EP1286590A2 - Peptide analogs and mimetics suitable for in vivo use in the treatment of diseases associated with abnormal protein folding into amyloid, amyloid-like deposits or beta-sheet rich pathological precursor thereof - Google Patents

Peptide analogs and mimetics suitable for in vivo use in the treatment of diseases associated with abnormal protein folding into amyloid, amyloid-like deposits or beta-sheet rich pathological precursor thereof

Info

Publication number
EP1286590A2
EP1286590A2 EP00976928A EP00976928A EP1286590A2 EP 1286590 A2 EP1286590 A2 EP 1286590A2 EP 00976928 A EP00976928 A EP 00976928A EP 00976928 A EP00976928 A EP 00976928A EP 1286590 A2 EP1286590 A2 EP 1286590A2
Authority
EP
European Patent Office
Prior art keywords
ala
pro
proψ
val
asp
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP00976928A
Other languages
German (de)
French (fr)
Other versions
EP1286590A4 (en
Inventor
Claudio Soto-Jara
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Axonyx Inc
Original Assignee
Axonyx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Axonyx Inc filed Critical Axonyx Inc
Publication of EP1286590A2 publication Critical patent/EP1286590A2/en
Publication of EP1286590A4 publication Critical patent/EP1286590A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • 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
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/34Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • 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
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0207Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -NH-(X)4-C(=0), e.g. 'isosters', replacing two amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated

Definitions

  • the present invention relates to peptide analogs and peptide mimetics of ⁇ -sheet
  • the present invention is a method for treating Alzheimer's and prion diseases. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease. More particularly, the present invention is a method for treating Alzheimer's and prion disease.
  • amyloid-like deposits or precursors thereof having a pathological beta-sheet structure amyloid-like deposits or precursors thereof having a pathological beta-sheet structure.
  • protein conformational diseases include Alzheimer's disease, prion-related disorders, systemic amyloidosis, serpin-deficiency disorders, Huntington's disease and Amyotrophic
  • the conformationally modified protein may be implicated in the
  • AD Alzheimer's disease
  • AD Alzheimer's AD is the most common dementia in elderly population. It is estimated that more than
  • AD Amyloid
  • AD Alzheimer's disease
  • amyloid aggregates, known as amyloid, in brain parenchyma and cerebral vessel walls.
  • amyloid beta-peptide A ⁇
  • APP precursor protein
  • amyloid plays an important role in
  • Amyloid is a generic term that describes fibrillar aggregates that have a common
  • tinctorial properties including the ability to emit a green birefringent glow after staining
  • Alzheimer's disease involving amyloidosis, it is possible to highlight Alzheimer's disease, prion-related disorders
  • amyloid fibrils are usually composed of proteolytic fragments of normal or mutant
  • amyloid is basically a problem of protein folding, whereby a mainly
  • hydrophobicity appears to be important to induce interaction of the monomers leading to
  • the 5-residue peptide is capable of preventing the neuronal death induced
  • ⁇ -sheet breaker peptide decreased cerebral A ⁇ accumulation and completely blocked the
  • ⁇ -sheet breaker peptides have also been designed to prevent and to revert
  • PrP c PrP sc conversion, but more interestingly to revert the infectious PrP sc conformer to a
  • Short peptides have been utilized extensively as drugs in medicine (Rao et al., C.
  • the present invention is an inhibitory peptide capable of inhibiting ⁇ pleated sheet formation in amyloid ⁇ -peptide, the inhibitory peptide being a ⁇ sheet breaker peptide analog
  • the peptide is altered chemically by: (1) modifications to the N- and C-terminal ends
  • the present invention also includes an inhibitory peptide capable of inhibiting
  • peptide being a ⁇ sheet breaker peptide analog designed by chemical modification of a ⁇ sheet
  • the present invention includes a peptide mimetic with the following
  • the peptide mimetic has the following structure:
  • the peptide mimetic has the following structure:
  • the present invention also includes a method for preventing, treating, or detecting
  • FIG. 1 is a schematic representation of the peptide bond and the potential target sites
  • FIG. 2 is a graph depicting the pharmacokinetics of a 11-residue ⁇ -sheet breaker
  • FIGS. 3A and 3B are representations of the tridimensional structure of Alzheimer's
  • FIG. 4a and 4b are graphs showing the bioavailability and stability of iAB5 and Ac-
  • FIG. 5a provides a graphical comparison of A ⁇ l-40 incubated with various other
  • FIG. 5b is a graph of amyloid formation vs. the Ac-iA ⁇ 5-Am concentration.
  • FIG. 5c is a graph of amyloid formation vs. the iA ⁇ 5 concentration.
  • FIG. 6 shows a model where there is an 83% dissolution of deposits in the ventricle
  • amygdala area and a 30% dissolution of amyloid plaque in the amygdala.
  • the inhibitory peptide is capable of
  • peptide is a ⁇ sheet breaker peptide analog designed by chemical modification of a ⁇ sheet
  • This invention also includes an inhibitory peptide capable of inhibiting conformational changes in prion PrP protein associated with amyloidosis, where the inhibitory peptide is a ⁇ sheet breaker peptide analog designed by chemical modification of a
  • Fig. 1 a generalized peptide backbone is shown, where possible targets for chemical modification are highlighted.
  • the possible targets include the following: (1)
  • Natural peptides are usually degraded by the concerted action of specialized
  • Endopeptidases and unspecific exopeptidases. Endopeptidases are often present in tissues
  • Exopeptidases are generally present in blood and peripheral organs and carry out the
  • exopeptidases recognize the atoms participating in the
  • peptide mimetics are usually completely resistant to proteolytic degradation and often can be
  • ⁇ -sheet breaker analogues designed by chemical modifications of the lead peptides.
  • DAPAAPAGPAVPV also denoted as Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro
  • Alzheimer's Inhibitors Prion Inhibitors ac-Leu Pro Phe Phe Asp-am ac-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-a des-Leu Pro Phe Phe Asp-am des-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-am ac-Leu Pro Phe Phe Asp-ale ac-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc des-Leu Pro Phe Phe Asp-ale des-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc des-Leu Pro Phe Phe Asp-ale des-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc
  • methylation of those residues in the inhibitor peptides is expected to enhance stability and potency.
  • the D-form showed virtually no degradation in the plasma after injection for 15 minutes.
  • the peptide was radio-iodinated using standard procedures. Peptide stability was
  • endopeptidase cleavage site is after this residue by an enzyme known as
  • prolylendopepti dase prolylendopepti dase .
  • Alzheimer's Inhibitors Prion Inhibitors leu pro phe phe asp asp ala pro ala ala pro ala gly pro ala val pro val leu Pro Phe Phe asp asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro val leu Pro phe Phe asp asp Ala Pro ala Ala Pro ala Gly Pro ala Val Pro val
  • Pseudopeptides or amide bond surrogates refers to peptides containing chemical modifications of some (or all) of the peptide bonds. Amide bond
  • Ala ⁇ [CH CH 2 ]Gly refers to the moiety
  • bonds to replace are those located at the end of the peptide to prevent exoprotease
  • endopeptidase cleavage site occurs after this residue by an enzyme known as
  • prolylendopeptidase Additional amide bonds that need to be protected are determined by
  • Alzheimer's Inhibitors Leu ⁇ [CH 2 CH 2 ]Pro ⁇ [CH 2 CH 2 ]Phe Phe ⁇ [CH 2 CH 2 ]Asp Leu ⁇ [CH 2 S]Pro ⁇ [CH 2 S]Phe Phe ⁇ [CH 2 S]Asp Prion inhibitors Asp ⁇ [CH 2 CH 2 ]Ala Pro ⁇ [CH 2 CH 2 ]Ala Ala Pro ⁇ [CH 2 CH 2 ]Ala Gly Pro ⁇ [CH 2 CH 2 ]Ala Val Pro ⁇ [CH 2 CH 2 ]Val
  • peptides successfully stabilized against proteolysis consist of a mixture of several types of
  • Alzheimer's Inhibitors Ac-Leu Pro ⁇ [CH 2 CH 2 ]Phe Phe Asp-Am Ac-Leu Pro ⁇ [CH 2 S]Phe Phe Asp-Am (Me)Leu Pro ⁇ [CH 2 CH 2 ]Phe Phe Asp-Am leu Pro ⁇ [CH 2 CH 2 ]Phe Phe asp leu Pro ⁇ [CH 2 S]Phe Phe asp Ac-Leu Aib Phe Phe Asp- Am (Me)Leu Aib Phe Phe Asp- Am Leu Pro ⁇ [CH 2 CH 2 ]Phe Phe asp
  • a peptide mimetic is a molecule
  • peptide mimetic has been used sometimes to describe molecules that are
  • Peptide mimetics are not derivatives of a parent peptide
  • the objective is to reconstruct the spatial position of
  • the objective is to reconstruct the position of the pharmaco-active groups using
  • the present invention is administered in an effective amount to a subject in need thereof,
  • prevention of a condition, such as Alzheimer's disease or
  • amyloidosis disorders in a subject involves administering the compound according to
  • development of a disorder or disease comprises "treatment" of the disease.
  • the invention is
  • the compound of the present invention may be administered by any means that
  • administration may be by a
  • parenteral routes including, but not limited to, subcutaneous,
  • transdermal, or buccal routes Parenteral administration can be bolus injection or by gradual
  • amyloid or amyloid-like deposits comprises either: (1) administration of an effective amount
  • the dosage administered will be dependent upon the age, sex, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of
  • amyloid or amyloid-like deposits or pathological beta-sheet precursors thereof
  • dosage may be dependent on the stability of the administered compound. A less stable
  • Preparations for parenteral administration include sterile aqueous or non-aqueous
  • solutions, suspensions, and emulsions which may contain auxiliary agents or excipients
  • compositions such as tablets and capsules can be any suitable pharmaceutical compositions which are known in the art.
  • Pharmaceutical compositions such as tablets and capsules can be any suitable pharmaceutical compositions such as tablets and capsules.
  • compositions may contain suitable, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted, unreacted
  • pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate
  • Suitable pharmaceutically acceptable vehicles are well known in the art and are described for
  • formulations for intravenous administration may include sterile aqueous solutions which
  • buffers may also contain buffers, diluents and other suitable additives.
  • Suitable formulations for parenteral administration include aqueous solutions of the
  • active compounds in water-soluble form for example, water-soluble salts.
  • water-soluble salts for example, water-soluble salts.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame
  • suspensions that may contain substances which increase the viscosity of the suspension
  • the suspension may also contain stabilizers.
  • amyloid-like deposits or into pathological beta-sheet-rich precursors of such deposits to be
  • Alzheimer's disease includes, but is not limited to, Alzheimer's disease, FAF, Down's syndrome, other
  • amyloidosis disorders human prion diseases, such as kuru, Creutzf el dt- Jakob Disease
  • CJD Gerstmann-Strausslet-Scheinker Syndrome
  • GSS Gerstmann-Strausslet-Scheinker Syndrome
  • neurodegenerative diseases as well as animal prion diseases such as scrapie, spongiform
  • GYLTVAAVFRG GYLTVAAVFRG, CP10: ISEVKMDAEF
  • short A ⁇ fragments such as A ⁇ 18-21, AB1- 16
  • amyloid-like deposit in the place of injection. These lesions have the same tinctorial (congo).
  • AD brain similar to that observed in AD brain, including extensive neuronal shrinkage, astrocytosis
  • peptide were precipitated in cold methanol (mix/MeOH, 4/5, v/v) for one hour at -20°C.
  • the precipitated proteins were pelleted by centrifugation (10 OOOg, 10 min, 4°C). The
  • cannula was slowly withdrawn. Following surgery the animals were placed on a heating pad
  • references cited within the references cited herein are also entirely incorporated by reference.

