WO2005123759A2 - Fgfr binding peptides - Google Patents

Fgfr binding peptides Download PDF

Info

Publication number
WO2005123759A2
WO2005123759A2 PCT/DK2005/000400 DK2005000400W WO2005123759A2 WO 2005123759 A2 WO2005123759 A2 WO 2005123759A2 DK 2005000400 W DK2005000400 W DK 2005000400W WO 2005123759 A2 WO2005123759 A2 WO 2005123759A2
Authority
WO
WIPO (PCT)
Prior art keywords
peptide sequence
fgfr
amino acid
peptide
seq
Prior art date
Application number
PCT/DK2005/000400
Other languages
English (en)
French (fr)
Other versions
WO2005123759A3 (en
WO2005123759A8 (en
Inventor
Elisabeth Bock
Vladimir Berezin
Original Assignee
Enkam Pharmaceuticals A/S
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 Enkam Pharmaceuticals A/S filed Critical Enkam Pharmaceuticals A/S
Priority to BRPI0512254-6A priority Critical patent/BRPI0512254A/pt
Priority to MXPA06014804A priority patent/MXPA06014804A/es
Priority to EP05748535A priority patent/EP1765861B1/en
Priority to CA002570267A priority patent/CA2570267A1/en
Priority to AT05748535T priority patent/ATE545655T1/de
Priority to US11/629,896 priority patent/US20090074774A1/en
Priority to AU2005254613A priority patent/AU2005254613A1/en
Priority to JP2007515781A priority patent/JP2008504236A/ja
Publication of WO2005123759A2 publication Critical patent/WO2005123759A2/en
Publication of WO2005123759A8 publication Critical patent/WO2005123759A8/en
Publication of WO2005123759A3 publication Critical patent/WO2005123759A3/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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to new peptide compounds derived from the sequence of the fibronectin type-Ill module 1 (F3, 1) of the neural cell adhesion molecule (NCAM) which are capable of binding to fibroblast growth factor receptor (FGFR).
  • F3, 1 the fibronectin type-Ill module 1
  • NCAM neural cell adhesion molecule
  • the invention also relates to pharmaceutical compositions comprising the compounds and uses thereof in treatment and/or prevention of different pathological conditions wherein FGFR and/or NCAM play a role in pathology and/or recovery from disease.
  • CAMs cell adhesion molecules
  • Candidate molecules include the amyloid precursor protein, which shares many properties of a classical CAM and beta- amyloid, which can masquerade as a pseudo CAM.
  • Beta-Amyloid serves as a nidus for the formation of senile plaques in Alzheimer's disease and like CAMs provides an environment for organising neurotrophic factors and other CAMs. Inflammatory responses evolve in this environment and can initiate a vicious cycle of perpetuated neuronal damage that is medicated by microglia, complement and other factors (Cotman et al. (1998) Prog Neurobiol. 55:659-69).
  • CAMs Neural cell adhesion molecules of the immunoglobulin superfamily nucleate and maintain groups of cells at key sites during early development and in the adult.
  • CAMs homophylic and heterophylic interac- tions can affect intracellular signalling. Their ability to influence developmental events, including cell migration, proliferation, and differentiation may therefore result both from their adhesive as well as their signalling properties.
  • NCAM neural cell adhesion molecule
  • Ig immu- noglobulin
  • F3 fibronectin type 111
  • NCAM assists both the cell-cell and cell-substratum interactions.
  • NCAM binds to various extracellular matrix components such as hepa- rin/heparan sulfate, chondroitin sulfate proteoglycans, and different types of collagen.
  • NCAM fibroblast growth factor receptor
  • Fibroblast growth factor receptors are the family of four closely related receptor protein tyrosine kinases consisting extracellularly of three Ig-like modules and intracellularly of a split tyrosine-kinase module (Powers et al. (2000) Endocr Relat Cancer 7:165-97).
  • the receptors are known as key regulators of morphogenesis, development, angiogenesis, and wound healing.
  • FGFR activation and signalling are dependent on dimerization of the receptor which is induced by a high affinity binding of its ligand, fibroblast growth factor (FGF), and it also requires participation of cell surface heparin or heparan sulphate proteoglycans.
  • FGF fibroblast growth factor
  • Fibroblast growth receptors play a prominent role in the functioning of the peripheral and central neural system (Jungnickel et al, (2004) Mol Cell Neurosci. 25:21-9; Reuss and von Bohlen und Halbach (2003) Cell Tissue Res. 313:139-57).
  • NCAM has been regarded as a member of a new class of putative alternative ligands of FGFR, and recently there has been obtained evidence for a direct interaction between NCAM and the receptor (Kiselyov et al. (2003) Structure (Camb) 11 :691-701 ; WO 04/056865).
  • the identified NCAM fragment which is directly in- volved in the interaction with FGFR has the sequence EVYVVAENQQGKSKA and derived from the second finronectin type-Ill (F3, 2) module of NCAM.
  • WO 01/96364 has disclosed the 13 amino acid residues sequence SIDRVEPYSSTAQ, wherein the motif DRVE is claimed to be essential for stimulating neurite outgrowth and the motif PYSSTA to be responsible for the capability of the sequence to stimulate cell survival.
  • sequence and peptide fragments thereof comprising the latter motifs were suggested for treatment of diseases associated with neural cell loss due to trauma or disease and as medicaments for stimulating neural cell differentiation and survival.
  • the authors of the present invention demonstrates herein that the 13 amino acid residues sequence of WO 01/96364 extended to 17 amino acid residues, wherein 4 additional amino acid residues are added to the C-terminal of the sequence, is not any more capable of stimulating neurite outgrowth or neuronal cell survival, however, surprisingly, the 17 amino acid residues sequence is capable of stimulating learning and memory. Moreover, the 17 amino acid sequence is also capable of direct binding and activating FGFR.
  • the authors of the present invention correlate these new features of the latter sequence with the presence of the additional C-terminal 4 amino acid residues which comprise an essential amino acid residue of the amino acid motif K/R-x p -D/E/N/Q-x p -Xo-S-XrX 2 -X 3 -D/E/N/Q-x p , wherein x p is any hydrophobic amino acid residue, x 0 is K, R or Y, and Xi, x 2) X 3 are indepently any amino acid residue.
  • this motif is essential for a capability of a peptide sequence to stimulate neural plasticity associated with learning and memory.
  • the invention relates to use of a contigous peptide sequence of 11 to 18 amino acid residues compising the amino acid motif of formula (I) K R-x p -D/E/N/Q-x p -x 0 -S-x 1 -x 2 -x 3 -D/E/N/Q-x p wherein x p is any hydrophobic amino acid residue, Xo is K, R or Y, and x x 2 , x 3 are independently any amino acid residue.
  • a sequence comprising the motif may be used for the manufacturing of a medicament for stimulating learning and memory.
  • the motif of formula (I) is also important for a direct binding of a peptide sequence to FGFR and activating the receptor, however, surprisingly, the authors of the present invention found out that the choice of a hydrophobic residue of the motif defines the array of biological activities that a peptide sequence comprising the motif has.
  • proline (P) residue at any x p position of the motif restricts ca- pabilities of the sequence to the capability of stimulating learning and memory only, whereas any other hydrophobic residue at position x p confers to the sequence both the capability of stimulating learning and memory, capability of stimulating neurite outgrowth and capability of stimulating neuronal cell survival.
  • the present invention relates to a compound comprising a contigous peptide sequence of 11 to 18 amino acid residues comprising the amino acid motif of the formula (II) K R-x p -D/E/N/Q-x p -Xo-S-XrX 2 -X 3 -D/E/N/Q-x p ' wherein x p is any hydrophobic amino acid residue with the except for P, x 0 is K, R or Y, and x 1t x 2 , X 3 are independently any amino acid residue
  • a peptide sequence comprising the latter motif is capable of stimulating neurite outgrowth, stimulating neural cell survival and stimulating neu- ral plasticity associated with learning and memory and it is capable of binding and activating FGFR.
  • Such sequence according to invention may advantageously be used for the manufacturing a medicament for treatment of a number of diseases wherein NCAM and/or FGFR play the major roles.
  • the invention relates also to pharmaceutical compositions and medicaments comprising peptide sequences of the invention. Further, the invention features a method of treatment comprising a step of administering a peptide sequence and/or pharmaceutical composition and/or medicament according to invention. Additionally, the invention relates to an antibody capable of recognising and binding to an epitope comprising an amino acid sequence of the formulas disclosed herein and use of said antibody for the modulating biological activity mediated by NCAM and/or FGFR.
  • Figure 1 A shows the effect of the CDL (SEQ ID NO: 1), ABL (SEQ ID NO: 2) and
  • EFL SEQ ID NO: 5 peptides on neurite outgrowth of rat cerebellar granular neurons (CGNs) in culture.
  • the CDL and EFL peptides stimulate neurite outgrowth from CGNs.
  • B. Neurite outgrowth stimulated by EFL is inhibited by SU5402, an inhibitor of fibroblast growth factor receptor 1 (FGFR1). Results from four independent ex- periments are in all cases expressed as a percentage ⁇ SEM, with non-treated control cells set at 100%. *P ⁇ 0.05, * * P ⁇ 0.01 , ***P ⁇ 0.001 , compared with control. Paired student's t-test. For experiments, CGNs from 7-old-day rats were treated with the peptides for 24 hr. The cultures were fixed in 4% paraformaldehyde, followed by blocking with 1 % BSA and then immunostained with rabbit anti-rat GAP-43 primary antibodies, and then with secondary Alexa Fluor®488 goat anti-rabbit antibodies..
  • Figure 2 A shows the effect of the dendrimeric forms of CDL (SEQ ID NO: 1) (CDLd) and ABL (SEQ ID NO: 2) (ABLd) peptides on survival of rat cerebellar granular neurons in culture.
  • the effect of the peptides is compared to the effect of glia-derived neurotrophic factor (GDNF) and dendrimeric form of the C3 peptide (C3d).
  • B demonstrates the effect of the EFL (SEQ ID NO: 5) dendrimer (EFLd) on on survival of rat cerebellar granular neurons in culture.
  • EFL SEQ ID NO: 5
  • EFLd EFL
  • Figure 3 shows the effect of the monomeric versions of CDL (SEQ ID NO: 1) (A) (CDIoop) and EFL (SEQ ID NO: 5) (B) (EFIoop) on caspase3-like activity in cultures of rat cerebellar granular neurons in low concentration of KCI (5mM) and high concentration of KCI (40 mM) in the medium.
  • the effect of the peptide is compared to the effect of high concentration of KCI. .
  • *P ⁇ 0.05, **P ⁇ 0.01 paired student's t-test
  • Figure 4 demonstrates the binding of four different fragments of NCAM Fn-lll,1 module having the "strand-loop-strand” structure to recombinant FGFR1 studied by means of surface plasmon resonance (SPR) analysis.
  • the binding is given as a response difference (Resp. Diff.) between the binding to the sensor chip with the immobilized FGFR1 modules and a blank sensor chip (unspecific binding).
  • AB - A strand-loop-B strand (corresponds to the ABL peptide of SEQ ID NO: 2); CD - C strand-loop-D strand (corresponds to the CDL peptide of SEQ ID NO: 1); EF - E strand-loop-F strand (corresponds to the EFL peptide of SEQ ID NO: 5); BC - B strand-loop-C strand of F3, 1 module of NCAM; DE - D strand-loop-E strand of F3, 1 module of NCAM; FG - F strand-loop-G strand of F3, 1 module of NCAM.
  • Figure 5 demonstrates the results of quantificative analysis of phosphorylation of FGFR-1 by the ABL, CDL and EFL peptides. Results from at least four independent experiments are expressed as a percentage of FGFR1 phosphorylation ⁇ SEM.
  • TREX-293 cells transfected with FGFR containing a C-terminal Strepll tag were stimulated with different concentrations of the peptides for 30 min. After stimulation, activated FGFR was immunopurified by anti-phosphtyrosine antibodies and then analyzed by western blotting by antibodies against the Strepll tag. The results from treated cells are compared with the resultas from non-treated cells (control, set to 100%). * P ⁇ 0.05, * *P ⁇ 0.01 , * * *P ⁇ 0.001 , paired student's t-test.
  • Figure 6 shows the effect of administration of the CDL (Cdl), and EFLpeptide in vivo on social bihavior rats (the social recognition test): A Time spent with social investigation at 1st meeting (T and second meeting (T 2 ) 1h after peptide/vehicle administration. Results from 8-9 animals expressed as a percentage ⁇ SEM. Paired t-test (*P ⁇ 0.05 and ***P ⁇ 0.001); B Time spent with social investigation at first meeting " H) and second meeting (T 2 ) 24h after peptide/vehicle administration. Results from 9 animals expressed as a percentage ⁇ SEM. Paired t-test (*P ⁇ 0.05). Detailed description of the invention
  • FGFR fibroblast growth factor receptor
  • NCAM neural cell adhesion molecule
  • the C-terminal amino acid of a peptide of the invention exists as the free carboxylic acid, this may also be specified as "-OH".
  • the C-terminal amino acid of a compound of the invention may be the amidated derivative, which is indicated as " ⁇ NH 2 ".
  • the N-terminal amino acid of a polypeptide comprise a free amino-group, this may also be specified as "H-”.
  • amino acid can be selected from any amino acid, whether naturally occurring or not, such as alfa amino acids, beta amino acids, and/or gamma amino acids. Accordingly, the group comprises but are not limited to: Ala, Val, Leu, lie, Pro, Phe, Trp, Met, Gly, Ser, Thr, Cys, Tyr, Asn, Gin, Asp, Glu, Lys, Arg, His Aib, Nal, Sar, Orn, Lysine analogues, DAP, DAPA and 4Hyp. Also, according to the invention modifications of the compounds/peptides may be performed, such as glycosylation and/or acetylation of the amino acids.
  • Basic amino acid residues are according to invention represented by the residues of amino acids Arg, Lys, and His, acidic amino acid residues - by the residues of amino acids Glu and Asp, hydrophobic amino acid residues - by the residues of amino acids Leu, lie, Val, Phe, Trp, Pro and Ala, and polar uncharged amino acid residues - by the residues of amino acids Tyr, The, Ser, Gin and Asn.
