Kd of at least about 10"11 M or 10"12 M or greater.
Diagnostic methods
In a further aspect, there is provided a method for diagnosing a neurodegenerative disease or condition, such as Alzheimer's disease. The method comprises determining the presence or absence of a beta amyloid plaque (i.e. neurodegenerative plaque) in the brain of a patient using an antibody as defined herein. Preferably in the method, the presence of a beta amyloid plaque in the subject's brain (or a sample thereof) is indicative for the subject being at risk of developing a neurodegenerative disease or condition such as Alzheimer's disease or indicative for the diagnosis of the neurodegenerative disease or condition, such as Alzheimer's disease. Preferably, such method is used for prognosing or diagnosing Alzheimer's disease in the brain of a patient. In the context of the invention, diagnosis means either a predictive risk assessment of a subject for developing later the Alzheimer's disease or preferably an assessment of the development of the Alzheimer's disease in a subject. In the context of the invention, a subject may be an animal or a human being. Preferably, a subject is a human being.
According to a preferred embodiment, the method is carried out in vitro or ex vzVo in a sample obtained from a subject. The sample preferably comprises brain tissue isolated from a subject. More preferably, the tissue is brain blood vessel.
Preferably, a detection of the presence of a beta amyloid plaque is revealed by the binding of an antibody of the invention to a brain sample as assayed by ELISA as explained in the example. The diagnosis method may be sequentially applied to a subject to monitor the development of the disease.
In this document and in its claims, the verb "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition the verb "to consist" may be replaced by "to consist essentially of meaning that a peptide or a composition as defined herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention. In addition, reference to an element by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements. The indefinite article "a" or "an" thus usually means "at least one".
All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety. The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way
Description of the figures
Figure 1. Model of an Aβl-42 fibril. A and B represent a dimeric cross-β unit and C the assembled fibril. [Credit: Olofsson et al. 2006 J. Biol. Chem. 281, 477-483].
Figure 2. Sequences of amyloid-derived cyclic peptides (K* is a modified lysine residue for conjugation purposes). Figure 3: IgG-antibody titres of pooled mice sera against homologous peptide-BSA conjugates. The titre is the 10log of the reciprocal serum dilution at 50% of the maximum optical density at 450 nm in ELISA.
Figure 4: OD450nm of pooled mice sera as a function of dilution with coating oligomeric or fibrillar Aβ 1-42.
Figure 5: IgG antibody titers of individual mice sera of groups 8 and 9 against oligomeric or fibrillar Aβ 1-42 The titre is the reciprocal serum dilution at 50% of the maximum optical density at 450 nm in ELISA.
Figure 6: Immunohistochemical staining of human brain section donor 99-30 (Braak 6) with a mouse serum (1 :300) immunized with cyclo[Aβ(22-28)-YNGK]/tetanus toxoid conjugate (a) and the control monoclonal 6E10 (1 :15.000) in (b). Similar pattern of plaques were recognized by the mouse sera as positive control. (Microscope: Leica DMRE fitted with a DC300 camera)
Examples Example 1 : synthesis strategy
We tested truncated peptides of Aβ, which do not comprise the immunodominant N-terminal B cell epitope. We aim at targeting an antibody response to early misfolded Aβ.
The sequence of Aβ 1-42 is:
DAE FR5HDS GY10EVHHQ15KLVFF2 0AEDVG25SNKGA3 0 I I GLM35VGGAAZ4 0 IA (SEQ ID NO:4).