Abstract

The present invention is an inhibitory peptide capable of inhibiting β pleated sheet formation in amyloid β-peptide. The inhibitory peptide is a βsheet breaker peptide analog designed by chemical modification of β sheet breaker peptide capable of inhibiting β pleated sheet formation in amyloid β-peptide. The present invention also includes an inhibitory peptide capable of inhibiting conformational changes in prion PrP protein associated with amyloidosis. The inhibitory peptide being a βsheet breaker peptide analog designed by chemical modification of a βsheet breaker peptide capable of inhibiting the conformational changes in prior PrP protein associated with amyloidosis. In addition, the present invention includes a peptide mimetic with the structure PMiAβ5. In another embodiment, the peptide mimetic has the structure PMiPrP13. In yet another embodiment, the peptide mimetic has the structure PMiPrP5.

Description

PEPTIDE ANALOGS AND MIMETICS SUITABLE FOR IN VIVO USE IN THE
TREATMENT OF DISEASES ASSOCIATED WITH ABNORMAL PROTEIN
FOLDING INTO AMYLOID, AMYLOID-LIKE DEPOSITS OR β-SHEET RICH
PATHOLOGICAL PRECURSOR THEREOF
This application claims priority from U.S. Provisional Application No. 60/163,911,
which was filed on November 5, 1999.
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention relates to peptide analogs and peptide mimetics of β-sheet
breaker peptides suitable for in vivo use in treating mammals with protein conformational
diseases such as Alzheimer's and prion disease. More particularly, the present invention is
directed to novel peptide analogs and mimetics, pharmaceutical compositions containing one
or a mixture of such peptide analogs and mimetics, and methods for preventing, treating, or
detecting disorders or diseases associated with abnormal protein folding into amyloid or
amyloid-like deposits or precursors thereof having a pathological beta-sheet structure.
Description of Related Art
Extensive evidence has been accumulated indicating that several diverse disorders
have the same molecular basis, i.e. a change in a protein conformation (Thomas et al.,
Trends Biochem. Sci. 20: 456-459, 1995; Soto, J. Mol. Med. 11: 412-418, 1999). These
protein conformational diseases include Alzheimer's disease, prion-related disorders, systemic amyloidosis, serpin-deficiency disorders, Huntington's disease and Amyotrophic
Lateral Sclerosis (Soto 1999, supra). The hallmark event in protein conformational disorders
is a change in the secondary and tertiary structure of a normal protein without alteration of
the primary structure. The conformationally modified protein may be implicated in the
disease by direct toxic activity, by the lack of biological function of normally-folded protein,
or by improper trafficking (Thomas et al., 1995, supra). In the cases where the protein is
toxic, it usually self- associates and becomes deposited as amyloid fibrils in diverse organs,
inducing tissue damage (Thomas et al., 1995, supra; Kelly, Curr. Opin. Struct. Biol. 6: 11-
17, 1996; Soto, 1999, supra).
Alzheimer's disease (AD) is a devastating neurodegenerative problem characterized
by loss of short-term memory, disorientation, and impairment of judgment and reasoning.
AD is the most common dementia in elderly population. It is estimated that more than
twenty-five million people worldwide are affected in some degree by AD (Teplow, Amyloid
5: 121-142, 1998). A hallmark event in AD is the deposition of insoluble protein
aggregates, known as amyloid, in brain parenchyma and cerebral vessel walls. The main
component of amyloid is a 4.3 KDa hydrophobic peptide, named amyloid beta-peptide (Aβ)
that is encoded on the chromosome 21 as part of a much longer precursor protein (APP)
(Selkoe, Science 275: 630-631, 1997). Genetic, biochemical, and neuropathological evidence
accumulated in the last 10 years strongly suggest that amyloid plays an important role in
early pathogenesis of AD and perhaps triggers the disease (Soto et al., J. Neurochem. 63:
1191-1198, 1994; Selkoe, 1997, supra; Teplow, 1998, supra; Sisodia and Price, FASEB J. 9: 366-370, 1995; Soto, Mol. Med. Today 5: 343-350, 1999).
Amyloid is a generic term that describes fibrillar aggregates that have a common
structural motif, i.e., the β-pleated sheet conformation (Serpell et al., Cell Mol. Life Sci. 53:
887, 1997; Sipe, Ann. Rev. Biochem. 61: 947-975, 1992). These aggregates exhibit specific
tinctorial properties, including the ability to emit a green birefringent glow after staining
with Congo red, and the capacity to bind the fluorochrome, thioflavin S (Sipe, 1992, supra;
Ghiso et al., Mol. Neurobiol. 8: 49-64, 1994). There are more than a dozen human diseases
of different etiology characterized by the extracellular deposition of amyloid in diverse
tissues, which lead to cell damage, organ dysfunction, and death. Among the diseases
involving amyloidosis, it is possible to highlight Alzheimer's disease, prion-related disorders
(also known as transmissible spongiform encephalopathy), and systemic amyloidosis (Table
1). The amyloid fibrils are usually composed of proteolytic fragments of normal or mutant
gene products. There are over 16 different proteins (Table 1) involved in amyloid deposition
in distinct tissues (Ghiso et al., 1994, supra).
The formation of amyloid is basically a problem of protein folding, whereby a mainly
random coil soluble peptide becomes aggregated, adopting a β-pleated sheet conformation
(Kelly, 1996, supra; Soto, 1999, supra). Amyloid formation proceeds by hydrophobic
interactions among conformationally altered amyloidogenic intermediates, which become
structurally organized into a β-sheet conformation upon peptide interaction. The
hydrophobicity appears to be important to induce interaction of the monomers leading to
aggregation, while the β-sheet conformation might determine the ordering of the aggregates in amyloid fibrils. In an attempt to inhibit amyloid fibril formation, these two properties were separated by designing short synthetic peptides bearing sequence homology and a
similar degree of hydrophobicity as the peptide domain implicated in the conformational
change, but having a very low propensity to adopt a β-sheet conformation (called β-sheet
breaker peptides) (Soto et al., 1996, supra; Soto et al., 1998, supra). The aim was to design a
peptide with the ability to bind specifically to the amyloidogenic peptide forming a complex
that stabilizes the physiological conformation and destabilizes the abnormal conformation of
the peptide (Soto, 1999, supra).
Table 1. Disorders related with amyloidosis and the protein component of the amyloid fibrils
DISEASE FIBRIL COMPONENT
Alzheimer's disease Amyloid-β protein
Primary systemic amyloidosis Immunoglobulin light chain or fragments thereof
Secondary systemic amyloidosis. Fragments of serum amyloid-A
Familial Mediterranean fever
Spongiform encephalopathy Fragments of pnon protein
Senile systemic amyloidosis, Transthyretin and fragments thereof
Familial amyloid polyneuropathy
Hemodialysis-related amyloidosis β2-mιcroglobulιn
Hereditary cerebral amyloid angiopathy, Icelandic type Cy stain C
Familial amyloidosis, Finnish type Gelsohn fragments
Type 11 diabetes Fragments of islet amyloid polypeptide
Familial amyloid polyneuropathy Fragments of apo poprotein A- l
Mcdullar carcinoma of the thyroid Fragments of calcitomn
Atπal amyloidosis Atπal natnuretic factor
Hereditary non-neuropathic systemic amyloidosis Lysozyme or fragments thereof
Hereditary renal amyloidosis Fibπnogen fragments
Islet amyloid Insulin
Amyloidosis in senescence Apohpoprotein A-I)
β-sheet breaker peptides have so far been designed to block the conformational
changes that occur in both Aβ and prion protein (PrP), which are implicated in the
pathogenesis of Alzheimer's and prion disease, respectively. The prior art has previously
shown that 11- and 5-residue β-sheet breaker peptides (namely, iAβl and iAB5, respectively)
homologous to the central hydrophobic region of Aβ inhibit peptide conformational changes
that result in amyloid formation and also dissolved preformed fibrils in vitro (Soto et al.,
Biochem. Biophys. Res. Commun. 226: 672-680, 1996; Soto et al., Nature Med. 4: 822-826,
1998). In addition, the 5-residue peptide is capable of preventing the neuronal death induced
by the formation of β-sheet rich oligomeric Aβ structures in cell culture experiments (Soto
et al., 1998, supra). Furthermore, by using a rat model of amyloidosis induced by
intracerebral injection of Aβl-42, the prior art has shown that co-injections of the 5-residue
β-sheet breaker peptide decreased cerebral Aβ accumulation and completely blocked the
deposition of fibrillar amyloid-like lesions in the rat brain (Soto et al., 1998, supra). Finally,
the β-sheet breaker peptide injected eight days after the injection of Aβ was able to
disassemble preformed Aβ fibrils in the rat brain in vivo, that leads to a reduction in the size
of amyloid deposits (Sigurdsson, Frangione, Soto, manuscript submitted). Interestingly,
removal of amyloid by the β-sheet breaker peptide reverts the associated cerebral histologic
damage, including neuronal shrinkage and microglial activation.
β-sheet breaker peptides have also been designed to prevent and to revert
conformational changes caused by prions (PrP). Based on the same principles and using as a