  • the present invention provides a peptide sequence of 11 to 18 amino acid residues compising the amino acid motif of formula (I) K/R-x p -D/E/N/Q-x -Xo-S-x 1 -x 2 -x 3 -D/E/N/Q-x p wherein x p is any hydrophobic amino acid residue, Xo is K, R or Y, and x-i, x 2 , x 3 are indepently any amino acid residue.
  • any hydrophobic residue is meant a residue is selected from the residues of amino acids A, I, L, P, V, F or W.
  • the residue x p may independently be selected from any hydrophobic residues.
  • the residue residue x p may independently be selected from A, P, V or W.
  • the term "independently" in the present context means that there are no any specific demands for the choice of an amino acid residue at any specified position.
  • the x p amino acid residue may be selected from A, V or W and the amino acid motif preferably is K R-V/A-D/E/N/Q-W-Xo-S- ⁇ -x 2 -X 3 -D/E/N/Q-V/A wherein Xo is K, R or Y, and X ⁇ , x 2 , x 3 are independently any amino acid residue.
  • the x p amino acid residue may be selected from A, V or P the amino acid motif preferably is K R-V/A-D/E/N/Q-P-x 0 -S-X ⁇ -x 2 -x 3 -D/E/N/Q-V/A wherein Xo is K, R or Y, and Xi , x 2 , x 3 are independently any amino acid residue.
  • the residues x ⁇ x 2 , x 3 of any of the above motifs are not of major importance for biological activity of peptide sequences comprising the mo- tifs of above and therefore said residues may independently be selected from any amino acid residues.
  • the residues A, G, L, I, S, T, and E may in some embodiments be preferred.
  • the sequence X ⁇ -x 2 -x 3 may be any sequence consisting of the latter residues contigously connected to each other, such as for example any of the sequences l-G-L, L-G-l, A-G-L, A-T-L, L-G-E, l-G-E, A-G-l, S-T-A or S-G-A.
  • the sequences S-T-A and L-G-E may be preferred.
  • a peptide sequence of the invention is a contiguous sequence of 11 to 18 amino acid residues comprising a motif as any of the motifs described above.
  • the invention relates to a sequence which is an isolated peptide sequence. This in the present context means that the peptide sequence exists as an individual chemical entity and is used for the purposes of the invention as an individual chemical entity.
  • the invention does not relate to peptide sequences comprising the above motif(s), which are integrated parts of natural or recombinant polypeptides, such as for example the intergrated parts of polypeptides or fragments of the neural cell adhesion molecule (NCAM).
  • NCAM neural cell adhesion molecule
  • a peptide sequence may have the length of 11 to 14 amino acid residues; in another embodiment the sequence may have the length of 15 to 18 amino acid residues.
  • the length of a sequence such as 11 , 12, 13, 14, 15, 16, 17 or 18 amino acid residues may be preferred depending on the embodiments.
  • the invention associates the structural features as described above with a capability of a peptide sequence to bind and activate FGFR, to stimulate learning and memory, neurite outgrowth and /or neuronal cell survival.
  • the invention discloses herein particular peptide sequences which are capable of stimulating memory and learning, namely the sequences KAEWKSLGEEAWHSK (SEQ ID NO: 1), SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) and SIDRVNPYSSTAQVQFD (SEQ ID NO: 3).
  • sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1) may be preferred, whereas in another embodiments the sequence SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) or sequence SIDRVNPYSSTAQVQFD (SEQ ID NO: 3) may be preferred.
  • Sequences KAEWKSLGEEAWHSK (SEQ ID NO: 1), SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) and SIDRVNPYSSTAQVQFD (SEQ ID NO: 3), all, are capable of direct binding to FGFR and stimulating the receptor, and they all are capable of stimulating learning and memory,
  • the sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1) is capable some additional biological activities in comparison to the sequences SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) and SIDRVNPYSSTAQVQFD (SEQ ID NO: 3), namely it is capable of stimulating nerite outgrowth and neural cell survival, which the sequences SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) and sequence SIDRVNPYSSTAQVQFD (SEQ ID NO: 3) are not capable.
  • a structural feature which confers to a sequence the capability of both binding and activating FGFR, stimulating learning and memory, stimulating neurite outgrowth and neural cell survival comprises the amino acid motif of the formula (II): K/R-x p -D/E/N/Q-x p -Xo-S-XrX 2 -x 3 -D/E/N/Q-x p , wherein P (proline residue) at position x p is excluded from the selection of hydrophobic amino acid residues.
  • the motif of formula (II) defined as K/R-x p -D/E/N/Q-x p -x 0 -S-x 1 -x 2 -x 3 -D/E/N/Q-x p I wherein x p is any hydrophobic amino acid residue with the except for P, Xo is K, R or Y, and Xi, x 2 , x 3 are indepently any amino acid residue, is a structural motif which confers to a peptide sequence comprising said motif the capability of binding and activating FGFR, stimulating learning and memory, stimulating neurite outgrowth and neural cell survival. In some embodiments, such sequence may have the residue Y at position x 0 .
  • residues K or R are preferred at this position.
  • Different embodiments for the residues Xi, x 2 , x 3 are discussed above.
  • Preferred embodiments for the residues of the positions X ⁇ , x 2 and x 3 are residues G, V, L, or E, wherein L or V are more preferred at position x ⁇ G at position x 2 and E at position x 3 .
  • residue P at position x p may restrict capabilities of a peptide sequence compisng the motif as above to the capabilities of binding and activating FGFR and stimulating learning and memory, such as for example the capabilities of the sequences SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) and SIDRVNPYSSTAQVQFD (SEQ ID NO: 3).
  • the present invention relates to fragments, variants and homologues of the peptide sequences comprising the motif of formula (II) K/R-x p -D/E/N/Q-x p -x 0 -S-x 1 -x 2 -x 3 -D/E/N/Q-x p , wherein x p is any hydrophobic amino acid residue with the except for P, Xo is K, R or Y, and Xi, x 2 , x 3 are indepently any amino acid residue, which is in detail described above.
  • the invention defines i) a fragment as a sequence which has at least 40%, more preferably at least 50%, more preferably at least 60%, more preferably at least 70%, more preferably at least 80%, more preferably at least 90%, more pref- erably at least 95% of the length of a sequence comprising the motif of above, in particular the sequence of SEQ ID NO: 1 ; ii) a variant as an amino acid sequence having at least 60 %, more preferably at least 70%, more preferably at least 80%, more preferably at least 90%, more preferably 95% homology to a sequence comprising the motif of above, in particular to the sequence of SEQ ID NO: 1 , or as an amino acid sequence having at least 60 %, more preferably at least 70%, more preferably at least 80%, more preferably at least 90%, more preferably 95% positive amino acid matches compared to a sequence comprising the motif of above, in particular to the sequence of SEQ ID NO: 1.
  • a posi- tive amino acid match is defined herein as the identity or similarity of physical and/or chemical properties of the amino acid residues having the same position in two compared sequences.
  • Preferred positive amino acid matches of the present invention are K to R, E to D, L to M, Q to E, I to V, I to L, A to S, Y to W, K to Q, S to T, N to S and Q to R.
  • Homology of one amino acid sequence with another amino acid is defined as the percentage of identical amino acids in the two collated sequences.
  • the homology of the sequences may be calculated using well known algorithms such as BLOSUM 30, BLOSUM 40, BLOSUM 45, BLOSUM 50, BLOSUM 55, BLOSUM 60, BLOSUM 62, BLOSUM 65, BLOSUM 70, BLOSUM 75, BLOSUM 80, BLOSUM 85, or BLOSUM 90; iii) a homologue as an amino acid sequence which has less then 60% but more then 30 %, such as 50-59%, for example 55%, such as 40-49%, for example 45%, such as 30-39%, for example 35% homology a sequence comprising the motif of above, in particular to the sequence of SEQ ID NO: 1.
  • a fragment, variant and homologue as defined above remain at least some biological activity of the original sequence, for example a capability of stimulating neural cell differentiation, memory and learning, neural cell survival, and/or binding and activating FGFR.
  • a preferable homologue of the invention is a homologue that has at least 40% ho- mology to the sequence of SEQ ID NO: 1, such as 40-44% homology, more preferably at least 45% homology, such as 45-49% homology, even more preferably at least 50%, such as 51 -54% homology, even more preferably at least 55% homology, such as 56%-59%.
  • a preferable fragment of the invention is a fragment that consists of at least 3 contigous amino acid residues of the sequence of SEQ ID NO:1 , more preferably 4 amino acid residues, even more preferably 5 amino acid residues, even more preferably 6 amino acid residues, even more preferably 7 amino acid residues, even more preferably 8 to 11 amino acid residues.
  • a preferable variant of the invention is a peptide sequence which has at least 70% homology to the sequence of SEQ ID NO: 1 , such as 71-74% homology, more preferably at least 75% homology, such as 76-79% homology, more preferably at least 80% ho- mology, such as 81 -84% homology, even more preferably at least 85% homology, such as 86-89% homology, even more preferably 90% homology, such as for example 91 -94% homology, even more preferably at least 95% homology, such as for example 96-99% homology.
  • an isolated peptide sequence as described above may be formulated as a compound,
  • a compound may contain a single copy of an individual amino acid sequence selected from any of the described above, or it may contain two or more copies of such amino acid sequences.
  • compound of the invention may be formulated as a monomer of a peptide sequence (monomeric compound), such as containing a single individual peptide sequence, or it may be formulated as a multimer of a peptide sequence (multimeric compound), i.e containing two or more individual peptide sequences, wherein said individual peptide sequences may be represented by two or more copies of the same sequence or by two or more different individual peptide sequences.
  • a multimer may also comprise a combination of the full-length sequence and one or more fragments thereof.
  • a compound may contain two amino acid sequences, such compound is defined herein as dimer, in another embodiment a compound may contain more then two amino acid sequences, such for example three, four or more sequences.
  • the present invention preferably relates to compounds containing two or four peptide sequences. However, compounds containing 3, 5, 6, 7, 8 or more sequences are also in the scope of the invention.
  • a multimeric compound may comprise the identical peptide sequences, or the sequences may be different.
  • One example of a compound wherein the sequences are different may be a compound containing SEQ ID NO: 1 and SEQ ID NO: 2, another example may be a compound containing SEQ ID NO: 1 and SEQ ID NO: 3. Any combination of the sequences of the invention may be made depending on different embodiments of the invention.
  • the sequences may be connected to each other via a bond, for example the peptide bond, or connected to each other through a linker molecule or grouping. Compounds wherein the peptide sequences are connected through a linker molecule or grouping are preferred.
  • compound may contain two or more identical copies of a sequence, such as for example two copies of a sequence selected from SEQ ID
  • linking grouping may be an achiral di-, tri- or tetracarboxylic acid. Suitable achiral di-, tri- or tetracarboxylic acids and a method of production such a compound, a ligand presentation assembly method (LPA), are discussed in detail further in the specification of the invention.
  • Another example of a possible linker may be the amino acid lysine.
  • Individual peptide sequences may be attached to a core molecule such as lysine forming thereby a dendritic multimer (dendrimer) of an individual peptide sequence(s).
  • dendrimers Production of dendrimers is well known in the art (PCT/US90/02039, Lu et al., (1991) Mol Immunol. 28:623-630; Defoort et al., (1992) Int J Pept Prot Res. 40:214- 221 ; Drijfhout et al. (1991) Int J Pept Prot Res. 37:27-32), and dedrimers are at present widely used in research and in medical applications.
  • a dendrimeric compound comprising four individual amino acid sequences attached to the lysine core molecule. It is also preferred that at least one of the four individual amino acid sequences comprises an amino acid sequence of the formula defined above. It is even more preferred if each of four individual amino acid sequences of a dendrimeric compound individually comprises an amino acid motif selected from the motifs discussed above.
  • Multimeric compounds of the invention such as LPA-dimers or Lysin-dendrimers, are preferred compounds of the invention.
  • other types of multimeric compounds comprising two or more individual sequences of the invention may be preferred depending on the embodiments.
  • Multimeric compounds which comprise one or more copies of an individual sequence of the invention, or fragment, variant or homologue thereof, and another biologically active entity, for example another peptide sequence, are also in the scope of the invention.
  • Such compounds most preferable are those which comprise at least one peptide sequence of the invention and a peptide sequence which has a capability of stimulating fibroblast growth factor receptor, for example such as peptide sequence EVYVVAENQQGKSKA (SEQ ID NO: 4) or peptide sequence TIMGLKPETRYAVR (SEQ ID NO: 5).
  • the multimeric compound of the invention is a dimer comprising two identical copies of an individual peptide sequence comprising a motif of the invention.