The structure of Aβl-42 fibrils has been resolved by NMR spectroscopy (Olofsson et al. 2006). Hydrogen/deuterium exchange experiments with fibrils showed that the regions GIu -GIy and Lys -Ala in the Aβ sequence are shielded from solvent, whereas the N-terminus Asp'-Tyr10 and the two-residue fragment Ser26-Asn27 are solvent-accessible. The NMR data are consistent: Asp'-Tyr10 and the two-residue fragment Ser26-Asn27 are solvent-accessible. The NMR data are consistent with a model of the fibril as depicted in Figure 1C. The predicted structure is a twisted cross-β spine.
Figure IA and IB show a section, the dimeric cross-β unit. Within the dimer, each monomer contains two antiparallel β sheets which are connected by a turn composed of
Ser26-Asn27.
Based on the amyloid fibril model (Figure 1), we decided to prepare a set of YNGK* -stabilized cyclic 10 and l lmer amyloid peptides. Figure 2 shows the aimed amyloid decapeptides. We hypothesized that we could stabilise the conformation of small Aβ peptides by adding an artificial sequence YNGK*, in which K* is a modified lysine residue for selective conjugation to a carrier protein, followed by main chain
("head to tail") amide cyclization (Oomen et al (2005)). Likewise, we prepared a small panel of cyclic decameric and undecameric peptides spanning six or seven residues from the region 21-31 of Aβ and YNGK* (see Table 1). Table 1 : synthetised peptides
Figure imgf000018_0001
Example 2: specificity of some of the modified cyclised peptides for a conformational epitope of folded Aβ 1-42
A tetanus toxoid conjugate of the cyclic peptide [Aβ(22-28)-YNGK*], i.e. eye Io [EDVGSNKYNGK*] or peptide defined as group 9 in Table 1, elicited antibodies that cross-react in vitro with Aβ (l-42)-oligomers (Figure 4 or 5) and Aβ(l-42)-fibrils (Figure 4 or 5). These antibodies also recognize Aβ deposits in post-mortem AD human brain tissue (hippocampus) see Figure 6. The corresponding conjugate of the linear N-acetylated peptide amide Ac-K*EDVGSNKYNG-NH2 induced good antipeptide antibody titers but the antibodies generated did not recognize oligomeric or fibrillar Aβ. Thus, the cyclic peptide mimics a conformational epitope in folded Aβ 1- 42 that normally does not induce an antibody response. At the time of filing testing of an antibody response to the conjugate with the peptide defined as group 10, the cyclic peptide [Aβ(21-27)- YNGK*] was still in progress.
Materials and methods
Peptide synthesis, purification, and conjugation
The α-(2,4-dimethoxybenzyl) ester of Nα-fluorenylmethoxycarbonyl-L-aspartic acid (Fmoc-Asp-ODmb) was coupled through its side-chain to a polymer for the synthesis of peptide amides (for later conversion of the starting Asp into Asn). The side-chain-protected resin-bound sequence GK*EDVGSNKYD(resin), in which K* is N ε-(5'-acetylmercaptoacetyl)lysyl, was then assembled as described earlier (Brugghe et al., 1994). The resin-bound linear peptide was converted to cyclo[GK*EDVGSNKYD(resin)]. After side-chain deprotection, except of Lys(SAMA), and cleavage from the resin cyclo[GK*EDVGSNKYN] ≡ cyclo [EDVGSNKYNGK*], the peptide from group 9 (Table 1), was obtained. The peptides from groups 4-8 were prepared similarly. The peptide from group 10, cyclo [AEDVGSNYNGK*], was prepared from the side-chain-protected resin-bound linear precursor YNGK* AED VGSD(resin). The peptides were purified by reverse- phase high performance liquid chromatography and characterized by ion-spray mass spectrometry (MH+ f0Und/caicuiated, see table 1). The purified peptides were coupled to either bromoacetylated tetanus toxoid or maleimidyl-modified bovine serum albumin (BSA) (modifying reagent: NHS-PEO2-Maleimide, Pierce) (Drijfhout JW et al., 2000).
Disaggregation of Aβ(l-42)