template the PrP sequence 114-122, the prior art has shown that when a set of β-sheet breaker peptides was synthesized, a 13-residue peptide (iPrP13) showed the greatest activity
(Soto, 1999, supra). Several in vitro cell culture and in vivo assays were used to test for
inhibitory activity and the results clearly indicated that it is possible not only to prevent the
PrPc — PrPsc conversion, but more interestingly to revert the infectious PrPsc conformer to a
biochemical and structural state similar to PrPc (Soto et al., manuscript submitted).
Short peptides have been utilized extensively as drugs in medicine (Rao et al., C.
Basava and G.M. Anantharamaiah, eds. Boston: Birkhauser, pp. 181-198, 1994). However,
the development of peptide drugs is strongly limited by their lack of oral bioavai lability and
their short duration of action resulting from enzymatic degradation in vivo (Fauchere and
Thurieau, Adv. Drug Res. 23: 127-159, 1992). Progress in recent years toward the
production of peptide analogs (such as pseudopeptides and peptide mimetics) with lower
susceptibility to proteolysis has increased the probability to obtain useful drugs structurally
related to their parent peptides (Fauchere and Thurieau, 1992, supra). Improving peptide
stability to proteases not only increases the half-life of the compound in the circulation but
also enhances its ability to be transported or absorbed at different levels, including intestinal
absorption and blood-brain barrier permeability, because transport and absorption appear to
be highly dependent upon the time of exposure of membranes or barriers to the bioactive
species (Fauchere and Thurieau, 1992, supra).
SUMMARY OF THE INVENTION
The present invention is an inhibitory peptide capable of inhibiting β pleated sheet formation in amyloid β-peptide, the inhibitory peptide being a βsheet breaker peptide analog
designed by chemical modification of a βsheet breaker peptide capable of inhibiting β
pleated sheet formation in amyloid β-peptide.
The peptide is altered chemically by: (1) modifications to the N- and C-terminal ends
of the peptide; (2) changes of the side-chain, which can involve amino acid substitutions; (3)
modification in the -carbon including methylations, alkylations and dehydrogenations; (4)
chirality changes by replacing D- for L-residue; (5) head-to-tail cyclizations; and (6)
introduction of amide bond replacements, i.e. changing the atoms participating in the peptide
(or amide) bond.
The present invention also includes an inhibitory peptide capable of inhibiting
conformational changes in prion PrP protein associated with amyloidosis, the inhibitory
peptide being a βsheet breaker peptide analog designed by chemical modification of a βsheet
breaker peptide capable inhibiting the conformational changes in prior PrP protein
associated with amyloidosis.
In addition, the present invention includes a peptide mimetic with the following
structure:
PMiAB5
In another embodiment, the peptide mimetic has the following structure:
PMiPrP13
In yet another embodiment, the peptide mimetic has the following structure:
PMiPrP5
The present invention also includes a method for preventing, treating, or detecting
disorders or diseases associated with abnormal protein folding into amyloid or amyloid-like
deposits or precursors thereof having a pathological beta-sheet structure is claimed.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is a schematic representation of the peptide bond and the potential target sites
for peptide modifications. FIG. 2 is a graph depicting the pharmacokinetics of a 11-residue β-sheet breaker
peptide inhibitor of Alzheimer's amyloidosis (Seq. RDLPFYPVPID) in its natural L-
configuration and in the non-natural D-form.
FIGS. 3A and 3B are representations of the tridimensional structure of Alzheimer's
and prion β-sheet breaker peptides iAβ5 and iPr P13, respectively.
FIG. 4a and 4b are graphs showing the bioavailability and stability of iAB5 and Ac-
iAB5-Am, respectively over time.
FIG. 5a provides a graphical comparison of Aβl-40 incubated with various other
peptides.
FIG. 5b is a graph of amyloid formation vs. the Ac-iAβ5-Am concentration.
FIG. 5c is a graph of amyloid formation vs. the iAβ5 concentration.
FIG. 6 shows a model where there is an 83% dissolution of deposits in the ventricle
area and a 30% dissolution of amyloid plaque in the amygdala.
DETAILED DESCRIPTION OF THE INVENTION
In the present invention, the bioavailability and stability of an inhibitory peptide is
improved by chemically modifying the parent peptide to produce a derivative more suitable
for in vivo use, which is preferably administered orally. The inhibitory peptide is capable of
inhibiting β pleated sheet formation in an amyloid β-peptide. Moreover, the inhibitory
peptide is a βsheet breaker peptide analog designed by chemical modification of a βsheet
breaker peptide capable of inhibiting β pleated sheet formation in amyloid β-peptide. This invention also includes an inhibitory peptide capable of inhibiting conformational changes in prion PrP protein associated with amyloidosis, where the inhibitory peptide is a βsheet breaker peptide analog designed by chemical modification of a
βsheet breaker peptide and is capable of inhibiting the conformational changes in prion PrP
protein associated with amyloidosis.
In Fig. 1, a generalized peptide backbone is shown, where possible targets for chemical modification are highlighted. The possible targets include the following: (1)
modifications to the N- and C-terminal ends of the peptide (targets a and b); (2) changes of
the side-chain (target c) which usually involve amino acid substitutions; (3) modification in
the α-carbon (target d) including methylations, alkylations and dehydrogenations; (4)
chirality changes by replacing D- for L-residue; (5) head-to-tail cyclizations; and (6) introduction of amide bond replacements (target e), i.e. changing the atoms participating in
the peptide (or amide) bond. The latter derivatives are known as pseudopeptides or amide
bond surrogates.
Natural peptides are usually degraded by the concerted action of specialized
endopeptidases and unspecific exopeptidases. Endopeptidases are often present in tissues
and cellular compartments and convert the peptide into two or more inactive fragments.
Exopeptidases are generally present in blood and peripheral organs and carry out the
degradation of the intact peptides or their fragments to the constituent amino acids and hence
contribute to the disappearance of the peptides from the circulation. Exopeptidases
recognize the free amino or carboxyl groups in peptides. Therefore, modification of those groups often diminish or abolish exopeptidase degradation. Head-to-tail peptide cyclization
results in the absence of free end-terminal groups, and hence also minimizes cleavage by
exopeptidases. On the other hand, endopeptidases recognize the atoms participating in the
amide bond. Thus, amide bond replacements dramatically decrease degradation by
endopeptidases. The same usually happens with modifications to the α-carbon. Since most
(if not all) of the exo- and endo-proteases are stereospecific, substitutions of the natural L-
amino acids by the D-stereoisomers result in a clear increase in peptide stability. Finally,
peptide mimetics are usually completely resistant to proteolytic degradation and often can be
administrated orally.
β-sheet breaker analogues designed by chemical modifications of the lead peptides.
Starting from the 5-residue Alzheimer's inhibitor peptide (iAB5, Seq. LPFFD - also
denoted as Leu Pro Phe Phe Asp) and the 13-residue prion inhibitor peptide (iPrP13, Seq.
DAPAAPAGPAVPV - also denoted as Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro
Val), the modifications described below are designed. The peptides used in the present
invention are synthesized using standard protocols as disclosed by Bergmann et al., and
incorporated herein by reference. (Bergmann & Zervas, Berichte der Deutschen Chemischen
Gesellschaft (1932) 65: 1192 - 1201)
a) N- and C-terminal modifications. Ν-terminal acetylation or desamination confers
protection against digestion by a number of aminopeptidases while the presence of amides or
alcohols replacing the C-terminal carboxyl group prevent splitting by several carboxypeptidases, including carboxypeptidases A and B. The altered peptide sequences
including these modifications are the following, where ac is acetylation, am is amidation, des
is desamination, and ale is alcoholization:
Alzheimer's Inhibitors Prion Inhibitors ac-Leu Pro Phe Phe Asp-am ac-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-a des-Leu Pro Phe Phe Asp-am des-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-am ac-Leu Pro Phe Phe Asp-ale ac-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc des-Leu Pro Phe Phe Asp-ale des-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc
b) Side-chain changes. The presence of non-natural amino acids usually increase
peptide stability. In addition, at least one of these amino acids (α-aminoisobutyric acid or
Aib) imposes significant constraints to model peptides diminishing their conformational
flexibility. In particular, the incorporation of Aib into β-sheet model peptides induces the