  • the multimeric compound of the invention is a dimer comprising two identical copies of an individual peptide sequence comprising a motif of the invention. Still, in another preferred embodiment the multimeric compound is a dimer comprising two different peptide sequences comprising a motif of the invention.
  • a preferred multimeric compound may also be a compound comprising a sequence comprising a motif of he invention and a sequence selected from . TIMGLKPETRYAVR (SEQ ID NO: 5) or EVYVVAENQQGKSKA (SEQ ID NO: 4).
  • the multimeric compound which is a dendrimer comprising four identical copies of a peptide sequence comprising a motif of the invention linked to a backbone structure of three lysine residues is also among preferred multimeric compounds of the invention.
  • Orientation of peptide sequences in a multimeric compound may be either N to C, C to N or mixed.
  • N to C orientation is meant that two predetermined sequences are connected to a linker through their N-terminal amino acid residue, such as a compound of the type:(COOH)peptide sequence(NH2)-Iinker- (NH2)peptide sequence(COOH).
  • C to N orientation is meant that two predetermined sequences are connected to a linker through their C-terminal amino acid residue, such as a compound of the type:(NH2)peptide sequence(COOH)- linker-(COOH)peptide sequence(NH2).
  • mixed orientation is meant that two predetermined sequences are connected to a linker either through their C- terminal amino acid residue or N-terminal, such as one peptide sequence though its N-terminal amino acid residue and another through its C-terminal amino acid residue, for example as a compound of the type:(NH2)peptide sequence(COOH)- linker-(NH2)peptide sequence(COOH).
  • peptide sequence as defined above and compound comprising said sequence are capable of binding and activation FGFR.
  • peptide sequences of the invention are capable of binding and activating a functional cell-surface receptor, which is a receptor of the fibroblast growth factor receptor (FGFR) family comprising fibroblast growth factor receptor-1 (FGFR-1), fibroblast growth factor receptor-2 (FGFR-2), fibroblast growth factor receptor-3
  • FGFR fibroblast growth factor receptor
  • FGFR-3 fibroblast growth factor receptor-4
  • FGFR-5 fibroblast growth factor receptor-5
  • Extracellular binding of a ligand to a receptor leads to receptor activation which means that the receptor is becoming capable of activating at least one intracellular signal transduction pathway resulting in one or another cellular response.
  • binding refers herein to a direct interaction of a peptide sequence of the invention with FGFR.
  • interaction is meant that a peptide sequence has a transient or permanent direct contact with FGFR.
  • activation of the receptor means that the receptor become capable of transmitting the effect of "ligand binding” into a cascade of biochemical reactions collectively termed “receptor signalling” or “signal transduction” inside the cell resulting in a physiological response of the cell.
  • Peptide sequences of the invention which are capable of binding and activating a cellular receptor are thus capable of mimicing the action of natural receptor ligands and, in this context, they should be understood as mimics of the receptor ligands, in particular ligands of FGFR. It is known that, a receptor may have a number of different ligands, binding of which to the receptor activates different signal transduction pathways and results in different cellular responces.
  • the invention preferably concerns FGFR-1 associated signal transduction pathways.
  • Activation of FGFR-1 is defined herein by the degree of FGFR-1 tyrosine phosphorylation, which is according to the present invention increases due to the binding of a sequence of the invention.
  • Activation of an FGFR-1 associated intracellular signal transduction pathway is defined herein as the presence of an activated molecule of one or more intracellular proteins involved in a FGFR-1 associated signal transduction pathway, such as for example activated molecules of proteins STAT1 , JNK, PLCy, ERK, STAT5, PI3K, PKC, FRS2 and/or GRB2.
  • the activated status of a molecule of one or more intracellular proteins involved in a FGFR-1 associated signal transduction pathway is defined herein as the degree of phosphory- lation of said molecule, which is according to the present invention increases or des- reases due to the binding of a sequence of the invention to FGFR.
  • the degree of phosphorylation is estimated as at least 20% above/below the control value, such as at least 20-200 %, for example at least 50-200%.
  • the control value is estimated as a degree of phosphorylation of the protein of interest when a compound capable of binding and activating FGFR is absent in cell environment.
  • the invention in particular relates to a cellular response selected from but not limited by i) induction of cell differentiation, such as for example differentiation of neu- ronal cells, stem cells or cancer cells, nerve regeneration, neurite out- growth and branching, inhibition of cell proliferation; ii) increased cell survival, such for example neuronal cell survival due body traumatic damage or due to disease, inhibition of apoptosis; iii) increased cellular plasticity, for example morphological plasticity, such as for example neural cell plasticity associated with the memory and learn- ing.
  • cell differentiation such as for example differentiation of neu- ronal cells, stem cells or cancer cells, nerve regeneration, neurite out- growth and branching, inhibition of cell proliferation
  • increased cell survival such for example neuronal cell survival due body traumatic damage or due to disease, inhibition of apoptosis
  • increased cellular plasticity for example morphological plasticity, such as for example neural cell plasticity associated with the memory and learn- ing.
  • a peptide sequence of the invention may derive from the neural cell adhesion molecule (NCAM), for example from NCAM polypeptide identified in the GenBank database under Swiss-Prot Ass Nos.: P13591 , P13592, P13596 or P13595.
  • NCAM neural cell adhesion molecule
  • An example of such peptide sequence may be sequence KAEWKSLGEEAWHSK (SEQ ID NO:
  • sequence SIDRVEPYSSTAQVQFD SEQ ID NO: 2
  • sequence SIDRVEPYSSTAQVQFD SEQ ID NO: 2
  • These peptide sequences are the integral sequences of NCAM polypeptide and when taken as isolated peptide sequences they are capable according to the invention of mimicking activity of NCAM associated with FGFR function, such as stimulating NCAM-FGFR- dependent neural cell differentiation, survival and plasticity.
  • the invention does not limit capability of a peptide sequence of the invention to mimicking the function of NCAM.
  • NCAM described herein and the peptide sequences of NCAM described in WO 04/056865 represent different parts of NCAM binding site for FGFR. Via this binding site NCAM interacts with a binding site on FGFR which is common for the low- affinity FGFR ligands described in WO 04/056865. Therefore, a peptide sequence of the present invention may be capable of mimicking the binding of a low-affinity ligand from the latter group of FGFR ligands to FGFR and thus mimicking biological activity of said ligand.
  • the invention includes in the scope any biological activity of the following proteins associated with FGFR function: Neural cell adhesion molecule L1 (of Swiss-Prot Ass.
  • FGFR2 Fibroblast Growth Factor Receptor 2 (FGFR2) (of Swiss-Prot Ass. Nos: Q96KM2, P21802 or Q63241 ),
  • FGFR3 Fibroblast Growth Factor Receptor 3 (FGFR3) (of Swiss-Prot Ass. Nos: Q95M13, AF487554 or Q99052),
  • FGFR4 Fibroblast Growth Factor Receptor 4
  • NRRKT-2 Neurotrophin Tyrosin Kinase Type-2
  • LAR-PTPRF Leukocyte Antigen Related Protein-Tyrosine Phosphatase
  • Phosphatase Receptor type S (PTPRS) (of Swiss-Prot Ass. Nos: Q64699, Q13332 or 075870),
  • R-PTP-kappa - Protein-Tyrosine Phosphatase Receptor type kappa
  • PPRD Protein-Tyrosine Phosphatase Receptor type D
  • Ephrin type-A receptor 8 EPHA8/Tyrosine-Protein Kinase Receptor EEK
  • - Ephrin type-A receptor 3 EPHA8/Tyrosine-Protein Kinase Receptor ETK- 1/CEK4
  • IGF-1 Insulin-like Growth Factor-1 Receptor
  • IRR Insulin-related Receptor
  • PDGFRB Platelet-Derived Growth Factor Receptor Beta
  • IL-6R - lnterleukin-6 Receptor
  • IL-23R - lnterleukin-23 Receptor
  • Beta-common cytokine receptor of IL-3, IL5 and GmCsf (of Swiss-Prot Ass. No: P32927) - Cytokine Receptor-Like molecule 3 (CRLF1 ) (of Swiss-Prot Ass. No: Q9JM58),
  • IFGP2 - Leukocyte Fc Receptor-like Protein
  • MSR2 Macrophage Scavenger Receptor 2
  • G-CSF-R Granulocyte Colony Stimulating Factor Receptor
  • TECAM - Metalloproteinase-desintegrin domain containing protein
  • TECAM - Metalloproteinase-desintegrin domain containing protein
  • TECAM - Metalloproteinase-desintegrin domain containing protein
  • - Metalloproteinase 1 of Swiss-Prot Ass. Nos: 095204 or Q9BSI6
  • Collagen type VII Swiss-Prot Ass. No: Q63870
  • Fibronectin of Swiss-Prot Ass. Nos: Q95KV4, Q95KV5, P07589, Q28377, U42594, 095609 or P11276
  • Tenascin-R of Swiss-Prot Ass. Nos: Q15568, 000531 , Q90995 of P10039
  • CCF-1 Cytokine-like factor-1
  • Any interaction between two molecules may be characterised by the affinity of interaction, which means the strength of attraction between two molecules.
  • the affinity of interaction is commonly expressed by the value of Kd, a dissociation equilibrium constant.
  • Kd reflects the ratio between the rate of dissociation and the rate of binding between two molecules and thus represents a measure of the strength of binding between two molecules. The stronger binding is reflected by the lower value of Kd.
  • the above FGFR ligands have a relatively low affinity binding to FGFR.
  • the low affinity interaction of the invention is characterised by Kd having a value in the range of 10 "3 to 10 "10 M, such for example in the range of 10 '4 to 10 "8 M.
  • the invention relates to affinity of interaction between a peptide sequence and FGFR in the range of 10 "3 to 10 "10 M, preferably in the range of 10 -4 to 10 "8 M.
  • the invention may relate to different biological activities of the peptide sequences of the invention.
  • the invention may relate to the capability of a peptide sequence to stimulate neural plasticity associated with learning and memory.
  • such peptide sequence may be selected from any peptide sequences described herein.
  • a preferred sequence may sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1), or sequence SIDRVEPYSSTAQVQFD (SEQ ID NO: 2).
  • the invention may relate to a capability of a peptide sequence to stimulate neuronal differentiation such as differentiation of neural precursor ceils or differentiation of neurons which includes neurite outgrowth and/or branching of the processes.
  • this activity is associated with a peptide sequence which comprises the motif K R-x p -D/E/N/Q-x p -Xo-S-X ⁇ -x 2 -x 3 - D/E/N/Q-x p , wherein x p is any hydrophobic amino acid residue with the except for P, Xo is K, R or Y, and X 1 . X 2 .
  • X 3 are indepently any amino acid residue
  • a pereferred sequence of such embodiment may be the sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1).
  • a peptide sequence comprising the above motif is also capable of stimulating cell survival, preferably neuronal cell survival and therefore some preferred embodiments of the invention may relate to a capability of a peptide sequence to stimulate cell survival, preferably neuronal cell survival.
  • a pereferred sequence of such embodiments may also be the sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1).
  • a sequence comprising the above motif may also be preferred when the invention relates to a sequence which is capable two or more biological activities selected from i) stimulating learning and memory , ii) stimulating neural cell survival, iii) stimulating neural cell differentiation.
  • a preferred sequence of such embodiments is also the sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1).
  • the peptides of the present invention may be prepared by any conventional synthetic methods, recombinant DNA technologies, enzymatic cleavage of full-length proteins which the peptide sequences are derived from, or a combination of said methods.
  • the peptides of the invention are produced by use of re- combinant DNA technologies.
  • the DNA sequence encoding a peptide or the corresponding full-length protein the peptide originates from may be prepared synthetically by established standard methods, e.g. the phosphoamidine method described by Beaucage and Caruthers, 1981, Tetrahedron Lett. 22:1859-1869, or the method described by Matthes et al., 1984, EMBO J. 3:801-805. According to the phosphoamidine method, oligonucleo- tides are synthesised, e.g. in an automatic DNA synthesiser, purified, annealed, ligated and cloned in suitable vectors.
  • the DNA sequence encoding a peptide may also be prepared by fragmentation of the DNA sequences encoding the corresponding full-length protein of peptide origin, using DNAase I according to a standard protocol (Sambrook et al., Molecular cloning: A Laboratory manual. 2 rd ed., CSHL Press, Cold Spring Harbor, NY, 1989).
  • the DNA encoding the full-length protein may alternatively be fragmented using specific restriction endonucleases.
  • the fragments of DNA are further purified using standard procedures described in Sambrook et al., Molecular cloning: A Laboratory manual. 2 rd ed., CSHL Press, Cold Spring Harbor, NY, 1989.
  • the DNA sequence encoding a full-length protein may also be of genomic or cDNA origin, for instance obtained by preparing a genomic or cDNA library and screening for DNA sequences coding for all or part of the full-length protein by hybridisation using synthetic oligonucleotide probes in accordance with standard techniques (cf. Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor, 1989).
  • the DNA sequence may also be prepared by polymerase chain re- action using specific primers, for instance as described in US 4,683,202 or Saiki et al., 1988, Science 239:487-491.
  • a recombinant expression vector which may be any vector, which may conveniently be subjected to recombinant DNA pro- cedures.