Lyophilized Aβ 1-42 (Anaspec) was dissolved in trifluoroacetic acid at a concentration of 1.0 mM, left to stand at room temperature for 1 h and dried under a stream of nitrogen and, thereafter, in a vacuum (1 mm Hg) for 15 min. The peptide was then redissolved in hexafluoroisopropanol at a concentration of 1.0 mM and, after 1 h of incubation at room temperature, dried as described above (Zeng et al., 2001). The peptide was stored at -20 0C for 18-20 h.
Preparation of oligomeric or fibrillar Aβ(l-42) Disaggregated of Aβ 1-42 was dissolved in dimethyl sulfoxide at a concentration of 5.0 mM, diluted 50-fold with either phosphate buffered saline (PBS), pH 7.2, or 10 mM hydrochloric acid. The solution in PBS was incubated at 4 0C for 24 h (to give oligomers), whereas the solution in 10 mM HCl was incubated at 37 0C for 24 h (to give fibrils) (Stine et al).
Immunization of mice
Groups of eight female Balb/c mice of 6-8 weeks of age were immunized subcutaneously on days 0 and 28 with either 25 μig Aβ 1-42 in PBS without adjuvant or with 50 μg peptide -TTd conjugate and 75 μg A1PO4 in PBS. Small serum samples were collected on day 0. The mice were bled on day 42.
ELISA Microtitre plates (Greiner 655092) were coated with Aβl-42 or peptide-BSA conjugates. Freshly prepared Aβ 1-42 oligomers or fibrils were diluted to a final concentration of 2.5 μM (11.3 μg/ml) in 0.04 M sodium carbonate/bicarbonate buffer, pH 9.7. Peptide-BSA conjugates in phosphate buffered saline, pH 7.2 (PBS), had a total protein concentration of 0.5 μg/ml. Aliquots (100 μl) of these solutions were transferred into wells of the plates. The plates were incubated for 90 min at 37 0C. The plates were further processed as described earlier (Westdijk, Van den Ijssel et al, 1997).
Immunohistochemical staining Human brain sections of the hippocampus of several donors with Alzheimer disease (Braak 5 or 6) were used (Netherlands Brain Bank). Cryosections (10 μm) were cut from unfixed directly frozen tissue, thaw-mounted, dried for 1 hour and stored in a sealed box at -20C. For immunostaining, sections were fixed in 4% PFA-PBS solution for 10 min, washed in 0.05 M phosphate buffer (PB) for 10 min with 2 exchanges and blocked with 10% normal donkey serum (NDS) + 0.4% TritonXIOO in 0.05M PB for 1 hour at RT. The blocking solution was discarded and diluted mouse sera (l :300;first antibody) in 3% NDS + 0.4% TritonXIOO in 0.05 M PB was added and incubated O/N at RT in a box with wet tissues. Sections were washed with 0.05M PB; at least 30 min with one or more exchanges. Then sections were incubated with Donkey-anti-Mouse -Cy 3 1 : 1400 in 0.05 M PB for 2 hours. Sections were washed with 0.05M PB; at least 30 min with one or more exchanges. Sections were sealed in Vectashield with Dapi (Vector). Mouse Monoclonal 6E10 to beta amyloid 1-17 (Abeam, Cambridge, UK) was used as positive control (1 :15,000). Reference list
Agadjanyan MG, Ghochikyan A, Petrushina IVasilevko V, Movsesyan N, Mkrtichyan M, Saing T, and Cribbs DH, 2005. Prototype Alzheimer's disease vaccine using the immunodominant B cell epitope from β-amyloid and promiscuous T cell epitope pan HLA DR-binding peptide. J. Immunol. 174, 1580-1586.
Beuvery, E. C, Roy, R., Kanhai, V., Jennings, H. J., 1986. Characteristics of two types of meningococcal group C polysaccharide conjugates using tetanus toxoid as carrier protein. Dev.Biol. Stand. 65, 197-204. Blennow K, de Leon MJ, and Zetterberg H, 2006. Alzheimer's disease. Lancet 368, 387-403.