complete disruption of this structure. The β-sheet blocking activity of Aib is comparable or
even greater than the natural residue proline used in the peptide as a β-sheet blocker.
Therefore, the introduction of Aib is expected to enhance peptide stability and inhibitory
activity at the same time.
Alzheimer's Inhibitors Prion inhibitors
Leu Aib Phe Phe Asp Asp Ala Aib Ala Ala Aib Ala Ala Aib Ala Gly Aib Ala Val Aib Val
c) Modifications in the a-carbon. The most commonly used α-carbon modification
to improve peptide stability is α-methylation. In addition, replacement of the hydrogen atom linked to the α-carbon of Phe, Val or Leu has been shown to favor the adoption of β-bend
conformation and strongly disfavor the formation of β-pleated sheet structures. According to
the present invention, methylation of those residues in the inhibitor peptides is expected to enhance stability and potency. Alzheimer's Inhibitors
(Me)Leu Pro Phe Phe Asp
Leu Pro (Me)Phe Phe Asp
Leu Pro Phe (Me)Phe Asp
(Me)Leu Pro (Me)Phe (Me)Phe Asp
Prion inhibitors
Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala (Me)Val Pro Val
Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro (Me)Val
Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala (Me)Val Pro (Me)Val
d) Chirality changes. Replacement of the natural L-residue by the D-enantiomers
dramatically increases resistance to proteolytic degradation. The increase in stability by
introduction of D-residue has already been demonstrated for the 1 l-residue β-sheet breaker
peptide (iAβl). In vivo studies showed that the peptide bearing the natural sequence rapidly
degraded in rat plasma. Indeed, approximately 90% of iAβl was degraded within minutes
after intravenous injection. Conversely, a derivative of iAβl containing all the residue in
the D-form showed virtually no degradation in the plasma after injection for 15 minutes. For detection, the peptide was radio-iodinated using standard procedures. Peptide stability was
evaluated after i.v bolus injection in rats by precipitation with trichloroacetic acid. Quantitation of the intact peptide was also done by paper chromatography. Thus, iAB5 and
iPrP13 peptides (FIGS. 3A and 3B, respectively) containing all-D residue as well as
peptides containing D-residue only at the N- and C-terminal ends to prevent exopeptidase
degradation are included in the compounds of the invention. In addition to the latter, D-
residue are used after each proline amino acid, since it has been reported that a frequent
endopeptidase cleavage site is after this residue by an enzyme known as
prolylendopepti dase .
Alzheimer's Inhibitors Prion Inhibitors leu pro phe phe asp asp ala pro ala ala pro ala gly pro ala val pro val leu Pro Phe Phe asp asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro val leu Pro phe Phe asp asp Ala Pro ala Ala Pro ala Gly Pro ala Val Pro val
Amino acids written with lower case letters denote D-residue.
e) Cyclic peptides. Conformationally constrained cyclic peptides represent better
drug candidates than linear peptides due to their reduced conformational flexibility and
improved resistance to exopeptidase cleavage. Two alternative strategies have been used to
convert a linear sequence into a cyclic structure. One is the introduction of cysteine residue to achieve cyclization through the formation of a disulfide bridge and the other is the side-
chain attachment strategy involving resin-bound head-to-tail cyclization. To avoid
modifications of the peptide sequence the latter approach is used, β-sheet breaker peptides
contain the ideal sequences for facilitating macrocyclization because proline, due to its
ability to promote turns and loops, is a constituent of many naturally occurring or artificially
synthesized cyclic peptides. Alzheimer's Inhibitors Prion inhibitors
*— Leu Pro Phe Phe Asp — -ι i— Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val — -ι
f) Pseudopeptides. Pseudopeptides or amide bond surrogates refers to peptides containing chemical modifications of some (or all) of the peptide bonds. Amide bond
replacements are usually represented by retaining the amino acid designation according to
the side-chain and specifying the changes that occur between the α-carbons, using the
nomenclature known as "psi-bracket."
For example, the term Alaψ[CH CH2]Gly refers to the moiety
NH2CH(CH3)CH2CH2CH2CO2H. Several amide bond surrogates have been described in Table 2 below.
Table 2. Some amide bond surrogates and their properties
Some of them are found in naturally occurring peptide analogs (such as ψ[CHOH],
ψ[CSNH], ψ[COO]) while others have been artificially synthesized The introduction of
amide bond surrogates not only decreases peptide degradation but also may significantly
modify some of the biochemical properties of the peptides, particularly the conformational
flexibility and hydrophobicity It is likely that an increase in conformational flexibility will
be beneficial for docking the inhibitor to the Aβ and PrP binding sites On the other hand,
since the interaction between the amyloidogenic proteins and the inhibitors seems to depend
to a great extent on hydrophobic interactions, it is likely that amide bond replacement
increasing hydrophobicity may enhance affinity and hence, potency of the inhibitors. In addition, increased hydrophobicity could also enhance transport of the peptide across
membranes and thus, improve barrier permeability (blood-brain barrier and intestinal
barrier). Therefore, to synthesize pseudopeptides amide bond replacement is used thereby
increasing flexibility and hydrophobicity, such as ψ[CH2CH2] and ψ[CH2S]. The amide
bonds to replace are those located at the end of the peptide to prevent exoprotease
degradation and after each of the prolines, since it has been described that a frequent
endopeptidase cleavage site occurs after this residue by an enzyme known as
prolylendopeptidase. Additional amide bonds that need to be protected are determined by
experimental studies involving the analysis of the degradation of β-sheet breaker peptides in
the plasma and tissue.
Alzheimer's Inhibitors Leuψ[CH2CH2]Proψ[CH2CH2]Phe Pheψ[CH2CH2]Asp Leuψ[CH2S]Proψ[CH2S]Phe Pheψ[CH2S]Asp Prion inhibitors Aspψ[CH2CH2]Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ[CH2CH2]Ala Val Proψ[CH2CH2]Val
Aspψ[CH2SlAla Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Proψ[CH2S]Val
g) Mixture of several modifications. By taking into account the features of the
peptide drugs on the market or under current development, it is clear that most of the
peptides successfully stabilized against proteolysis consist of a mixture of several types of
the above described modifications. This conclusion makes sense in the light of the
knowledge that many different enzymes are implicated in peptide degradation. The following structures contain combinations of different types of chemical modifications:
Alzheimer's Inhibitors Ac-Leu Proψ[CH2CH2]Phe Phe Asp-Am Ac-Leu Proψ[CH2S]Phe Phe Asp-Am (Me)Leu Proψ[CH2CH2]Phe Phe Asp-Am leu Proψ[CH2CH2]Phe Phe asp leu Proψ[CH2S]Phe Phe asp Ac-Leu Aib Phe Phe Asp- Am (Me)Leu Aib Phe Phe Asp- Am Leu Proψ[CH2CH2]Phe Phe asp
j — Leu Aib Phe Phe Asp — i
j — Leu Proψ [CH2CH2] Phe Phe Asp — ι
Ac-Leu pro Phe Phe Asp- Am
Ac-Leu Proψ[CH2CH2]Phe phe Asp- Am
Ac-Leu Proψ[CH2SJPhe phe Asp- Am
Ac-Leu Proψ[CH2CH2]Phe (Me)Phe Asp-Am
Ac-Leu Proψ[CH2CH2]Phe (Me)Phe asp Ac-Leu Pro phe phe Asp-Am
Ac-Leu Pro (Me)Phe phe Asp- Am leu Proψ[CH2CH2]Phe phe asp leu Pro (Me)Phe phe asp
Ac-Leu Aib Phe phe Asp- Am Prion inhibitors Ac-Asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ [CH2CH2] Ala Val Pro Val-Am asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ[CH2CH2]Ala Val Pro val Ac-Asp Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Pro Val-Am asp Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Pro val Ac-Asp Ala Aib Ala Ala Aib Ala Gly Aib Ala Val Pro Val-Am
Ac-Asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ [CH2CH2] Ala Val Pro (Me) Val Ac-Asp Ala pro Ala Ala Proψ[CH2CH2]Ala Gly pro Ala Val Pro Val-Am asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ[CH2CH2]Ala Val Pro (Me) Val asp Ala Aib Ala Ala Proψ[CH2CH2]Ala Gly pro Ala Val Pro (Me) Val asp Ala Aib Ala Ala Proψ[CH2S]Ala Gly pro Ala Val Pro (Me) Val asp Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Pro (Me) Val Ac-Asp Ala Aib Ala Ala Proψ[CH2CH2]Ala Gly Aib Ala Val Pro (Me)Val
j A Asspp A Allaa pprroo A Allaa A Allaa P Prrooψψ[[CCHH22CCHH22]J A Allaa G Gllyy pprroo A Allaa V Vaall P Prroo V Vaall- 1
j Asp Al Aib Ala Ala Proψ[CH2 CH2] Ala Gly Aib Ala (Me) Val Pro Val 1
Ac-Asp Ala Proψ[CH2S]Ala ala Pιoψ[CH2S]Ala gly Proψ[CH2S]Ala (Me)Val Pro Val-Am Ac-Asp Ala Aib ala Ala Proψ[CH2CH2]Ala Gly pro Ala Val Pro (Me) Val asp Ala Aib Ala Ala Proψ[CH2CH2]Ala Gly Aib ala Val Pro Val-Am Ac-Asp Ala pro Ala Ala Proψ[CH2CH2]Ala gly pro Ala (Me) Val Pro Val-Am asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala gly Proψ[CH2CH2] Ala val Pro val
Ac-Asp Ala pro Ala ala Aib Ala gly pro Ala (Me) Val Pro Val-Am
Asp Ala pro Ala Ala Proψ[CH2 CH2] Ala Gly pro Ala Val Pro Val
Asp Ala Aib Ala Ala Proψ[CH2 CH2] Ala Gly Aib Ala (Me) Val Pro Val Another approach to improve stability, which also may result in the generation of
orally active compounds, is to produce a peptide mimetic. A peptide mimetic is a molecule