  • the choice of vector will often depend on the host cell into which it is to be introduced.
  • the vector may be an autonomously replicating vector, i.e. a vector that exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g. a plasmid.
  • the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated.
  • the DNA sequence encoding a peptide or a full-length protein should be operably connected to a suitable promoter sequence.
  • the promoter may be any promoter sequence.
  • DNA sequence which shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell.
  • suitable promoters for directing the transcription of the coding DNA sequence in mammalian cells are the SV 40 promoter (Subramani et al., 1981, Mol. Cell Biol. 1:854-864), the MT-1 (metallothionein gene) promoter
  • a suitable promoter for use in insect cells is the polyhedrin promoter (Vasu- vedan et al., 1992, FEBS Lett. 311:7-11).
  • Suitable promoters for use in yeast host cells include promoters from yeast glycolytic genes (Hitzeman et al., 1980, J. Biol.
  • Suitable promoters for use in filamentous fungus host cells are, for instance, the ADH3 promoter (McKnight et al., 1985, EMBO J. 4:2093-2099) or the tpiA promoter.
  • the coding DNA sequence may also be operably connected to a suitable terminator, such as the human growth hormone terminator (Palmiter et al., op. cit.) or (for fungal hosts) the TPI1 (Alber and Kawasaki, op. cit.) or ADH3 (McKnight et al., op. cit.) promoters.
  • the vector may further comprise elements such as polyadenylation sig- nals (e.g. from SV 40 or the adenovirus 5 Elb region), transcriptional enhancer sequences (e.g. the SV 40 enhancer) and translational enhancer sequences (e.g. the ones encoding adenovirus VA RNAs).
  • the recombinant expression vector may further comprise a DNA sequence enabling the vector to replicate in the host cell in question.
  • a DNA sequence enabling the vector to replicate in the host cell in question.
  • An example of such a sequence is the SV 40 origin of replication.
  • the vector may also comprise a selectable marker, e.g. a gene the product of which complements a defect in the host cell, such as the gene coding for dihydrofolate reductase (DHFR) or one which confers resistance to a drug, e.g. neomycin, hydromycin or methofrexate.
  • DHFR dihydrofolate reductase
  • the coding DNA sequences may be usefully fused with a second peptide coding sequence and a protease cleavage site coding sequence, giving a DNA construct encoding the fusion protein, wherein the protease cleavage site coding sequence positioned between the HBP fragment and second peptide coding DNA, inserted into a recombinant expression vector, and expressed in recombinant host cells.
  • said second peptide selected from, but not limited by the group comprising glutathion-S-reductase, calf thymosin, bacterial thioredoxin or human ubiquitin natural or synthetic variants, or peptides thereof.
  • a peptide sequence comprising a protease cleavage site may be the Factor Xa, with the amino acid sequence IEGR, en- terokinase, with the amino acid sequence DDDDK, thrombin, with the amino acid sequence LVPR/GS, or Acharombacter lyticus, with the amino acid sequence X X, cleavage site.
  • the host cell into which the expression vector is introduced may be any cell which is capable of expression of the peptides or full-length proteins, and is preferably a eukaryotic cell, such as invertebrate (insect) cells or vertebrate cells, e.g. Xenopus laevis oocytes or mammalian cells, in particular insect and mammalian cells.
  • a eukaryotic cell such as invertebrate (insect) cells or vertebrate cells, e.g. Xenopus laevis oocytes or mammalian cells, in particular insect and mammalian cells.
  • Exam- pies of suitable mammalian cell lines are the HEK293 (ATCC CRL-1573), COS (ATCC CRL-1650), BHK (ATCC CRL-1632, ATCC CCL-10) or CHO (ATCC CCL- 61 ) cell lines.
  • fungal cells may be used as host cells.
  • suitable yeast cells include cells of Saccharomyces spp. or Schizosaccharo- myces spp., in particular strains of Saccharomyces cerevisiae.
  • Other fungal cells are cells of filamentous fungi, e.g. Aspergillus spp. or Neurospora spp., in particular strains of Aspergillus oryzae ot Aspergillus niger.
  • Aspergillus spp. for the expression of proteins is described in, e.g., EP 238 023.
  • the medium used to culture the cells may be any conventional medium suitable for growing mammalian cells, such as a serum-containing or serum-free medium containing appropriate supplements, or a suitable medium for growing insect, yeast or fungal cells. Suitable media are available from commercial suppliers or may be prepared according to published recipes (e.g. in catalogues of the American Type Culture Collection).
  • the peptides or full-length proteins recombinantly produced by the cells may then be recovered from the culture medium by conventional procedures including separating the host cells from the medium by centrifugation or filtration, precipitating the proteinaceous components of the supernatant or filtrate by means of a salt, e.g. ammonium sulphate, purification by a variety of chromatographic procedures, e.g. HPLC, ion exchange chromatography, affinity chromatography, or the like.
  • a salt e.g. ammonium sulphate
  • Peptides may for example be synthesised by using Fmoc chemistry and with Acm- protected cysteins. After purification by reversed phase HPLC, peptides may be further processed to obtain for example cyclic or C- or N-terminal modified isoforms.
  • the methods for cyclization and terminal modification are well-known in the art and described in detail in the above-cited manuals.
  • the peptide sequences of the invention are produced synthetically, in particular, by the Sequence Assisted Peptide Synthesis (SAPS) described in the above manuals. Otherwise, the synthesis of an individual peptide sequence of the invention may be ordered and purchased from a commercial manufacturer, such as for example Sigma-Genosys (USA).
  • SAPS Sequence Assisted Peptide Synthesis
  • the LPA method is disclosed in WO 00/18791. The method essentially comprises the following steps:
  • step (c1 ) if present, deprotecting any N-protected groups originating from the carboxylic acid used in step (c),
  • the method provides i.a. LPAs presenting desired sequences of the invention with N to C orientation (step (c)). And also simultaneously sequences with C to n orien- tation (step (c2))
  • two peptide chains attached to a solid phase are to be assembled by means of achiral di-, tri- or tetracarboxylic acids.
  • Suitable achiral di-, tri- or tetracarboxylic acids to be used in the present method have the general formula X[(A)nCOOH][(B)mCOOH]
  • n and m independently are an integer of from 1 to 20, X is HN, A and B independently are a substituted or unsubstituted CM O alkyl, a substituted or unsubstituted C - ⁇ o alkenyl, a substituted or unsubstituted cyclic moiety, a substituted or unsubstituted heterocyclic moiety, a substituted or unsubstituted aromatic moiety, or A and B together form a substituted or unsubstituted cyclic moiety, substituted or unsubstituted heterocyclic moiety, substituted or unsubstituted aromatic moiety.
  • suitable achiral di-, tri- or tetracarboxylic acids to be used in the present method have the general formula
  • n and m are 0 or an integer of from 1 to 20,
  • X is H 2 N(CR 2 )pCR, or RHN(CR 2 )pCR, wherein p is 0 or integer of from 1 to 20, wherein each R is H, a substituted or unsubstituted CM O alkyl, a substituted or unsubstituted C 2- ⁇ o alkenyl, a substituted or unsubstituted cyclic moiety, a substituted or unsubstituted heterocyclic moiety, a substituted or unsubstituted aromatic moiety, or A and B together form a substituted or unsubstituted cyclic moiety, substituted or unsubstituted heterocyclic moiety, substituted or unsubstituted aromatic moiety.
  • suitable achiral di-, tri- or tetracarboxylic acids to be used in the present method have the general formula
  • n and m are 0 or an integer of from 1 to 20,
  • X is HO(CR 2 )pCR, HS(CR 2 )pCR, halogen-(CR 2 )pCR, HOOC(CR 2 )pCR, ROOC(CR 2 )pCR,
  • each R independently is H or a substituted or unsubstituted CM O alkyl, a substituted or unsubstituted C 2- ⁇ 0 alkenyl, a substituted or unsubstituted cyclic moiety, a substituted or unsubstituted heterocyclic moiety, a substituted or unsubstituted aromatic moiety, or A and B together form a substituted or unsubstituted cyclic moiety, substituted or unsubstituted heterocyclic moiety, substituted or unsubstituted aromatic moiety.
  • suitable achiral di-, tri- or tetracarboxylic acids to be used in the present method have the general formula
  • n and m are 0 or an integer of from 1 to 20,
  • X is H 2 N(CR 2 )p, RHN(CR 2 )p, HO(CR 2 )p, HS(CR 2 )p, halogen-(CR 2 )p, HOOC(CR 2 )p, ROOC(CR 2 )p, HCO(CR 2 )p, RCO(CR 2 )p, or [HOOC(A)n][HOOC(B)m](CR 2 )p, wherein p is 0 or integer of from 1 to 20, each R independently is H or a substituted or unsubstituted CM O alkyl, a substituted or unsubstituted C 2-10 alkenyl, a substituted or unsubstituted cyclic moiety, a substituted or unsubstituted heterocyclic moiety, a substituted or unsubstituted aromatic moiety, or A and B together form a substituted or unsubstituted
  • C 1-10 alkyl is meant straight or branched chain alkyl groups having 1 - 10 carbon atoms, e.g. methyl, ethyl, isopropyl, butyl, and tertbutyl.
  • C 2- ⁇ o alkenyl is meant straight or branched chain alkenyl groups having 2-10 carbon atoms, e.g. ethynyl, propenyl, isopropenyl, butenyl, and tert-butenyl.
  • cyclic moiety is meant cyclohexan, and cyclopentane.
  • aromatic moiety is meant phenyl
  • a and B forms a cyclic, heterocyclic or aromatic moiety denotes cyclohexan, piperidine, benzene, and pyridine.
  • sequence is in the present content meant a peptide comprising naturally occurring and/or non-naturally occurring amino acids, a PNA-sequence, or pep- tidomimetic.
  • naturally occurring amino acids is meant L- and D-forms of the 20 acids found in nature. Non-naturally occurring amino acids are e.g. modified naturally occurring amino acids.
  • sequence is further intended to comprise one or more of such sequences. Examples of suitable peptidomimetics are described in Marshall G.R.,(1993) Tetrahedron, 49:3547-3558.
  • chemical moieties denotes an entity enhancing the solubility or biological activity of the LPA, and entity for directing the LPA to its target or a marker. Preferred embodiments for the sequences are described above.
  • the group X permits directly or indirectly continued stepwise synthesis or a fragment coupling of the same sequence, or of one or more different sequences and/or moie- ties.
  • Orientation of peptide fragments (N to C or C to N) in LPA is defined as desired.
  • the present invention features LPAs with N to C orientation, in another embodiment it concerns the compounds with simultaneous N to C and C to N presentation of the sequences, and in yet another embodiment the sequences have C to N orientation.
  • X comprises a temporally protected amino function
  • synthesis or coupling can be carried out directly after protection. Suitable activation of all car- boxyl-containing groups providing effective formation of the ring system (on step (c), see above) can be ensured using half-equivalent carboxy acid.
  • the activated carboxy group may further be derivatised with a diamine such as ethylenediamine or an amine suitably functionalised for further reactions such as mercapto-, an oxy-, an oxo or carboxyl group.
  • diamine peptide synthesis or fragment coupling can be continued directly according to the desired sequence or chemical moiety.
  • the Fmoc- protection strategy is used, but any amino protection group may be used depending on the synthesis or coupling strategy. Examples are the Boc-protection group strategy.
  • Favourable di-, tri- and tetracarboxiiyc acids for providing the ring structure may be selected from imino diacetic acid, 2-amino malonic acid, 3-amino glutaric acid, 3- methylamino glutaric acid, 3-chloro glutamic acid, 3-methoxy-carbonyl glutaric acid, 3-acetyl glutaruc acid, glutaric acid, tricarballylic acid,3,4-bis-carboxymethyl adipic acid, 4-(2-carboxyethyi)-pimelic acid, (3,5-bis-carboxymethyl-phenyl)-acetic acid, 3,4-bis-carboxymethyl-adipic acid, benzene-1 ,2,4,5-tetra carboxylic acid, 4-(3- carboxy-allylamino)-but-2-enoic acid, 4,4-imino-dibenzoic acid, 1 ,4-dihydropyridine- 3,5-dicarboxylic acid, 5-amin
  • Fragment coupling may be performed according to standard procedures, e.g. as described in Peptide Synthesis protocols, Methods in Molecular Biology Vol. 35, Chapter 15, 303-316, Nyfeler R, Pennington MW and Dunne BM Eds., Humana Press, 1994. Accordingly, fragments may be syn- thesised on a solid phase, cleaved from the solid phase with full preservation of protecting groups, purified and characterised as described above. Suitable fragments may also be obtained by other techniques described above.
  • the antibody is an antibody that recognizes and binds to an epitope comprising the motif K/R-x p - D/E/N/Q-x p -x 0 -S-X ⁇ -x 2 -x 3 -D/E/N/Q-x p , wherein x p is any hydrophobic amino acid residue, x o is K, R or Y, and Xi, x 2 , x 3 are indepently any amino acid residue.
  • an antibody is able to recognize and bind to an epitope on NCAM, said epitope comprising the motif K/R-x p -D/E/N/Q-x p -x o -S-XrX 2 - x 3 -D/E/N/Q-x p , wherein x p is W or P, x o is K, R or Y, and Xi, x 2 , x 3 are independently any amino acid residue.
  • an antibody which recognize and bind to an epitope comprising or being comprised by sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1) and/or sequence SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) of NCAM.
  • epitope is meant the specific group of atoms (on an antigen molecule) that is recognized by (that antigen's) antibodies (thereby causing an immune response).
  • epitope is the equivalent to the term “antigenic determinant”.
  • the epitope may comprise 3 or more amino acid residues, such as for example 4, 5, 6, 7, 8 amino acid residues, located in close proximity, such as within a contiguous amino acid sequence, or located in distant parts of the amino acid sequence of an antigen, but due to protein folding have been approached to each other.