Brugghe HF, Timmermans HAM, van Unen LMA, ten Hove GJ, van de Werken G, Poolman JT, and Hoogerhout P, 1994. Simultaneous multiple synthesis and selective conjugation of cyclized peptides, derived from a surface-loop of a meningococcal class 1 outer membrane protein. Int. J. Peptide Protein Res. 43, 166-172. Check E, 2002. Nerve inflammation halts trial for Alzheimer's drug. Nature 415, 462. Claesson, B. A., Trollfors, B., Lagergard, T., Knutsson, N., Schneerson, R., Robbins, J. B., 2005. Antibodies against Haemophilus influenzae type b capsular polysaccharide and tetanus toxoid before and after a booster dose of the carrier protein nine years after primary vaccination with a protein conjugate vaccine. Pediatr.Infect.Dis.J. 24, 463-464. Drijfhout JW and Hoogerhout P, 2000. Methods of preparing peptide-carrier conjugates. In: Fmoc Solid Phase Peptide Synthesis: A Practical Approach (W.C. Chan and P.D. White, eds.). Oxford University Press, pp. 229-241. Editorials Nature Med. 2006, Vol. 12, # 7 July 2006. Gelinas DS, DaSilva K, Fenili D, St. George-Hyslop P, and McLaurin J, 2004. Immunotherapy for Alzheimer's disease. Proc. Natl. Acad. Sci. USA 101, 14657-14662. Gevorkian G., Petrushina I, Manoutcharian K, Ghochikyan A, Acero G, Vasilevko V, Cribbs DH, and Agadjanyan MA, 2004. Mimotopes of conformational epitopes in fibrillar β-amyloid. J. Neuroimmunol. 156, 10-20. Ghochikyan A, Mkrtichyan M, Petrushina I, Movsesyan N, Karapetyan A, Cribbs DH, and Agadjanyan MG, 2006. Prototype Alzheimer's disease epitope vaccine induced strong Th2-type anti-Aβ antibody response with Alum to Quil A adjuvant switch. Vaccine 24, 2275-2282. Gilman S, Roller M, Black RS, Jenkins L, Griffith SG, Fox NC, Eisner L, Kirby L, Boada Rovira M, Forette F, and Orgogozo J-M, for the AN1792(QS-21)-201 study team, 2005. Clinical effects of Aβ immunization (AN 1792) in patients with AD in an interrupted trial. Neurology 64, 1553-1562. Haass,C, Selkoe,D.J., 2007. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide. Nat.Rev.Mol.Cell Biol. 8, 101-112. Henriksen,G., Yousefi,B.H., Drzezga,A., Wester,H.J., 2008. Development and evaluation of compounds for imaging of beta-amyloid plaque by means of positron emission tomography. Eur. J.Nucl.Med.Mol. Imaging 35 Suppl 1, S75-S81. Hock C, Konietzko U, Papassotiropoulos A, Wollmer A, Streffer J, von Rotz RC, Davey G, Moritz E, and Nitsch RM, 2002. Generation of antibodies specific for beta- amyloid by vaccination of patients with Alzheimer disease. Nature Med. 8, 1270-1275. Hutchinson, E. G., Sessions, R. B., Thornton, J. M., Woolfson, D. N., 1998. Determinants of strand register in antiparallel beta-sheets of proteins. Protein Sci. 7, 2287-2300.
Klein WL, Stine WB, and Teplow DB, 2004. Small assemblies of unmodified amyloid β-protein are the proximate neurotoxin in Alzheimer's disease. Neurobiol. Aging 25,
569-580.
Lambert,M.P., Velasco,P.T., Chang,L., Viola,K.L., Fernandez,S., Lacor,P.N., Khuon,D., Gong,Y., Bigio,E.H., Shaw,P., De Felice,F.G., Krafft,G.A., Klein,W.L., 2007. Monoclonal antibodies that target pathological assemblies of Abeta. J.Neurochem. 100, 23-35.
Lee,M., Bard,F., Johnson- Wood,K., Lee,C, Hu,K., Griffith, S. G., Black,R.S., Schenk,D., Seubert,P., 2005. Abeta42 immunization in Alzheimer's disease generates Abeta N-terminal antibodies. Ann.Neurol. 58, 430-435.
Lemere CA, Maier M, Jiang L, Peng Y, and Seabrook TJ, 2006. Amyloid-beta immunotherapy for the prevention and treatment of Alzheimer's disease: lessons from mice, monkeys, and humans. Rejuvenation Res. 9, 77-84. Lemere,C.A., Maier,M., Peng,Y., Jiang,L., SeabrookJ.J., 2007. Novel Abeta immunogens: is shorter better? Curr.Alzheimer Res. 4, 427-436.
Maier M, Seabrook TJ, Lazo ND, Jiang L, Das P, Janus C, and Lemere CA, 2006. Short amyloid β (Aβ) immunogens reduce cerebral Aβ load and learning deficits in an Alzheimer's disease mouse model in the absence of an Aβ-specifϊc cellular immune response. J. Neurosci. 26, 4717-4728.
McDonnell J, Redekop WK, Van der Roer N, Goes E, Ruitenberg A, Busschbach JJ,
Breteler MM, and Rutten FF, 2001. The cost of treatment of Alzheimer's disease in The Netherlands: a regression-based simulation model. Pharmacoeconomics 19, 379-390.
McLaurin J, Cecal R, Kierstead ME, Tian X, Phinney AL, Manea M, French JE,
