WO1997010836A1 - Peptides and peptidomimetics inhibiting the oncogenic action of p21 ras - Google Patents

Peptides and peptidomimetics inhibiting the oncogenic action of p21 ras Download PDF

Info

Publication number
WO1997010836A1
WO1997010836A1 PCT/US1996/015098 US9615098W WO9710836A1 WO 1997010836 A1 WO1997010836 A1 WO 1997010836A1 US 9615098 W US9615098 W US 9615098W WO 9710836 A1 WO9710836 A1 WO 9710836A1
Authority
WO
WIPO (PCT)
Prior art keywords
lys
val
asp
leu
ala
Prior art date
Application number
PCT/US1996/015098
Other languages
French (fr)
Inventor
Joseph J. Hlavka
Matthew R. Pincus
John Fowler Noble
Henry Baxter Abajian
Andrew S. Kende
Original Assignee
Innapharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/531,525 external-priority patent/US5840683A/en
Application filed by Innapharma, Inc. filed Critical Innapharma, Inc.
Priority to AU73668/96A priority Critical patent/AU7366896A/en
Publication of WO1997010836A1 publication Critical patent/WO1997010836A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to peptides effective in inhibiting oncogenesis, particularly as related to inhibition of p21 ras and adenocarcinomas of the colon, pancreatic carcinomas, neuroblastomas, and other cancers which express the transformed sequence of the ras gene product .
  • ras protooncogenes are activated by characteristic point mutations in a wide variety of malignancies.
  • the expressed p21 ras proteins are oncogenic by virtue of single substituted amino acids, usually at position 12 or 61 of the 189-residue p21 ras gene product, ras proteins act as membrane-associated molecular switches that bind GTP and GDP and slowly hydrolyze GTP to GDP.
  • Pancreatic carcinoma has a high incidence of ⁇ -ras mutations. Mutated K-ras sequences which can be identified by polymerase chain reaction utilizing allele-specific primers can even be found in the plasma or serum from patients with pancreatic carcinoma.
  • the c-Ki-ras oncogene is activated by point mutations involving codon 12 in 72%-100% of primary pancreatic adenocarcinomas, but the gene is not activated in nonneoplastic tissues. Cancer of the exocrine pancreas is rarely curable.
  • Lung cancers also frequently involve ras mutations.
  • Point mutations in codon 12 of the K-ras protooncogene occur more frequently in lung adenocarcinomas from smokers (30%) than they do in lung adenocarcinomas from nonsmokers (7%) , suggesting that smoking is an important factor in the induction of these mutations.
  • the ras oncogene may thus be a specific target of the mutagenic activity of tobacco smoke, and suggest that DNA alterations at this site can occur early and irreversibly during the development of adenocarcinomas of the lung. Mutations in the ras protooncogenes are the most frequently observed molecular alteration in acute myeloid leukemia (AMD .
  • AMD acute myeloid leukemia
  • ras mutations occur as late or relatively early events in the multistep process of myeloid transformation, remains an open question.
  • the ras oncogene plays a role in experimental mammary carcinogenesis; the evidence in human breast cancer, however, is more limited.
  • the ras oncogene plays a role in nitrosoamine-induced esophageal tumors in rats, but in human esophageal cancers ras gene mutations are more rarely found.
  • the ras proteins are key regulators of the growth of eukaryotic cells. Some of the direct targets are unknown. These target proteins include raf-1, gap, phosphatidylinositol-3- hydroxykinase and, very recently, two nuclear proteins, C-JUN and its kinase (JNK) .
  • the three-dimensional x-ray crystal structure for a ras-related protein bound to a domain of raf-1 has been elucidated.
  • the ras-related protein (_______________) binds to raf directly, utilizing residues contained in a sequence involving amino acids 35-37.
  • All of the contact residues in the ras- related protein are homologous to those in the corresponding segment of ras-p-21.
  • One of the inventors has shown that the p- 21 ras protein (35-47 segment) selectively inhibits the mitogenic effects of oncogenic ras-p-21.
  • the activation of ras proteins is a key step in the signal transduction pathways triggered by ligand-bound cell surface receptors, such as the insulin receptor.
  • the classical target of the ras protein is the GTPase activating protein GAP.
  • GAP GTPase activating protein
  • This target protein is thought to play an essential role in the regulation of ras activity by increasing the GTPase activity of wild type, but not transformed ras .
  • GAP-related proteins which includes pl20-GAP.
  • Other target proteins besides mammalian gap itself include (1) IRA1 and IRA2 , the functional equivalents of GAP in yeast.
  • NF1 Human neurofibromitosis
  • NF1 is associated with type 1 neurofibromatosis, one of the most frequently inherited genetic diseases characterized, in part, by multiple neural tumors.
  • NF1 has been shown genetically and biochemically to interact with and stimulate the GTPase activity of ras,- (5) Drosophila Gapl, which acts as a negative regulator of signalling by the Sevenless (SOS) receptor tyrosine kinase involved in eye development.
  • SOS Sevenless
  • Human SOSl and SOS2 genes have also been recently identified which encode proteins that control GDP-->GTP exchange on ras proteins and are involved in signal transduction by tyrosine kinase receptors.
  • si tu hybridization shows that SOSl maps to 2p22-->pl6 and SOS2 to 14q21-->q22 in the human genome.
  • raf Another important target of ras is raf .
  • the protein encoded by the c-raf-1 protooncogene is thought to function downstream of p21 ras because disruption of raf blocks signalling by ras in a number of systems.
  • a highly-conserved 81 residue region of the N-terminus of raf protein has been to be shown to be critical as the ras protein interaction region.
  • the raf gene product interacts with both wild-type and activated ras protein.
  • approximately 50% of the clones identified as interacting with ras were encoded portions of the c-raf and A-raf serine/threonine kinases.
  • ras and the N-terminal region of raf protein associate directly in vi tro and this interaction is dependent on GTP bound to ras .
  • ras-related GTP-binding proteins Within the superfamily of ras-related GTP-binding proteins, only the ras protein itself has been shown to act as an oncogenic protein. Many other proteins, however, have substantial amino acid homology to ras. This ras superfamily of GTP-binding proteins (> 50 members) regulates a diverse spectrum of intracellular processes. These include cellular proliferation and differentiation, intracellular vesicular trafficking, cytoskeletal control, NADPH oxidase function, as well as others. Some of these homologs may have biological activities which are related to ras. For example, rhoA encodes a ras-related GTP-binding protein that was thought principally to play a role in cytoskeletal organization.
  • This small molecule induces the rrg gene, which encodes a proteinase sequence showing 90% amino acid sequence identity to lysyl oxidase.
  • the precursor of the ras oncoprotein must undergo farnesylation or similar modification of the cysteine residue located in a carboxyl-terminal tetrapeptide.
  • These C-terminal lipid modifications are essential for the interaction of ras-related proteins with membranes. While all ras proteins are farnesylated and some palmitoylated, the majority of other ras-related proteins are geranylgeranylated.
  • EP 203587 describes new ras oncogene polypeptides which are used for producing antibodies for immunogenic assays.
  • these sequences are derived from ras and its homologs in the carboxyl terminal domain (residues 170 - 189 in SEQ ID NO:5) and are thus physically distant from and completely unrelated to any sequences claimed herein.
  • these sequences were claimed for the production of antibodies, preferably by linking to an immunogenic carrier, and a claim for direct therapeutic application was not made.
  • peptides constructed from ras and its homologs for therapeutic application namely by interfering with downstream or upstream actions of r ⁇ s_ itself, are useful.
  • the method of identification of said peptides utilizing calculational approaches is believed novel and has unexpectedly led us to these cyclic peptides and peptidomimetics disclosed herein.
  • the present invention provides peptides, cyclized peptides and peptidomimetics capable of inhibiting the oncogenic action of p21 ras .
  • the oncogenic ras-inhibiting cyclized peptides correspond to domains of the oncogenic ras protein which are most flexible and important in " interacting with target proteins upstream and downstream from ras.
  • the peptidomimetics are obtained by molecular modeling, including the structural minimization techniques of molecular dynamics.
  • the peptides are designated by the formulas: Val-Val-lie, Lys-Arg-Val, Ile-Lys-Arg-Val-Lys-Asp (SEQ ID NO:1), Lys-Cys-Asp- Leu-Ala (SEQ ID NO:2) , Cys-Asp-Leu-Ala-Ala-Arg-Thr (SEQ ID NO:3) , Asp-Leu-Ala-Ala (SEQ ID NO:4) or physiologically acceptable salts of the foregoing peptides.
  • cyclic analogues of the above peptides and certain peptides and cyclic peptides are also provided in the present invention.
  • R(l) R(2) , R(3) and R(4) represent, in the most general case, any amino acid which can serve as an amino acid residue linker.
  • Amino acid residue linkers are usually at least one residue and can be most often two to four residues, more often 1 to 10 residues, both ranges being inclusive.
  • Typical amino acid residues useful for linking are tyrosine, cysteine, lysine, and glutamic and aspartic acid.
  • [R(D , R(2)] and [R(3) , R(4)] are each independently selected from either the group consisting of Glu, Gin, Asp, Asn or from the group consisting of Lys, Arg, Orn.
  • the symbol - represents a bond between the carboxyl and amino termini by which R(l) and R(4) can be interconnected to each other via an lower alkenyl or lower alkynyl group, but most preferably by a branched or unbranched methylene bridge of type --(CH 2 ) m --or -- (CH 2 ) m --M-- (CH 2 ) m ,-- .
  • m and m' are integers from 1 to 6, inclusive, and preferably from 1 to , inclusive; and M is NH, N[R(5)] , 0, S or CH-R(5), wherein R(5) is lower alkyl, cycloalkyl or aryl and is preferably methyl, ethyl, propyl, phenyl, X-phenyl, or heterocyclic, wherein X is Cl-, CF 3 -, F-, substituted at the o-, m- , or p- positions on the phenyl group M can contain a part of another diamino acid within the same peptide, e.g., the omega amino group of the one residue can be so linked to such an unnatural amino acid residue in a terminal residue.
  • any amino acid in the sequences provided hereinabove may be replaced with its D-analogue, with the proviso that not more than 50% of the total amino acids are so replaced.
  • a homologous conservative substitution for any amino acid is within the bounds of the present invention provided that substitution does not eliminate the oncogenic ras p21-inhibiting activity.
  • peptides according to the invention it is also possible to envisage intercalating between several amino acids, or even between all the amino acids, of the peptides defined above, dextrorotatory amino acids, and in particular dextrorotatory phenylalanine or dextrorotatory tryptophan, capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity.
  • Another modification in this sense consists in replacing certain amino acids, for example of the isoleucine type, by leucine.
  • a subject polypeptide can differ, unless otherwise specified, from the natural sequences shown above by the sequence being modified by terminal -NH 2 acylation, e.g., acetylation, or by terminal-carboxylamidation, e.g., with ammonia, alkylamines, and the like.
  • This invention further relates to peptidomimetics which model the critical semi-extended conformation of at least one peptide of or cyclic peptide of the present invention, exemplified by the compounds of Structure 1:
  • each of X and Y is H.
  • An exemplary compound falling within Structure 1 is 3 malonoxy-6- (2-aminoethyl) aminocyclopentanoperhydrophenanthrene (Structure 2) .
  • the natural sequence of the human oncogenic ras p21 is given in SEQ ID NO:5.
  • the crystal X-ray structure has been determined at high resolution for that portion of the human ras protein corresponding to residues 1 to 166 of SEQ ID NO: 5.
  • the regions of the p21 protein that are the most likely to change their conformations upon activation of the protein, e.g. by oncogenic amino acid substitutions have been computed using two different methods. Both methods are based on the principle that the linear sequence of amino acids in a protein determines its unique three-dimensional structure. Given an amino acid sequence of a polypeptide or protein, therefore, it should be possible to predict its three-dimensional structure. This task can be accomplished by using the principle that the observed three-dimensional structure of a protein is the one of lowest free energy. There are a vast number of possible structures a given polypeptide chain can adopt, but essentially only one of these is observed.
  • the interatomic interactions in the protein chain must greatly stabilize its final folded form, i.e., lower its conformational energy substantially with respect to that of any other competing structure.
  • the structure of lowest conformational energy so computed is then predicted to be the observed structure of the protein. This structure may be the one determined by x-ray crystallography or by 2- or 3-dimensional nuclear magnetic resonance (NMR) techniques.
  • Equation 1 A set of potential energy functions, in the computer program ECEPP (Empirical Conformational Energy of Peptides Program) , have been developed that accurately compute the conformational energies of given conformations of proteins.
  • the conformational energy of a peptide can be expressed in Equation 1.
  • E tot is the total conformational energy of the protein
  • the Q's are the charges on the i th and j th atoms
  • R 1 is the distance between the i th and j th atoms
  • D is the dielectric constant
  • e 13 and 1D are the lowest non-bonded (Lennard-Jones) energy and the distance at this lowest energy between atoms i and j in the protein
  • a k is the torsional barrier to rotation around specific bonds
  • ⁇ k is the k ch dihedral angle in the protein
  • n is a degeneracy factor, i.e., 3 for single bonds and 2 for double bonds
  • the sign in the last summation term is positive for single bonds and negative for double bonds such as occur in the peptide bond units.
  • This equation shows the total conformational energy as the sum of three terms: the pairwise electrostatic interactions between the individual atoms of a protein, each of which has a partial charge, (first sum) ; a non-bonded energy term (second term) that consists of an attractive term that varies as the inverse sixth power (tenth power for hydrogen-bonding atoms) of the distance between the atoms (from an induced dipole-induced dipole interaction term) and a repulsive term, from the overlap of electron shells, that varies as the inverse twelfth power of the interatomic distance; and finally a torsional term (third sum) that depends upon the bonds about which rotation takes place.
  • Molecular dynamics is based on the principle that the positions of the atoms of a molecule can be predicted as a function of time by solving Newton's equations of motion for the molecule.
  • the force on the molecule is the negative of the first derivative of the potential function with respect to the coordinates of each of the atoms.
  • Newton's equations of motion are then integrated, using the Verlet algorithm, over a trajectory such that the low energy regions around the starting structure are computed.
  • the trajectories are computed over time intervals such that the total energy converges to a low, constant value.
  • the structures whose energies have converged are then used to compute an average structure.
  • the present inventors have identified important peptide regions of the protein that are involved in the signal transduction process, and these peptides can be used to design anti-cancer agents, as taught herein.
  • these peptides can be used to design anti-cancer agents, as taught herein.
  • these domains contain unique extended structures and/or short beta-bend structures which are hypothesized to account in large part for their biological uniqueness. This suggested that cyclization of the peptide structures to force the beta-bend conformation in place would serve to enhance therapeutic activity.
  • a domain of particular interest is the domain from residues 35 through 47 of SEQ ID NO:5, i.e., Thr-Ile-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val- Ile-Asp (SEQ ID NO:6)
  • the peptide corresponding to residues 44 to 46 in SEQ ID N0:5, i.e., Val-Val-He of still more interest is the sequence from residues 96 to 110 of SEQ ID NO:5, i.e., Tyr-Arg-Glu-Gln-He-Lys-Arg-Val- Lys-Asp-Ser-Asp-Asp-Val-Pro
  • SEQ ID NO:7 of even more particular interest is the sequence from residues 101-103 in SEQ ID NO:5, i.e., Lys-Arg-Val; and the sequence corresponding to residues 100 to 105 in SEQ ID NO:5, i.e.
  • sequences of closely related ras proteins can be derived by one skilled in the art from the sequences of closely related ras proteins . Such sequences may possess enhanced therapeutic activity.
  • Nonlimiting examples of such proteins closely related to the ras gene product which represent the parent sequences having identical or nearly identical three dimensional structures and from which homologs of the sequences given in the preceding paragraph can be derived by one normally skilled in the art are:
  • ras-related protein Ara-3 [Arabidopsis thaliana (mouse ear cress) ] (SEQ ID NO: 10) ;
  • ras-related protein Rab-IA [Lvmnea stagnalis (great pond snail) ] SEQ ID NO:14
  • ras-related protein Rab-2 [Homo sapiens (human) ] SEQ ID NO: 15;
  • Rab-2 [Orvctolagus cuniculus (rabbit)] SEQ ID NO:17;
  • ras-related protein Rab-3 [Drosophila melanogaster (fruitfly)] SEQ ID NO:19;
  • Rab-7 [Canis familiaris (dog) ] SEQ ID NO:23;
  • ras-related protein Rab-7 [Dictvostelium discoideum (slime mold) ] SEQ ID NO:24;
  • RabC ras-related protein RabC [EL . discoideum] SEQ ID NO:26;
  • ras-related protein Ral-B [IL. sapiens] SEQ ID NO:32;
  • Rap-1 [IK. discoideum] SEQ ID NO:37;
  • Rap-2A H. sapiens
  • Rap-2B [IK. sapiens] SEQ ID NO:39;
  • Rap-IA [H__ sapiens] SEQ ID NO:41;
  • Rap-IB [IL. sapiens] SEQ ID NO:42;
  • ras-like protein rasA [P. discoideum] SEQ ID NO:44;
  • ras-like protein rasB [P. discoideum] SEQ ID NO:45;
  • ras-like protein rasC [D. discoideum] SEQ ID NO:46;
  • ras-like protein rasG [D. discoideum] SEQ IP NO:47;
  • ras-like protein F54C8.5 [CL . elegansl SEQ ID NO:48; ras . -like protein CC-ras [Coprinus cinereus (inky cap fungus) ]
  • ras-like protein [Geodia cydonium (sponge)] SEQ ID NO:50;
  • cyclized peptides useful for inhibiting the oncogenic activity of the ras protein, said peptides having formulas as given below:
  • R(l) R(2) , R(3) and R(4) represent, in the most general case, any amino acid, such that they serve as amino acid residue linkers.
  • Amino acid residue linkers are usually at least one residue and can be most often two to four residues, more often 1 to 10 residues.
  • Typical amino acid residues used for linking are tyrosine, cysteine, lysine, glutamic and aspartic acid.
  • [R(D, R(2)] and [R(3) , R(4)] independently are selected from either the groups [Glu, Gin, Asp, Asn] or [Lys, Arg, Orn] .
  • - represents a bond between the carboxyl and amino termini by which R(l) and R(4) can be interconnected to each other via an lower alkyl, alkenyl or lower alkynyl group, but most preferably by a branched or unbranched methylene bridge of type --(CH 2 ) m --or -- (CH 2 ) m --M-- (CH 2 ) m . -- .
  • m and m' are integers from 1 to 6, inclusive, and preferably from 1 to 3, inclusive; and M is NH, N[R(5)] , O, S or CH-R(5) , wherein R(5) is lower alkyl, cycloalkyl or aryl and is preferably methyl, ethyl, propyl, phenyl, X-phenyl, or heterocyclic, wherein X is C1-, CF 3 , F- , substituted at the o- , m- , or p- positions on the phenyl group.
  • M can contain a part of another diamino acid within the same peptide, e.g., the omega amino group of the one residue can be so linked to such an unnatural amino acid residue in a terminal residue.
  • any amino acid in the cyclized peptide sequences (I)-(X) recited herein may be replaced with its D- analogue, insofar as not more than 50% of the total amino acids are so replaced.