that mimics the biological activity of the peptides, but is no longer a peptide in chemical
nature. The term peptide mimetic has been used sometimes to describe molecules that are
partially peptide in nature, such as pseudopeptides, semi-peptides or peptoids, but a strict
definition and the one that is used in the present application is an organic molecule that no
longer contains any peptide bonds. Peptide mimetics are not derivatives of a parent peptide,
but rather are chemically synthesized de novo trying to mimic the structural and functional
properties of the peptide. The rational design of peptide mimetics requires a sufficient
knowledge of the pharmacophoric groups that are responsible for the activity and detailed
structural information of the peptide. The objective is to reconstruct the spatial position of
the pharmaco-active groups using an organic template to mount them. Selection of the
template is important and has to take into consideration the size and flexibility based on the
conformational model of the peptide.
Peptide mimetics designed to imitate β-sheet breaker peptide properties.
The rational design of peptide mimetics requires a sufficient knowledge of the
chemical groups that are responsible for the activity and detailed structural information of
the peptide. The objective is to reconstruct the position of the pharmaco-active groups using
an organic template to mount them. Selection of the template is important and has to take into consideration the size and flexibility based on the conformational model of the peptide.
From the study of the activity of different β-sheet breaker sequences bearing single amino
acid substitutions, the residues that are key for inhibition have been determined. In addition,
the tri dimensional structure of the lead Alzheimer's and prion β-sheet breaker peptides
(FIGS. 3 A and 3B) were either modeled or experimentally determined. The 5-residue
inhibitor of Aβ fibrillogenesis was modeled by energy minimization and Monte Carlo
simulations using the computer program ICM. The structure of the 13-residue inhibitor of
prion protein conformational changes was experimentally calculated by 2D-NMR.
There are numerous approaches to the design and synthesis of peptide mimetics as
described in recent reviews by Joachim Gante and Iwao Ojima et al. of which are
incorporated herein by reference.
The peptide mimetics shown below represent a further aspect of this invention.
Alzheimer's Inhibitors
PMiAB5 Prion inhibitors
PMiPrP13
PMiPrP5
The latter (PMiPrP5) is a shorter and easier to synthesize version that contains the
chemically active groups and is analog to a 5-residue prion β-sheet breaker peptide.
As a method of preventing or treating a disorder or disease associated with amyloid
or amyloid-like deposits or pathological beta-sheet-rich precursors thereof, the compound of
the present invention is administered in an effective amount to a subject in need thereof,
where the subject can be human or animal. Likewise, a method of detecting such disorders
or diseases also includes administering a sufficient amount of the designed compound to
visualize its binding to fibril deposits or precursors thereof by well-known imaging techniques. As used herein, the term "prevention" of a condition, such as Alzheimer's disease or
other amyloidosis disorders, in a subject involves administering the compound according to
the present invention prior to the clinical onset of the disease. "Treatment" involves
administration of the protective compound after the clinical onset of the disease. For
example, successful administration of the compound of the present invention, after
development of a disorder or disease comprises "treatment" of the disease. The invention is
useful in the treatment of humans as well as for veterinary uses in animals.
The compound of the present invention may be administered by any means that
achieves its intended purpose, preferably oral. For example, administration may be by a
number of different parenteral routes including, but not limited to, subcutaneous,
intravenous, intradermal, intramuscular, intraperitoneal, intracerebral, intranasal, oral,
transdermal, or buccal routes. Parenteral administration can be bolus injection or by gradual
perfusion over time.
A typical regimen for preventing, suppressing, or treating a condition associated with
amyloid or amyloid-like deposits, comprises either: (1) administration of an effective
amount in one or two doses of a high concentration of the compound in the range of 0.5 to
10 mg, more preferably 0.5 to 5 mg, or (2) administration of an effective amount of the
compound administered in multiple doses of lower concentrations in the range of 10 -
10,000 μg, more preferably 50 - 500 μg over a period of time up to and including several
months to several years.
It is understood that the dosage administered will be dependent upon the age, sex, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of
treatment, and the nature of the effect desired. The total dose required for each treatment
may be administered by multiple doses or in a single dose. By "effective amount," it is
meant a concentration of the compound which is capable of slowing down or inhibiting the
formation of amyloid or amyloid-like deposits, or pathological beta-sheet precursors thereof,
or of dissolving preformed fibril deposits. Such concentrations can be routinely determined
by those of skill in the art. It will also be appreciated by those of skill in the art that the
dosage may be dependent on the stability of the administered compound. A less stable
compound may required administration in multiple doses.
Preparations for parenteral administration include sterile aqueous or non-aqueous
solutions, suspensions, and emulsions, which may contain auxiliary agents or excipients
which are known in the art. Pharmaceutical compositions such as tablets and capsules can
also be prepared according to routine methods.
Pharmaceutical compositions comprising the compound of the invention include all
compositions wherein the compound is contained in an amount effective to achieve its
intended purpose. In addition, the pharmaceutical compositions may contain suitable
pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate
processing of the active compounds into preparations which can be used pharmaceutically.
Suitable pharmaceutically acceptable vehicles are well known in the art and are described for
example in Gennaro, Alfonso, Ed., Remington's Pharmaceutical Sciences, 18l Edition 1990, Mack Publishing Co., Easton, PA, a standard reference text in this field. Pharmaceutically acceptable vehicles can be routinely selected in accordance with the mode
of administration and the solubility and stability of the compound. For example,
formulations for intravenous administration may include sterile aqueous solutions which
may also contain buffers, diluents and other suitable additives.
Suitable formulations for parenteral administration include aqueous solutions of the
active compounds in water-soluble form, for example, water-soluble salts. In addition,
suspension of the active compound as appropriate oily injections suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame
oil, or synthetic fatty acid esters, for example ethyl oleate or triglycerides. Aqueous injection
suspensions that may contain substances which increase the viscosity of the suspension
include, for example, sodium carboxymethyl cellulose, sorbitol, and/or destran. Optionally,
the suspension may also contain stabilizers.
Disorders or diseases associated with abnormal protein folding into amyloid or
amyloid-like deposits or into pathological beta-sheet-rich precursors of such deposits to be
treated or prevented by administering the pharmaceutical composition of the invention
includes, but is not limited to, Alzheimer's disease, FAF, Down's syndrome, other
amyloidosis disorders, human prion diseases, such as kuru, Creutzf el dt- Jakob Disease
(CJD), Gerstmann-Strausslet-Scheinker Syndrome (GSS), prion associated human
neurodegenerative diseases as well as animal prion diseases such as scrapie, spongiform
encephalopathy, transmissible mink encephalopathy and chronic wasting disease of mule
deer and elk. EXAMPLES
One of the major drawbacks for the use of peptides as drugs is their rapid proteolytic
degradation in biological fluids and tissues. In in vitro experiments, iAB5, (Seq LPFFD- also
depicted as Leu Pro Phe Phe Asp herein) degraded very quickly in vitro after incubation
with fresh human plasma. As shown in Figure 4a , fifty percent of the peptide iAB5
disappeared in approximately 5 minutes in the presence of plasma. Since it was not possible
to identify any metabolic fragments as a result of the proteolytic digestion, it seems likely
that the degradation is mainly done by unspecific exopeptidases. This conclusion is
supported by the finding that protection of amino- and carboxy-terminus of the peptide by
acetylation and amidation, respectively, (to form Ac-iAβ5-Am — also depicted as Ac-Leu
Pro Phe Phe Asp-Am herein) dramatically increases the stability of the peptide in vitro. As
shown in Fig. 4b, the end-protected modified peptide of the present invention (Ac-iAβ5-Am)
remained stable for a period of more than 24 hours in human plasma. (The modified peptide
was also slowly metabolized in vitro in human and rat liver microsomes, in which after one