Lambermon MHL, Darabie AA, Brown ME, Janus C, Chishti MA, Home P, Westaway
D, Fraser PE, Mount HTJ, Przybylski M, and St George-Hyslop P, 2002.
Therapeutically effective antibodies against amyloid-beta peptide target amyloid-beta residues 4-10 and inhibit cytotoxicity and fϊbrillogenesis. Nature Med. 1263-1269.
Meyer-Luehmann,M., Spires- Jones,T. L., Prada,C, Garcia-Alloza,M., de,C.A.,
Rozkalne,A., Koenigsknecht-TalbooJ., Holtzman,D.M., Bacskai,B.J., Hyman,B.T.,
2008. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of
Alzheimer's disease. Nature 451, 720-724. Movsesyan,N., Ghochikyan,A., Mkrtichyan,M., Petrushina,L, Davtyan,H.,
Olkhanud,P.B., Head,E., Biragyn,A., Cribbs,D.H., Agadjanyan,M.G., 2008. Reducing
AD-like pathology in 3xTg-AD mouse model by DNA epitope vaccine - a novel immunotherapeutic strategy. PLoS. ONE. 3, e2124.
O'BrienJ.T., 2007. Role of imaging techniques in the diagnosis of dementia. Br. J.Radiol. 80 Spec No 2, S71-S77.
Olofsson A, Sauer-Eriksson AE, and Ohman A, 2006. The solvent protection of
Alzheimer amyloid-β-(l-42) fibrils as determined by solution NMR spectroscopy. J.
Biol Chem. 281, 477-483.
Oomen,C.J., Hoogerhout,P., Bonvin,A.M., Kuipers,B., Brugghe,H., Timmermans,H., Haseley,S.R., van,A.L., Gros,P., 2003. Immunogenicity of peptide -vaccine candidates predicted by molecular dynamics simulations. J.Mol.Biol. 328, 1083-1089.
Oomen CJ, Hoogerhout P, Kuipers B, Vidarsson G, van Alphen L, and Gros P, 2005.
Crystal structure of an anti -meningococcal subtype P 1.4 PorA antibody provides basis for peptide -vaccine design. J. MoI. Biol. 351, 1070-1080. Sadowski M and Wisniewski T, 2004. Vaccines for conformational disorders. Expert
Rev. Vaccines 3, 279-290. Sawaya MR, Sambashivan S, Nelson R, Ivanova MI, Sievers SA, Apostol M, Thompson MJ, Balbirnie M, Wiltzius JJW, McFarlane HT, Madsen A0, Riekel C, and Eisenberg D, 2007. Atomic structures of amyloid cross-beta spines reveal varied steric zippers. Nature 447, 453-457. Schenk, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, Hu K, Huang J, Johnson- Wood K, Khan K, Kholodenko D, Lee M, Liao Z, Lieberburg I, Motter R, Mutter L, Soriano F, Shopp G, Vasquez N, Vandevert C, Walker S, Wogulis M, Yednock T, Games D, and Seubert P, 1999. Immunization with amyloid-beta attenuates Alzheimer- disease-like pathology in the PDAPP mouse. Nature 400, 173-177. Sigurdsson EM, Knudsen E, Asuni A, Fitzer-Attas C, sage D, Quartermain D, Goni F, Frangione B, and Wisniewski T, 2004. An attenuated immune response is sufficient to enhance cognition in an Alzheimer's disease mouse model immunized with amyloid-β derivatives. J. Neurosci. 24, 6277-6282. Stine W B, Jr, Dahlgren KN, Rrafft GA, LaDu MJ, 2003. J. Biol. Chem., 278:11612. Tomiyama,T., Nagata,T., Shimada,H., Teraoka,R., Fukushima,A., Kanemitsu,H., Takuma,H., Kuwano,R., Imagawa,M., Ataka,S., Wada,Y., Yoshioka,E., Nishizaki,T., Watanabe,Y., Mori,H., 2008. A new amyloid beta variant favoring oligomerization in Alzheimer's-type dementia. Ann.Neurol. 63, 377-387. Vas CJ, Rajkumar S, Tanyakitpisal P, and Chandra V, 2001. Alzheimer's disease: the brain killer. Report of the World Health Organization.
Walsh,D.M., Selkoe,D.J., 2007. A beta oligomers - a decade of discovery. J.Neurochem. 101, 1172-1184.
WestdijkJ., Van den Ijssel, J., Thalen,M., Beuvery,C, Jiskoot,W., 1997. Quantification of cell-associated and free antigens in Bordetella pertussis suspensions by antigen binding ELISA. J.Immunoassay 18, 267-284.
Woolfson, D. N., Evans, P. A., Hutchinson, E. G., Thornton, J. M., 1993. Topological and stereochemical restrictions in beta-sandwich protein structures. Protein Eng 6, 461-
470.
Zeng H, Zhang Y, Peng L-J, Shao H, Menon NK, Yang J, Salomon AR, Freidland RP, and Zagorski MG, 2001. Nicotine and amyloid formation. Biol. Psychiatry 49, 248- 257.