  • a homologous conservative substitution for any amino acid is within the bounds of the present invention.
  • Conservative substitutions include Glu for Asp, Gin for Asn and Val for He, among others, as well-known to the art.
  • dextrorotatory amino acids capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity.
  • dextrorotatory amino acids capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity.
  • Another modification in this sense consists in replacing certain amino acids, for example of the proline type, by D-tryptophan.
  • a subject polypeptide can differ, unless otherwise specified, from any of the natural sequences shown herein above by the sequence being modified by terminal -NH 2 acylation, e.g., acetylation or by terminal-carboxylamidation, e.g., with ammonia, alkylamines, and the like.
  • the placement of hydrophobic amino acid residues is highly dependent on the peptide sequence. For example, for the 35-47 peptide sequence, corresponding to amino acids 35-47 of SEQ ID NO: 5, there is a distinct hydrophobic region for the amino acid residues corresponding to amino acids 44-46 of SEQ ID NO:5.
  • the bridge in Compound (X) occurs at what corresponds in structure to amino acids 44-46 in SEQ ID NO:5. It is possible to extend this hydrophobic segment without sacrificing activity.
  • the carboxyl terminal Asp residue can be replaced with one or more hydrophobic residues such as Val or He, and the result is greater efficiency in crossing cell membranes.
  • Short half-lives of peptides can be at least partially extended by the addition of D-amino acids to either or both of the amino and carboxyl terminal ends of the peptide.
  • D-amino acid residues block the action of exo- proteases that degrade peptides from their amino or carboxyl ends.
  • the cyclization of the peptide further renders the peptide less susceptible to proteolysis.
  • Recent advances in the field of peptides have been directed towards the stabilization of these peptides against enzymatic or hydrolytic degradation. It would be extremely valuable to stabilize these peptides from degradation by proteolytic enzymes in order to improve their pharmacokinetic properties.
  • Peptidomimetics are generally preferable as therapeutic agents to peptides owing to their enhanced bioavailability and relative lack of attack from proteolytic enzymes.
  • the present inventors have used the techniques of molecular modeling supra to design a peptidomimetic which mimics the critical beta-bend aspects of the peptide corresponding in sequence to amino acids 96-110 of SEQ ID NO:5 (p21 ____£) .
  • the bend structure occurs at amino acids 102-103 in the p21 ras protein. These residues have been implicated in the binding of ras p21 to SOS.
  • Peptidomimetric compounds which inhibit the oncogenic or transforming activity of the p21 ras protein are provided by the compounds of Structure I :
  • each of x and y is H.
  • the instant invention comprises novel peptides of medicinal importance most particularly for the treatment of adenocarcinomas of the colon, pancreatic carcinomas, neuroblastomas, and other cancers of undefined germ cell origin which express the transformed sequence of the ras protein.
  • These peptide sequences were unexpectedly obtained by the use of molecular dynamic simulations on ras p21 to define which domains of the protein were most flexible and were thus most important in interacting with target proteins upstream and downstream from ras .
  • peptides are identified by the following amino acid sequences: Thr-Ile-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val-Ile-Asp (SEQ ID NO:6) , Val-Val-He, Tyr-Arg-Glu-Gln-He-Lys-Arg-Val-Lys-Asp-Ser- Asp-Asp-Val-Pro (SEQ ID NO:7) , Lys-Arg-Val, Ile-Lys-Arg-Val-Lys- Asp (SEQ ID NO:l), Gly-Asn-Lys-Cys-Asp-Leu-Ala-Ala-Arg-Thr-Val- Glu (SEQ ID NO:8), Lys-Cys-Asp-Leu-Ala (SEQ ID NO:2) , Cys-Asp- Leu-Ala-Ala-Arg-Thr (SEQ ID NO:9), and Asp-Leu-A
  • R(l) R(2), R(3) and R(4) represent, in the most general case, any amino acid, such that they serve as amino acid residue linkers.
  • Amino acid residue linkers are usually at least one residue and can be most often two to four residues, more often 1 to 10 residues.
  • Typical amino acid residues used for linking are tyrosine, cysteine, lysine, glutamic and aspartic acid, or the like.
  • [R(D , R(2)] and [R(3) , R(4)] independently are selected from either the groups [Glu, Gin, Asp, Asn] or [Lys, Arg, Orn] .
  • the symbol - represents a bond between the carboxyl and amino termini by which R(l) and R(4) can be interconnected to each other via an lower alkenyl or lower alkynyl group, but most preferably by a branched or unbranched methylene bridge of type --(CH 2 ) m --or -- (CH 2 ) m --M-- (CH 2 ) m .-- .
  • m and m' are integers from 1 to 6 and preferably from 1 to 3; and M is NH, N[R(5)] , O, S CH-R(5) or does not exist, wherein R(5) is lower alkyl, cycloalkyl or aryl and is preferably methyl, ethyl, propyl, phenyl, X-phenyl, or heterocyclic, wherein X is C1-, CF 3 - , F- , substituted at the o-, m- , or p- positions on the phenyl.
  • M can contain a part of another diamino acid within the same peptide, e.g., the omega amino group of the one residue can be so linked to such an unnatural amino acid residue in a terminal residue.
  • any amino acid in the sequences provided may be replaced with its D-analogue, insofar as not more than 50% of the total amino acids are so replaced.
  • Conservative substitutions include Glu for Asp, Gin for Asn and Val for He, among others, as is well known to those of ordinary skill in the art.
  • a homologous conservative substitution for any amino acid is within the bounds of the present invention.
  • dextrorotatory amino acids capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity.
  • dextrorotatory amino acids capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity.
  • Another modification in this sense consists in replacing certain amino acids, for example of the proline type, by D-tryptophan.
  • a subject polypeptide can differ, unless otherwise specified, from the natural sequences shown above by the sequence being modified by terminal -NH 2 acylation, e.g., acetylation, or by terminal-carboxylamidation, e.g., with ammonia, alkylamines, and the like.
  • the instant invention also comprises a method of use of the peptides supra for the treatment of adenocarcinomas of the colon, pancreatic carcinomas, neuroblastomas, and other cancers of undefined germ cell origin which express the transformed sequence of the ras protein.
  • amino acid residues described herein are preferred to be in the "L" isomeric form.
  • residues in the "D" isomeric form can be substituted for any L-amino acid residue, as long as the desired functional property is retained by the polypeptide.
  • NH 2 refers to the free amino group present at the amino terminus of a polypeptide.
  • COOH refers to the free carboxyl group present at the carboxy terminus of a polypeptide.
  • variable amino acids capable of participating in the composition of this peptide is as follows: Y, Tyr, tyrosine; G, Gly, glycine; F, Phe, phenylalanine; M, Met, methionine; A, Ala, alanine; S, Ser, serine; I, He, isoleucine; L, Leu, leucine; T, Thr, threonine; V, Val, valine; P, Pro, proline; K, Lys, lysine; H, His, histidine; Q, Gin, glutamine; E, Glu, glutamic acid; W, Trp, tryptophan; R, Arg, arginine; D, Asp, aspartic acid; N, Asn, asparagine; C, Cys, cysteine.
  • amino acid residue sequences are presented herein in the conventional left-to-right direction of amino-terminus to carboxy-terminus.
  • amino acid residue is broadly defined to include the amino acids listed hereinabove, and modified and unusual amino acids, such as those listed in 37 C.F.R. 1.822(b) (4) , incorporated herein by reference.
  • a dash at the beginning or end of an amino acid residue sequence indicates either a peptide bond to a further sequence of one or more amino acid residues or a covalent bond to an amino or hydroxyl end group.
  • Polypeptide and peptide are terms used interchangeably herein to designate a linear series of amino acid residues connected one to the other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues.
  • Protein is a term used herein to designate a linear series of greater than about 20 amino acid residues connected one to the other as in a polypeptide.
  • synthetic peptide refers to a chemically produced chain of amino acid residues linked together by peptide bonds that is free of naturally occurring proteins and fragments thereof.
  • peptide encompasses linear and cyclic peptides .
  • (D,L), (D) , or (L) preceding the amino acid designation means that this amino acids exists in that specific isomeric form, i.e. (D,L) Phe means that the amino acid phenylalanine exists as a racemic mixture; (D) Phe or D-Phe means that the amino acid phenylalanine exists as the D-stereoisomer or implied R configuration; (L) Phe means that the amino acid phenylalanine exists as the L stereoisomer or implied S configuration.
  • Alkyl as used herein means methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1, 1-dimethylpropyl, n-hexyl, 1-methylpentyl, 2-methylpentyl, 3 -methylpentyl , 4 -methylpentyl , 3 , 3 -dimethylbutyl , 2,2-dimethylbutyl, 1, 1-dimethylbutyl, 2-ethylbutyl, 1-ethylbutyl, 1, 3-dimethylbutyl, n-heptyl, 5-methylhexyl, 4- ethylhexyl, 3-methylhexyl, 2-methylhexyl, 1-methylhexyl, 3-eth
  • Cycloalkyl refers to a hydrocarbon ring having from 3 to 7 carbon atoms, inclusive.
  • Examples of cycloalkyl groups are cyclopropyl, cyclopentyl, cycloheptyl, cyclooctyl, cyclononyl, and the like.
  • aryl refers to aromatic groups which have at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl aralkyl, and biaryl groups, all of which may be optionally substituted.
  • Heterocyclic groups means groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and their heterocyclic compounds can include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl, and the like, all optionally substituted.
  • Substituted heterocyclic refers to any heterocyclic aryl group substituted by a alkyl, aryl, cycloalkyl, halo, sulfonate, or trifluoromethyl group.
  • alkyl amino refers to the groups --NRR' wherein respectively, (a) R is alkyl and R' is hydrogen or alkyl; (b) R is aryl and R' is hydrogen or aryl, (c) R is cycloalkyl and R' is hydrogen or alkyl, (d) R is hydrogen and R' is itself linear aminoalkyl, (e) R is alkyl and R' is itself linear aminoalkyl .
  • aminoalkyl refers to the groups - (CH 2 ) m -NRR' , wherein m is an integer from 1 to 6, inclusive and -NRR' is alkyl amino, as defined supra .
  • Halo encompasses fluoro, chloro, bromo and iodo.
  • protecting group means substituents which protect the reactive functional group from undesirable chemical reactions. Examples of such protecting groups include esters of carboxylic acids, ethers of alcohols and acetals and ketals of aldehydes and ketones.
  • N-protecting group or N-protected as used herein means those groups intended to protect the N-terminus of an amino acid or peptide, to protect an amino group against undesirable reactions during synthetic procedures and includes, but is not limited to, sulfonyl, acetyl, pivaloyl, t-butyloxycarbonyl (Boc) , carbonylbenzyloxy (Cbz) , benzoyl and an L- or D-aminoacyl residue, which may itself be N-protected similarly.
  • the deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl, alkoxycarbonyl or an aroyl group may be removed for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an acyl group such as a t-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid such as hydrochloric, sulfuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-charcoal.
  • COOH-protecting group or carboxyl-protecting group is, an esterifying group, for example an alkyl group
  • an esterifying group such as an alkyl or arylmethyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an esterifying group such as an arylmethyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-charcoal using either hydrogen or ammonium formate as a hydrogen source by methods well-known to those skilled in the art.
  • Electrolyte means a solution that has sufficient acid strength to render a basic starting material essentially protonated.
  • Chemical derivative refers to a subject polypeptide having one or more residues chemically derivatized by reaction of a functional side group.
  • derivatized molecules include for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups.
  • Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides.
  • Free hydroxyl groups may be derivatized to form O-acyl or 0-alkyl derivatives.
  • the imidazole nitrogen of histidine may be derivatized to form N-imidazolyl- benzylhistidine.
  • chemical derivatives those peptides which contain one or more naturally occurring amino acid derivatives of the twenty standard amino acids.
  • 4-hydroxyproline may be substituted for proline
  • 5-hydroxylysine may be substituted for lysine
  • 3-methylhistidine may be substituted for histidine
  • homoserine may be substituted for serine
  • ornithine may be substituted for lysine.
  • Polypeptides of the present invention also include any polypeptide having one or more additions and/or deletions or residues relative to the sequence of a polypeptide whose sequence is shown herein, so long as the requisite activity is maintained.
  • fragment means any subject peptide or polypeptide having an amino acid residue sequence shorter than that of a peptide or polypeptide whose full length amino acid residue sequence is shown herein.
  • a pharmaceutically acceptable salt is one which is prepared by contacting a compound of formulas (I) - (X) according to the specifications therein with an acid whose anion is generally considered suitable for human consumption.
  • pharmacologically acceptable acid addition salts include the hydrochloride, hydrobromide, hydroiodide, sulfate, phosphate, acetate, propionate, lactate, maleate, alate, succinate, and tartrate salts. All of these salts may be prepared by conventional means by reacting, for example, the appropriate acid with the corresponding compound of structure of Formulas (I) - (X) .
  • the preparation methods disclosed herein result in product distributions which include all possible structural isomers. It is understood that physiological response may vary according to stereochemical structure.
  • the isomers may be separated by conventional means such as fractional crystallization or High Pressure Liquid Chromatography (HPLC) .
  • HPLC High Pressure Liquid Chromatography
  • the absolute configuration of a compound relates to how its substituents are oriented in space about a central atom. This notion becomes significant when coupled with the rigors of chirality. Chirality involves the identity of the substituents " about that central atom.
  • a compound is said to be chiral when four distinctly different groups are bound to a central carbon atom. These groups may be spatially aligned in more than one manner without repeating their individual orientations. That is, a chiral compound may exhibit a mirror image which is also chiral . These mirror images are termed meso configurations, and are each absolute configurations of a chiral compound.
  • compositions according to the present invention comprise one or more peptides and/or peptidomimetics of the invention in association with a pharmaceutically acceptable carrier or excipient, adapted for use in human or veterinary medicine.
  • the compositions may contain from 0.001-99% of the active material. Such compositions may be presented for use in conventional manner in admixture with one or more physiologically acceptable carriers of excipients.
  • the pharmaceutical compositions according to the invention may also contain other active ingredients such as antimicrobial agents, or preservatives.
  • the compositions may optionally further contain one or more other therapeutic agents which may, if desired, be a chemotherapeutic antiviral agent.
  • compositions of the peptides of this invention may be formed conventionally by reaction with an appropriate acid.
  • the addition salts so formed from addition by acid may be identified by hydrochloric, hydrobromic, phosphoric, acetic, fumaric, salicylic, citric, lactic, mandelic, tartaric, oxalic, methanesulfonic, and the like.
  • the peptides and peptidomimetics according to the present invention may be formulated for oral, buccal, parenteral, topical or rectal administration.
  • these peptides and peptidomimetics may be formulated for injection or for infusion and may be presented in unit dose form in ampoules or in multidose containers with an added preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • the present invention further provides a process for preparing a pharmaceutical composition which comprises bringing a peptide and/or peptidomimetic of the invention into association with a pharmaceutically acceptable excipient or carrier.
  • the daily dosage as employed for treatment of an adult human of approximately 70 kg body weight will range from 0.01 mg to 10 mg of each active ingredient, preferably 0.1 to 5 mg, which may be administered in 1 to 4 doses, for example, depending on the route of administration and the condition of the patient.
  • the dosage of the peptide used in the treatment will vary, depending on the seriousness of the disorder, the weight of the patient, the relative efficacy of the peptide and the judgment of the treating physician.
  • suitable unit dosages in humans may be between about 0.05 mg to about 100 mg.
  • a unit dosage may be from between about 0.2 mg to about 50 mg.
  • Such a unit dosage, described hereinabove may be administered more than once a day, e g., two or three times a day.
  • the total daily dosage is in the range of about 0.01 mg to 10 mg/kg.
  • Such therapy may extend for several weeks, in an intermittent or uninterrupted manner, until the patient's symptoms are eliminated.
  • compositions which comprise a pharmaceutically effective amount of the one or more peptides and/or peptidomimetics of this invention, or pharmaceutically acceptable salts thereof, and, preferably, a pharmaceutically acceptable carrier or adjuvant.
  • Therapeutic methods of this invention comprise the step of treating patients in a pharmaceutically acceptable manner with those peptides or compositions.
  • Such compositions may be in the form of tablets, capsules, caplets, powders, granules, lozenges, suppositories, reconstitutable powders, or liquid preparations, such as oral or sterile parenteral solutions or suspensions.
  • a composition ' of the invention is in the form of a unit dose.
  • the unit dose presentation forms for oral administration may be tablets and capsules and may contain conventional expedients.
  • binding agents such as acacia, gelatin, sorbitol, or polyvinylpyrrolidone
  • fillers such as lactose, sugar, maize-starch, calcium phosphate, sorbitol or glycine
  • tabletting lubricants such as magnesium stearate
  • disintegrants such as starch, polyvinylpyrrolidone, sodium starch glycolate or microcrystalline cellulose
  • pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.
  • the solid oral compositions may be prepared by conventional methods of blending, filling, tabletting, or the like. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are, of course, conventional in the art.
  • the tablets may be coated according to methods well-known in normal pharmaceutical practice, in particular with an enteric coating.
  • Oral liquid preparations may be in the form of emulsions, syrups, or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may or may not contain conventional additives.
  • suspending agents such as sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminum stearate gel, or hydrogenated edible fats
  • emulsifying agents such as sorbitan monooleate or acacia
  • non-aqueous vehicles which may include edible oils
  • non-aqueous vehicles such as almond oil, fractionated coconut oil, oily esters selected from the group consisting of glycerine, propylene glycol, ethylene glycol, and ethyl alcohol
  • preservatives for instance methyl para-hydroxybenzoate, ethyl para-hydroxybenzoate, n-propyl parahydroxybenzoate, or n-butyl parahydroxybenzoate or sorbic acid
  • conventional flavoring or coloring agents such as sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminum stearate gel, or hydrogenated edible fats
  • emulsifying agents
  • fluid unit dosage forms may be prepared by utilizing the peptide and a sterile vehicle, and, depending on the concentration employed, may be either suspended or dissolved in the vehicle.
  • the peptides of this invention may be dissolved in water, whereas opiates used heretofore showed only marginal solubility in aqueous media or physiological fluids.
  • the peptide may be injected and filter sterilized before filling a suitable vial or ampoule and subsequently sealing the carrier or storage package.
  • Adjuvants such as a local anaesthetic, a preservative or a buffering agent, may be dissolved in the vehicle prior to use.
  • Stability of the pharmaceutical composition may be enhanced by freezing the composition after filling the vial and removing the. water under vacuum, e.g., freeze drying the composition.
  • Parenteral suspensions may be prepared in substantially the same manner, except that the peptide should be suspended in the vehicle rather than being dissolved.
  • a surfactant or wetting solution may be advantageously included in the composition to facilitate uniform distribution of the peptide.
  • the stability of the peptides and cyclized peptides of the present invention exceeds that of naturally occurring peptides if substitution is made with D-amino acids in at least 20%, but not more than 50%, of those residues which are naturally present in the (L) configuration.
  • the enhanced stability of the peptides of this invention may also be the result of the introduction of modifications of traditional peptide linkages.
  • the introduction of a cyclic ring within the peptide backbone may confer enhanced stability in order to circumvent the effect of many proteolytic enzymes known to digest small peptides in the stomach or other digestive organs and in serum.