  • Antibody molecules belong to a family of plasma proteins called immunoglobulins, whose basic building block, the immunoglobulin fold or domain, is used in various forms in many molecules of the immune system and other biological recognition systems.
  • a typical immunoglobulin has four polypeptide chains, containing an antigen binding region known as a variable region and a non-varying region known as the constant region.
  • Native antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end.
  • VH variable domain
  • VL variable domain at one end
  • the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains (Novotny J, & Haber E. Proc Natl Acad Sci U S A. 82(14):4592-6, 1985).
  • immunoglobulins can be assigned to different classes. There are at least five (5) major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g. lgG-1 , lgG-2, lgG-3 and lgG-4; lgA-1 and lgA-2.
  • the heavy chains constant domains that correspond to the different classes of immunoglobulins are called alpha ( ⁇ ), delta ( ⁇ ), epsilon ( ⁇ ), gamma ( ⁇ ) and mu ( ⁇ ), respectively.
  • the light chains of antibodies can be assigned to one of two clearly distinct types, called kappa (K) and lambda ( ⁇ ), based on the amino sequences of their constant domain.
  • K kappa
  • lambda
  • the subunit structures and three- dimensional configurations of different classes of immunoglobulins are well known.
  • variable in the context of variable domain of antibodies, refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies.
  • the variable domains are for binding and determine the specificity of each particular antibody for its particular antigen.
  • variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs) also known as hypervariable regions both in the light chain and the heavy chain variable domains.
  • CDRs complementarity determining regions
  • variable domains The more highly conserved portions of variable domains are called the framework (FR).
  • the variable domains of native heavy and light chains each comprise four FR regions, largely a adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen- binding site of antibodies.
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity.
  • an antibody that is contemplated for use in the present invention thus can be in any of a variety of forms, including a whole immunoglobulin, an antibody fragment such as Fv, Fab, and similar fragments, a single chain antibody which includes the variable domain complementarity determining regions (CDR), and the like forms, all of which fall under the broad term "antibody”, as used herein.
  • the present invention contemplates the use of any specificity of an antibody, polyclonal or monoclonal, and is not limited to antibodies that recognize and immunoreact with a specific antigen.
  • an antibody or fragment thereof is used that is immuno- specific for an antigen or epitope of the invention.
  • antibody fragment refers to a portion of a full-length antibody, generally the antigen binding or variable region.
  • antibody fragments include Fab, Fab', F(ab') 2 and Fv fragments.
  • Papain digestion of antibodies produces two identical antigen binding fragments, called the Fab fragment, each with a single antigen binding site, and a residual "Fc" fragment, so-called for its ability to crystallize read- ily.
  • Pepsin treatment yields an F(ab') 2 fragment that has two antigen binding fragments that are capable of cross-linking antigen, and a residual other fragment (which is termed pFc').
  • Additional fragments can include diabodies, linear antibodies, single-chain antibody molecules, and multispecific antibodies formed from antibody fragments.
  • “functional fragment” with respect to antibodies refers to Fv, F(ab) and F(ab') 2 fragments.
  • antibody fragment is used herein interchangeably with the term “antigen binding fragment”.
  • Antibody fragments may be as small as about 4 amino acids, 5 amino acids, 6 amino acids, 7 amino acids, 9 amino acids, about 12 amino acids, about 15 amino acids, about 17 amino acids, about 18 amino acids, about 20 amino acids, about 25 amino acids, about 30 amino acids or more.
  • an antibody fragment of the invention can have any upper size limit so long as it is has similar or immunological properties relative to antibody that binds with specificity to an epitope comprising a peptide sequence selected from any of the sequences identified herein as SEQ ID
  • antibody fragment is identical to term “antigen binding fragment”.
  • Antibody fragments retain some ability to selectively bind with its antigen or receptor.
  • Some types of antibody fragments are defined as follows: (1 ) Fab is the fragment that contains a monovalent antigen-binding fragment of an antibody molecule. A Fab fragment can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain. (2) Fab' is the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain. Two Fab' fragments are obtained per antibody molecule. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region. (3) (Fab') is the fragment of an antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction. (4) F(ab') 2 is a dimer of two Fab' fragments held together by two disulfide bonds
  • Fv is the minimum antibody fragment that contains a complete antigen recognition and binding site. This region consists of a dimer of one heavy and one light chain variable domain in a tight, non-covalent association (V H -V L dimer). It is in this con- figuration that the three CDRs of each variable domain interact to define an antigen binding site on the surface of the V H -V dimer. Collectively, the six CDRs confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • Single chain antibody defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
  • Such single chain antibodies are also referred to as "single-chain Fv” or “sFv” antibody fragments.
  • the Fv polypeptide further comprises a poly- peptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to a small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (VH-VL).
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VH-VL polypeptide chain
  • the invention contemplate both polyclonal and monoclonal antibody, antigen binding fragments and recombinant proteins thereof which are capable of binding an epitope according to the invention.
  • polyclonal antibodies The preparation of polyclonal antibodies is well-known to those skilled in the art. See, for example, Green et al. 1992. Production of Polyclonal Antisera, in: Immuno- chemical Protocols (Manson, ed.), pages 1 -5 (Humana Press); Coligan, et al., Production of Polyclonal Antisera in Rabbits, Rats Mice and Hamsters, in: Current Protocols in Immunology, section 2.4.1 , which are hereby incorporated by reference.
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation tech- niques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography.
  • the monoclonal antibodies for use with the present invention may also be isolated from phage antibody libraries using the techniques described in Clackson et al., 1991, Nature 352: 624- 628, as well as in Marks et al., 1991 , J Mol Biol 222: 581-597.
  • Another method involves humanizing a monoclonal antibody by recombinant means to generate anti- bodies containing human specific and recognizable sequences. See, for review, Holmes, et al., 1997, J Immunol 158:2192-2201 and Vaswani, et al., 1998, Annals Allergy, Asthma & Immunol 81 :105-115.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In additional to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier "monoclonal" indicates the character of the antibody indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies (immu- noglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or sub- class, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (US 4,816,567); Morrison et al., 1984, Proc Natl Acad Sci 81 : 6851 -6855.
  • chimeric antibodies immunoglobulins
  • Antibody fragments of the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab') 2 .
  • This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab' monovalent fragments.
  • a thiol reducing agent optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages
  • an enzymatic cleavage using pepsin produces two monovalent Fab' fragments and an Fc fragment directly.
  • Fv fragments comprise an association of V H and V L chains. This association may be noncovalent or the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde.
  • the Fv fragments comprise V H and V L chains connected by a peptide linker.
  • These single-chain antigen binding proteins are prepared by constructing a structural gene comprising DNA sequences encoding the V H and V ⁇ _ domains connected by an oligonucleo- tide.
  • the structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli.
  • the recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains.
  • CDR peptides (“minimal recognition units") are often involved in antigen recognition and binding. CDR peptides can be obtained by cloning or constructing genes encoding the CDR of an antibody of interest.
  • Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells. See, for example, Larrick, et al., Methods: a Companion to Methods in Enzymology, Vol. 2, page 106 (1991).
  • the invention contemplates human and humanized forms of non-human (e.g. mur- ine) antibodies.
  • humanized antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) that contain a minimal sequence derived from non-human immunoglobulin, such as the eitope recognising sequence.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a nonhuman species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementary determining region
  • donor antibody nonhuman species
  • humanized antibodies may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance.
  • humanized antibodies will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • antibodies may be achieved by any standard methods of the art for producing polyclonal and monoclonal antibodies using peptide fragments comprising the motifs described above, such as for example fragments of sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1) and/or sequence SIDRVEPYSSTAQVQFD (SEQ ID NO: 2).
  • Such antibodies may be also generated using variants or homo- logues of peptide sequences of SEQ ID NOs: 1 and 2.
  • SEQ ID NO: 1 for the production of the antibody
  • the antibodies may also be produced in vivo by the individual to be treated, for example, by administering an immunogenic fragment according to the invention to said individual. Accordingly, the present invention further relates to a vaccine comprising an immunogenic fragment described above.
  • the application also relates to a method for producing an antibody of the invention said method comprising a step of providing of an immunogenic fragment described above.
  • the invention relates both to 1 ) an antibody, which is capable of modulating, such as enhancing or attenuating, biological function of human NCAM and/or FGFR and/or FGFR ligands described herein, in particular a function related to cell differentiation and survival, and/or neural plasticity associated with learning and memory, and 2) an antibody, which can recognise and specifically bind to NCAM without modulating biological activity thereof. It is preferred that such antibody is produced by using an immunogenic peptide sequences described above.
  • the invention relates to use of the described above antibodies for 1) therapeutic applications involving the modulation of activity of FGFR and/or NCAM and/or FGFR ligands described herein; 2) detecting and/or monitoring NCAM polypeptides or fragments thereof in vitro and/or in vivo for diagnostic/prognostic purposes, 4) research purposes.
  • the invention relates to a pharmaceutical composition comprising an antibody described above.
  • the invention features compounds that promote survival of neural cells and can be used as medicaments for the treatment of conditions involving neural cell death.
  • a compound of the invention may also be used as a medicament for promotion of survival of another type of cells, e.g. different type of muscle cells, or, alternatively, for enhancing cell death of still another type of cells, e.g. cancer cells, as the FGFR signalling has been shown to be a survival factor for both muscle and cancer cells (Ozen et al. (2001) J Nat Cancer Inst. 93:1783-90; Miyamoto et al. (1998) J Cell Physiol. 177:58-67; Detiliiux et al.
  • Another approach in the strategy aimed to achieve a compensation for functional cell loss is to create a new pool of said functional cells, for example by commiting the progenitor (stem) cells to differentiate to a new population of differentiated cells, or to initiate regenerating processes in damaged cells.
  • FGFRs play an important role in the mechanisms triggering differentiation of a variety of progenitor cell types (Es- warakumar et al. (2005) Cytokine Growth Factor Rev. 16(2): 139-49), cancer cells (St Bernard et al. (2005) Endocrinology 146(3):1145-53) and neural cells (Sapieha et al. (2003) Mol Cell Neurosci. 24(3):656-72).
  • FGFR signalling leads to development of a number of different of pathologic conditions, e.g. diabetes (Hart et al., Nature 2000, 408:864-8).
  • Activation of FGF receptors is involved as in normal, as in pathologic angiogenesis (Slavin, Cell Biol Int 1995, 19:431-44). It is important for development, proliferation, functioning and survival skeletal muscle cells, cardiomyocytes and neurons (Merle at al., J Biol Chem 1995, 270:17361-7; Cheng and Mattson, Neuron 1991 , 7:1031-41 ; Zhu et al., Mech Ageing Dev 1999, 108:77-85).
  • kidney structure It plays a role in maintenance of normal kidney structure (Cancilla et al., Kidney Int 2001 , 60:147-55), and it is implicated in mound heal- ing and cancer disease (Powers et al., Endocr Relat Cancer. 2000, 7:165-97).
  • the present invention provides compounds capable of modulating the activity of FGFRs, in particular stimulating activity of FGFRs. Consequently, said compounds are concerned by the invention for the production of a medicament for the treatment of diseases, wherein stimulating biological activity dependent on the activity of FGFRs is considered to be beneficial for treatment.