Claims

Claims
1. A cyclic peptide consisting of the formula XiX2X3VGSN-Z, X2X3VGSNK-Z or X3VGSNKG-Z, wherein X1 is A or G, X2 is E, G, Q or K, X3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX2X3VGSN, X2X3VGSNK or X3VGSNKG.
2. A peptide according to claim 1, wherein Z is a peptide fragment of 4-8 amino acid residues or a steroid scaffold.
3. A peptide according to claim 2, wherein the peptide fragment is selected from the group consisting of YNGK, TCGV, CGNT, LCGT, LKGT, GAIK, GAIC, AIIK, AIIC, TcGV, CGNT, LcGT and LkGT, wherein c = D-Cys and wherein k = D-Lys; or, wherein the steroid scaffold is a bile acid or a derivative thereof, preferably selected from the group consisting of cholic acid, deoxycholic acid and methyl 7-α-acetoxy-3α- amino- 12«-amino-5/?-cholan-24-oate.
4. A peptide according to any one of claims 1 - 3, wherein the peptide is conjugated to an immunogenic carrier molecule.
5. A peptide according to claim 4, wherein the peptide is conjugated to the immunogenic carrier molecule through a selective covalent linkage of Z to the immunogenic carrier molecule.
6. A peptide according to claim 4 or 5, wherein the immunogenic carrier molecule is tetanus toxoid.
7. A peptide according to any one of claims 1 - 6, for use as a medicament.
8. A peptide according to any one of claims 1 - 6, for use in the treatment or prevention of Alzheimer' s disease.
9. Use of a peptide according to any one of claims 1 - 6, for the manufacture of a medicament the treatment or prevention of Alzheimer's disease.
10. A method for prevention, delaying the onset and/or treatment of Alzheimer's disease, the method comprising the step of administering an effective amount of a peptide according to any one of claims 1 - 6 to a patient in a need thereof.
11. A method for producing a cyclic peptide as defined in any one of claims 1 - 6, wherein the method comprising the steps of: a) synthesising a cyclic peptide consisting of the sequence X1X2X3 VGSN-Z, X2X3VGSNK-Z or X3VGSNKG-Z, wherein Xi is A or G, X2 is E, G, Q or K, X3 is D or N, and Z is an agent stabilizing the bend present within the peptide sequence XiX2X3VGSN, X2X3VGSNK or X3VGSNKG; and b) optionally, conjugating an immunogenic carrier molecule to the cyclic peptide obtained in b), preferably through linkage of Z to the immunogenic carrier molecule.
12. Antibody directed against the peptide as defined in any one of claim 1 - 6.
13. An in vitro method for diagnosing a neurodegenerative disease or condition, wherein the method comprises the step of determining in a sample of a patient, the presence or absence of a beta amyloid plaque, using an antibody as defined in claim 12.
14. An in vitro method according to claim 12, for diagnosing Alzheimer's disease.
PCT/NL2009/050387 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate WO2010002251A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
RU2011103448/10A RU2546234C2 (en) 2008-07-01 2009-07-01 Vaccine against intermediate with amyloid coagulation
CA2729414A CA2729414C (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
CN2009801306774A CN102112488A (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
AU2009266552A AU2009266552B2 (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
EP09773784A EP2297196B1 (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
MX2011000134A MX2011000134A (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate.
BRPI0914925A BRPI0914925A2 (en) 2008-07-01 2009-07-01 cyclic peptide, use of a peptide, methods for producing a cyclic peptide and for diagnosing a disease or condition, and antibody
ES09773784T ES2397641T3 (en) 2008-07-01 2009-07-01 Vaccine against the amyloid folding intermediary
NZ590324A NZ590324A (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
JP2011516186A JP5722770B2 (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
KR1020117002526A KR101694788B1 (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
US13/002,174 US9045555B2 (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate
IL210364A IL210364A (en) 2008-07-01 2010-12-29 Cyclic peptide analogue of the ß-amyloid peptide and uses thereof in the treatment or diagnosis of neurodegenerative diseases or conditions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US7726408P 2008-07-01 2008-07-01
EP08159385 2008-07-01
US61/077,264 2008-07-01
EP08159385.7 2008-07-01

Publications (1)

Publication Number Publication Date
WO2010002251A1 true WO2010002251A1 (en) 2010-01-07

Family

ID=39944379

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2009/050387 WO2010002251A1 (en) 2008-07-01 2009-07-01 Vaccine against amyloid folding intermediate

Country Status (14)

Country Link
US (1) US9045555B2 (en)
EP (1) EP2297196B1 (en)
JP (1) JP5722770B2 (en)
KR (1) KR101694788B1 (en)
CN (1) CN102112488A (en)
AU (1) AU2009266552B2 (en)
BR (1) BRPI0914925A2 (en)
CA (1) CA2729414C (en)
ES (1) ES2397641T3 (en)
IL (1) IL210364A (en)
MX (1) MX2011000134A (en)
NZ (1) NZ590324A (en)
RU (1) RU2546234C2 (en)
WO (1) WO2010002251A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011106885A1 (en) * 2010-03-03 2011-09-09 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
WO2012072611A1 (en) * 2010-11-29 2012-06-07 Philipps-Universität Marburg SYNTHETIC LIGANDS FOR HUMAN ANTI-Aβ ANTIBODIES
AU2013205000B2 (en) * 2010-03-03 2015-05-28 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
EP2780082A4 (en) * 2011-11-10 2015-08-05 Cangene U S Inc Compositions and methods for treating alzheimer's disease
CN108350053A (en) * 2015-11-09 2018-07-31 英属哥伦比亚大学 Amyloid beta epitope and its antibody
CN108350052A (en) * 2015-11-09 2018-07-31 英属哥伦比亚大学 Epitope in amyloid beta intermediate region and its conformation antibodies selective
CN108368160A (en) * 2015-11-09 2018-08-03 英属哥伦比亚大学 C- terminal epitopes in amyloid beta and its conformation antibodies selective
EP3374382A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia Epitopes in amyloid beta and conformationally-selective antibodies thereto
EP3374379A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
US10751382B2 (en) 2016-11-09 2020-08-25 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
EP3861015A4 (en) * 2018-10-07 2022-11-23 The University of British Columbia Conformation-specific epitopes in alpha-synuclein, antibodies thereto and methods related thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI571207B (en) 2011-06-26 2017-02-21 安麗托克斯公司 Cold weather formulation for conditioning animal feed
US10195257B2 (en) 2013-07-28 2019-02-05 Qantu Therapeutics, Inc. Vaccine formulations comprising quillaja desacylsaponins and beta amyloid peptides or tau protein to induce a Th2 immune response