  • the compounds of the " present invention are initially synthesized by either solution or by solid phase techniques. Specific exemplary syntheses are described in the examples hereinbelow.
  • the peptides of this invention may be prepared by initially reacting a first appropriately protected amino acid with a second appropriately protected amino acid in an organic solvent inert to the reactants, in the presence of a suitable peptide coupling agent according to the following scheme:
  • Z (l)NH-AA(l) -COOH + coupling agent + NH 2 -AA(2) -COOZ (2) Z(l) -NH-AA(l) -CONH-AA(2) -COOZ (2) wherein Z(l) is a suitable nitrogen protecting group and Z(2) is a suitable carboxyl protecting group and AA represents any natural or unnatural amino acid residue.
  • the desired peptides may be prepared by utilizing the appropriate amino acids and repeating this reaction sequence as required until a peptide with three to ten amino acid residues has been prepared. A suitable deprotection method is then employed to remove specified or all of the remaining protecting groups or the peptide from the resin.
  • the first appropriately protected amino acid and, for instance, an appropriately protected tyrosine may be reacted together in the presence of a suitable peptide coupling agent in a suitably inert organic solvent with stirring, shaking, or agitation to form a protected tyrosine containing dipeptide.
  • a suitable peptide coupling agent in a suitably inert organic solvent with stirring, shaking, or agitation to form a protected tyrosine containing dipeptide.
  • Introducing this dipeptide to appropriate protecting group removal conditions affords a selectively deprotected dipeptide which is well-suited for continued peptide synthesis.
  • Contacting this mono-deprotected tyrosine containing dipeptide with an appropriately protected amino acid having a side chain represented as above, in the presence of a suitable peptide coupling agent in a suitably inert organic solvent with stirring, shaking, or agitation forms a protected tyrosine containing tripeptide. This method may be repeated as many times as necessary to achieve
  • the method of preparation for peptide synthesis requires specific functional groups to react with other substituents to link amino acid residues in a desired manner to form a peptide possessing a known and desired sequence of amino acid residues. Since amino acids possess at least two reactive functional groups, suitable protection, blocking, or masking of these groups is required to ensure that reaction will occur only at specifically desired sites.
  • protecting groups should be introduced to the moiety efficaciously while their removal should be performed under conditions which do not affect other portions of the molecule. In this manner, certain reactions and modifications may be performed on the amino acid, peptide, or other compound, with assurance that the protected functionality will not interfere with the desired reaction. Further, by choosing a protecting group that is sensitive and labile to certain reactive conditions, a reaction scheme may be outlined to advantageously utilize these characteristics to effectively remove the protecting group once the synthesis is complete.
  • N-protecting groups and COOH-protecting groups may be used within the scope of this invention.
  • a variety of protecting groups known in the field of peptide synthesis and recognized by conventional abbreviations therein, may be found in T. Greene, Protective Groups In Organic Synthesis, Academic Press (1981) .
  • the preferred protecting groups that may be utilized for suitable protection of reactive nucleophilic substituents include, for example, benzyl (Bz) , carbobenzyloxy (Cbz) , t-butoxycarbonyl (Boc) , or 9-fluorenylmethyloxy-carbonyl (Fmoc) .
  • Coupling of amino acids which may be the same or different as those described above, to yield small peptides in route to peptides comprised of greater numbers of amino acid residues may be accomplished by employing established techniques in the field of peptide chemistry. A broad range of suitable reactions are described in E. Gross and J. Meinhofer, The Peptides: Analysis, Synthesis, Biology; Modern Techniques of Peptide and Amino Acid Analysis, John Wiley & Sons, (1981) and M. Bodanszky, Principles Of Peptide Synthesis, Springer-Verlag (1984) .
  • the peptide coupling agents which may be used to assist condensation of amino and carboxyl i c ac i d mo i et i e s inc lude
  • CDI 1-hydroxy benzotriazole
  • HOBt 1-hydroxy benzotriazole
  • EEDQ 2-dihydroquinoline
  • BOP benzotriazoyl-oxy-tris- (dimethyl) amino-phosphonium hexafluoro phosphate
  • the DCC reaction is often performed at room temperature but may be carried out from about -78 °C to gentle reflux in a variety of solvents that are inert with respect to the reactants.
  • the solvents are normally organic solvents which are polar and aprotic.
  • Preferred solvents include, for example, dichloromethane, chloroform, diethyl ether, tetrahydrofuran
  • the coupling reaction may be carried out at atmospheric pressure a temperature of -78 °C to reflux for a period of between 1 and 48 hours.
  • the reaction is carried out at about -10° C to 25° C with stirring, shaking or agitation, over a period of between 4 and 6 hours.
  • synthesis may be achieved prepared using solid phase synthesis, such as that described by Merrifield, J Am. Chem. Soc. , 85, p 2149 (1964), although other equivalent chemical syntheses known in the art can also be used.
  • Solid-phase synthesis is commenced from the C-terminus of the peptide by coupling a protected alpha-amino acid to a suitable resin as generally set forth in U.S. Pat. No. 4,244,946.
  • a protected alpha-amino acid to a suitable resin as generally set forth in U.S. Pat. No. 4,244,946.
  • He protected by BOC is coupled to the a BHA resin using methylene chloride and dimethylformamide .
  • the alpha-amino protecting group is removed, as by using trifluoroacetic acid
  • TFA in methylene chloride
  • TFA alone or with HCl in dioxane Preferably 50 volume % TFA in methylene chloride is used with 0-5 weight % 1,2 ethanedithiol.
  • the deprotection is carried out at a temperature between about 0° C and room temperature.
  • Other standard cleaving reagents and conditions for removal of specific alpha-amino protecting groups may be used as described in Schroder & Lubke, The Peptides, pp 72-75 (Academic Press 1965) .
  • the remaining alpha-amino- and side chain-protected amino acids are coupled step-wise in the desired order to obtain the intermediate compound defined hereinbefore.
  • some of them may be coupled to one another prior to addition to the solid phase reactor. The selection of an appropriate coupling reagent is within the skill of the art.
  • coupling reagents are N,N' -dicyclohexyl carbodiimide (DCC) and N, N ' -diisopropyl carbodiimide ( D I C I ) , or N-ethyl-N' - (3-dimethylaminopropyl) carbodiimide.
  • DCC N,N' -dicyclohexyl carbodiimide
  • D I C I N, N ' -diisopropyl carbodiimide
  • N-ethyl-N' - (3-dimethylaminopropyl) carbodiimide N-ethyl-N' - (3-dimethylaminopropyl) carbodiimide.
  • Other activating reagents and their use in peptide coupling are described by Schroder & Lubke, supra , in Chapter III and by Kapoor (1970) J. Phar. Sci. .59:127.
  • Each protected amino acid or amino acid sequence is introduced into the solid phase reactor in about a fourfold excess, and the coupling is carried out in a medium of dimethylformamide: dichloromethane (1:1) or in DMF or dichloromethane alone.
  • the coupling is carried out manually, the success of the coupling reaction at each stage of the synthesis is monitored by the ninhydrin reaction, as described by E. Kaiser et al . (1970) Anal. Biochem. 3_4:595.
  • the coupling procedure is repeated before removal of the alpha-amino protecting group prior to the coupling of the next amino acid.
  • the coupling reactions can be performed automatically, as on a Applied Biosystems automatic synthesizer.
  • the intermediate peptide is removed from the resin support by treatment with a reagent, such as liquid hydrogen fluoride, which not only cleaves the peptide from the resin but also cleaves all remaining side chain protecting groups and the alpha-amino protecting group (unless it is an acyl group which is intended to be present in the final peptide) to obtain the peptide.
  • a reagent such as liquid hydrogen fluoride
  • anisole or cresol and methylethyl sulfide are included in the reaction vessel as scavengers.
  • the BOC protecting group may be cleaved with trifluoroacetic acid (TFA) /ethanedithiol prior to cleaving the peptide from the resin to eliminate potential S-alkylation.
  • the synthesis of the peptide of SEQ ID NO:l is conducted in a stepwise manner on a MBHA hydrochloride resin, such as available from Bachem, Inc. (Torrance, CA) having a substitution range of about 0.1 to 0.5 mmoles/gm. resin.
  • a MBHA hydrochloride resin such as available from Bachem, Inc. (Torrance, CA) having a substitution range of about 0.1 to 0.5 mmoles/gm. resin.
  • DIPEA DIPEA
  • PIP piperidine
  • DCC dicyclohexylcarbodiimide
  • HOBt 1-hydroxybenzotriazole
  • BOP [benzotriazole-1-yl- oxy-tris (dimethyl) phosphonium hexafluorophosphate]
  • the synthesis is performed on an Applied Biosystems peptide synthesizer (Foster City, CA) using a suitable program, preferably as follows:
  • the peptide chain is built stepwise on the resin. Generally, one to two mmol. of BOC-protected amino acid in methylene chloride is used per gram of resin, plus one equivalent of 2 M DCC in methylene chloride, for two hours.
  • BOC-Arg(Tos) is being coupled, a mixture of 50% DMF and methylene chloride is used.
  • Bzl is used as the hydroxyl side-chain protecting group for Ser and Thr.
  • p-nitrophenyl ester (ONp) can be used to activate the carboxyl end of Asn or Gin; for example, BOC-Asn(ONp) can be coupled overnight using one equivalent of HOBt in a 50% mixture of DMF and methylene chloride.
  • the amido group of Asn or Gin is protected by Xan when DCC coupling is used instead of the active ester method.
  • 2-Cl-CBZ is used as the protecting group for the Lys side chain.
  • Tos is used to protect the guanidine group of Arg and the imidazole group of His, and the side-chain carboxyl group of Glu or Asp is protected by OBzl.
  • the resin-peptide is washed alternately with dry diethyl ether and chloroform, and the peptides are then extracted with degassed 2 N aqueous acetic acid and separated from the resin by filtration on a Hirsch funnel.
  • the peptide is purified by gel permeation followed by preparative HPLC as described in Marki et al . (1981) J. Am. Chem. Soc. 103 :3178; Rivier, et al. (1984) J. Chromatography 288 : 303-328 ; and Hoeger, et al . (1987) BioChromatography 2 . :134-142.
  • the chromatographic fractions are carefully monitored by HPLC (see below) , and only the fractions showing substantial purity are pooled.
  • the peptide is hydrolyzed in sealed evacuated tubes containing constant boiling HCl, 3 ⁇ l of thioglycol/ml and 1 nmol of Nle (as an internal standard) for 9 hours at 140 °C.
  • Amino acid analysis of the hydrolysates using a Beckman 121 amino acid analyzer to determine amino acid ratios allows confirmation that the desired peptide structure has been obtained.
  • Cyclization "traps" the bioactive conformation of the peptide by making the active conformation part of a ring system that allows it much less conformational flexibility.
  • aspartate or glutamate residues are introduced into the sequence either in place of non-essential amino acid residues or as added residues in the chain.
  • the new peptide is then subjected to electro-oxidation in which the two residues are decarboxylated, in an intramolecular Kolbe electro-oxidative coupling reaction, resulting in the joining of their respective - CH 2 groups, forming a ring as shown in Fig. 1.
  • This method has been used to make a cyclized /S-bend of the dipeptide, Pro-Gly, by placing a glutamic acid residue on the amino and carboxyl ends of this dipeptide and then performing the Kolbe electro-oxidation to form the tetra- (CH 2 ) -bridge.
  • the Pro-Gly peptide which has a variety of conformations in solution, when cyclized, was found to adopt the ⁇ -bend structure uniquely (Joran, A. , "Conformationally restricted biologically active peptides, methods for their production and uses thereof," U.S. Patent No.
  • Scheme I illustrates an exemplary result of using electrolytic decarboxylation to cyclize peptides to trap them in their active conformations.
  • Either two gultamate, two aspartate, or one glutamate and one aspartate residues are introduced in the chain represented by the wavy line.
  • a tetramethylene bridge forms cyclizing the region of the peptide that is to be held fixed in its bioactive conformation.
  • the two connected residues shown constitute the diamino suberic acid moiety. It should be noted that, in the synthesis of this new peptide, there are possibly other aspartate and glutamate amino acid residues that can undergo the oxidative decarboxylation.
  • the electrooxidative coupling reaction used to prepare the cyclic peptides of the invention can be performed in a divided or an undivided cell such as a standard glass H-cell, as described in Organic Electrochemistry (2nd Ed.) , M. Baizer and H. Lund, eds., Marcel Dekker, New York, 1983, Chap. 5, p 168.
  • the reaction can be carried out in a plate and frame flow cell as described in Technique of Electroorqanic Synthesis. Part III, N. Weinberg and B. Tilak, ed. , John Wiley & Sons, New York, 1982, Chap. Ill, p 179.
  • Cathode materials useful for the preparation of the compounds of the invention include, but are not limited to, high hydrogen overvoltage materials such as mercury, lead or cadmium.
  • Anode materials include, but are not limited to, materials such as mercury, lead, graphite, or graphite paste, which are stable under electrolysis conditions.
  • the electrooxidative coupling can occur in aqueous, or aqueous organic electrolytes, comprising solutions of Bronsted acids, such as sulfuric, fluoroboric, and trifluoroacetic acids. Any electrolyte may be selected that has sufficient acid strength to render a basic starting material protonated. A dilute solution of trifluoroacetic acid is most preferred.
  • Typical current densities are between 1 and 5000 illiamps (mA) /cm 2 , preferably between 10 and 100 mA/cm 2 .
  • the reaction is preferably carried out at a temperature in the range of about 0 °C to 37 °C, more preferably about 10 °C.
  • a standard glass H-cell 200 ml volume, glass frit separator was equipped with a mercury pool cathode 12 cm 2 area) , a magnetic stirrer, and a platinum foil anode.
  • the cell reservoir was filled with 40 mM trifluoroacetic acid (110 ml) and placed in a water bath maintained at 10 °C.
  • the catholyte was purged with nitrogen.
  • the starting peptide (20 mg) was added to the catholyte and constant current electrolysis was initiated at 0.1 A.
  • the reaction progress was followed by HPLC and after passage of 1,060 coulombs, all the substrate had been consumed and the electrolysis was terminated.
  • the catholyte was recovered and adjusted to pH 8 with NaOH.
  • the pH-adjusted catholyte was extracted with chloroform (2 times 70 ml) .
  • the extract was freeze dried and the resultant powdery material extracted with acetonitrile (HPLC grade) .
  • Buffer A is an aqueous 0.1% (vol/vol) trifluoroacetic acid solution (1.0 ml of TFA per 1000 Ml solution) ; Buffer B is 100% acetonitrile. The determination is run at room temperature with a gradient from 15.5% Buffer B to 75% Buffer B over a 30 min. The flow rate is 2.2 ml per minute, and the retention time is 25.0 min.
  • the structure was confirmed by 300 MHz X H NMR, 13 C NMR, and electrospray mass spectroscopy.
  • the amounts of the reactants and the conditions required to facilitate reaction and encourage efficient completion of the aforementioned Examples may vary widely. However, in general, the amounts of material employed to induce reaction in the processes discussed above will be substantially stoichiometric, unless otherwise specified. In the following examples, reaction concentrations are generally held at 0.1 M for the reactants, unless a higher concentration or dilution would be particularly useful for influencing the direction of a specific reaction. In practice, the amounts used will depend upon variations in reaction conditions and the nature of the reactants as readily apparent to one of ordinary skill in the art .
  • the desired products may be isolated from the reaction mixture by crystallization.
  • chromatographic techniques including, but not limited to, normal phase, reverse phase, ion-exchange, affinity, or gel permeation, may be employed, as well as electrophoresis or extraction or other means.
  • test peptides, cyclized peptides and/or peptidomimetics are injected into immature oocytes at various doses.
  • the oocytes are co-injected with recombinant transforming ras p21 obtained from the National Cancer Institute of Japan.
  • the oncogenic ras p21 can be prepared by the ordinary skilled artisan without the expense of undue experimentation as described in Chung et al . (1991) supra and in Chung et al . (1992) Exp. Cell . Res .
  • the maturation of the oocytes is evaluated microscopically at low power (20 X) , using a Nikon Diaphot microscope, for example. Percent inhibition is calculated based on comparisons with oocytes which are injected with 0.05 mg/ml oncogenic ras p21. The following results were obtained using a dose of each peptide equivalent to an internal oocyte concentration of 50 nM:
  • the peptidomimetics and cyclic peptides of the present invention will be similarly effective in inhibiting oocyte maturation in response to the oncogenic ras p21 protein, and in inhibiting oncogenesis.
  • a representative peptidomimetic of the present invention is synthesized according to Scheme II, as further described hereinbelow.
  • R' tBiiQ-CCH-C O
  • Steroid 1 was commercially available, and used without further purification.
  • Mono-t-butylmalonate was prepared by literature methods [Brunwin, D.M.; et al. (1971) J. Chem . Soc . C. 3756] .
  • THF was distilled from sodium/benzophenone under Ar.
  • Methylene chloride and ethanol (absolute) were distilled from CaH2 under Ar.
  • Benzylchloroformate and n-propyl alcohol are commercially available, and used without further purification or drying. All reactions performed under an atmosphere of Ar unless otherwise noted.
  • the quenched reaction was extracted twice with CHCI 3 (50 mL) , and the extracts were washed with brine, dried over Na 2 SO Reference, filtered, and concentrated.
  • the yield of a vanilla colored solid was 723 mg.
  • the material was purified by flash chromatrography on silica in CHCl 3 /MeOH (3:2) to give 577 mg (79%) of a vanilla colored solid.
  • the product can be recrystallized from EtOAc.
  • N- (benzyloxy carbonyl) -5- ⁇ -6- ⁇ -amino-3-j3-hydroxycholestane (4) The amine 3 (465 mg. 1.15 mmol) and anhydrous K 2 C0 3 (457 mg, 3.3 mmol) were stirred in dry THF (4 mL) , and cooled to 0°C in an ice-water bath. Benzyl-chloroformate (0.16 mL, 1.15 mmol) was added dropwise, and the reaction was stirred at 0°C for 15 min, then at room temperature for 6h. The reaction was diluted with water and poured into saturated ⁇ aHC0 3 (15 mL) . A thick, flocculent white precipitate formed.
  • the aqueous mixture was extracted with CHC1 3 (20 mL) , and the aqueous layer was saturated with NaCl and extracted with CHC1 3 (5x20 mL) .
  • the organic extracts were washed with brine (40 mL) , dried over Na 2 S0 4 , filtered, and concentrated.
  • the yield of crude material was 649 mg of a tan solid.
  • the material was flash chromatographed on silica in Hex/EtOAc (1:1) , (using a little CHC1 3 to help dissolve material) to give 546 mg (88%) of a white solid.
  • N- (benzyloxy carbonyl) -5- -6-or-aunino-3-3-mono-t-butyl malonyl cholestane (5) The amine 4 (144.4 mg, 0.27 mmol), mono-t-butyl malonate (107.7 mg, 0.67 mmol) , and DMAP were dissolved in dry CH 2 C1 2 (1.2 mL) , and stirred efficiently at room temperature. DCC (67.4 dmg, 0.33 mmol) was added in one portion under a stream of Ar. The reaction was stirred at room temperature for 23h, then diluted with ether, and filtered through a pad of Celite (a white solid remains on the pad) .