  • the medicament of the invention may be used for prevention and/or treatment of 1 ) diseases and conditions of the central and peripheral nervous system, or of the muscles or of various organs, and/or 2) diseases or conditions of the central and peripheral nervous system, such as postoperative nerve damage, traumatic nerve damage, impaired myelination of nerve fibers, postischaemic damage, e.g.
  • Parkinson's disease Alzheimer's disease, Huntington's disease, dementias such as multiin- farct dementia, sclerosis, nerve degeneration associated with diabetes mellitus, disorders affecting the circadian clock or neuro-muscular transmission, and schizophrenia, mood disorders, such as manic depression; for treatment of diseases or conditions of the muscles including conditions with impaired function of neuro-muscular connections, such as after organ transplantation, or such as ge- netic or traumatic atrophic muscle disorders; or for treatment of diseases or conditions of various organs, such as degenerative conditions of the gonads, of the pancreas such as diabetes mellitus type I and II, of the kidney such as nephrosis and of the heart, liver and bowel, and/or 3) postoperative nerve damage, traumatic nerve damage, impaired myelination of nerve fibers, postischaemic, e.g.
  • Parkinson's disease Alzheimer's disease, Huntington's disease, dementias such as multiinfarct dementia, sclerosis, nerve degeneration associated with diabetes mellitus, disorders affecting the circadian clock or neuro-muscular transmission, and schizo- phrenia, mood disorders, such as manic depression, and/or
  • the invention concerns the cancer being any type of solid tumors requiring neoan- giogenesis. Cancers of neural system are of particular interest of the invention.
  • the invention concerns prion diseases selected from the group consisting of sera- pie, Creutzfeldt-Jakob disease. It has been shown that FGFRs plays a distinct role in prion diseases (Castelnau et al. (1994) Exp Neurobiol. 130:407-10; Ye and Carp (2002) J Mol Neurosci. 18:179-88).
  • a compound of the invention may be used for the manufacture of a medicament for 1) promotion of wound-healing, and/or 2) prevention of cell death of heart muscle cells, such as after acute myocardial infarction, or after angiogenesis, and/or
  • a compound of the invention may be used for the manufacture of a medicament for treatment of normal, degenerated or damaged cells which normally in vivo express one or more FGFR low-affinity ligands selected from the group consisting Neural Cell Adhesion Molecule (NCAM), - Neural cell adhesion molecule L1 , - Neural Cell Adhesion Molecule-2 (NCAM-2), - Neuron-glia Cell Adhesion Molecule (Ng-CAM), - Neural cell adhesion molecule CALL, - Neuroglian, - Nr-CAM (HBRAVO, NRCAM, NR-CAM 12), - Axonin-1/TAG-1 , - Axonal-associated Cell Adhesion Molecule (AxCAM), - Myelin-Associated Glycoprotein (MAG), - Neural cell adhesion molecule BIG-1 , - Neural cell adhesion molecule BIG-2, - Fasci
  • NCAM Neural Cell Adhesion Molecule
  • R-PTP-kappa Protein-Tyrosine Phosphatase Receptor type kappa
  • Phosphatase Receptor type D Phosphatase Receptor type D
  • Ephrin type-A receptor 8 EPHA ⁇ /Tyrosine-Protein Kinase Receptor EEK
  • Ephrin type-A receptor 3 EPHA ⁇ /Tyrosine-Protein Kinase Receptor ETK- 1/CEK4
  • IGF-1 Insulin-like Growth Factor-1 Receptor
  • IRR Insulin-related Receptor
  • Tie-1 Tetra-Protein Kinase Receptor Tie-1
  • CHRNA3 Neuronal nicotinic acetylcholine receptor alpha 3 subunit
  • PDGFRB Platelet-Derived Growth Factor Receptor Beta
  • IL-6R - lnterleukin-6 Receptor
  • IL-23R - lnterleukin-23 Receptor
  • Beta-common cytokine receptor of IL-3, IL5 and GmCsf Beta-common cytokine receptor of IL-3, IL5 and GmCsf
  • IFGP2 - Leukocyte Fc Receptor-like Protein
  • - ADAM-1 A Fertilin alpha, - ADAM-15, - Metalloproteinase-desintegrin domain containing protein (TECAM), - Metalloproteinase 1 , - Collagen type VII, - Fibronectin, - - Tenascin-R, - - Cytokine-like factor-1 (CLF-1).
  • the above listed proteins are natural low-affinity ligands of FGFR and a compound of the invention comprising at least one peptide fragment of the invention capable of binding to FGFR and activating FGFR signalling, may advantageously be used as a mimic of any of the above proteins in conditions wherein the function of said proteins is impaired.
  • such compound is useful for treatment normal, degenerated or damaged NCAM presenting cells.
  • a medicament according to the invention comprises an effective amount of one or more of peptide sequences or compounds as defined above, or a pharmaceutical composition comprising one or more compound of the invention and pharmaceutically acceptable additives.
  • Biological activity associated with specific structural features of the peptide se- quences of the invention should be taken in the account when manufacturing a medicament or preparing a pharmaceutical composition.
  • it may be preffered to use sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1) for the manufacturing a medicament
  • sequence SIDRVEPYSSTAQVQFD SEQ ID NO: 2
  • sequence SIDRVEPYSSTAQVQFD SEQ ID NO: 2
  • a medicament and/or pharmaceutical composition of the invention may be for stimulating of learning and memory
  • a pharmaceutical composition may be for stimulating neuronal survival
  • a pharmaceutical composition may be for stimulating neural cell differentiation
  • a pharmaceutical composition may be i) for stimulating memory and learning and stimulating cell survival, or ii) for stimulating memory and learning and stimulating neurite outgrowth, or iii) stimulating neurite outgrowth and cell survival.
  • a pharmaceutical composition may be preferably used for enhancing or attenuating a function/biological activity of said ligand, which may be different from the biological activities mentioned above.
  • the invention also relates to a medicament and pharmaceutical composition com- prising an antibody of the invention as described above.
  • a further aspect of the invention is a process of producing a pharmaceutical composition, comprising mixing an effective amount of one or more of the compounds of the invention, or a pharmaceutical composition according to the invention with one or more pharmaceutically acceptable additives or carriers.
  • the compounds are used in combination with a prosthetic device, wherein the device is a prosthetic nerve guide.
  • the present invention relates to a prosthetic nerve guide, characterised in that it comprises one or more of the compounds or the pharmaceutical composition as defined above. Nerve guides are known in the art.
  • the invention relates to the use a medicament and/or pharmaceutical composition comprising the compound of invention for the treatment or prophylaxis of any of the diseases and conditions mentioned below.
  • Such medicament and/or pharmaceutical composition may suitably be formulated for oral, percutaneous, intramuscular, intravenous, intracranial, intrathecal, in- tracerebroventricular, intranasal or pulmonal administration.
  • Injectables are usually prepared either as liquid solutions or suspensions, solid forms suitable for solution in, or suspension in, liquid prior to injection.
  • the prepara- tion may also be emulsified.
  • the active ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient.
  • Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like, and combinations thereof.
  • the preparation may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH- buffering agents, or which enhance the effectiveness or transportation of the preparation.
  • Formulations of the compounds of the invention can be prepared by techniques known to the person skilled in the art.
  • the formulations may contain pharmaceuti- cally acceptable carriers and excipients including microspheres, liposomes, micro- capsules, nanoparticles or the like.
  • the preparation may suitably be administered by injection, optionally at the site, where the active ingredient is to exert its effect.
  • Additional formulations which are suitable for other modes of administration include suppositories, nasal, pulmonal and, in some cases, oral formulations.
  • traditional binders and carriers include polyalkylene glycols or triglycerides.
  • Such suppositories may be formed from mixtures containing the active ingredient(s) in the range of from 0.5% to 10%, preferably 1 -2%.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and generally contain 10-95% of the active ingredient(s), preferably 25-70%.
  • formulations are such suitable for nasal and pulmonal administration, e.g. inhalators and aerosols.
  • the active compound may be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include acid addition salts (formed with the free amino groups of the peptide compound) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic acid, oxalic acid, tartaric acid, mandelic acid, and the like. Salts formed with the free carboxyl group may also be derived from inorganic bases such as, for example, sodium, po- tassium, ammonium, calcium, or ferric hydroxides, and such organic bases as iso- propylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • the preparations are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective.
  • the quantity to be ad- ministered depends on the subject to be treated, including, e.g. the weight and age of the subject, the disease to be treated and the stage of disease. Suitable dosage ranges are per kilo body weight normally of the order of several hundred ⁇ g active ingredient per administration with a preferred range of from about 0.1 ⁇ g to 5000 ⁇ g per kilo body weight.
  • the suitable dos- ages are often in the range of from 0.1 ⁇ g to 5000 ⁇ g per kilo body weight, such as in the range of from about 0.1 ⁇ g to 3000 ⁇ g per kilo body weight, and especially in the range of from about 0.1 ⁇ g to 1000 ⁇ g per kilo body weight.
  • the suitable dosages are often in the range of from 0.1 ⁇ g to 1000 ⁇ g per kilo body weight, such as in the range of from about 0.1 ⁇ g to 750 ⁇ g per kilo body weight, and especially in the range of from about 0.1 ⁇ g to 500 ⁇ g per kilo body weight such as in the range of from about 0.1 ⁇ g to 250 ⁇ g per kilo body weight.
  • Administration may be performed once or may be followed by subsequent administrations.
  • the dosage will also depend on the route of administration and will vary with the age and weight of the subject to be treated.
  • a preferred dosage of multimeric forms would be in the interval 1 mg to 70 mg per 70 kg body weight.
  • a localised or substantially localised application is preferred.
  • intranasal application is preferred.
  • the preparation further comprises phar- maceutically acceptable additives and/or carriers.
  • additives and carriers will be known in the art.
  • Administration may be a continuous infusion, such as intraventricular infusion or administration in more doses such as more times a day, daily, more times a week, weekly, etc. It is preferred that administration of the medicament is initiated before or shortly after the individual has been subjected to the factor(s) that may lead to cell death. Preferably the medicament is administered within 8 hours from the factor onset, such as within 5 hours from the factor onset. Many of the compounds exhibit a long term effect whereby administration of the compounds may be conducted with long intervals, such as 1 week or 2 weeks.
  • the administration may be continuous or in small portions based upon controlled release of the active compound(s).
  • precursors may be used to control the rate of release and/or site of release.
  • Other kinds of implants and well as oral administration may similarly be based upon controlled release and/or the use of precursors.
  • Treatment by the use of the compounds/compositions according to the invention is in one embodiment useful for inducing differentiation, modulating proliferation, stimulating regeneration, neuronal plasticity and survival of cells, for example cells being implanted or transplanted. This is particularly useful when using compounds having a long term effect.
  • the treatment may be for stimulation of survival of cells which are at risk of dying due to a variety of factors, such as traumas and injuries, acute diseases, chronic diseases and/or disorders, in particular degenerative diseases normally leading to cell death, other external factors, such as medical and/or surgical treatments and/or diagnostic methods that may cause formation of free radicals or otherwise have cytotoxic effects, such as X-rays and chemotherapy.
  • factors such as traumas and injuries, acute diseases, chronic diseases and/or disorders, in particular degenerative diseases normally leading to cell death
  • other external factors such as medical and/or surgical treatments and/or diagnostic methods that may cause formation of free radicals or otherwise have cytotoxic effects, such as X-rays and chemotherapy.
  • cytotoxic effects such as X-rays and chemotherapy.
  • the FGFR binding compounds according to the invention are useful in cancer treatment of all cancer cells presenting FGFRs.
  • the treatment comprises treatment and/or prophylaxis of cell death in relation to diseases or conditions of the central and peripheral nervous system, such as postoperative nerve damage, traumatic nerve damage, e.g. resulting from spinal cord injury, impaired myelination of nerve fibers, postischaemic damage, e.g. result- ing from a stroke, multiinfarct dementia, multiple sclerosis, nerve degeneration associated with diabetes mellitus, neuro-muscular degeneration, schizophrenia, Alzheimer's disease, Parkinson's disease, or Huntington's disease.
  • diseases or conditions of the central and peripheral nervous system such as postoperative nerve damage, traumatic nerve damage, e.g. resulting from spinal cord injury, impaired myelination of nerve fibers, postischaemic damage, e.g. result- ing from a stroke, multiinfarct dementia, multiple sclerosis, nerve degeneration associated with diabetes mellitus, neuro-muscular degeneration, schizophrenia, Alzheimer's disease, Parkinson's disease, or Huntington's disease.