hour of incubation at 37° a 81.5% and 76.3% of the peptide remained intact, in human and
rat tissue homogenate, respectively. )
Additional in vitro studies showed that Ac-iAβ5-Am has similar activity as iAB5 in inhibiting amyloid formation (see Fig. 5a) and the effect followed a similar dose-dependency
as the activity of the unmodified peptide as shown in Figures Fig. 5b and 5c. Returning to
Fig. 5a, it can be seen that modification of the N-terminus by Boc also retains the in vitro activity exhibited by iAB5 while several unrelated peptides (CPLVHVSEEGTEPA, CP2:
GYLTVAAVFRG, CP10: ISEVKMDAEF) or short Aβ fragments (such as Aβ 18-21, AB1- 16) at the same concentrations had no effect on fibrillogenesis or slightly increased amyloid
formation probably by incorporation into the fibrils.
To evaluate the effect of Ac-iAB5-Am in vivo, we used a rat model in which amyloidosis was induced by intracerebral injection of non-aggregated AB1-42. After some
time, the peptide aggregates inside the rat brain resulting in the formation of a single
amyloid-like deposit in the place of injection. These lesions have the same tinctorial (congo
red birefringence and thioflavine S binding) and translucent (fibrillar structure under electron
microscopy) properties than Alzheimer's amyloid plaques and induce some cerebral damage
similar to that observed in AD brain, including extensive neuronal shrinkage, astrocytosis
and microglial activation. Using this model, we have shown previously that co-injection of
the unprotected iAB5 with AB1-42 induce a 50% inhibition of amyloid plaque formation and
i. c. injection of iAB5 in animals already containing amyloid plaques produced a 67%
dissolution of preformed deposits. (Sigurdsson, E.M., Permanne, B. Soto, C. , Wisniewski,
T. & Frangione, B. (2000) In vivo disassembly of amyloid-β deposits in rat brain. J.
Neuropath. Exp. Neurol. 59: 11-17) In the previous experiments, the unprotected peptide
was injected directly in the brain region where the amyloid was located. In the present
experiment the amyloid-B5 peptide was injected into the amygdala of the rats. After 7
days, which is the time required to have fully formed the amyloid deposits, one-hundred μL of a solution containing 13 mg/ml of the Ac-iAB5-Am were infused for a period of three
weeks using an ALZET infusion pump connected to the lateral ventricle. The animals were
sacrificed and the brain analyzed for the presence of amyloid deposits by immunohistochemistry. In this model, a compacted amyloid plaque was obtained in the
place where the solution containing ABl-42 was deposited (amygdala) and also several
smaller amyloid deposits were observed throughout the canula track in regions closer to the
ventricle (Fig. 6, left panel). The results show that infusion of the peptide induces a 30%
dissolution of preformed amyloid plaque in the amygdala and 83% dissolution of the
deposits located near the ventricle (Fig. 6).
Experimental Procedures
In vitro assays of peptide stability. Peptides were prepared as a 1 μg/μl solution in water.
20 μl of the peptide solution was diluted in 80 μl of fresh human plasma. The solution was
incubated at 37°C for different time periods and the reaction was stopped by adding a
complete cocktail of protease inhibitors. The bulk of the plasma proteins (none of the
peptide) were precipitated in cold methanol (mix/MeOH, 4/5, v/v) for one hour at -20°C.
The precipitated proteins were pelleted by centrifugation (10 OOOg, 10 min, 4°C). The
supernatant, containing the peptide, was concentrated 5 times under vacuum and separated
by reverse-phase HPLC. The peak area corresponding to the intact peptide was measured
and compared with an equivalent sample incubated without plasma. In vitro assays of activity. Amyloid formation was quantitatively evaluated by the
fluorescence emission of thioflavine T (ThT) bound to amyloid fibrils. Aliquots of Aβ at a
concentration of 0.5 mg/ml prepared in 0.1M Tris, pH 7.4 were incubated for 7 days at 37°C
in the absence or in the presence of different concentrations of iAB5 and derivatives. At the
end of the incubation period, 50 mM glycine, pH 9.2 and 2 μM ThT were added in a final
volume of 2 ml. Fluorescence was measured at: excitation 435 nm and emission 485 nm in
a Perkin Elmer, model LS50B fluorescence spectrometer.
In vivo studies using an animal model of cerebral AP deposition. Male Fischer-344 rats
weighed 250-300g and were 3-4 months of age at the time of arrival. The animals were
housed 2 per cage, maintained on a 12 hour light-dark cycle with access to food and water ad
libitum and were habituated to their new environment for 2-3 weeks prior to surgery.
Surgery was performed under sodium pentobarbital (50 mg/kg, i.p.) anesthesia. Atropine
sulfate (0.4 mg/kg) and ampicillin sodium salt (50 mg/kg) were injected subcutaneously
once the animals were anesthetized. Aβl-42 was dissolved in dimethylsulfoxide (DMSO)
and then diluted with water to a 16.7% DMS. The animal received a bilateral injection of
5.0 nmol Aβl-42 into each amygdala by using a Kopf stereotaxic instrument with the incisor
bar set at 3.3 mm below the interaural line. Injection coordinates measured from the bregma
and the surface of the skull (AP -3.0, ML ± 4.6 DV -8.8) were empirically determined based
on the atlas of Paxinos and Watson. A volume of 3.0 μl was administered over 6 min (flow rate 0.5 μl/min) using a CMA/100 micrasyringe pump. The cannula was left in situ for 2 min
following injection, then it was withdrawn 0.2 mm and left for 3 min, and after 5 min the
cannula was slowly withdrawn. Following surgery the animals were placed on a heating pad
until they regained their righting reflex. To evaluate the effect of Ac-iB5-AM the animals
were subjected to a second surgery one week after the first one, in which an ALZET infusion
pump was connected to the cerebral ventricle following the manufacturer indications. A
total of 1.3 mg of peptide in 100 μl of PBS/10% DMSO was delivered into the lateral
ventricle over a period of 3 weeks. After this time, the animals were sacrificed by an
overdose of sodium pentobarbital (150 mg/kg, i.p.), perfused transaortically. For histology,
serial coronal sections (40 μm) of the brain were cut, placed in ethylene glycol
cryoprotectant and stored at -20°C until stained. Tissue sections were stained with anti ABl-
42 antibodies as described in Soto, C, Sigursson, E., Morelli, L., Kumar, R.A., Castano,
E.M. and Frangione, B.(1998) β-sheet breaker peptides inhibit fibrillogencsis in a rat brain
model of amyloidosis: Implications for Alzheimer's therapy. Nature med. 4: 822-826. An
image analysis system was used to determine the size of the amyloid deposits. The data was
analyzed by a two-way ANOVA followed by a Newman-Keuls' multiple range test for/705/
hoc comparisons. Total brain deposition was analyzed using an unpaired t-test, two tailed.
Having now fully described this invention, it will be appreciated by those skilled in
the art that the same can be performed within a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention
and without due experimentation.
While this invention has been described in connection with specific embodiments
thereof, it will be understood that it is capable of further modifications. This application is
intended to cover any variations, uses, or adaptations of the inventions following, in general,
the principles of the invention and including such departures from the present disclosure as
come within known or customary practice within the art to which the invention pertains and
as may be applied to the essential features hereinbefore set forth as follows in the scope of
the appended claims.
All references cited herein, including journal articles or abstracts, published or
corresponding U.S. or foreign patent applications, issued U.S. or foreign patents, or any
other references, are entirely incorporated by reference herein, including all data, tables,
figures, and text presented in the cited references. Additionally, the entire contents of the
references cited within the references cited herein are also entirely incorporated by reference.
Reference to known method steps, conventional methods steps, known methods or
conventional methods is not in any way an admission that any aspect, description or
embodiment of the present invention is disclosed, taught or suggested in the relevant art.
The foregoing description of the specific embodiments will so fully reveal the
general nature of the invention that others can, by applying knowledge within the skill of the
art (including the contents of the references cited herein), readily modify and/or adapt for
various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present invention. Therefore, such adaptations and
modifications are intended to be within the meaning and range of equivalents of the
disclosed embodiments, based on the teaching and guidance presented herein. It is to be
understood that the phraseology or terminology herein is for the purpose of description and
not of limitation, such that the terminology or phraseology of the present specification is to
be interpreted by the skilled artisan in light of the teachings and guidance presented herein,
in combination with the knowledge of one of ordinary skill in the art.