  • N-CBZ group of 5 was cleanly removed by hydrogenolysis using 10% palladium on carbon under a hydrdogen atmosphere to give 6 in 93% yield.
  • Trials employing 1,4-cyclohexadiene as the hydrogen source [Felix et al . (1978) J " . Org. Chem . 4 . 3:4194] produced 6 in only 23% yield, even when a large excess of the diene was used. Longer reaction time did not improve the yield.
  • 5-a-6-a-amino-3-3-mono-t-butyl malonyl cholestane (6) 5 (200.7 mg, 0.3 mmol) and 10% palladium on carbon (194 mg) were stirred vigorously in abs.
  • MOLECULE TYPE peptide
  • HYPOTHETICAL NO
  • SEQUENCE DESCRIPTION SEQ ID NO:l: lie Lys Arg Val Lys Asp 1 5
  • MOLECULE TYPE protein
  • HYPOTHETICAL NO
  • Val Phe Ala lie Asn Asn Thr Lys Ser Phe Glu Asp lie His Gin Tyr 85 90 95
  • ORGANISM Arabidopsis thaliana
  • MOLECULE TYPE protein
  • HYPOTHETICAL NO
  • ORGANISM Arabidopsis thaliana
  • ORGANISM Arabidopsis thaliana
  • xi SEQUENCE DESCRIPTION: SEQ ID NO:12:
  • ORGANISM Discopyge ommata
  • ORGANISM Oryctolagus cuniculus
  • ORGANISM Caenorhabditis elegans
  • xi SEQUENCE DESCRIPTION: SEQ ID NO:21:
  • ORGANISM Dictyosteliu discoideum
  • ORGANISM Dictyostelium discoideum
  • ORGANISM Caenorhabditis elegans
  • ORGANISM Dictyostelium discoideum
  • ORGANISM Dictyostelium discoideum
  • ORGANISM Dictyostelium discoideum
  • Lys Lys Met Ser Glu Asn Lys Asp Lys Asn Gly Lys Lys Ser Ser Lys 180 185 190
  • ORGANISM Discopyge ommata

Abstract

The present invention provides peptides, cyclized peptides and peptidomimetics which inhibit the oncogenic and/or transforming activity of the p21 ras protein, pharmaceutical compositions containing at least one of the ras-inhibiting peptides, cyclized peptides and peptidomimetics, and methods for inhibiting the ras-mediated oncogenic and/or transformation process in mammalian cells or tissues.

Description

PEPTIDES AND PEPTIDOMIMETICS INHIBITING THE ONCOGENIC ACTION
OF P21 RAS
Field of the Invention
This invention relates to peptides effective in inhibiting oncogenesis, particularly as related to inhibition of p21 ras and adenocarcinomas of the colon, pancreatic carcinomas, neuroblastomas, and other cancers which express the transformed sequence of the ras gene product .
Background of the Invention ras protooncogenes are activated by characteristic point mutations in a wide variety of malignancies. The expressed p21 ras proteins are oncogenic by virtue of single substituted amino acids, usually at position 12 or 61 of the 189-residue p21 ras gene product, ras proteins act as membrane-associated molecular switches that bind GTP and GDP and slowly hydrolyze GTP to GDP.
Mutations in ras are associated with the vast majority of adenocarcinomas of the colon. Cancer of the colon is a highly treatable and often curable disease when it remains localized to the bowel. It is the second most frequently diagnosed malignancy in the United States as well as the second most common cause of cancer death. Surgery is the primary treatment and results in cure in approximately 50% of patients. Adenocarcinoma is the primary lesion in the majority of cases. Recurrence following surgery is a major problem and often is the ultimate cause of death. The prognosis for colon cancer patients is clearly related to the degree of penetration of the tumor through the bowel wall and the presence or absence of nodal involvement. For locally advanced disease, the role of radiation therapy in colon cancer is under clinical evaluation. There is no standard therapy for advanced colon cancer and no evidence that chemotherapy improves survival, although short-term palliation may be achieved in approximately 10-20% of patients. Pancreatic carcinoma has a high incidence of κ-ras mutations. Mutated K-ras sequences which can be identified by polymerase chain reaction utilizing allele-specific primers can even be found in the plasma or serum from patients with pancreatic carcinoma. The c-Ki-ras oncogene is activated by point mutations involving codon 12 in 72%-100% of primary pancreatic adenocarcinomas, but the gene is not activated in nonneoplastic tissues. Cancer of the exocrine pancreas is rarely curable. The highest cure rate (4%-12%) occurs if the tumor is truly localized to the pancreas. Unfortunately, this stage of disease accounts for fewer than 20% of cases and, even with surgical resection, results in little more than a 5% 5-year survival rate. For small cancers (less than 2 cm) in the head of the pancreas with no lymph node metastases and no extension beyond the "capsule" of the pancreas, the survival rate following resection of the head of the pancreas approaches 20%. Overall survival rate of all stages is less than 2% at 5 years with most patients dying within one year. Worldwide, very few patients with cancers of the pancreatic tail or uncinate process have been cured.
Lung cancers also frequently involve ras mutations. Point mutations in codon 12 of the K-ras protooncogene occur more frequently in lung adenocarcinomas from smokers (30%) than they do in lung adenocarcinomas from nonsmokers (7%) , suggesting that smoking is an important factor in the induction of these mutations. The ras oncogene may thus be a specific target of the mutagenic activity of tobacco smoke, and suggest that DNA alterations at this site can occur early and irreversibly during the development of adenocarcinomas of the lung. Mutations in the ras protooncogenes are the most frequently observed molecular alteration in acute myeloid leukemia (AMD . Whether ras mutations occur as late or relatively early events in the multistep process of myeloid transformation, remains an open question. There is significant evidence that the ras oncogene plays a role in experimental mammary carcinogenesis; the evidence in human breast cancer, however, is more limited. Similarly, there is significant evidence that the ras oncogene plays a role in nitrosoamine-induced esophageal tumors in rats, but in human esophageal cancers ras gene mutations are more rarely found. However, it is probable that there is a significant role of mutated ras genes in both cell proliferation and malignant transformation of human esophageal cells.
Certain human neuroblastomas also show a high incidence of oncogenic ras mutations. Indeed, one study suggested that expressions of the oncogene N-myc and p21 together as detected by immunohistochemical staining could be among the most reliable prognostic indicators in neuroblastoma patients.
The ras proteins are key regulators of the growth of eukaryotic cells. Some of the direct targets are unknown. These target proteins include raf-1, gap, phosphatidylinositol-3- hydroxykinase and, very recently, two nuclear proteins, C-JUN and its kinase (JNK) . The three-dimensional x-ray crystal structure for a ras-related protein bound to a domain of raf-1 has been elucidated. The ras-related protein (_____________) binds to raf directly, utilizing residues contained in a sequence involving amino acids 35-37. All of the contact residues in the ras- related protein are homologous to those in the corresponding segment of ras-p-21. One of the inventors has shown that the p- 21 ras protein (35-47 segment) selectively inhibits the mitogenic effects of oncogenic ras-p-21. In addition to its role as an oncogene, the activation of ras proteins is a key step in the signal transduction pathways triggered by ligand-bound cell surface receptors, such as the insulin receptor.
The classical target of the ras protein is the GTPase activating protein GAP. This target protein is thought to play an essential role in the regulation of ras activity by increasing the GTPase activity of wild type, but not transformed ras . On the other hand, there is a considerable superfamily of these GAP- related proteins, which includes pl20-GAP. Other target proteins besides mammalian gap itself include (1) IRA1 and IRA2 , the functional equivalents of GAP in yeast. They regulate the ras-cyclic AMP pathway, controlling cell growth; (2) sari, the fission yeast protein that regulates rasl in that organism; (3) BUD2, a yeast protein that activates BUD1/RSR1 which participates in the regulation of bud-site selection; (4) Human neurofibromitosis (gene NF1) . NF1 is associated with type 1 neurofibromatosis, one of the most frequently inherited genetic diseases characterized, in part, by multiple neural tumors. NF1 has been shown genetically and biochemically to interact with and stimulate the GTPase activity of ras,- (5) Drosophila Gapl, which acts as a negative regulator of signalling by the Sevenless (SOS) receptor tyrosine kinase involved in eye development. Human SOSl and SOS2 genes have also been recently identified which encode proteins that control GDP-->GTP exchange on ras proteins and are involved in signal transduction by tyrosine kinase receptors. In si tu hybridization shows that SOSl maps to 2p22-->pl6 and SOS2 to 14q21-->q22 in the human genome.
Another important target of ras is raf . The protein encoded by the c-raf-1 protooncogene is thought to function downstream of p21 ras because disruption of raf blocks signalling by ras in a number of systems. A highly-conserved 81 residue region of the N-terminus of raf protein has been to be shown to be critical as the ras protein interaction region. Importantly, the raf gene product interacts with both wild-type and activated ras protein. In one study, approximately 50% of the clones identified as interacting with ras were encoded portions of the c-raf and A-raf serine/threonine kinases. Thus, ras and the N-terminal region of raf protein associate directly in vi tro and this interaction is dependent on GTP bound to ras .
Within the superfamily of ras-related GTP-binding proteins, only the ras protein itself has been shown to act as an oncogenic protein. Many other proteins, however, have substantial amino acid homology to ras. This ras superfamily of GTP-binding proteins (> 50 members) regulates a diverse spectrum of intracellular processes. These include cellular proliferation and differentiation, intracellular vesicular trafficking, cytoskeletal control, NADPH oxidase function, as well as others. Some of these homologs may have biological activities which are related to ras. For example, rhoA encodes a ras-related GTP-binding protein that was thought principally to play a role in cytoskeletal organization. Recent evidence, however, has suggested both that rhoA could act either as a dominant oncogene, since transfection of both normal and activated rho genes confer a transformed phenotype on fibroblast cells in culture, or as a recessive tumor suppressor gene, by virtue, in part, of its chromosomal location at 3p21, a site deleted in many human malignancies. Thus, it is important to consider these ras homologs as potentially involved in cell growth and transformation.
Azatyrosine strongly inhibits oncogenic ras-p-21. This small molecule induces the rrg gene, which encodes a proteinase sequence showing 90% amino acid sequence identity to lysyl oxidase. To acquire transforming potential, the precursor of the ras oncoprotein must undergo farnesylation or similar modification of the cysteine residue located in a carboxyl-terminal tetrapeptide. These C-terminal lipid modifications are essential for the interaction of ras-related proteins with membranes. While all ras proteins are farnesylated and some palmitoylated, the majority of other ras-related proteins are geranylgeranylated. Thus selective peptide and peptidomimetic inhibitors of ras lipidation have found potential utility as anti-oncogenic agents. In view of the foregoing, there is there a longfelt need in the art for agents which inhibit the transforming ability of ras . As described above, selective peptide and peptidomimetic inhibitors or ras lipidation have found potential utility as anti-oncogenic agents (Kohl et al . (1993) Science 26.0: 1934-1937; James et al . (1993) Science 260 :1937-1942) . Similarly, FR patents 2694296 and 2690162 teach that peptides derived from the GAP protein may serve to inhibit ras . However, neither '694296 nor '690162 describes peptides derived from the ras protein itself. EP 203587 describes new ras oncogene polypeptides which are used for producing antibodies for immunogenic assays. However, these sequences are derived from ras and its homologs in the carboxyl terminal domain (residues 170 - 189 in SEQ ID NO:5) and are thus physically distant from and completely unrelated to any sequences claimed herein. Furthermore, these sequences were claimed for the production of antibodies, preferably by linking to an immunogenic carrier, and a claim for direct therapeutic application was not made.
Thus, peptides constructed from ras and its homologs for therapeutic application, namely by interfering with downstream or upstream actions of r^s_ itself, are useful. Furthermore, the method of identification of said peptides utilizing calculational approaches is believed novel and has unexpectedly led us to these cyclic peptides and peptidomimetics disclosed herein.
Summary of the Invention
The present invention provides peptides, cyclized peptides and peptidomimetics capable of inhibiting the oncogenic action of p21 ras . The oncogenic ras-inhibiting cyclized peptides correspond to domains of the oncogenic ras protein which are most flexible and important in "interacting with target proteins upstream and downstream from ras. The peptidomimetics are obtained by molecular modeling, including the structural minimization techniques of molecular dynamics.
The peptides are designated by the formulas: Val-Val-lie, Lys-Arg-Val, Ile-Lys-Arg-Val-Lys-Asp (SEQ ID NO:1), Lys-Cys-Asp- Leu-Ala (SEQ ID NO:2) , Cys-Asp-Leu-Ala-Ala-Arg-Thr (SEQ ID NO:3) , Asp-Leu-Ala-Ala (SEQ ID NO:4) or physiologically acceptable salts of the foregoing peptides.
Also provided in the present invention are cyclic analogues of the above peptides and certain peptides and cyclic peptides:
cyclo [- R(l) R(2) Thr lie Glu Asp Ser Tyr Arg Lys Gin Val Val He Asp R(3) R(4)-] (I) ;
cyclo [-R(l) R(2) Val Val He R(3) R(4)-] (II) ;
cyclo [-R(l) R(2) Tyr Arg Glu Gin He Lys Arg Val Lys Asp Ser Asp Asp Val Pro R(3) R(4)-] (III) ; cyclo [-R(l) R(2) Lys Arg Val R(3) R(4)-] (IV) ;
cyclo [-R(l) R(2) He Lys Arg Val Lys Asp R(3) R(4)-] (V) ;
cyclo [-R(l) R(2) Gly Asn Lys Cys Asp Leu Ala Ala Arg Thr Val Glu R(3) R(4) -] (VI) ;
cyclo [-R(l) R(2) Lys Cys Asp Leu Ala R(3) R(4)-] (VII) ;
cyclo [-R(l) R(2) Cys Asp Leu Ala Ala Arg Thr R(3) R(4)] (VIII) ;
cyclo [-R(l) R(2) Asp Leu Ala Ala R(3) R(4)-] (IX) ; and
NH2
I 0 Asp
II I
H2N-Thr-He-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-NH-CH2-C-Val-NH-CH (X)
I I CH2 CH- 2
or physiologically acceptable salts thereof.
In cyclized peptide formulas (I) -(IX) , R(l) R(2) , R(3) and R(4) represent, in the most general case, any amino acid which can serve as an amino acid residue linker. Amino acid residue linkers are usually at least one residue and can be most often two to four residues, more often 1 to 10 residues, both ranges being inclusive. Typical amino acid residues useful for linking are tyrosine, cysteine, lysine, and glutamic and aspartic acid. Most preferably [R(D , R(2)] and [R(3) , R(4)] are each independently selected from either the group consisting of Glu, Gin, Asp, Asn or from the group consisting of Lys, Arg, Orn.
The symbol - represents a bond between the carboxyl and amino termini by which R(l) and R(4) can be interconnected to each other via an lower alkenyl or lower alkynyl group, but most preferably by a branched or unbranched methylene bridge of type --(CH2)m --or -- (CH2)m--M-- (CH2)m,-- . In such an moiety, m and m' are integers from 1 to 6, inclusive, and preferably from 1 to , inclusive; and M is NH, N[R(5)] , 0, S or CH-R(5), wherein R(5) is lower alkyl, cycloalkyl or aryl and is preferably methyl, ethyl, propyl, phenyl, X-phenyl, or heterocyclic, wherein X is Cl-, CF3-, F-, substituted at the o-, m- , or p- positions on the phenyl group M can contain a part of another diamino acid within the same peptide, e.g., the omega amino group of the one residue can be so linked to such an unnatural amino acid residue in a terminal residue. Furthermore, any amino acid in the sequences provided hereinabove may be replaced with its D-analogue, with the proviso that not more than 50% of the total amino acids are so replaced. Similarly, a homologous conservative substitution for any amino acid is within the bounds of the present invention provided that substitution does not eliminate the oncogenic ras p21-inhibiting activity. Thus, depending on the applications for which the peptides according to the invention are intended, it is also possible to envisage intercalating between several amino acids, or even between all the amino acids, of the peptides defined above, dextrorotatory amino acids, and in particular dextrorotatory phenylalanine or dextrorotatory tryptophan, capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity. Another modification in this sense consists in replacing certain amino acids, for example of the isoleucine type, by leucine.
In addition, a subject polypeptide can differ, unless otherwise specified, from the natural sequences shown above by the sequence being modified by terminal -NH2 acylation, e.g., acetylation, or by terminal-carboxylamidation, e.g., with ammonia, alkylamines, and the like.
This invention further relates to peptidomimetics which model the critical semi-extended conformation of at least one peptide of or cyclic peptide of the present invention, exemplified by the compounds of Structure 1:
Figure imgf000011_0001
STRUCTURE 1
wherein the sidechain R attached at the carbon atom numbered 6 on the sterol nucleus can be NH-CH2-CH2NH3 +, alkylamino, arylamino, or aralkylamino group, and wherein the sidechain attached at the carbon number 3 can be replaced with -0-C(=0)- - (CH2)m--COOH, where m is an integer from 1 to 6, inclusive, preferably from 1 to 3, inclusive, and more preferably 2, and one of X and Y at each position independently, can be one H, a small alkyl group of Cx to C3, preferably C1 ; a halogen, preferably F, or an amino group where the other of one of X and Y is H. Preferably, each of X and Y is H.
12
17
11 ' 13-
16 1 6 14 /
! I I
/ I
CzzO NH / I
" OOC- CH- CH2
CH2
+NH3
STRUCTURE 2
An exemplary compound falling within Structure 1 is 3 malonoxy-6- (2-aminoethyl) aminocyclopentanoperhydrophenanthrene (Structure 2) .
Detailed Description of the Invention
The natural sequence of the human oncogenic ras p21 is given in SEQ ID NO:5. The crystal X-ray structure has been determined at high resolution for that portion of the human ras protein corresponding to residues 1 to 166 of SEQ ID NO: 5.
The regions of the p21 protein that are the most likely to change their conformations upon activation of the protein, e.g. by oncogenic amino acid substitutions have been computed using two different methods. Both methods are based on the principle that the linear sequence of amino acids in a protein determines its unique three-dimensional structure. Given an amino acid sequence of a polypeptide or protein, therefore, it should be possible to predict its three-dimensional structure. This task can be accomplished by using the principle that the observed three-dimensional structure of a protein is the one of lowest free energy. There are a vast number of possible structures a given polypeptide chain can adopt, but essentially only one of these is observed. To allow folding to occur, therefore, the interatomic interactions in the protein chain must greatly stabilize its final folded form, i.e., lower its conformational energy substantially with respect to that of any other competing structure. Thus, to compute the lowest energy form of a protein, it is necessary to be able first to compute the conformational energy of a given conformation of the protein and then, second, to generate its low energy conformations, or a representative sampling of them. The structure of lowest conformational energy so computed is then predicted to be the observed structure of the protein. This structure may be the one determined by x-ray crystallography or by 2- or 3-dimensional nuclear magnetic resonance (NMR) techniques.
A set of potential energy functions, in the computer program ECEPP (Empirical Conformational Energies of Peptides Program) , have been developed that accurately compute the conformational energies of given conformations of proteins. The conformational energy of a peptide can be expressed in Equation 1.