  • the compounds accord- ing to the invention may be used for inducing differentiation, modulating proliferation, stimulate regeneration, neuronal plasticity and survival , i.e. stimulating survival.
  • the compound and/or pharmaceutical composition may be for preventing cell death of heart muscle cells, such as after acute myocardial infarction, in or- der to induce angiogenesis.
  • the compound and/or pharmaceutical composition is for the stimulation of the survival of heart muscle cells, such as survival after acute myocardial infarction.
  • the compound and/or pharmaceutical composition is for revascularisation, such as after injuries.
  • the present compounds are capable of stimulating angiogenesis and thereby promote the wound healing process.
  • the invention further discloses the use of the compound and/or pharmaceutical composition in the treatment of cancer. Regulation of activation of FGFR is important for tumor agiogenesis, proliferation and spreading.
  • the use of the compound and/or pharmaceutical composition is for the stimulation of the ability to learn and/or of the short and/or long term memory, as FGFR activity is important for differentiation of neural cells.
  • a compound and/or pharmaceutical composition of the invention is for treatment of body damages due to alcohol consumption.
  • Develop- mental malformations of foetuses, long-term neurobehavioral alterations, alcoholic liver disease is in particular concerned.
  • Therapeutic treatment of prion diseases including using a compound and/or phar- maceutical composition is still another embodiment of the invention.
  • Neoplasms such as malignant neoplasms, benign neoplasms, carcinoma in situ and neoplasms of uncertain behavior
  • diseases of endocrine glands such as diabetes mellitus
  • psychoses such as senile and presenile organic psychotic conditions
  • alcoholic psychoses drug psychoses
  • transient organic psychotic conditions Alzheimer's disease, cerebral lipi- doses, epilepsy, general paresis [syphilis], hepatolenticular degeneration, Hunting- ton's chorea, Jakob-Creutzfeldt disease, multiple sclerosis, Pick's disease of the brain, syphilis.
  • Schizophrenic disorders affective psychoses, neurotic disorders, personality disorders, including character neurosis, nonpsychotic personality disorder associated with organic brain syndromes, paranoid personality disorder, fanatic personality, paranoid personality (disorder), paranoid traits, sexual deviations and disorders,
  • Inflammatory and toxic neuropathy including acute infective polyneuritis, Guillain-Barre syndrome, Postinfectious polyneuritis, polyneuropathy in collagen vascular disease, disorders affecting multi- pie structures of eye, purulent endophthalmitis, diseases of the ear and mastoid process, chronic rheumatic heart disease, ischaemic heart disease, arrhythmia, diseases in the pulmonary system, abnormality of organs and soft tissues in newborn, including in the nerve system, complications of the administration of anesthetic or other sedation in labor and delivery, diseases in the skin including infection, insufficient circulation problem, injuries, including after surgery, crushing injury, burns.
  • Injuries to nerves and spinal cord including division of nerve, lesion in continuity (with or without open wound), traumatic neuroma (with or without open wound), traumatic transient paralysis (with or without open wound), accidental puncture or laceration during medical procedure, injury to optic nerve and pathways, optic nerve injury, second cranial nerve, injury to optic chiasm, injury to optic pathways, injury to visual cortex, unspecified blindness, injury to other cranial nerve(s), injury to other and unspecified nerves.
  • diseases or conditions of various organs such as degenerative conditions of the gonads, of the pancreas, such as diabetes mellitus type I and II, of the kidney, such as nephrosis.
  • Scrapie Creutzfeldt-Jakob disease, Gerstmann-Straussler-Sheinker (GSS) disease.
  • the treatment and/or prevention of the above conditions and symptoms comprises a step of administering an effective amount of a compound and/or pharmaceutical composition of the invention to an individual in need.
  • NCAM TIMGLKPETRYAVR SEQ ID NO: 5
  • EFL peptide EFL peptide
  • EVYVVAENQQGKSKA SEQ ID NO: 4
  • Sequence KAEWKSLGEEAWHSK (SEQ ID NO: 1 ) is termed herein as the CDL peptide and sequence SIDRVEPYSSTAQVQFD (SEQ ID NO: 2) is termed herein as the ABL peptide.
  • Cerebellar granule neurons were prepared from postnatal day seven Wistar rats largely as previously described (Neiiendam et al, (2004) J Neurochem. 91 (4):920-35). Cerebellar tissue was dissected in modified Krebs-Ringer solution kept on ice, and treated as described for the hippocampal neurons above. All cell cultures were incubated at 37°C in a humidified atmosphere containing 5 % C0 2 . All animals were handled in accordance with the national guidelines for animal welfare.
  • Dissociated cells were plated at a density of 10,000 cells/cm 2 on uncoated 8-well permanox Lab-Tek chamber slides in Neurobasal medium supplemented with 0.4 % (w/v) bovine serum albumin (BSA; Sigma-Aldrich), 2 % (v/v) B27 Neurobasal supplement, 1 % (v/v) glutamax, 100 U/ml penicillin, 100 g/ml streptomycin and 2 % 1 M HEPES (all from Gibco, BRL). Peptide solutions without or with inhibitors of vari- ous signal transduction pathways were added to a total volume of 300 ⁇ l/cm 2 , and the slides were incubated at 37°C.
  • BSA bovine serum albumin
  • Peptide solutions without or with inhibitors of vari- ous signal transduction pathways were added to a total volume of 300 ⁇ l/cm 2 , and the slides were incubated at 37°C.
  • the neurons were fixed with 4 % (v/v) formaldehyde for 20 minutes and thereafter immunostained using primary rabbit antibodies against GAP-43 and Alexa Fluor secondary goat anti-rabbit antibodies.
  • Images of at least 200 neurons for each group in each individual experiment were obtained systematically by using computer assisted fluorescence microscopy as previously described (R ⁇ nn et al., 2000 op. cit.). Briefly, a Nikon Diaphot inverted microscope with a Nikon Plan 20x objective (Nikon, Tokyo, Japan) coupled to a video camera (Grundig Electronics, Germany) was used for recordings. The same software package as described above for the dopaminergic neurite outgrowth assay was used to process the recorded images.
  • the EFL and CDL peptides of NCAM F3, 1 are strong stimulators of neurite outgrowth neurons grown in culture without substrate, whereas the ABL peptide is inactive in these culture conditions.
  • the effect of the EFL peptide can be abolished by application of SU 5402, a known inhibitor of FGFR, indicating that the stimulation is dependent on activation of the receptor.
  • CGN Primary cultures of CGN were plated at a density of 100,000 cells/cm 2 on poly-L- iysine coated 8-well permanox slides in Neurobasal-A medium (Gibco, BRL) supplemented with 2 % (v/v) B27, 0.5 % (v/v) glutamax, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin and KCI, making the final concentration of KCI in the medium 40 mM.
  • Neurobasal-A medium Gibco, BRL
  • cytosine- ⁇ -D-arabinofuranoside (Ara-C; Sigma-Aldrich) was added to a final concentration of 10 ⁇ M to avoid proliferation of glial cells, after which the neurons were allowed to differentiate for further six days at 37°C.
  • Apoptotic cell death was induced by washing twice and changing the medium to Basal Medium Eagle (BME; Gibco BRL) supplemented with 1 % (v/v) glutamine, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin, 3.5 g D-glucose/l and 1 % (v/v) sodium pyruvate (Gibco BRL) together with various concentrations of peptide.
  • Figure 2A and 2B correspondingly significantly promote survival of CGN in culture, and the effect is higher then the effect of well-known cell survival promoters such as the C3 peptide (Berezin V and Bock E. (2004) J Mol Neurosci 22(1-2):33-39) and GDNF (Krieglstein K (2004) Cell Tissue Res 318(1):73-80) ( Figure 2A).
  • the ABL peptide does not influence survival of CGN neurons in culture.
  • Figure 3 demonstrates that the CDL and EFL peptides inhibit the apoptosis- associated Caspase-3-like activity in cells maintained in culture maximn depleted of KCI and that the effect is comparable with the effect of high KCI (40 mM).
  • CD - C strand-loop-D strand corresponds to the CDL peptide SEQ ID NO: 1
  • EF - E strand-loop-F strand corresponds to the EFL peptide SEQ ID NO: 5
  • BC - B strand-loop-C strand DE - D strand-loop-E strand
  • FG - F strand-loop-G strand have been prepared synthetically.
  • the FGFR Ig module 3 and modules 2, 3 were expressed in Drosophila S2 cells (Invitrogen, USA) according to the manufacturer's instructions. All the proteins were purified by affinity chromatography using Ni 2+ -NTA resin (Qiagen, USA) and/or ion exchange chromatography and gel filtration.
  • Binding analysis was performed using a BIAcoreX instrument (Biosensor AB) at 25 °C using 10 mM sodium phosphate pH 7.4, 150 mM NaCI as running buffer. The flow-rate was 5 ⁇ l/min. Data were analysed by non-linear curve-fitting using the manufacture's software.
  • the FGFR Ig modules 2, 3 were immobilized on a sensor chip CM5 using amine coupling kit (Biosensor AB) as follows: 1 ) the two halves of the chip (designated Fc1 and Fc2) were activated by 20 ⁇ l activation solution; 2) the protein was immobilized on Fc1 using 12 ⁇ l 20 ⁇ g/ml protein in 10 mM sodium phosphate buffer pH 6.0; 3) Fc1 and Fc2 were blocked by 35 ⁇ l blocking solution. Binding of various compounds to the immobilized FGFR modules was studied as follows: A compound was injected simultaneously into Fc1 (with the immobilized FGFR modules) and Fc2 (with nothing immobilized).
  • Example 5 Phosphorylation of FGFR TREX-293 cells (Invitrogen) were stably transfected with human FGFR1 having a C- terminal Strepll-tag (IBA biotech), using the Flp-ln system (Invitrogen). For the study of phosphorylation: -2x10 7 cells were starved overnight before stimulation for 20 min with the specified compounds. Cells were lysed in PBS with 1% NP-40 and phosphatase inhibitors cocktail set II (Calbiochem). The cleared cell lysates were incu- bated with 50 ⁇ l agarose-coupled anti-phosphotyrosine antibodies (4G10-AC, Upstate Biotechnologies) for 3 hrs at 4°C.
  • the rats were handled for 120 seconds every day for 3 days preceding the first day of the experiment. On day 1 (training), the rats received a 1 -second shock (uncondi- tioned stimuli) each minute for two minutes. Animals were injected immediately after training with 5 ⁇ l EFL, ABL, CDL or control peptide, or the vehicle. Each treatment group consisted of 11-14 rats. In total, 39 animals were evaluated. Animals were tested on days 2, 7 and 30 by placing them in the chamber used for conditioning, but in the absence of a shock, for 8 minutes. Each rat was rated as either "freezing" or "active", every 2 seconds, using a time-sampling procedure.
  • the animals were evaluated using the SRT, 1 hour and 24 hours after subcutaneous injection of the peptide or vehicle.
  • Habituation session On the day before the social recognition test (SRT), one ha- bituation session was performed under similar conditions as the test session for adaptation to the experimental room, time when the test was performed, test cage, and contact with juvenile rats (a different juvenile was used during habituation session and social recognition test).
  • the inter-trial interval for the habituation session was of 1 hour.
  • Experimental session Each experimental session consisted of an initial and a test trial separated by an inter-trial (retention) interval of 2 hours.
  • Test trial The retest was performed after a delay of 2 hours under essentially the same experimental conditions as the initial trial. The test was conducted with an unfamiliar juvenile, thus each adult was exposed to a juvenile they had not previ- ously experienced.
  • Juvenile recognition was determined to be present during the second exposure, if the adult spend significantly less time investigating the previously exposed juvenile. A lack of difference between the times spent investigating the juvenile during the first and second exposures, indicated that the adult failed to recognize the familiar juvenile. Further, the recognition ratio (RR): T 2 /(T ⁇ +T 2 ), T ⁇ and T 2 being the time spent on investigating the juvenile animal during the first (initial) and the second (test) trial respectively (Kogan et al., 2000) was used as an index of social recognition memory. If the adult rat demonstrated aggressive, passive (investigating time less than 25% of total exposure time) or sexual behaviour towards the juvenile rat, the rat was not scored.
  • and T 2 were carefully entered into a computerised database (GRAPH PAD version 4). The following parameters were analysed: 1 ) Duration of Ti and T 2 : a) A significant difference between Ti and T 2 of a same animal (obtained using paired t-test analysis) was taken as an indication of presence of social memory. b) A significant difference between T, of different treatments groups, (obtained by unpaired t-test in case of comparison between 2 groups, or by one-way ANOVA followed by Neuman-Keuls post-hoc test in case of comparison between more than 2 groups), was taken as an indication that a treatment was affecting the investigative behaviour of the animals.
  • test peptides ABL and EFL
  • vehicle water
PCT/DK2005/000400 2004-06-18 2005-06-17 Fgfr binding peptides WO2005123759A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
BRPI0512254-6A BRPI0512254A (pt) 2004-06-18 2005-06-17 peptìdeos ligantes ao fgfr
MXPA06014804A MXPA06014804A (es) 2004-06-18 2005-06-17 Peptidos de union del receptor del factor de crecimiento fibroblastico.
EP05748535A EP1765861B1 (en) 2004-06-18 2005-06-17 Fgfr binding peptides
CA002570267A CA2570267A1 (en) 2004-06-18 2005-06-17 Fgfr binding peptides
AT05748535T ATE545655T1 (de) 2004-06-18 2005-06-17 Fgfr-bindende peptide
US11/629,896 US20090074774A1 (en) 2004-06-18 2005-06-17 Fgfr binding peptides
AU2005254613A AU2005254613A1 (en) 2004-06-18 2005-06-17 FGFR binding peptides
JP2007515781A JP2008504236A (ja) 2004-06-18 2005-06-17 Fgfr結合性ペプチド