Claims

WHAT IS CLAIMED IS:
1. An inhibitory peptide capable of inhibiting β pleated sheet formation in amyloid β-
peptide said inhibitory peptide being a βsheet breaker peptide analog designed by chemical
modification of a βsheet breaker peptide capable of inhibiting β pleated sheet formation in
amyloid β-peptide.
2. The inhibitory peptide of claim 1 wherein said βsheet breaker peptide is a 5 residue
Alzheimer inhibitor peptide ιAβ5 (Seq Leu-Pro-Phe-Phe-Asp)
3. The inhibitory peptide of claim 2 wherein said chemical modification is achieved by
a process selected from the group consisting of alteration of the N- and C- terminal ends of
said Alzheimer inhibitor peptide ιAβ5; replacing at least one residue of said Alzheimer
inhibitor peptide I Aβ5wιth α-aminoisobuiπc acid (Aib); methylation of the α carbon of at
least one residue of said Alzheimer inhibitor peptide ιAβ5, replacing at least one L-
enantiomeπc residue of said Alzheimer inhibitor peptide ιAβ5 with a D-enantiomeπc
residue, forming head to tail cyclization of said Alzheimer inhibitor peptide ιAβ5, replacing
amide bonds in said Alzheimer inhibitor peptide ιAβ5 with an amide bond surrogate, and
combinations thereof.
4. The inhibitory peptide of claim 3 said alteration of the N- and C- terminal ends of said Alzheimer inhibitor peptide iAβ5 is achieved by a process selected from
acetylation, amidation, desamination, alcoholization and combinations thereof.
5. The compound of Claim 4 wherein inhibitory peptide is selected from the group
consisting of: ac-Leu Pro Phe Phe Asp-am, des-Leu Pro Phe Phe Asp-am, ac-Leu Pro Phe Phe Asp-ale, and des-Leu Pro Phe Phe Asp-ale,
6. The inhibitory peptide of Claim 5 wherein said inhibitory peptide is ac-Leu Pro Phe
Phe Asp-am.
7. The inhibitory peptide of claim 3 wherein said inhibitory peptide is selected from the
group consisting of Leu Aib Phe Phe Asp; (Me)Leu Pro Phe Phe Asp; Leu Pro (Me)Phe Phe Asp, Leu Pro
Phe (Me)Phe Asp; (Me)Leu Pro (Me)Phe (Me)Phe Asp, leu pro phe phe asp, leu Pro Phe Phe asp, leu Pro phe Phe asp,
Leuψ[CH2CH2]Proψ[CH2CH2]Phe Pheψ[CH2CH2]Asp; Leuψ[CH2S]ProψtCH2S]PhePheψ[CH2S]Asp; Ac-Leu Proψ[CH2CH2]Phe Phe Asp- Am;
Ac-Leu Proψ[CH2S]Phe Phe Asp-Am;
(Me)Leu Proψ[CH2CH2lPhe Phe Asp-Am; leu Proψ[CH2CH2]Phe Phe asp; leu Proψ[CH2S]Phe Phe asp; Ac-Leu Aib Phe Phe Asp-Am;
(Me)Leu Aib Phe Phe Asp- Am;
Leu Proψ[CH2CH2]Phe Phe asp;
j — Leu Proψ [CH2CH2] Phe Phe Asp — j
Ac-Leu pro Phe Phe Asp- Am; Ac-Leu Proψ[CH2CH ]Phe phe Asp-Am; Ac-Leu Proψ[CH2S]Phe phe Asp- Am; Ac-Leu Proψ[CH2CH2]Phe (Me)Phe Asp- Am; Ac-Leu Proψ[CH2CH2]Phe (Me)Phe asp; Ac-Leu Pro phe phe Asp- Am; Ac-Leu Pro (Me)Phe phe Asp- Am; leu Proψ[CH2CH2]Phe phe asp; leu Pro (Me)Phe phe asp; Ac-Leu Aib Phe phe Asp- Am; and
8. An inhibitory peptide capable of inhibiting conformational changes in prion PrP
protein associated with amyloidosis, said inhibitory peptide being a βsheet breaker peptide
analog designed by chemical modification of a βsheet breaker peptide capable inhibiting said
conformational changes in prior PrP protein associated with amyloidosis.
9. The inhibitory peptide of claim 8 wherein said βsheet breaker peptide is 13 residue
prion inhibitor peptide iPrP13 (Seq. Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val).
10. The inhibitory peptide of claim 9 wherein said chemical modification is achieved by
a process selected from the group consisting of: alteration of the N- and C- terminal ends of
said prion inhibitor peptide iPrP13; replacing at least one residue of said prion inhibitor
peptide iPrP13with α-aminoisobuiric acid (Aib); methylation of the α carbon of at least one
residue of said prion inhibitor peptide iPrP13; replacing at least one L-enantiomeric
residue of said prion inhibitor peptide iPrP13 with a D-enantiomeric residue, forming head
to tail cyclization of said prion inhibitor peptide iPrP13, replacing amide bonds in said
prion inhibitor peptide iPrP13 with an amide bond surrogate; and combinations thereof.
11. The inhibitory peptide of claim 10 wherein said alteration of the N- and C-
terminal ends of said prion inhibitor peptide iPrP13 is achieved by a process selected from
acetylation, amidation, desamination, alcoholization and combinations thereof.
12. The compound of Claim 11 wherein said inhibitory peptide is selected from the
group consisting of: ac-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-am, des-Asp Ala
Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-am, ac-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc, and des-Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro Val-alc.
13. The inhibitory peptide of claim 10 wherein said inhibitory peptide is selected from
the group consisting of
Asp Ala Aib Ala Ala Aib Ala Ala Aib Ala Gly Aib Ala Val Aib Val;
Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala (Me) Val Pro Val; Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro (Me) Val;
Asp Ala Pro Ala Ala Pro Ala Gly Pro Ala (Me) Val Pro (Me) Val; asp ala pro ala ala pro ala gly pro ala val pro val; asp Ala Pro Ala Ala Pro Ala Gly Pro Ala Val Pro val; asp Ala Pro ala Ala Pro ala Gly Pro ala Val Pro val;
Aspψ[CH2CH2]Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2] Ala Gly Proψ[CH2CH2]Ala
ValProψ[CH2CH2]Val;
Aspψ[CH2S]Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Proψ[CH2S]Val,;
Ac-Asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ[CH2CH2]Ala Val Pro Val-Am; asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ[CH2CH2]Ala Val Pro val;
Ac-Asp Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Pro Val-Am; asp Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Pro val;
Ac-Asp Ala Aib Ala Ala Aib Ala Gly Aib Ala Val Pro Val-Am;
Ac-Asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala Gly Proψ[CH2CH2]Ala Val Pro (Me)Val; Ac-Asp Ala pro Ala Ala Proψ [CH2CH2] Ala Gly pro Ala Val Pro Val-Am; asp Ala Proψ[CH2CH2]AIa Ala Proψ[CH2CH2]Ala Gly Proψ [CH2CH2] Ala Val Pro (Me) Val; asp Ala Aib Ala Ala Proψ[CH2CH2]Ala Gly pro Ala Val Pro (Me) Val; asp Ala Aib Ala Ala ProψfCH2S]Ala Gly pro Ala Val Pro (Me)Val; asp Ala Proψ[CH2S]Ala Ala Proψ[CH2S]Ala Gly Proψ[CH2S]Ala Val Pro (Me) Val; Ac-Asp Ala Aib Ala Ala Proψ [CH2CH2] Ala Gly Aib Ala Val Pro (Me) Val; j — Asp Ala pro Ala Ala Proψ[CH2CH2] Ala Gly pro Ala Val Pro Val — |
Asp Al Aib Ala Ala Proψ[CH2 CH2] Ala Gly Aib Ala (Me) Val Pro Val — |
Ac-Asp Ala Proψ[CH2S]Ala ala Proψ[CH2S]Ala gly Proψ[CH2S]Ala (Me)Val Pro Val-Am; Ac-Asp Ala Aib ala Ala Proψ[CH2CH2]Ala Gly pro Ala Val Pro (Me) Val, asp Ala Aib Ala Ala Proψ[CH2CH2]Ala Gly Aib ala Val Pro Val-Am,
Ac-Asp Ala pro Ala Ala Proψ[CH2CH2lAla gly pro Ala (Me)Val Pro Val-Am, asp Ala Proψ[CH2CH2]Ala Ala Proψ[CH2CH2]Ala gly Proψ[CH2CH2]Ala val Pro val,
Ac-Asp Ala pro Ala ala Aib Ala gly pro Ala (Me) Val Pro Val-Am,
Asp Ala pro Ala Ala Proψ[CH2 CH2] Ala Gly pro Ala Val Pro Val,
Asp Ala Aib Ala Ala Proψ[CH2 CH2] Ala Gly Aib Ala (Me) Val Pro Val; and ,
Asp Ala Pro Ala Ala Pro Ala Gly pro Ala Val Pro Val
14. A peptide mimetic with the following structure:
PMιAB5
15. A peptide mimetic with the following structure-
PMιPrP13
16. A peptide mimetic with the following structure:
PMiPrP5
17. A method for reducing the formation of amyloid or amyloid like deposits involving
abnormal folding into β sheet structure of amyloid β peptide or for reducing the amount of
said amyloid β peptide which has already formed into a beta sheet structure comprising
bringing into the presence of said amyloid β peptide either prior to or after the abnormal
folding thereof into a β sheet structure, an effective amount of the peptide of claim 1.
18. A method for reducing the formation of amyloid or amyloid like deposits involving
conformational changes in prion Pr protein or reducing the amount of said prion Pr protein
which has already formed into amyloid or amyloid-like deposits comprising bringing into the
presence of said prion Pr protein either prior to or after said conformational changes thereof
into amyloid deposits an effective amount of the peptide of claim 8.
19. A method for reducing the formation of amyloid or amyloid like deposits by administation of a peptide mimetic selected from one of the group consisting of :
PMiAB5,
PMiPrP13 and
PMiPrP5
EP00976928A 1999-11-05 2000-11-04 Peptide analogs and mimetics suitable for in vivo use in the treatment of diseases associated with abnormal protein folding into amyloid, amyloid-like deposits or beta-sheet rich pathological precursor thereof Withdrawn EP1286590A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16391199P 1999-11-05 1999-11-05
US163911P 1999-11-05
PCT/US2000/030416 WO2001034631A2 (en) 1999-11-05 2000-11-04 PEPTIDE ANALOGS AND MIMETICS SUITABLE FOR IN VIVO USE IN THE TREATMENT OF DISEASES ASSOCIATED WITH ABNORMAL PROTEIN FOLDING INTO AMYLOID, AMYLOID-LIKE DEPOSITS OR β-SHEET RICH PATHOLOGICAL PRECURSOR THEREOF

Publications (2)

Publication Number Publication Date
EP1286590A2 true EP1286590A2 (en) 2003-03-05
EP1286590A4 EP1286590A4 (en) 2005-05-11

Family

ID=22592138

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00976928A Withdrawn EP1286590A4 (en) 1999-11-05 2000-11-04 Peptide analogs and mimetics suitable for in vivo use in the treatment of diseases associated with abnormal protein folding into amyloid, amyloid-like deposits or beta-sheet rich pathological precursor thereof

Country Status (11)