Figure imgf000013_0001
where Etot is the total conformational energy of the protein, the Q's are the charges on the ith and jth atoms; R1 is the distance between the ith and jth atoms, D is the dielectric constant, e13 and 1D are the lowest non-bonded (Lennard-Jones) energy and the distance at this lowest energy between atoms i and j in the protein; Ak is the torsional barrier to rotation around specific bonds; θk is the kch dihedral angle in the protein; n is a degeneracy factor, i.e., 3 for single bonds and 2 for double bonds; and the sign in the last summation term is positive for single bonds and negative for double bonds such as occur in the peptide bond units.
This equation shows the total conformational energy as the sum of three terms: the pairwise electrostatic interactions between the individual atoms of a protein, each of which has a partial charge, (first sum) ; a non-bonded energy term (second term) that consists of an attractive term that varies as the inverse sixth power (tenth power for hydrogen-bonding atoms) of the distance between the atoms (from an induced dipole-induced dipole interaction term) and a repulsive term, from the overlap of electron shells, that varies as the inverse twelfth power of the interatomic distance; and finally a torsional term (third sum) that depends upon the bonds about which rotation takes place. All of the constants in these terms have been determined from experimental crystal packing data and reproduce the lattice constants of all of the crystal structures of small molecules to which they have been applied and, where measured, the sublimation energies of these crystals These potential functions have been used to compute the low energy minima for single terminally blocked amino acid residues, simple peptides, oligopeptides, polypeptides, and proteins with excellent agreement between the lowest energy predicted structures and the structures determined experimentally. These potentials have therefore been well- tested, are based on experimental data, and have proved to be reliable in prediction of structure from sequence.
These potential functions have been used to compute the average structure for the ras-p-21 protein in its normal and in its oncogenic form using the perturbation method called the electrostatically-driven Monte Carlo method (EDMC) . Specific regions of the oncogenic p21 protein undergo large conformational changes compared with the structure of the normal, inactive protein. One of these regions has been found to be residues 35- 47. All of the segments that change conformation in the oncogenic protein were found to be the most flexible in the normal, inactive protein.
Of considerable significance has been the finding that a completely different method, viz. molecular dynamics, based upon a completely different set of potential functions, i.e. the program DISCOVER, yields identical results for the p21 protein.
Molecular dynamics is based on the principle that the positions of the atoms of a molecule can be predicted as a function of time by solving Newton's equations of motion for the molecule. The force on the molecule is the negative of the first derivative of the potential function with respect to the coordinates of each of the atoms. Newton's equations of motion are then integrated, using the Verlet algorithm, over a trajectory such that the low energy regions around the starting structure are computed. The trajectories are computed over time intervals such that the total energy converges to a low, constant value. The structures whose energies have converged are then used to compute an average structure. Comparison of the coordinates of the atoms of this average structure with those of the starting structure reveals regions of the protein whose conformations may change significantly. Furthermore, if the variance of the coordinates of regions of the low energy structures from the corresponding coordinates of the average structure are high, these regions can be identified as being flexible, i.e., are the ones most likely to be parts of effector domains. Within this algorithm, for the p21 protein, up to 2000 water molecules have been generated around the protein in the molecular dynamics simulations performed thus far.
Using these novel calculational approaches, the present inventors have identified important peptide regions of the protein that are involved in the signal transduction process, and these peptides can be used to design anti-cancer agents, as taught herein. We have found that most particularly the 35-47, 96-110 and 115-126 peptides have strong and specific anti- oncogenic p21 activity. Even more particularly, we found that these domains contain unique extended structures and/or short beta-bend structures which are hypothesized to account in large part for their biological uniqueness. This suggested that cyclization of the peptide structures to force the beta-bend conformation in place would serve to enhance therapeutic activity.
The results of these studies indicate that a domain of particular interest is the domain from residues 35 through 47 of SEQ ID NO:5, i.e., Thr-Ile-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val- Ile-Asp (SEQ ID NO:6) , of even more particular interest the peptide corresponding to residues 44 to 46 in SEQ ID N0:5, i.e., Val-Val-He, of still more interest is the sequence from residues 96 to 110 of SEQ ID NO:5, i.e., Tyr-Arg-Glu-Gln-He-Lys-Arg-Val- Lys-Asp-Ser-Asp-Asp-Val-Pro (SEQ ID NO:7) , of even more particular interest is the sequence from residues 101-103 in SEQ ID NO:5, i.e., Lys-Arg-Val; and the sequence corresponding to residues 100 to 105 in SEQ ID NO:5, i.e., Ile-Lys-Arg-Val-Lys-Asp (SEQ ID NO:l) ; the sequence corresponding to residues 115 to 126 of SEQ ID NO:5, i.e., Gly-Asn-Lys-Cys-Asp-Leu-Ala-Ala-Arg-Thr- Val-Glu (SEQ ID NO: 8) ; and most particularly the sequence corresponding to residues 117 to 121 of SEQ ID NO: 5, i.e, Lys- Cys-Asp-Leu-Ala (SEQ ID NO:2) and the sequence corresponding to residues 118 to 124 of SEQ ID NO: 5, i.e., Cys-Asp-Leu-Ala-Ala- Arg-Thr (SEQ ID NO: 9) ; and the sequence corresponding to residues 119 to 122 of SEQ ID NO:5, i.e., Asp-Leu-Ala-Ala (SEQ ID NO:4) .
Additional sequences homologous to the various preferred sequences recited hereinabove can be derived by one skilled in the art from the sequences of closely related ras proteins . Such sequences may possess enhanced therapeutic activity. Nonlimiting examples of such proteins closely related to the ras gene product which represent the parent sequences having identical or nearly identical three dimensional structures and from which homologs of the sequences given in the preceding paragraph can be derived by one normally skilled in the art are:
ras-related protein Ara-3 [Arabidopsis thaliana (mouse ear cress) ] (SEQ ID NO: 10) ;
ras-related protein Ara-2 [A^. thaliana] SEQ ID NO:11;
ras-related protein Ara-1 [A _ thaliana] SEQ ID NO: 12;
ras-related protein OraB-1 fDiscopyge ommata (electric ray) ] SEQ ID NO: 13;
ras-related protein Rab-IA [Lvmnea stagnalis (great pond snail) ] SEQ ID NO:14; ras-related protein Rab-2 [Homo sapiens (human) ] SEQ ID NO: 15;
ras-related protein Rab-2 [L^_ stagnalisl SEQ ID NO: 16;
ras-related protein Rab-2 [Orvctolagus cuniculus (rabbit)] SEQ ID NO:17;
ras-related protein Rab-2 [Rattus norvegicus (rat)] SEQ ID NO:18;
ras-related protein Rab-3 [Drosophila melanogaster (fruitfly)] SEQ ID NO:19;
ras-related protein Rab-4 [R^_ norvegicusl SEQ ID NO:20;
ras-related protein Rab-6 [Caenorhabditis elegans] SEQ ID NO:21;
ras-related protein Rab-6 [IL. sapiens] SEQ ID NO:22;
ras-related protein Rab-7 [Canis familiaris (dog) ] SEQ ID NO:23;
ras-related protein Rab-7 [Dictvostelium discoideum (slime mold) ] SEQ ID NO:24;
ras-related protein Rab-8 [C_^ familiaris! SEQ ID NO:25;
ras-related protein RabC [EL. discoideum] SEQ ID NO:26;
ras-related protein Rac-1 [C_^ elegansl SEQ ID NO:27;
ras-related protein Rac-IA [D^. discoideum! SEQ ID NO:28;
ras-related protein RacB [D^ discoideum] SEQ ID NO:29; ras-related protein RacC [D__. discoideum] SEQ ID NO:30;
ras-related protein Ral-A [IL. sapiens] SEQ ID NO:31;
ras-related protein Ral-B [IL. sapiens] SEQ ID NO:32;
ras-related protein O-Ral [I _ ommatal SEQ ID NO:33;
ras-related protein Ora-1 [D^. ommatal SEQ ID NO:34;
ras-related protein Ora-2 [p__. ommatal SEQ ID NO:35;
ras-related protein Ora-3 [IX. ommatal SEQ ID NO:36;
ras-related protein Rap-1 [IK. discoideum] SEQ ID NO:37;
ras-related protein Rap-2A [H. sapiens] SEQ ID NO:38;
ras-related protein Rap-2B [IK. sapiens] SEQ ID NO:39;
ras-related protein O-KREV [IX. ommata] SEQ ID NO:40;
ras-related protein Rap-IA [H__ sapiens] SEQ ID NO:41;
ras-related protein Rap-IB [IL. sapiens] SEQ ID NO:42;
ras-like protein GNROR3 [EL. melanogaster] SEQ ID NO:43;
ras-like protein rasA [P. discoideum] SEQ ID NO:44;
ras-like protein rasB [P. discoideum] SEQ ID NO:45;
ras-like protein rasC [D. discoideum] SEQ ID NO:46;
ras-like protein rasG [D. discoideum] SEQ IP NO:47;
ras-like protein F54C8.5 [CL. elegansl SEQ ID NO:48; ras.-like protein CC-ras [Coprinus cinereus (inky cap fungus) ]
SEQ ID NO:49;
ras-like protein [Geodia cydonium (sponge)] SEQ ID NO:50;
ras-related protein Rab-10 [CK. familiaris] SEQ ID NO:51;
ras-related protein Rab-11 [H__ sapiens! SEQ ID NO:52.
In addition, as described hereinabove, the therapeutic activity of these sequences is enhanced by cyclization. Cyclization forces and maintains the conformations of these peptides in unique structures like beta-bends. The following are representative, nonlimiting examples of cyclized peptides useful for inhibiting the oncogenic activity of the ras protein, said peptides having formulas as given below:
cyclo [- R(l) R(2) THR ILE GLU ASP SER TYR ARG LYS GLN VAL VAL ILE ASP R(3) R(4)-] (I)
cyclo [-R(l) R(2) VAL VAL ILE R(3) R{4)-] (II)
cyclo [-R(l) R(2) TYR ARG GLU GLN ILE LYS ARG VAL LYS ASP SER ASP ASP VAL PRO R(3) R(4)-] (III)
cyclo [-R(l) R(2) LYS ARG VAL R(3) R(4)-] (IV)
cyclo [-R(l) R(2) ILE LYS ARG VAL LYS ASP R{3) R(4)-] (V)
cyclo [-R(l) R{2) GLY ASN LYS CYS ASP LEU ALA ALA ARG THR VAL GLU R(3) R(4)-] (VI)
cyclo t-R(l) R(2) LYS CYS ASP LEU ALA R(3) R(4)-] (VII)
cyclo [-R(l) R(2) CYS ASP LEU ALA ALA ARG THR R (3) R(4)-] (VIII) cyclo [ -R ( l ) R ( 2 ) ASP LEU ALA ALA R ( 3 ) R ( 4 ) - ] ( IX) ; and
NH2
I 0 Asp II I
H2N-Thr- Ile -Glu-Asp-Ser-Tyr-Arg-Lys -Gln-NH-CH2-C-Val -NH-CH (X)
I I
CH2 CH2
or a physiologically acceptable salt thereof.
In the aforementioned cyclized peptide formulas I-IX, R(l) R(2) , R(3) and R(4) represent, in the most general case, any amino acid, such that they serve as amino acid residue linkers. Amino acid residue linkers are usually at least one residue and can be most often two to four residues, more often 1 to 10 residues. Typical amino acid residues used for linking are tyrosine, cysteine, lysine, glutamic and aspartic acid. Most preferably [R(D, R(2)] and [R(3) , R(4)] independently are selected from either the groups [Glu, Gin, Asp, Asn] or [Lys, Arg, Orn] . The term - represents a bond between the carboxyl and amino termini by which R(l) and R(4) can be interconnected to each other via an lower alkyl, alkenyl or lower alkynyl group, but most preferably by a branched or unbranched methylene bridge of type --(CH2)m --or -- (CH2)m--M-- (CH2) m. -- . In such an moiety, m and m' are integers from 1 to 6, inclusive, and preferably from 1 to 3, inclusive; and M is NH, N[R(5)] , O, S or CH-R(5) , wherein R(5) is lower alkyl, cycloalkyl or aryl and is preferably methyl, ethyl, propyl, phenyl, X-phenyl, or heterocyclic, wherein X is C1-, CF3, F- , substituted at the o- , m- , or p- positions on the phenyl group. M can contain a part of another diamino acid within the same peptide, e.g., the omega amino group of the one residue can be so linked to such an unnatural amino acid residue in a terminal residue.
Furthermore, any amino acid in the cyclized peptide sequences (I)-(X) recited herein may be replaced with its D- analogue, insofar as not more than 50% of the total amino acids are so replaced. Similarly, a homologous conservative substitution for any amino acid is within the bounds of the present invention. Conservative substitutions include Glu for Asp, Gin for Asn and Val for He, among others, as well-known to the art. Depending on the applications for which the peptides according to the invention are intended, it is also possible to intercalate between several amino acids, or even between all the amino acids, of the peptides defined above, dextrorotatory amino acids, and in particular dextrorotatory phenylalanine or dextrorotatory tryptophan, capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity. Another modification in this sense consists in replacing certain amino acids, for example of the proline type, by D-tryptophan. In addition, a subject polypeptide can differ, unless otherwise specified, from any of the natural sequences shown herein above by the sequence being modified by terminal -NH2 acylation, e.g., acetylation or by terminal-carboxylamidation, e.g., with ammonia, alkylamines, and the like. The placement of hydrophobic amino acid residues is highly dependent on the peptide sequence. For example, for the 35-47 peptide sequence, corresponding to amino acids 35-47 of SEQ ID NO: 5, there is a distinct hydrophobic region for the amino acid residues corresponding to amino acids 44-46 of SEQ ID NO:5. The bridge in Compound (X) occurs at what corresponds in structure to amino acids 44-46 in SEQ ID NO:5. It is possible to extend this hydrophobic segment without sacrificing activity. For example, the carboxyl terminal Asp residue can be replaced with one or more hydrophobic residues such as Val or He, and the result is greater efficiency in crossing cell membranes.
Short half-lives of peptides, a major problem, can be at least partially extended by the addition of D-amino acids to either or both of the amino and carboxyl terminal ends of the peptide. These D-amino acid residues block the action of exo- proteases that degrade peptides from their amino or carboxyl ends. In addition, the cyclization of the peptide further renders the peptide less susceptible to proteolysis. Recent advances in the field of peptides have been directed towards the stabilization of these peptides against enzymatic or hydrolytic degradation. It would be extremely valuable to stabilize these peptides from degradation by proteolytic enzymes in order to improve their pharmacokinetic properties. Enhanced resistance to enzymatic degradation would increase the usefulness of these peptides as therapeutic agents. However, since they only exhibit short half lives in vivo, large amounts of such peptides must typically be administered to a subject in order to achieve the desired effect. Alternatively, smaller quantities may be prescribed to an individual, but more frequent dosages would be required to achieve the same level of potency.
It is further well-known to those normally skilled in the art that it is possible to replace peptides with peptidomimetics. Peptidomimetics are generally preferable as therapeutic agents to peptides owing to their enhanced bioavailability and relative lack of attack from proteolytic enzymes. The present inventors have used the techniques of molecular modeling supra to design a peptidomimetic which mimics the critical beta-bend aspects of the peptide corresponding in sequence to amino acids 96-110 of SEQ ID NO:5 (p21 ____£) . The bend structure occurs at amino acids 102-103 in the p21 ras protein. These residues have been implicated in the binding of ras p21 to SOS.
Peptidomimetric compounds which inhibit the oncogenic or transforming activity of the p21 ras protein are provided by the compounds of Structure I :
Figure imgf000022_0001
wherein the sidechain R attached at the carbon atom numbered 6 on the sterol nucleus can be NH-CH2-CH2NH3K alkyl amino, arylamino, or aralkylamino group, and wherein the sidechain attached at the carbon number 3 can be replaced with -0-C(=0)- - (CH2)m--COOH, where is an integer from 1 to 6, inclusive, preferably from 1 to 3, inclusive, and more preferably 2, and one of x and y at each position independently, can be one H, a small alkyl group of Cx to C3, preferably Cα; a halogen, preferably F, or an amino group where the other of one of x and y is H. Preferably, each of x and y is H.
Without wishing to be bound by any particular theory, the structure believed to be the optimally designed ras-inhibiting peptidomimetic is illustrated below in Structure II:
12
17
11 "13-
\ 16
14
O"
Figure imgf000023_0001
/ C=_0 NH
/ I
OOC- CH. CH.
I ' CH. I ' NH.
STRUCTURE 2
The instant invention comprises novel peptides of medicinal importance most particularly for the treatment of adenocarcinomas of the colon, pancreatic carcinomas, neuroblastomas, and other cancers of undefined germ cell origin which express the transformed sequence of the ras protein. These peptide sequences were unexpectedly obtained by the use of molecular dynamic simulations on ras p21 to define which domains of the protein were most flexible and were thus most important in interacting with target proteins upstream and downstream from ras . These peptides are identified by the following amino acid sequences: Thr-Ile-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val-Ile-Asp (SEQ ID NO:6) , Val-Val-He, Tyr-Arg-Glu-Gln-He-Lys-Arg-Val-Lys-Asp-Ser- Asp-Asp-Val-Pro (SEQ ID NO:7) , Lys-Arg-Val, Ile-Lys-Arg-Val-Lys- Asp (SEQ ID NO:l), Gly-Asn-Lys-Cys-Asp-Leu-Ala-Ala-Arg-Thr-Val- Glu (SEQ ID NO:8), Lys-Cys-Asp-Leu-Ala (SEQ ID NO:2) , Cys-Asp- Leu-Ala-Ala-Arg-Thr (SEQ ID NO:9), and Asp-Leu-Ala-Ala (SEQ ID NO:4) .
Including the cyclic analogues of the above peptides, namely:
cyclo [- R(l) R(2) Thr-Ile-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val- Ile-Asp-R(3) R(4) -] (I) ;
cyclo [-R(l) R(2) Val-Val-Ile-R(3) R(4)-] (II) ;
cyclo [-R(l) R(2) Tyr-Arg-Glu-Gln-Ile-Lys-Arg-Val-Lys-Asp-Ser- Asp-Asp-Val-Pro-R(3) R(4)-] (III) ;
cyclo [-R(l) R(2) Lys-Arg-Val R(3) R(4)-] (IV) ;
cyclo [-R(l) R(2) He-Lys-Arg-Val-Lys-Asp R(3) R(4)-] (V) ;
cyclo [-R(l) R(2) Gly-Asn-Lys-Cys-Asp-Leu-Ala-Ala-Arg-Thr-Val-Glu R(3) R(4)-] (VI);
cyclo [-R(l) R(2) Lys-Cys-Asp-Leu-Ala R(3) R(4)-] (VII) ;
cyclo [-R(l) R{2) Cys-Asp-Leu-Ala-Ala-Arg-Thr R(3) R(4)-] (VIII) ;
cyclo [-R(l) R(2) Asp-Leu-Ala-Ala R(3) R(4)-] Z (IX) ; and NH2 0 Asp
II I H2N-Thr- Ile -Glu-Asp-Ser-Tyr-Arg-Lys-Gln-NH-CH2 -C-Val -NH-CH (X)
I I
CH2 CH2
or a physiologically acceptable salt thereof.