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002055552A2 (en) * 2001-01-13 2002-07-18 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Soluble cyclic analogues of beta amyloid peptide
WO2006121656A2 (en) * 2005-05-05 2006-11-16 Merck & Co., Inc. Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1284998B1 (en) * 2000-05-22 2004-12-29 New York University Synthetic immunogenic but non-amyloidogenic peptides homologous to amyloid beta for induction of an immune response to amyloid beta and amyloid deposits
EP2338510A1 (en) * 2002-07-19 2011-06-29 Novartis Pharma AG Vaccine compositions containing amyloid beta1-6 antigen arrays
RU2391341C2 (en) * 2002-09-25 2010-06-10 Мемори Фармасьютиклз Корпорейшн Indazoles, bentothiazoles, benzoisothiazoles, production and use thereof
CN100409896C (en) * 2003-03-31 2008-08-13 姚志彬 Senile dementia vaccinum and preparing method thereof
EP1676859A1 (en) * 2004-12-30 2006-07-05 Pevion Biotech Ltd. Immunogenic compositions of cyclic peptides derived from the beta-amyloid peptide
MX2011000975A (en) * 2008-07-25 2011-05-25 Abbott Lab Amyloid 㟠peptide analogues, oligomers thereof, processes for preparing and compositions comprising said analogues or oligomers, and their uses.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002055552A2 (en) * 2001-01-13 2002-07-18 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Soluble cyclic analogues of beta amyloid peptide
WO2006121656A2 (en) * 2005-05-05 2006-11-16 Merck & Co., Inc. Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CRUZ LUIS ET AL: "Solvent and mutation effects on the nucleation of amyloid beta-protein folding.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 102, no. 51, 20 December 2005 (2005-12-20), pages 18258 - 18263, XP002504493, ISSN: 0027-8424 *
MATSUZAKI KATSUMI ET AL: "Design, synthesis, and biophysical properties of a helical Abeta1-42 analog: Inhibition of fibrillogenesis and cytotoxicity.", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 11 JUL 2008, vol. 371, no. 4, 8 May 2008 (2008-05-08), pages 777 - 780, XP022688462, ISSN: 1090-2104 *
SCIARRETTA KIMBERLY L ET AL: "Abeta40-Lactam(D23/K28) models a conformation highly favorable for nucleation of amyloid.", BIOCHEMISTRY, vol. 44, no. 16, 26 April 2005 (2005-04-26), pages 6003 - 6014, XP002504492, ISSN: 0006-2960 *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2013205000B2 (en) * 2010-03-03 2015-05-28 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
CN102858796A (en) * 2010-03-03 2013-01-02 不列颠哥伦比亚大学 Oligomer-specific amyloid beta epitope and antibodies
EP2542571A1 (en) * 2010-03-03 2013-01-09 The University of British Columbia Oligomer-specific amyloid beta epitope and antibodies
JP2013524774A (en) * 2010-03-03 2013-06-20 ザ・ユニバーシティ・オブ・ブリティッシュ・コロンビア Oligomer-specific amyloid beta epitopes and antibodies
EP2542571A4 (en) * 2010-03-03 2013-07-31 Univ British Columbia Oligomer-specific amyloid beta epitope and antibodies
WO2011106885A1 (en) * 2010-03-03 2011-09-09 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
US9216217B2 (en) 2010-03-03 2015-12-22 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
EP3056510A1 (en) * 2010-03-03 2016-08-17 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
JP2016193914A (en) * 2010-03-03 2016-11-17 ザ・ユニバーシティ・オブ・ブリティッシュ・コロンビア Oligomer-specific amyloid beta epitope and antibodies
US9849165B2 (en) 2010-03-03 2017-12-26 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
RU2644335C2 (en) * 2010-03-03 2018-02-08 Зе Юниверсити Оф Бритиш Коламбия Epitope, specific to amyloid beta oligomer and antibodies
WO2012072611A1 (en) * 2010-11-29 2012-06-07 Philipps-Universität Marburg SYNTHETIC LIGANDS FOR HUMAN ANTI-Aβ ANTIBODIES
EP2780082A4 (en) * 2011-11-10 2015-08-05 Cangene U S Inc Compositions and methods for treating alzheimer's disease
CN108350052A (en) * 2015-11-09 2018-07-31 英属哥伦比亚大学 Epitope in amyloid beta intermediate region and its conformation antibodies selective
EP3374379A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
CN108368160A (en) * 2015-11-09 2018-08-03 英属哥伦比亚大学 C- terminal epitopes in amyloid beta and its conformation antibodies selective
EP3374382A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia Epitopes in amyloid beta and conformationally-selective antibodies thereto
EP3374380A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia C-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
EP3374383A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia Amyloid beta epitopes and antibodies thereto
EP3374381A4 (en) * 2015-11-09 2019-05-15 The University Of British Columbia Epitopes in amyloid beta mid-region and conformationally-selective antibodies thereto
CN108350053A (en) * 2015-11-09 2018-07-31 英属哥伦比亚大学 Amyloid beta epitope and its antibody
US11970522B2 (en) 2015-11-09 2024-04-30 The University Of British Columbia Cyclic compound/peptide comprising an A-beta15-18 peptide and a linker that is covalently coupled to the n-terminus residue and the c-terminus residue of the A-BETA15-18 peptide
US10759837B2 (en) 2015-11-09 2020-09-01 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US10772969B2 (en) 2015-11-09 2020-09-15 The University Of British Columbia N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
US10774120B2 (en) 2015-11-09 2020-09-15 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US11905318B2 (en) 2015-11-09 2024-02-20 The University Of British Columbia Cyclic compound/peptide comprising an A-beta13-16 peptide and a linker that is covalently coupled to the n-terminus residue and the c-terminus residue of the A-beta13-16 peptide
US11779629B2 (en) 2016-11-09 2023-10-10 The University Of British Columbia Compositions comprising cyclic peptides derived from an A-beta peptide
US10751382B2 (en) 2016-11-09 2020-08-25 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
EP3861015A4 (en) * 2018-10-07 2022-11-23 The University of British Columbia Conformation-specific epitopes in alpha-synuclein, antibodies thereto and methods related thereof