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200400963 2004-06-18
DKPA200400963 2004-06-18

Publications (3)

Publication Number Publication Date
WO2005123759A2 true WO2005123759A2 (en) 2005-12-29
WO2005123759A8 WO2005123759A8 (en) 2006-03-09
WO2005123759A3 WO2005123759A3 (en) 2006-05-04

Family

ID=35510321

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2005/000400 WO2005123759A2 (en) 2004-06-18 2005-06-17 Fgfr binding peptides

Country Status (11)

Country Link
US (1) US20090074774A1 (pt-PT)
EP (1) EP1765861B1 (pt-PT)
JP (1) JP2008504236A (pt-PT)
CN (1) CN101027320A (pt-PT)
AT (1) ATE545655T1 (pt-PT)
AU (1) AU2005254613A1 (pt-PT)
BR (1) BRPI0512254A (pt-PT)
CA (1) CA2570267A1 (pt-PT)
MX (1) MXPA06014804A (pt-PT)
WO (1) WO2005123759A2 (pt-PT)
ZA (1) ZA200610546B (pt-PT)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007110079A2 (en) * 2006-03-29 2007-10-04 Enkam Pharmaceuticals A/S Targeted delivery of fgfr ligands into the brain
EP2412727A3 (en) * 2006-02-14 2012-04-25 University Of Tasmania Through The Menzies Research Institute Metallothionein-derived peptide fragments
WO2013184824A1 (en) 2012-06-05 2013-12-12 The Board Of Trustees Of The Leland Stanford Junior University Methods for treating psychiatric disorders or symptoms thereof using ncam peptide mimetics

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2004275929A1 (en) * 2003-09-30 2005-04-07 Enkam Pharmaceuticals A/S A method of modulating cell survival, differentiation and/or synaptic plasticity
CN101619093B (zh) * 2009-05-26 2011-12-07 中国人民解放军第三军医大学野战外科研究所 具有促进成纤维细胞生长因子受体3活性的多肽及其筛选方法和应用
EP2483406A2 (en) 2009-09-30 2012-08-08 President and Fellows of Harvard College Methods for modulation of autophagy through the modulation of autophagy-inhibiting gene products
CN109867724B (zh) * 2019-04-19 2020-09-15 温州医科大学 一种fgfr抗体及其在脊髓损伤治疗中的用途

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001096364A2 (en) * 2000-06-16 2001-12-20 Imperial College Innovations Limited Peptides that stimulate cell survival and axon regeneration
US20020104122A1 (en) * 1994-06-17 2002-08-01 Kirin Beer Kabushiki Kaisha Methods for producing a plant with enhanced resistance to pathogenic fungi
WO2003016351A2 (en) * 2001-08-17 2003-02-27 Enkam Pharmaceuticals A/S N - cam related compounds modulating cell groth
WO2005049641A2 (en) * 2003-11-21 2005-06-02 Ace Biosciences A/S Surface-located campylobacter jejuni polypeptides

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4036945A (en) * 1976-05-03 1977-07-19 The Massachusetts General Hospital Composition and method for determining the size and location of myocardial infarcts
US4331647A (en) * 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4599311A (en) * 1982-08-13 1986-07-08 Kawasaki Glenn H Glycolytic promotersfor regulated protein expression: protease inhibitor
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5693488A (en) * 1994-05-12 1997-12-02 The Rockefeller University Transmembrane tyrosine phosphatase, nucleic acids encoding the same, and methods of use thereof
EP2275553B1 (en) * 1999-10-29 2015-05-13 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigenic peptides

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020104122A1 (en) * 1994-06-17 2002-08-01 Kirin Beer Kabushiki Kaisha Methods for producing a plant with enhanced resistance to pathogenic fungi
WO2001096364A2 (en) * 2000-06-16 2001-12-20 Imperial College Innovations Limited Peptides that stimulate cell survival and axon regeneration
WO2003016351A2 (en) * 2001-08-17 2003-02-27 Enkam Pharmaceuticals A/S N - cam related compounds modulating cell groth
WO2005049641A2 (en) * 2003-11-21 2005-06-02 Ace Biosciences A/S Surface-located campylobacter jejuni polypeptides

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2412727A3 (en) * 2006-02-14 2012-04-25 University Of Tasmania Through The Menzies Research Institute Metallothionein-derived peptide fragments
EP2412726A3 (en) * 2006-02-14 2012-05-23 University Of Tasmania Through The Menzies Research Institute Metallothionein-derived peptide fragments
US9518089B2 (en) 2006-02-14 2016-12-13 University Of Tasmania Metallothionein-derived peptide fragments
US10100100B2 (en) 2006-02-14 2018-10-16 University Of Tasmania Metallothionein-derived peptide fragments
WO2007110079A2 (en) * 2006-03-29 2007-10-04 Enkam Pharmaceuticals A/S Targeted delivery of fgfr ligands into the brain
WO2007110079A3 (en) * 2006-03-29 2008-03-20 Enkam Pharmaceuticals As Targeted delivery of fgfr ligands into the brain
WO2013184824A1 (en) 2012-06-05 2013-12-12 The Board Of Trustees Of The Leland Stanford Junior University Methods for treating psychiatric disorders or symptoms thereof using ncam peptide mimetics

Also Published As

Publication number Publication date
CN101027320A (zh) 2007-08-29
AU2005254613A1 (en) 2005-12-29
ZA200610546B (en) 2008-06-25
EP1765861B1 (en) 2012-02-15
ATE545655T1 (de) 2012-03-15
CA2570267A1 (en) 2005-12-29
JP2008504236A (ja) 2008-02-14
WO2005123759A3 (en) 2006-05-04
US20090074774A1 (en) 2009-03-19
MXPA06014804A (es) 2007-03-23
WO2005123759A8 (en) 2006-03-09
EP1765861A2 (en) 2007-03-28
BRPI0512254A (pt) 2008-02-19

Similar Documents

Publication Publication Date Title
US8008259B2 (en) Neurotrophin-derived peptide sequences
US8637465B2 (en) Fibroblast growth factor receptor-derived peptides binding to NCAM
US8329652B2 (en) Neuritogenic peptides
EP1765861B1 (en) Fgfr binding peptides
WO2007019860A2 (en) Gdnf derived peptides
WO2007045243A2 (en) Novel fgf receptor binding compounds comprising peptide fragments of neural cell adhesion molecule l1
US20080226639A1 (en) Heparin Binding Peptide
WO2008022645A2 (en) Ncam fibronectin type 3 binding peptides
US20150252075A1 (en) Neuroplastin derived peptides
US20100168382A1 (en) Neuroplastin derived peptides
EP1940875A2 (en) Fibroblast growth factor receptor-derived peptides
WO2009100724A1 (en) Novel peptide derived from ncam (bcl)
WO2008086813A2 (en) Peptides derived from proteins of the insulin super-family

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 52/2005 UNDER (72, 75) THE NAME SHOULD READ "BOCK, ELISABETH"

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2570267

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 200610546

Country of ref document: ZA

Ref document number: PA/a/2006/014804

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2007515781

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 2005254613

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005748535

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 202/CHENP/2007

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2005254613

Country of ref document: AU

Date of ref document: 20050617

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005254613

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580028412.5

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005748535

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11629896

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0512254

Country of ref document: BR