Wherein for cyclized peptide formulas designated by (I) - (IX) hereinabove, R(l) R(2), R(3) and R(4) represent, in the most general case, any amino acid, such that they serve as amino acid residue linkers. Amino acid residue linkers are usually at least one residue and can be most often two to four residues, more often 1 to 10 residues. Typical amino acid residues used for linking are tyrosine, cysteine, lysine, glutamic and aspartic acid, or the like. Most preferably [R(D , R(2)] and [R(3) , R(4)] independently are selected from either the groups [Glu, Gin, Asp, Asn] or [Lys, Arg, Orn] .
The symbol - represents a bond between the carboxyl and amino termini by which R(l) and R(4) can be interconnected to each other via an lower alkenyl or lower alkynyl group, but most preferably by a branched or unbranched methylene bridge of type --(CH2)m --or -- (CH2)m--M-- (CH2)m.-- . In such an moiety, m and m' are integers from 1 to 6 and preferably from 1 to 3; and M is NH, N[R(5)] , O, S CH-R(5) or does not exist, wherein R(5) is lower alkyl, cycloalkyl or aryl and is preferably methyl, ethyl, propyl, phenyl, X-phenyl, or heterocyclic, wherein X is C1-, CF3- , F- , substituted at the o-, m- , or p- positions on the phenyl. M can contain a part of another diamino acid within the same peptide, e.g., the omega amino group of the one residue can be so linked to such an unnatural amino acid residue in a terminal residue.
Furthermore, any amino acid in the sequences provided may be replaced with its D-analogue, insofar as not more than 50% of the total amino acids are so replaced. Conservative substitutions include Glu for Asp, Gin for Asn and Val for He, among others, as is well known to those of ordinary skill in the art. Similarly, a homologous conservative substitution for any amino acid is within the bounds of the present invention. Depending on the applications for which the peptides according to the invention are intended, it is also possible to envisage intercalating between several amino acids, or even between all the amino acids, of the peptides defined above, dextrorotatory amino acids, and in particular dextrorotatory phenylalanine or dextrorotatory tryptophan, capable of preventing the action of the degradative enzymes in the cell environment and thus of increasing their activity. Another modification in this sense consists in replacing certain amino acids, for example of the proline type, by D-tryptophan.
In addition, a subject polypeptide can differ, unless otherwise specified, from the natural sequences shown above by the sequence being modified by terminal -NH2 acylation, e.g., acetylation, or by terminal-carboxylamidation, e.g., with ammonia, alkylamines, and the like.
The instant invention also comprises a method of use of the peptides supra for the treatment of adenocarcinomas of the colon, pancreatic carcinomas, neuroblastomas, and other cancers of undefined germ cell origin which express the transformed sequence of the ras protein.
It is also an object of the present invention to provide peptides and cyclized peptide homologs from the sequences listed in SEQ ID NOS:10-52.
The amino acid residues described herein are preferred to be in the "L" isomeric form. However, residues in the "D" isomeric form can be substituted for any L-amino acid residue, as long as the desired functional property is retained by the polypeptide. NH2 refers to the free amino group present at the amino terminus of a polypeptide. COOH refers to the free carboxyl group present at the carboxy terminus of a polypeptide. In keeping with standard polypeptide nomenclature described in J. Biol. Chem. 243.:3552-3559 (1969) and adopted at 37 C.F.R. 1.822(b) (2)) , The list of variable amino acids, capable of participating in the composition of this peptide is as follows: Y, Tyr, tyrosine; G, Gly, glycine; F, Phe, phenylalanine; M, Met, methionine; A, Ala, alanine; S, Ser, serine; I, He, isoleucine; L, Leu, leucine; T, Thr, threonine; V, Val, valine; P, Pro, proline; K, Lys, lysine; H, His, histidine; Q, Gin, glutamine; E, Glu, glutamic acid; W, Trp, tryptophan; R, Arg, arginine; D, Asp, aspartic acid; N, Asn, asparagine; C, Cys, cysteine.
Amino acid residue sequences are presented herein in the conventional left-to-right direction of amino-terminus to carboxy-terminus. In addition, the phrase "amino acid residue" is broadly defined to include the amino acids listed hereinabove, and modified and unusual amino acids, such as those listed in 37 C.F.R. 1.822(b) (4) , incorporated herein by reference. Furthermore, it should be noted that a dash at the beginning or end of an amino acid residue sequence indicates either a peptide bond to a further sequence of one or more amino acid residues or a covalent bond to an amino or hydroxyl end group.
Polypeptide and peptide are terms used interchangeably herein to designate a linear series of amino acid residues connected one to the other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues. Protein is a term used herein to designate a linear series of greater than about 20 amino acid residues connected one to the other as in a polypeptide.
The term synthetic peptide refers to a chemically produced chain of amino acid residues linked together by peptide bonds that is free of naturally occurring proteins and fragments thereof. The term peptide encompasses linear and cyclic peptides .
(D,L), (D) , or (L) preceding the amino acid designation means that this amino acids exists in that specific isomeric form, i.e. (D,L) Phe means that the amino acid phenylalanine exists as a racemic mixture; (D) Phe or D-Phe means that the amino acid phenylalanine exists as the D-stereoisomer or implied R configuration; (L) Phe means that the amino acid phenylalanine exists as the L stereoisomer or implied S configuration.
Alkyl as used herein means methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1, 1-dimethylpropyl, n-hexyl, 1-methylpentyl, 2-methylpentyl, 3 -methylpentyl , 4 -methylpentyl , 3 , 3 -dimethylbutyl , 2,2-dimethylbutyl, 1, 1-dimethylbutyl, 2-ethylbutyl, 1-ethylbutyl, 1, 3-dimethylbutyl, n-heptyl, 5-methylhexyl, 4- ethylhexyl, 3-methylhexyl, 2-methylhexyl, 1-methylhexyl, 3-ethylpentyl, 2 -ethylpentyl , 1 -ethylpentyl , 4 , 4 -dimethylpentyl , 3, 3-dimethylpentyl, 2,2-dimethylpentyl, 1, 1-dimethylpentyl, n-octyl, 6-methylheptyl, 5-methylheptyl, 4-methylheptyl, 3-methylheptyl, 2-methylheptyl, 1-methylheptyl, 1-ethylhexyl, 1 -propylpentyl , 3 -ethylhexyl , 5 , 5 -dimethylhexy1 , 4 , 4-dimethylhexyl, 2 , 2-diethylbutyl , 3 , 3-diethylbutyl, 1-methyl-1-propylbutyl .
Cycloalkyl refers to a hydrocarbon ring having from 3 to 7 carbon atoms, inclusive. Examples of cycloalkyl groups are cyclopropyl, cyclopentyl, cycloheptyl, cyclooctyl, cyclononyl, and the like.
The term aryl refers to aromatic groups which have at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl aralkyl, and biaryl groups, all of which may be optionally substituted.
Heterocyclic groups means groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and their heterocyclic compounds can include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl, and the like, all optionally substituted.
Substituted heterocyclic refers to any heterocyclic aryl group substituted by a alkyl, aryl, cycloalkyl, halo, sulfonate, or trifluoromethyl group.
The term alkyl amino refers to the groups --NRR' wherein respectively, (a) R is alkyl and R' is hydrogen or alkyl; (b) R is aryl and R' is hydrogen or aryl, (c) R is cycloalkyl and R' is hydrogen or alkyl, (d) R is hydrogen and R' is itself linear aminoalkyl, (e) R is alkyl and R' is itself linear aminoalkyl . The term aminoalkyl refers to the groups - (CH2)m-NRR' , wherein m is an integer from 1 to 6, inclusive and -NRR' is alkyl amino, as defined supra .
Halo encompasses fluoro, chloro, bromo and iodo. The phrase protecting group, as used herein, means substituents which protect the reactive functional group from undesirable chemical reactions. Examples of such protecting groups include esters of carboxylic acids, ethers of alcohols and acetals and ketals of aldehydes and ketones. The phrase N-protecting group or N-protected as used herein means those groups intended to protect the N-terminus of an amino acid or peptide, to protect an amino group against undesirable reactions during synthetic procedures and includes, but is not limited to, sulfonyl, acetyl, pivaloyl, t-butyloxycarbonyl (Boc) , carbonylbenzyloxy (Cbz) , benzoyl and an L- or D-aminoacyl residue, which may itself be N-protected similarly. The deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group. Thus, for example, an acyl group such as an alkanoyl, alkoxycarbonyl or an aroyl group may be removed for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide. Alternatively an acyl group such as a t-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid such as hydrochloric, sulfuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-charcoal.
The phrase COOH-protecting group or carboxyl-protecting group is, an esterifying group, for example an alkyl group
(especially methyl or ethyl) or an arylmethyl group (especially benzyl) . The deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group.
Thus, for example, an esterifying group such as an alkyl or arylmethyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide. Alternatively an esterifying group such as an arylmethyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-charcoal using either hydrogen or ammonium formate as a hydrogen source by methods well-known to those skilled in the art. Electrolyte means a solution that has sufficient acid strength to render a basic starting material essentially protonated.
Chemical derivative refers to a subject polypeptide having one or more residues chemically derivatized by reaction of a functional side group. Such derivatized molecules include for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides. Free hydroxyl groups may be derivatized to form O-acyl or 0-alkyl derivatives. The imidazole nitrogen of histidine may be derivatized to form N-imidazolyl- benzylhistidine. Also included as chemical derivatives are those peptides which contain one or more naturally occurring amino acid derivatives of the twenty standard amino acids. For examples, 4-hydroxyproline may be substituted for proline; 5-hydroxylysine may be substituted for lysine; 3-methylhistidine may be substituted for histidine; homoserine may be substituted for serine; and ornithine may be substituted for lysine. Polypeptides of the present invention also include any polypeptide having one or more additions and/or deletions or residues relative to the sequence of a polypeptide whose sequence is shown herein, so long as the requisite activity is maintained. As used herein, fragment means any subject peptide or polypeptide having an amino acid residue sequence shorter than that of a peptide or polypeptide whose full length amino acid residue sequence is shown herein.
A pharmaceutically acceptable salt is one which is prepared by contacting a compound of formulas (I) - (X) according to the specifications therein with an acid whose anion is generally considered suitable for human consumption. Examples of pharmacologically acceptable acid addition salts include the hydrochloride, hydrobromide, hydroiodide, sulfate, phosphate, acetate, propionate, lactate, maleate, alate, succinate, and tartrate salts. All of these salts may be prepared by conventional means by reacting, for example, the appropriate acid with the corresponding compound of structure of Formulas (I) - (X) .
Unless otherwise indicated, the preparation methods disclosed herein result in product distributions which include all possible structural isomers. It is understood that physiological response may vary according to stereochemical structure. The isomers may be separated by conventional means such as fractional crystallization or High Pressure Liquid Chromatography (HPLC) . Briefly, the absolute configuration of a compound relates to how its substituents are oriented in space about a central atom. This notion becomes significant when coupled with the rigors of chirality. Chirality involves the identity of the substituents" about that central atom. Thus, in general, a compound is said to be chiral when four distinctly different groups are bound to a central carbon atom. These groups may be spatially aligned in more than one manner without repeating their individual orientations. That is, a chiral compound may exhibit a mirror image which is also chiral . These mirror images are termed meso configurations, and are each absolute configurations of a chiral compound.
Pharmaceutical compositions according to the present invention comprise one or more peptides and/or peptidomimetics of the invention in association with a pharmaceutically acceptable carrier or excipient, adapted for use in human or veterinary medicine. The compositions may contain from 0.001-99% of the active material. Such compositions may be presented for use in conventional manner in admixture with one or more physiologically acceptable carriers of excipients. The pharmaceutical compositions according to the invention may also contain other active ingredients such as antimicrobial agents, or preservatives. The compositions may optionally further contain one or more other therapeutic agents which may, if desired, be a chemotherapeutic antiviral agent.
Pharmaceutically acceptable salts of the peptides of this invention may be formed conventionally by reaction with an appropriate acid. The addition salts so formed from addition by acid may be identified by hydrochloric, hydrobromic, phosphoric, acetic, fumaric, salicylic, citric, lactic, mandelic, tartaric, oxalic, methanesulfonic, and the like.
Thus, the peptides and peptidomimetics according to the present invention may be formulated for oral, buccal, parenteral, topical or rectal administration. In particular, these peptides and peptidomimetics may be formulated for injection or for infusion and may be presented in unit dose form in ampoules or in multidose containers with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
The present invention further provides a process for preparing a pharmaceutical composition which comprises bringing a peptide and/or peptidomimetic of the invention into association with a pharmaceutically acceptable excipient or carrier. For administration by injection or infusion, the daily dosage as employed for treatment of an adult human of approximately 70 kg body weight will range from 0.01 mg to 10 mg of each active ingredient, preferably 0.1 to 5 mg, which may be administered in 1 to 4 doses, for example, depending on the route of administration and the condition of the patient. The dosage of the peptide used in the treatment will vary, depending on the seriousness of the disorder, the weight of the patient, the relative efficacy of the peptide and the judgment of the treating physician. However, suitable unit dosages in humans may be between about 0.05 mg to about 100 mg. For example, a unit dosage may be from between about 0.2 mg to about 50 mg. Such a unit dosage, described hereinabove, may be administered more than once a day, e g., two or three times a day. Thus, the total daily dosage is in the range of about 0.01 mg to 10 mg/kg. Such therapy may extend for several weeks, in an intermittent or uninterrupted manner, until the patient's symptoms are eliminated.
The present invention also provides pharmaceutical compositions which comprise a pharmaceutically effective amount of the one or more peptides and/or peptidomimetics of this invention, or pharmaceutically acceptable salts thereof, and, preferably, a pharmaceutically acceptable carrier or adjuvant. Therapeutic methods of this invention comprise the step of treating patients in a pharmaceutically acceptable manner with those peptides or compositions. Such compositions may be in the form of tablets, capsules, caplets, powders, granules, lozenges, suppositories, reconstitutable powders, or liquid preparations, such as oral or sterile parenteral solutions or suspensions.
In order to obtain consistency of administration, it is preferred that a composition' of the invention is in the form of a unit dose. The unit dose presentation forms for oral administration may be tablets and capsules and may contain conventional expedients. For example binding agents, such as acacia, gelatin, sorbitol, or polyvinylpyrrolidone; fillers, such as lactose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricants such as magnesium stearate; disintegrants, such as starch, polyvinylpyrrolidone, sodium starch glycolate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.
The solid oral compositions may be prepared by conventional methods of blending, filling, tabletting, or the like. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are, of course, conventional in the art. The tablets may be coated according to methods well-known in normal pharmaceutical practice, in particular with an enteric coating. Oral liquid preparations may be in the form of emulsions, syrups, or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may or may not contain conventional additives. For example suspending agents, such as sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminum stearate gel, or hydrogenated edible fats; emulsifying agents, such as sorbitan monooleate or acacia; non-aqueous vehicles (which may include edible oils) , such as almond oil, fractionated coconut oil, oily esters selected from the group consisting of glycerine, propylene glycol, ethylene glycol, and ethyl alcohol; preservatives, for instance methyl para-hydroxybenzoate, ethyl para-hydroxybenzoate, n-propyl parahydroxybenzoate, or n-butyl parahydroxybenzoate or sorbic acid; and, if desired, conventional flavoring or coloring agents.
For parenteral administration, fluid unit dosage forms may be prepared by utilizing the peptide and a sterile vehicle, and, depending on the concentration employed, may be either suspended or dissolved in the vehicle. In preparing solutions, the peptides of this invention may be dissolved in water, whereas opiates used heretofore showed only marginal solubility in aqueous media or physiological fluids. Once in solution, the peptide may be injected and filter sterilized before filling a suitable vial or ampoule and subsequently sealing the carrier or storage package. Adjuvants, such as a local anaesthetic, a preservative or a buffering agent, may be dissolved in the vehicle prior to use. Stability of the pharmaceutical composition may be enhanced by freezing the composition after filling the vial and removing the. water under vacuum, e.g., freeze drying the composition. Parenteral suspensions may be prepared in substantially the same manner, except that the peptide should be suspended in the vehicle rather than being dissolved. A surfactant or wetting solution may be advantageously included in the composition to facilitate uniform distribution of the peptide. The stability of the peptides and cyclized peptides of the present invention exceeds that of naturally occurring peptides if substitution is made with D-amino acids in at least 20%, but not more than 50%, of those residues which are naturally present in the (L) configuration. Without being bound by theory, we believe that the increased resistance to enzymatic degradation over of the peptides of the present invention as compared to natural peptides is due to the presence of D-amino acids in the peptides. This switch from L to D amino acids neutralizes the digestion capabilities of many of the ubiquitous peptidases found in the digestive tract. Alternatively, the enhanced stability of the peptides of this invention may also be the result of the introduction of modifications of traditional peptide linkages. For example, the introduction of a cyclic ring within the peptide backbone may confer enhanced stability in order to circumvent the effect of many proteolytic enzymes known to digest small peptides in the stomach or other digestive organs and in serum.
The compounds of the " present invention are initially synthesized by either solution or by solid phase techniques. Specific exemplary syntheses are described in the examples hereinbelow. The peptides of this invention may be prepared by initially reacting a first appropriately protected amino acid with a second appropriately protected amino acid in an organic solvent inert to the reactants, in the presence of a suitable peptide coupling agent according to the following scheme:
Z (l)NH-AA(l) -COOH + coupling agent + NH2-AA(2) -COOZ (2) = Z(l) -NH-AA(l) -CONH-AA(2) -COOZ (2) wherein Z(l) is a suitable nitrogen protecting group and Z(2) is a suitable carboxyl protecting group and AA represents any natural or unnatural amino acid residue. The desired peptides may be prepared by utilizing the appropriate amino acids and repeating this reaction sequence as required until a peptide with three to ten amino acid residues has been prepared. A suitable deprotection method is then employed to remove specified or all of the remaining protecting groups or the peptide from the resin.
The first appropriately protected amino acid and, for instance, an appropriately protected tyrosine may be reacted together in the presence of a suitable peptide coupling agent in a suitably inert organic solvent with stirring, shaking, or agitation to form a protected tyrosine containing dipeptide. Introducing this dipeptide to appropriate protecting group removal conditions affords a selectively deprotected dipeptide which is well-suited for continued peptide synthesis. Contacting this mono-deprotected tyrosine containing dipeptide with an appropriately protected amino acid having a side chain represented as above, in the presence of a suitable peptide coupling agent in a suitably inert organic solvent with stirring, shaking, or agitation forms a protected tyrosine containing tripeptide. This method may be repeated as many times as necessary to achieve the desired peptide.
The method of preparation for peptide synthesis requires specific functional groups to react with other substituents to link amino acid residues in a desired manner to form a peptide possessing a known and desired sequence of amino acid residues. Since amino acids possess at least two reactive functional groups, suitable protection, blocking, or masking of these groups is required to ensure that reaction will occur only at specifically desired sites.
These protecting groups should be introduced to the moiety efficaciously while their removal should be performed under conditions which do not affect other portions of the molecule. In this manner, certain reactions and modifications may be performed on the amino acid, peptide, or other compound, with assurance that the protected functionality will not interfere with the desired reaction. Further, by choosing a protecting group that is sensitive and labile to certain reactive conditions, a reaction scheme may be outlined to advantageously utilize these characteristics to effectively remove the protecting group once the synthesis is complete.