Also Published As

Publication number Publication date
RU2546234C2 (en) 2015-04-10
IL210364A0 (en) 2011-03-31
AU2009266552A1 (en) 2010-01-07
IL210364A (en) 2016-09-29
KR101694788B1 (en) 2017-01-13
JP5722770B2 (en) 2015-05-27
EP2297196A1 (en) 2011-03-23
NZ590324A (en) 2012-08-31
JP2011526885A (en) 2011-10-20
RU2011103448A (en) 2012-08-10
BRPI0914925A2 (en) 2015-10-20
KR20110043635A (en) 2011-04-27
ES2397641T3 (en) 2013-03-08
CA2729414C (en) 2017-08-22
CA2729414A1 (en) 2010-01-07
US9045555B2 (en) 2015-06-02
MX2011000134A (en) 2011-04-05
CN102112488A (en) 2011-06-29
EP2297196B1 (en) 2012-11-14
US20110182928A1 (en) 2011-07-28
AU2009266552B2 (en) 2014-01-23

Similar Documents

Publication Publication Date Title
US9045555B2 (en) Vaccine against amyloid folding intermediate
JP6174727B2 (en) Β1-42 specific monoclonal antibodies with therapeutic properties
CA2633399C (en) Therapeutic vaccine
TW200906852A (en) Monoclonal antibody
KR20110071117A (en) Modified amyloid beta peptide
JP2007300856A (en) Amyloid protein imitation
US9770496B2 (en) Method for treating amyloid disease

Legal Events

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

Ref document number: 200980130677.4

Country of ref document: CN

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

Ref document number: 09773784

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2729414

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2011516186

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 210364

Country of ref document: IL

Ref document number: 2010122221

Country of ref document: EG

Ref document number: 2009773784

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009266552

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 590324

Country of ref document: NZ

Ref document number: MX/A/2011/000134

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2009266552

Country of ref document: AU

Date of ref document: 20090701

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117002526

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011103448

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 13002174

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0914925

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20101230