Country Link
EP (1) EP1286590A4 (en)
JP (1) JP2004513873A (en)
KR (2) KR20020079731A (en)
CN (1) CN1437442A (en)
AU (1) AU781044B2 (en)
BR (1) BR0015513A (en)
CA (1) CA2389041A1 (en)
EA (1) EA004739B1 (en)
IL (1) IL149392A0 (en)
MX (1) MXPA02004468A (en)
WO (1) WO2001034631A2 (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8003612B2 (en) 1997-10-08 2011-08-23 Proteotech Inc. Small peptides for the treatment of Alzheimer's disease and other beta-amyloid protein disorders
DE10101430B4 (en) 2001-01-13 2008-10-02 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Soluble cyclic analogues for the modulation of amyloidogenesis
US20040052928A1 (en) 2002-09-06 2004-03-18 Ehud Gazit Peptides and methods using same for diagnosing and treating amyloid-associated diseases
AU2004203461B2 (en) * 2002-01-31 2009-09-03 Tel Aviv University Future Technology Development L.P. Peptides Antibodies Directed Thereagainst and Methods Using Same for Diagnosing and Treating Amyloid-Associated Diseases
DE10230141B4 (en) * 2002-07-04 2004-07-15 Priontype Gmbh Method and kit for the enrichment and detection of modified prion proteins (PrPSc)
EP1532170A2 (en) * 2002-07-08 2005-05-25 Applied Research Systems ARS Holding N.V. Beta-sheet breaking peptides
JP2006520321A (en) * 2002-12-02 2006-09-07 アプライド リサーチ システムズ エーアールエス ホールディング ナームロゼ フェンノートシャップ Aza-peptide
EP2719394B1 (en) * 2003-06-30 2015-07-29 Tel Aviv University Future Technology Development L.P. Peptides for treating amyloid-associated diseases
AU2005265182B2 (en) * 2004-06-17 2012-06-21 Mannkind Corporation Epitope analogs
EP1858912B1 (en) * 2005-02-28 2012-04-11 Alphabeta AB Compounds for reducing aggregation of amyloid beta-peptide
US7772177B2 (en) 2005-05-18 2010-08-10 Aegera Therapeutics, Inc. BIR domain binding compounds
EP1746422A1 (en) * 2005-07-19 2007-01-24 Universita'degli Studi Di Milano Method for the identification of proteins folding inhibitors
WO2008015384A1 (en) * 2006-08-04 2008-02-07 Lonza Biologics Plc Method for predicting protein aggregation and designing aggregation inhibitors
TW201225972A (en) 2010-11-15 2012-07-01 Univ Ramot Dipeptide analogs for treating conditions associated with amyloid fibril formation
SG187271A1 (en) * 2011-07-07 2013-02-28 Agency Science Tech & Res Anti-amyloidogenic, alpha-helix breaking ultra-small peptide therapeutic
EP2776055B1 (en) * 2011-08-17 2016-12-14 Ironwood Pharmaceuticals, Inc. Treatments for gastrointestinal disorders
DE102013016002A1 (en) * 2013-09-26 2015-03-26 Forschungszentrum Jülich GmbH Cyclic amyloid beta-binding peptides and their use
DK3049099T3 (en) * 2013-09-26 2020-09-21 Priavoid Gmbh AMYLOID BETA-BINDING PEPTIDES AND ITS USE FOR THERAPY AND DIAGNOSIS OF ALZHEIMER'S DEMENTIA
MY179462A (en) * 2013-10-11 2020-11-06 Malaysian Palm Oil Board Protective effects of oil palm composition on alzheimer?s disease
KR101627106B1 (en) 2014-03-26 2016-06-03 연세대학교 산학협력단 polypeptide having multiple directionality and self-assembled nanostructure containing the same
DE102015003676A1 (en) * 2015-03-20 2016-09-22 Forschungszentrum Jülich GmbH Fachbereich Patente Specifically A-beta species-binding peptides for the therapy and / or diagnosis of Alzheimer's dementia
JP2021524864A (en) 2018-05-15 2021-09-16 ロイド ハン ロイ トランLloyd Hung Loi Tran Therapeutic compositions for the treatment of mild cognitive impairment, depression, and psychiatric disorders and their use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5854204A (en) * 1995-03-14 1998-12-29 Praecis Pharmaceuticals, Inc. Aβ peptides that modulate β-amyloid aggregation

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5948763A (en) * 1995-06-07 1999-09-07 New York University Peptides and pharmaceutical compositions thereof for treatment of disorders or diseases associated with abnormal protein folding into amyloid or amyloid-like deposits
EP1586584A1 (en) * 1996-08-27 2005-10-19 Praecis Pharmaceuticals Incorporated Modulators of beta-amyloid peptide aggregation comprising D-amino acids

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5854204A (en) * 1995-03-14 1998-12-29 Praecis Pharmaceuticals, Inc. Aβ peptides that modulate β-amyloid aggregation

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
ADESSI ET AL.: JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 278, 2003, pages 13905-13911, *
ALI ET AL.: ACTA CHIMICA SCANDICANICA, vol. 27, 1973, pages 1509-1518, *
BASU ET AL: BIOPOLYMERS, vol. 31, 1991, pages 1763-1774, XP002056915 *
FAUCHERE J-L ET AL: "Evaluation of the stability of peptides and pseudopeptides as a tool in peptide drug design" ADVANCES IN DRUG RESEARCH 1992 UNITED KINGDOM, vol. 23, 1992, pages 127-159, XP009036334 ISSN: 0065-2490 *
GANTE J: "PEPTIDOMIMETICS-TAILORED ENZYME INHIBITORS" ANGEWANDTE CHEMIE. INTERNATIONAL EDITION, VERLAG CHEMIE. WEINHEIM, DE, vol. 33, no. 17, 16 September 1994 (1994-09-16), pages 1699-1720, XP000465275 ISSN: 0570-0833 *
PODUSLO J F ET AL: "BETA-SHEET BREAKER PEPTIDE INHIBITOR OF ALZHEIMER'S AMYLOIDOGENESIS WITH INCREASED BLOOD-BRAIN BARRIER PERMEABILITY AND RESISTANCE TO PROTEOLYTIC DEGRADATION IN PLASMA" JOURNAL OF NEUROBIOLOGY, JOHN WILEY AND SONS., NEW YORK, NY, US, vol. 39, no. 3, 5 June 1999 (1999-06-05), pages 371-382, XP009008758 ISSN: 0022-3034 *
ROSSI ET AL.: BIOPOLYMERS, vol. 40, 1996, pages 465-478, XP009074244 *
See also references of WO0134631A2 *
SOTO C ET AL: "BETA-SHEET BREAKER PEPTIDES INHIBIT FIBRILLOGENESIS IN A RAT BRAIN MODEL OF AMYLOIDOSIS: IMPLICATIONS FOR ALZHEIMER'S THERAPY" NATURE MEDICINE, NATURE PUBLISHING, CO, US, vol. 4, no. 7, 1998, pages 822-826, XP000914747 ISSN: 1078-8956 *
SOTO C: "Alzheimer's and prion disease as disorders of protein conformation: implications for the design of novel therapeutic approaches" JOURNAL OF MOLECULAR MEDICINE, SPRINGER VERLAG, DE, vol. 77, no. 5, May 1999 (1999-05), pages 412-418, XP002203443 ISSN: 0946-2716 *
TONIOLO ET AL.: PEPTIDE RESEARCH, vol. 5, 1992, pages 56-61, *

Also Published As

Publication number Publication date
JP2004513873A (en) 2004-05-13
WO2001034631A2 (en) 2001-05-17
EA004739B1 (en) 2004-08-26
MXPA02004468A (en) 2004-09-10
KR20020079731A (en) 2002-10-19
AU1463501A (en) 2001-06-06
KR20060133114A (en) 2006-12-22
BR0015513A (en) 2004-07-06
IL149392A0 (en) 2002-11-10
EP1286590A4 (en) 2005-05-11
WO2001034631A3 (en) 2002-12-27
CA2389041A1 (en) 2001-05-17
CN1437442A (en) 2003-08-20
AU781044B2 (en) 2005-05-05
EA200200536A1 (en) 2002-12-26

Similar Documents

Publication Publication Date Title
US6689753B1 (en) β sheet breaker peptide analogs that inhibit β pleated sheet formation in amyloid β-peptide
AU781044B2 (en) Peptide analogs and mimetics suitable for in vivo use in the treatment of diseases associated with abnormal protein folding into amyloid, amyloid-like deposits or beta-sheet rich pathological precursor thereof
EP0843516B1 (en) Peptides and pharmaceutical compositions thereof for treatment of disorders or diseases associated with abnormal protein folding into amyloid or amyloid-like deposits
Adessi et al. Beta‐sheet breaker strategy for the treatment of Alzheimer's disease
Sun et al. A survey of peptides with effective therapeutic potential in Alzheimer’s disease rodent models or in human clinical studies
Estrada et al. Inhibition of protein misfolding and aggregation by small rationally-designed peptides
US7060671B1 (en) Peptides containing N-substituted D-amino acids for preventing β-strand association
US7125838B1 (en) Peptides containing N-substituted L-amino acids for preventing β-strand association
Granic et al. LPYFDa neutralizes amyloid-β-induced memory impairment and toxicity
US20050181998A1 (en) Prion inhibiting peptides and derivatives thereof
US20070293422A1 (en) Beta-Amyloid Inhibitors and Use Thereof
JP6168998B2 (en) Amyloid β protein-specific production inhibitory polypeptide
US20070155955A1 (en) Amylin aggregation inhibitors and use thereof
Rostagno et al. Protein misfolding, aggregation, and fibril formation: common features of cerebral and non-cerebral amyloidosis
CA2489754A1 (en) .beta.-sheet breaking peptides

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020531

AK Designated contracting states

Kind code of ref document: A2

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

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

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

A4 Supplementary search report drawn up and despatched

Effective date: 20050330

17Q First examination report despatched

Effective date: 20050705

17Q First examination report despatched

Effective date: 20050705

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20070522