Both N-protecting groups and COOH-protecting groups (see definitions) may be used within the scope of this invention. A variety of protecting groups known in the field of peptide synthesis and recognized by conventional abbreviations therein, may be found in T. Greene, Protective Groups In Organic Synthesis, Academic Press (1981) . Among the preferred protecting groups that may be utilized for suitable protection of reactive nucleophilic substituents include, for example, benzyl (Bz) , carbobenzyloxy (Cbz) , t-butoxycarbonyl (Boc) , or 9-fluorenylmethyloxy-carbonyl (Fmoc) .
Coupling of amino acids, which may be the same or different as those described above, to yield small peptides in route to peptides comprised of greater numbers of amino acid residues may be accomplished by employing established techniques in the field of peptide chemistry. A broad range of suitable reactions are described in E. Gross and J. Meinhofer, The Peptides: Analysis, Synthesis, Biology; Modern Techniques of Peptide and Amino Acid Analysis, John Wiley & Sons, (1981) and M. Bodanszky, Principles Of Peptide Synthesis, Springer-Verlag (1984) . The peptide coupling agents which may be used to assist condensation of amino and carboxyl i c ac i d mo i et i e s inc lude
N,N' -dicyclohexylcarbodiimide (DCC) , N,N' -carbonyl diimidazole
(CDI) , 1-hydroxy benzotriazole (HOBt) , ethyl chloroformate, b e n z y l c h l o r o f o r m a t e , 1- (ethoxycarbonyl) -2-ethoxy-l, 2-dihydroquinoline (EEDQ) , benzotriazoyl-oxy-tris- (dimethyl) amino-phosphonium hexafluoro phosphate (BOP) and the like. A preferred technique uses DCC as the coupling reagent. The DCC method may be used with or without catalytic additives such as 4-dimethylaminopyridine (DMAP) , copper (II) chloride or HOBt to hasten the reaction and suppress the racemization of the desired compound.
The DCC reaction is often performed at room temperature but may be carried out from about -78 °C to gentle reflux in a variety of solvents that are inert with respect to the reactants. The solvents are normally organic solvents which are polar and aprotic. Preferred solvents include, for example, dichloromethane, chloroform, diethyl ether, tetrahydrofuran
(THF) , N,N' -dimethylformamide (DMF) , and the like. Particularly preferred solvents are dichloromethane and DMF. In general, the coupling reaction may be carried out at atmospheric pressure a temperature of -78 °C to reflux for a period of between 1 and 48 hours. Preferably, the reaction is carried out at about -10° C to 25° C with stirring, shaking or agitation, over a period of between 4 and 6 hours.
Alternatively, synthesis may be achieved prepared using solid phase synthesis, such as that described by Merrifield, J Am. Chem. Soc. , 85, p 2149 (1964), although other equivalent chemical syntheses known in the art can also be used. Solid-phase synthesis is commenced from the C-terminus of the peptide by coupling a protected alpha-amino acid to a suitable resin as generally set forth in U.S. Pat. No. 4,244,946. As an example, He protected by BOC is coupled to the a BHA resin using methylene chloride and dimethylformamide . Following the coupling of BOC-He to the resin support, the alpha-amino protecting group is removed, as by using trifluoroacetic acid
(TFA) in methylene chloride, TFA alone or with HCl in dioxane. Preferably 50 volume % TFA in methylene chloride is used with 0-5 weight % 1,2 ethanedithiol. The deprotection is carried out at a temperature between about 0° C and room temperature. Other standard cleaving reagents and conditions for removal of specific alpha-amino protecting groups may be used as described in Schroder & Lubke, The Peptides, pp 72-75 (Academic Press 1965) .
After removal of the alpha-amino protecting group of He, the remaining alpha-amino- and side chain-protected amino acids are coupled step-wise in the desired order to obtain the intermediate compound defined hereinbefore. As an alternative to adding each amino acid separately in the synthesis, some of them may be coupled to one another prior to addition to the solid phase reactor. The selection of an appropriate coupling reagent is within the skill of the art. Particularly suitable as coupling reagents are N,N' -dicyclohexyl carbodiimide (DCC) and N, N ' -diisopropyl carbodiimide ( D I C I ) , or N-ethyl-N' - (3-dimethylaminopropyl) carbodiimide. Other activating reagents and their use in peptide coupling are described by Schroder & Lubke, supra , in Chapter III and by Kapoor (1970) J. Phar. Sci. .59:127. Each protected amino acid or amino acid sequence is introduced into the solid phase reactor in about a fourfold excess, and the coupling is carried out in a medium of dimethylformamide: dichloromethane (1:1) or in DMF or dichloromethane alone. In instances where the coupling is carried out manually, the success of the coupling reaction at each stage of the synthesis is monitored by the ninhydrin reaction, as described by E. Kaiser et al . (1970) Anal. Biochem. 3_4:595. In cases where incomplete coupling occurs, the coupling procedure is repeated before removal of the alpha-amino protecting group prior to the coupling of the next amino acid. The coupling reactions can be performed automatically, as on a Applied Biosystems automatic synthesizer.
After the desired amino acid sequence has been completed, the intermediate peptide is removed from the resin support by treatment with a reagent, such as liquid hydrogen fluoride, which not only cleaves the peptide from the resin but also cleaves all remaining side chain protecting groups and the alpha-amino protecting group (unless it is an acyl group which is intended to be present in the final peptide) to obtain the peptide. When using hydrogen fluoride for cleaving, anisole or cresol and methylethyl sulfide are included in the reaction vessel as scavengers. When Met is present in the sequence, the BOC protecting group may be cleaved with trifluoroacetic acid (TFA) /ethanedithiol prior to cleaving the peptide from the resin to eliminate potential S-alkylation.
All patents and publications referred to in the examples, and throughout the specification, are incorporated herein by reference, without admission that such is prior art.
The following nonlimiting examples are provided to illustrate the invention. The skilled artisan will recognize that there may be substitutions and variations of the exemplified methods and compositions which are apparent and can be practiced without departing from the essence of the invention.
EXAMPLES Example 1. Peptide Synthesis
The synthesis of the peptide of SEQ ID NO:l (Thr-Ile-Glu- Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val-He-Asp) is conducted in a stepwise manner on a MBHA hydrochloride resin, such as available from Bachem, Inc. (Torrance, CA) having a substitution range of about 0.1 to 0.5 mmoles/gm. resin.
All equipment employed in the examples is commercially available. Unless otherwise indicated, all starting materials employed in the examples are commercially available. Sources for these materials include Sigma Chemical Co. (St. Louis, MO) ,
Aldrich Chemical Co. (Milwaukee, WI) , Lancaster Synthesis
(Windham, NH) , Fisher Scientific (Pittsburgh, PA) , Boehringer Mannheim Biochemicals (Indianapolis, IN) , Fluka Chemical Corp. (Ronkonkoma, NY) and Chemical Dynamics Corp. (South Plainfield, NJ) . Most of the starting materials were obtained from Aldrich Chemical Co. (Milwaukee, WI) .
All solvents used in the peptide preparations described herein, e.g. methylene chloride dichloromethane, 2-propanol, dimethylformamide (DMF), and methanol, were Burdick and Jackson
"distilled in glass" grade and used without additional distillation. Trifluoroacetic acid (TFA) , diisopropylethylamine
(DIPEA) , piperidine (PIP) , dicyclohexylcarbodiimide (DCC) , 1-hydroxybenzotriazole (HOBt) , and [benzotriazole-1-yl- oxy-tris (dimethyl) phosphonium hexafluorophosphate] (BOP) were purchased from Chemical Dynamics Corp. and were "sequenal" grade purity. 1,2-ethanedithiol (EDT) was purchased from Sigma Chemical Co. and used without further purification. All protected amino acids were of the L-configuration unless otherwise indicated and were obtained from Bachem (Torrance, CA) .
The synthesis is performed on an Applied Biosystems peptide synthesizer (Foster City, CA) using a suitable program, preferably as follows:
Figure imgf000041_0001
Note: All wash and mix times three minutes exceptwhere noted.
Coupling of BOC-ASP(OBz) results in the substitution of about 0.35 mmol ASP per gram of resin. All solvents that are used are carefully degassed, preferably by sparging with an inert gas, e.g., helium or nitrogen, to insure the absence of oxygen.
After deprotection and neutralization, the peptide chain is built stepwise on the resin. Generally, one to two mmol. of BOC-protected amino acid in methylene chloride is used per gram of resin, plus one equivalent of 2 M DCC in methylene chloride, for two hours. When BOC-Arg(Tos) is being coupled, a mixture of 50% DMF and methylene chloride is used. Bzl is used as the hydroxyl side-chain protecting group for Ser and Thr. p-nitrophenyl ester (ONp) can be used to activate the carboxyl end of Asn or Gin; for example, BOC-Asn(ONp) can be coupled overnight using one equivalent of HOBt in a 50% mixture of DMF and methylene chloride. The amido group of Asn or Gin is protected by Xan when DCC coupling is used instead of the active ester method. 2-Cl-CBZ is used as the protecting group for the Lys side chain. Tos is used to protect the guanidine group of Arg and the imidazole group of His, and the side-chain carboxyl group of Glu or Asp is protected by OBzl.
To cleave and deprotect the resulting protected peptide-resin, it is treated with 1.5 ml anisole, 0.5 ml of methylethylsulfide and 15 ml liquid hydrogen fluoride (HF) per gram of peptide-resin, first at -20 °C for 20 min and then at 0 °C for 30 min. This reaction must be performed with great care owing to the highly toxic and corrosive nature of hydrogen fluoride. This reaction is performed in a commercially available teflon apparatus (Peninsula Research, Inc., Richmond, CA) . After complete elimination of HF under high vacuum using a KOH trap, the resin-peptide is washed alternately with dry diethyl ether and chloroform, and the peptides are then extracted with degassed 2 N aqueous acetic acid and separated from the resin by filtration on a Hirsch funnel.
The peptide is purified by gel permeation followed by preparative HPLC as described in Marki et al . (1981) J. Am. Chem. Soc. 103 :3178; Rivier, et al. (1984) J. Chromatography 288 : 303-328 ; and Hoeger, et al . (1987) BioChromatography 2.:134-142. The chromatographic fractions are carefully monitored by HPLC (see below) , and only the fractions showing substantial purity are pooled.
To confirm that the desired sequence is achieved, the peptide is hydrolyzed in sealed evacuated tubes containing constant boiling HCl, 3 μl of thioglycol/ml and 1 nmol of Nle (as an internal standard) for 9 hours at 140 °C. Amino acid analysis of the hydrolysates using a Beckman 121 amino acid analyzer to determine amino acid ratios allows confirmation that the desired peptide structure has been obtained.
Example 2. Cyclization of Active Peptides
Cyclization "traps" the bioactive conformation of the peptide by making the active conformation part of a ring system that allows it much less conformational flexibility. In this procedure, aspartate or glutamate residues are introduced into the sequence either in place of non-essential amino acid residues or as added residues in the chain. The new peptide is then subjected to electro-oxidation in which the two residues are decarboxylated, in an intramolecular Kolbe electro-oxidative coupling reaction, resulting in the joining of their respective - CH2 groups, forming a ring as shown in Fig. 1. This method has been used to make a cyclized /S-bend of the dipeptide, Pro-Gly, by placing a glutamic acid residue on the amino and carboxyl ends of this dipeptide and then performing the Kolbe electro-oxidation to form the tetra- (CH2) -bridge. The Pro-Gly peptide, which has a variety of conformations in solution, when cyclized, was found to adopt the β-bend structure uniquely (Joran, A. , "Conformationally restricted biologically active peptides, methods for their production and uses thereof," U.S. Patent No. 5,364,851.) This method has been used quite recently to synthesize cyclized forms of the peptide vasopressin; these forms have been tested in an in vi tro adenylate cyclase system and have been found to have prolonged half-lives and greater activity than the native peptide. Therefore, this cyclization procedure may result in enhanced peptide inhibition and in increased half-life. Introduction of the cyclizing rigidifying agent reduces the flexibility of the peptide and concurrently introduces non-polar aliphatic groups into the peptide (such as the tetra-methylene bridge shown in Scheme I) that help promote transport of the peptide through the cell membrane.
Scheme I
Figure imgf000044_0001
Kolbe electrolytic decarboxvlation
Di-aminosuberic acid ring
Scheme I illustrates an exemplary result of using electrolytic decarboxylation to cyclize peptides to trap them in their active conformations. Either two gultamate, two aspartate, or one glutamate and one aspartate residues are introduced in the chain represented by the wavy line. Under electro-oxidation shown here for two glutamate residues, a tetramethylene bridge forms cyclizing the region of the peptide that is to be held fixed in its bioactive conformation. The two connected residues shown constitute the diamino suberic acid moiety. It should be noted that, in the synthesis of this new peptide, there are possibly other aspartate and glutamate amino acid residues that can undergo the oxidative decarboxylation. To prevent these reactions from occurring, these Asp and Glu residues are protected as esters during the solid phase synthesis of the peptide. The free Glu residues at positions 44 and 46 are then allowed to undergo the cyclization reaction, after which the protected acid groups are then deprotected.
This cyclization procedure can be performed on other regions of this peptide and on the other two active peptides. The electrooxidative coupling reaction used to prepare the cyclic peptides of the invention can be performed in a divided or an undivided cell such as a standard glass H-cell, as described in Organic Electrochemistry (2nd Ed.) , M. Baizer and H. Lund, eds., Marcel Dekker, New York, 1983, Chap. 5, p 168. For large scale runs, the reaction can be carried out in a plate and frame flow cell as described in Technique of Electroorqanic Synthesis. Part III, N. Weinberg and B. Tilak, ed. , John Wiley & Sons, New York, 1982, Chap. Ill, p 179.
Cathode materials useful for the preparation of the compounds of the invention include, but are not limited to, high hydrogen overvoltage materials such as mercury, lead or cadmium. Anode materials include, but are not limited to, materials such as mercury, lead, graphite, or graphite paste, which are stable under electrolysis conditions. The electrooxidative coupling can occur in aqueous, or aqueous organic electrolytes, comprising solutions of Bronsted acids, such as sulfuric, fluoroboric, and trifluoroacetic acids. Any electrolyte may be selected that has sufficient acid strength to render a basic starting material protonated. A dilute solution of trifluoroacetic acid is most preferred.
Although the preferred method of electrolysis to obtain the compounds of this invention takes place under constant current conditions, the oxidative coupling could also be performed using controlled potential electrolysis, as understood by those skilled in the art. Typical current densities are between 1 and 5000 illiamps (mA) /cm2, preferably between 10 and 100 mA/cm2. The reaction is preferably carried out at a temperature in the range of about 0 °C to 37 °C, more preferably about 10 °C. A standard glass H-cell (200 ml volume, glass frit separator) was equipped with a mercury pool cathode 12 cm2 area) , a magnetic stirrer, and a platinum foil anode. The cell reservoir was filled with 40 mM trifluoroacetic acid (110 ml) and placed in a water bath maintained at 10 °C. The catholyte was purged with nitrogen. The starting peptide (20 mg) was added to the catholyte and constant current electrolysis was initiated at 0.1 A. The reaction progress was followed by HPLC and after passage of 1,060 coulombs, all the substrate had been consumed and the electrolysis was terminated. The catholyte was recovered and adjusted to pH 8 with NaOH. The pH-adjusted catholyte was extracted with chloroform (2 times 70 ml) . The extract was freeze dried and the resultant powdery material extracted with acetonitrile (HPLC grade) . This was filtered through a sintered- glass filter (fine porosity) and was reduced in volume on a rotary evaporator using a mechanical vacuum pump to a volume of 2 ml. This material was purified by reversed-phase high pressure liquid chromatography using a Waters HPLC system with a 0.46 x 0.25 cm column packed with 5 μm C18 silica, 300 A pore size. Buffer A is an aqueous 0.1% (vol/vol) trifluoroacetic acid solution (1.0 ml of TFA per 1000 Ml solution) ; Buffer B is 100% acetonitrile. The determination is run at room temperature with a gradient from 15.5% Buffer B to 75% Buffer B over a 30 min. The flow rate is 2.2 ml per minute, and the retention time is 25.0 min.
The structure was confirmed by 300 MHz XH NMR, 13C NMR, and electrospray mass spectroscopy. The amounts of the reactants and the conditions required to facilitate reaction and encourage efficient completion of the aforementioned Examples may vary widely. However, in general, the amounts of material employed to induce reaction in the processes discussed above will be substantially stoichiometric, unless otherwise specified. In the following examples, reaction concentrations are generally held at 0.1 M for the reactants, unless a higher concentration or dilution would be particularly useful for influencing the direction of a specific reaction. In practice, the amounts used will depend upon variations in reaction conditions and the nature of the reactants as readily apparent to one of ordinary skill in the art .
In any of the methods described hereinabove, the desired products may be isolated from the reaction mixture by crystallization. Alternatively, chromatographic techniques including, but not limited to, normal phase, reverse phase, ion-exchange, affinity, or gel permeation, may be employed, as well as electrophoresis or extraction or other means.
Example 3. Oocyte Maturation Assay
Using the method described in Chung et al . (1991) Anticancer Res. JH:1373-1378, test peptides, cyclized peptides and/or peptidomimetics are injected into immature oocytes at various doses. The oocytes are co-injected with recombinant transforming ras p21 obtained from the National Cancer Institute of Japan. Alternatively, the oncogenic ras p21 can be prepared by the ordinary skilled artisan without the expense of undue experimentation as described in Chung et al . (1991) supra and in Chung et al . (1992) Exp. Cell . Res . 203 :329-335 The maturation of the oocytes is evaluated microscopically at low power (20 X) , using a Nikon Diaphot microscope, for example. Percent inhibition is calculated based on comparisons with oocytes which are injected with 0.05 mg/ml oncogenic ras p21. The following results were obtained using a dose of each peptide equivalent to an internal oocyte concentration of 50 nM:
Figure imgf000048_0001
The peptidomimetics and cyclic peptides of the present invention will be similarly effective in inhibiting oocyte maturation in response to the oncogenic ras p21 protein, and in inhibiting oncogenesis.
Example 4. Synthesis of Peptidomimetic p21 ras Inhibitor
A representative peptidomimetic of the present invention is synthesized according to Scheme II, as further described hereinbelow.
Scheme I I
Na, πPrOH reflux, 79%
Figure imgf000049_0001
= CHaCHfCH-JaCHfCH^
R' = tBiiQ-CCH-C O
Steroid 1 was commercially available, and used without further purification. Mono-t-butylmalonate was prepared by literature methods [Brunwin, D.M.; et al. (1971) J. Chem . Soc . C. 3756] . THF was distilled from sodium/benzophenone under Ar. Methylene chloride and ethanol (absolute) were distilled from CaH2 under Ar. Benzylchloroformate and n-propyl alcohol are commercially available, and used without further purification or drying. All reactions performed under an atmosphere of Ar unless otherwise noted.
5-α-3-/S-hydroxy-6-oximinocholestane (2) : Cholestanone 1 (1.00 g. 2.5 mmol) , NaOAc (352 mg, 4.3 mmol) and hydroxylamine hydrochloride (197 mg, 2.8 mmol) were heated at reflux in absolute ethanol (15 ml) . The reaction was efficiently stirred under Ar for llh at reflux. The reaction was cooled, and the solvent removed on the rotary evaporator. The resulting white solid was dissolved in CHC13, and washed once with brine (80 ml) . The organic layer was dried over Na2S04, filtered, and concentrated in vacuo. The yield of crude material was 1.068 g of a lightly colored solid. The material was recrystallized from absolute ethanol (15.5 mL) to give 752 mg (72%) of white needles, mp = 202-204°C (dec) . XH NMR(CDC13) : 63.60 (m, IH) , 3.33 (dd, J = 4.37, 13.6 Hz, IH) , 2.10-1.75 (broad m, 6H) , 1.70-1.45 (broad m, 5H) , 1.42-1.28 (broad m, 8H) , 1.14 (m, 9H) , 0.92 (d, J = 6.87 Hz, 3H) , 0.88 (d, J = 6.54 Hz, 6H) , 0.77 (s, 3H) , 0.67 (s, 3H) . 13C NMR (CDC13) : 6159.55, 70.77, 56.39, 55.91, 54.04, 49.26, 42.60, 39.42, 39.20, 38.57, 35.92, 35.84, 35.63, 35.43, 31.24, 30.40, 29.39, 27.87, 27.70, 23.80, 23.53, 22.51, 22.26, 21.17, 18.36, 12.34, 11.79, IR (neat film) : 3354, 2941, 1667, 1467, 1065, 978 cm-1. 5-α-6-α-amino-3-/3-hydroxycholestane (3) : Oxime 2 (752 mg, 1.8 mmol) was dissolved with good stirring in boiling n-propyl alcohol (32 mL) . The flask was removed from the oil bath, and small pieces of freshly prepared sodium wire (2.898 g, 126 mmol) were added at a rate sufficient to maintain the reflux. After the addition of the sodium was complete, the flask was lowered into the bath, and stirred at reflux for 2h (a thick, white crust forms) . The reaction was cooled to room temperature, and carefully quenched by slow, dropwise addition of water under an inert atmosphere. The quenched reaction was extracted twice with CHCI3 (50 mL) , and the extracts were washed with brine, dried over Na2SO„, filtered, and concentrated. The yield of a vanilla colored solid was 723 mg. The material was purified by flash chromatrography on silica in CHCl3/MeOH (3:2) to give 577 mg (79%) of a vanilla colored solid. Alternatively, the product can be recrystallized from EtOAc. "H NMR (CDC13) : 63.59 (m, IH) , 2.60 (dd, J = 2.87, 10.35, 20.73Hz, IH) , 2.09 (d, J = 11.79Hz, IH) , 1.99 (d, J = 12.94Hz, IH) , 1.85 (tn, 3H) , 1.78 - 1.45 (broad m, 10H) , 1.36 (m, 5H) , 1.24 - 0.96 (m, 9H) , 0.89 (m, 10H) , 0.83 (s, 3H) , 0.77 (m, 2H) , 0.67 (s, 3H) .
N- (benzyloxy carbonyl) -5-α-6-α-amino-3-j3-hydroxycholestane (4) : The amine 3 (465 mg. 1.15 mmol) and anhydrous K2C03(457 mg, 3.3 mmol) were stirred in dry THF (4 mL) , and cooled to 0°C in an ice-water bath. Benzyl-chloroformate (0.16 mL, 1.15 mmol) was added dropwise, and the reaction was stirred at 0°C for 15 min, then at room temperature for 6h. The reaction was diluted with water and poured into saturated ΝaHC03 (15 mL) . A thick, flocculent white precipitate formed. The aqueous mixture was extracted with CHC13 (20 mL) , and the aqueous layer was saturated with NaCl and extracted with CHC13 (5x20 mL) . The organic extracts were washed with brine (40 mL) , dried over Na2S04, filtered, and concentrated. The yield of crude material was 649 mg of a tan solid. The material was flash chromatographed on silica in Hex/EtOAc (1:1) , (using a little CHC13 to help dissolve material) to give 546 mg (88%) of a white solid. XH NMR (CDC13) : δ7.88 (m, 4H) , 7.23 (s, IH) , 5.08 (d, J = 3.08Hz, 2H) , 4.86 (d, J = 9.46Hz, IH) , 3.53 (m, IH) , 3.50 (s, IH; overlaps with the multiplet at 3.53) , 2.00 (d, J = 12.35Hz, 2H) , 1.85 (m, 3H) , 1.53 (m, 4H) , 1.49 - 1.22 (broad m, 9H) , 1.20 - 0.98 (broad m, 10H) , 0.90 (m, 9H) , 0.87 (s, 3H) , 0.77 ( , 2H) , 0.66 (s, 3H) . IR (neat film) : 3346 (broad) , 1691, 1544, 1022 cm'1.
N- (benzyloxy carbonyl) -5- -6-or-aunino-3-3-mono-t-butyl malonyl cholestane (5) : The amine 4 (144.4 mg, 0.27 mmol), mono-t-butyl malonate (107.7 mg, 0.67 mmol) , and DMAP were dissolved in dry CH2C12 (1.2 mL) , and stirred efficiently at room temperature. DCC (67.4 dmg, 0.33 mmol) was added in one portion under a stream of Ar. The reaction was stirred at room temperature for 23h, then diluted with ether, and filtered through a pad of Celite (a white solid remains on the pad) . The filtrate was washed with 10% citric acid (20 mL) , saturated ΝaHC03 (20 mL) , and brine (20 mL) . The organic layer was dried over Na2S04, filtered and concentrated. The yield of crude material was 200 mg. The material was flash chromatographed in Hex/EtOAc (6.7:1) to give 168.7 mg (92%) of a yellow-gold residue XH NMR (CDC13) : 67.36 (s, 4H) , 7.29 (s, IH) , 5.08 (q, J = 12.25, 27.09Hz, 2H) , 4.70 (m, IH) , 4.38 (d, J" = 9.56Hz, IH) , 3.27 (s, 2H) , 2.00 (m, 2H) , 1.85 (m, 4H) , 1.54 (m, 3H) , 1.48 (s, 9H) , 1.35 (M, 8H) , 1.11 (m, HH) , 0.89 9(m, 12H) , 0.75 (m, 2H) , 0.66 (s, 3H) .
The N-CBZ group of 5 was cleanly removed by hydrogenolysis using 10% palladium on carbon under a hydrdogen atmosphere to give 6 in 93% yield. Trials employing 1,4-cyclohexadiene as the hydrogen source [Felix et al . (1978) J". Org. Chem . 4.3:4194] produced 6 in only 23% yield, even when a large excess of the diene was used. Longer reaction time did not improve the yield. 5-a-6-a-amino-3-3-mono-t-butyl malonyl cholestane (6) : 5 (200.7 mg, 0.3 mmol) and 10% palladium on carbon (194 mg) were stirred vigorously in abs. EtOH (1.8 mL) . The system was flushed with a balloon of hydrogen, and an atmosphere of hydrogen was maintained by 2 balloons of hydrogen. The reaction was stirred for 20h at room temperature, then vented with Ar, and suction-filtered through a tighly packed pad of Celite. The flask and filter cake were thoroughly washed with 1:1 EtOH/THF. The filtrate was concentrated in vacuo to give 167.8 mg of material. The crude product was flash chromatographed first in Hex/EtOAc (3:2) to elute off minor byproducts, then CHCl3/MeOH (9:1) to give the product in a yield of 152.3 mg (93%) of a golden residue. JH ΝMR (CDClj) : 64.76 ( , IH) , 3.27 (s, 2H) , 2.60 (m, IH) , 2.22 (m, IH) , 2.05 - 1.71 (broad m, 6H) , 1.51 (s, 3H) , 1.49 (s, 9H) , 1.34 (broad m, 8H) , 1.24 - 0.97 (broad m, 12H) , 0.92 (d, J = 6.55Hz, 3H) , 0.88 (d, J = 6.64Hz, 6H) , 0.85 (s, 3H) , 0.75 (m, 2H) , 0.67 (s, 3H) , IR (neat film) : 3368, 2946, 2868, 1747, 1729, 1144, 1008 cm"1.
Reductive amination of Ν(BOC) aminoacetaldehyde [Buchardt et al. (1993) Org. Prep . Proc . Int . .25.:457] is promoted by the use of NaBH3CN and catalytic acetic acid. t-Butylester and BOC groups are removed. The general plan is given in Scheme III. Scheme III
R--.H02CCH2CO
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: INNAPHARMA, INC.
(ii) TITLE OF INVENTION: Peptides and Peptidomimetics Inhibiting the Oncogenic Action of P21 Ras
(iii) NUMBER OF SEQUENCES: 52
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Greenlee, Winner and Sullivan, P.C.
(B) STREET: 5370 Manhattan Circle, Suite 201
(C) CITY: Boulder
(D) STATE: Colorado
(E) COUNTRY: US
(F) ZIP: 80303
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.30
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: PCT/US96/15098
(B) FILING DATE: 20-SEP-1996
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/531,525
(B) FILING DATE: 21-SEP-1995
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 60/004,091
(B) FILING DATE: 21-SEP-1995
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Ferber, Donna M.
(B) REGISTRATION NUMBER: 33,878
(C) REFERENCE/DOCKET NUMBER: 37-9 A WO
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (303) 499-8080
(B) TELEFAX: (303) 499-8089
(2) INFORMATION FOR SEQ ID NO:1 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 6 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS:
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l: lie Lys Arg Val Lys Asp 1 5
(2) INFORMATION FOR SEQ ID NO:2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 5 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Lys Cys Asp Leu Ala 1 5
(2) INFORMATION FOR SEQ ID NO: 3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS:
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3
Cys Asp Leu Ala Ala Arg Thr l 5
(2) INFORMATION FOR SEQ ID NO:4 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 4 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4 :
Asp Leu Ala Ala
1
(2) INFORMATION FOR SEQ ID NO:5 : (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 188 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5 :
Met Thr Glu Tyr Lys Leu Val Val Val Gly Ala Gly Gly Val Gly Lys 1 5 10 15
Ser Ala Leu Thr lie Gin Leu lie Gin Asn His Phe Val Asp Glu Tyr 20 25 30
Asp Pro Thr lie Glu Asp Ser Tyr Arg Lys Gin Val Val lie Asp Gly 35 40 45
Glu Thr Cys Leu Leu Asp lie Leu Asp Thr Ala Gly Gin Glu Glu Tyr 50 55 60
Ser Ala Met Arg Asp Gin Tyr Met Arg Thr Gly Glu Gly Phe Leu Cys 65 70 75 80
Val Phe Ala lie Asn Asn Thr Lys Ser Phe Glu Asp lie His Gin Tyr 85 90 95
Arg Glu Gin lie Lys Arg Val Lys Asp Ser Asp Asp Val Pro Met Val 100 105 110
Leu Val Gly Asn Lys Cys Asp Leu Ala Ala Thr Val Glu Ser Arg Gin 115 120 125
Ala Gin Asp Leu Ala Arg Ser Tyr Gly lie Pro Tyr lie Glu Thr Ser 130 135 140
Ala Lys Thr Arg Gin Gly Val Glu Asp Ala Phe Tyr Thr Leu Val Arg 145 150 155 160
Glu lie Arg Gin His Lys Leu Arg Lys Leu Asn Pro Pro Asp Glu Ser 165 170 175
Gly Pro Gly Cys Met Ser Cys Lys Cys Val Leu Ser 180 185
(2) INFORMATION FOR SEQ ID NO:6 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 13 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6 : Thr He Glu Asp Ser Tyr Arg Lys Gin Val Val He Asp 1 5 10
(2) INFORMATION FOR SEQ ID NO:7 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(Xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7:
Tyr Arg Glu Gin He Lys Arg Val Lys Asp Ser Asp Asp Val Pro
1 5 10 15
(2) INFORMATION FOR SEQ ID NO: 8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8 :
Gly Asn Lys Cys Asp Leu Ala Ala Arg Thr Val Glu 1 5 10
(2) INFORMATION FOR SEQ ID NO: 9 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
Cys Asp Leu Ala Ala Arg Thr 1 5
(2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 215 amino acids (B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Arabidopsis thaliana
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:
Met Ala Ala Pro Pro Ala Arg Ala Arg Ala Asp Tyr Asp Tyr Leu He 1 5 10 15
Lys Leu Leu Leu He Gly Asp Ser Gly Val Gly Lys Ser Cys Leu Leu 20 25 30
Leu Arg Phe Ser Asp Gly Ser Phe Thr Thr Ser Phe He Thr Thr He 35 40 45
Gly He Asp Phe Lys He Arg Thr He Glu Leu Asp Gly Lys Arg He 50 55 60
Lys Leu Gin He Trp Asp Thr Ala Gly Gin Glu Arg Arg Thr He Thr 65 70 75 80
Thr Ala Tyr Tyr Arg Gly Ala Met Gly He Leu Leu Val Tyr Asp Val 85 90 95
Thr Asp Glu Ser Ser Phe Asn Asn He Arg Asn Trp He Arg Asn He 100 105 no
Glu Gin His Ala Ser Asp Asn Val Asn Lys He Leu Val Gly Asn Lys 115 120 125
Ala Asp Met Asp Glu Ser Lys Arg Ala Val Pro Thr Ala Lys Gly Gin 130 135 140
Ala Leu Ala Asp Glu Tyr Gly He Lys Phe Phe Glu Thr Ser Ala Lys 145 150 155 160
Thr Asn Leu Asn Val Glu Glu Val Phe Phe Ser He Gly Arg Asp He 165 170 175
Lys Gin Arg Leu Ser Asp Thr Asp Ser Arg Ala Glu Pro Ala Thr He 180 185 190
Lys He Ser Gin Thr Asp Gin Ala Ala Gly Ala Gly Gin Ala Thr Gin 195 200 205
Lys Ser Ala Cys Cys Gly Thr 210 215
(2) INFORMATION FOR SEQ ID NO:11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 213 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Arabidopsis thaliana
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:
Met Ala Gly Tyr Ala Asp Glu Glu Tyr Asp Tyr Leu Phe Lys Leu Val 1 5 10 15
Leu He Gly Asp Ser Gly Val Gly Lys Ser Asn Leu Leu Ser Arg Phe 20 25 30
Thr Lys Asn Phe Asn Leu Glu Ser Lys Ser Thr He Gly Val Glu Phe 35 40 45
Ala Thr Lys Thr Thr Lys Val Glu Gly Lys Val Val Lys Ala Gin He 50 55 60
Trp Asp Thr Ala Gly Gin Glu Arg Tyr Arg Ala He Thr Ser Ala Tyr 65 70 75 80
Tyr Arg Gly Ala Val Gly Ala Leu Leu He Tyr Asp Val Thr Arg His 85 90 95
Ala Thr Phe Glu Asn Ala Ala Arg Trp Leu Arg Glu Leu Arg Gly His 100 105 110
Thr Asp Pro Asn He Val Val Met Leu He Gly Asn Lys Cys Asp Leu 115 120 125
Arg His Leu Val Ala Val Lys Thr Glu Glu Ala Lys Ala Phe Ala Glu 130 135 140
Arg Glu Ser Leu Tyr Phe Met Glu Thr Ser Ala Leu Asp Ala Thr Asn 145 150 155 160
Val Glu Asn Ala Phe Thr Glu Val Leu Thr Gin He His Lys He Val 165 170 175
Ser Lys Arg Ser Val Asp Gly Gly Gly Ser Ala Asp Leu Pro Gly Lys 180 185 190
Gly Glu Thr He Asn Val Lys Glu Asp Gly Ser Val Leu Lys Arg Met 195 200 205
Gly Cys Cys Ser Asn 210
(2) INFORMATION FOR SEQ ID NO:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 213 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Arabidopsis thaliana (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:
Met Ser Ser Asp Asp Glu Gly Arg Glu Glu Tyr Phe Lys He Val Val 1 5 10 15
He Gly Asp Ser Ala Val Gly Lys Ser Asn Leu Leu Ser Arg Tyr Ala 20 25 30
Arg Asn Glu Phe Ser Ala Asn Ser Lys Ala Thr He Gly Val Glu Phe 35 40 45
Gin Thr Gin Ser Met He Glu Gly Lys Glu Val Lys Ala Gin He Trp 50 55 60
Asp Thr Ala Gly Gin Glu Phe Arg Ala Val Thr Ser Tyr Tyr Arg Gly 65 70 75 80
Ala Val Gly Ala Leu Val Val Tyr Asp He Thr Arg Arg Thr Thr Phe 85 90 95
Glu Ser Val Gly Arg Trp Leu Asp Glu Leu Lys He His Ser Asp Thr 100 105 110
Thr Val Ala Arg Met Leu Val Gly Asn Lys Cys Asp Leu Glu Asn He 115 120 125
Arg Ala Val Ser Val Glu Glu Gly Lys Ala Leu Ala Glu Glu Glu Gly 130 135 140
Leu Phe Phe Val Glu Thr Ser Ala Leu Asp Ser Thr Asn Val Lys Thr 145 150 155 160
Ala Phe Glu Met Val He Leu Asp He Tyr Asn Asn Val Ser Arg Lys 165 170 175
Gin Leu Asn Ser Asp Thr Tyr Lys Asp Glu Leu Thr Val Arg Val Ser 180 185 190
Leu Val Lys Asp Asp Asn Ser Ala Ser Lys Gin Ser Ser Gly Phe Ser 195 200 205
Cys Cys Ser Ser Thr 210
(2) INFORMATION FOR SEQ ID NO: 13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 201 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Discopyge ommata
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
Met Asn Pro Glu Tyr Asp Tyr Leu Phe Lys Leu Leu Leu He Gly Asp 1 5 10 15
Ser Gly Val Gly Lys Ser Cys Leu Leu Leu Arg Phe Ala Asp Asp Thr 20 25 30
Tyr Thr Glu Ser Tyr He Ser Thr He Gly Val Asp Phe Lys He Arg 35 40 45
Thr He Glu Leu Asp Gly Lys Thr He Lys Leu Gin He Trp Asp Thr 50 55 60
Ala Gly Gin Glu Arg Phe Arg Thr He Thr Ser Ser Tyr Tyr Arg Gly 65 70 75 80
Ala His Gly He He Val Val Tyr Asp Val Thr Asp Gin Glu Ser Phe 85 90 95
Asn Asn Val Lys Gin Trp Leu Gin Glu He Asp Arg Tyr Ala Ser Glu 100 105 110
Asn Val Asn Lys Leu Leu Val Gly Asn Lys Cys Asp Leu Thr Thr Lys 115 120 125
Lys Val Val Asp Tyr Thr Thr Lys Glu Phe Ala Asp Ser Leu Gly He 130 135 140
Pro Phe Leu Glu Thr Ser Ala Lys Asn Ala Thr Asn Val Glu Gin Ala 145 150 155 160
Phe Met Thr Met Ala Ala Glu He Lys Lys Arg Met Gly Pro Gly Ala 165 170 175
Thr Ser Gly Gly Ser Glu Lys Ser Asn Val Asn He Gin Ser Thr Pro 180 185 190
Val Lys Ser Ser Gly Gly Gly Cys Cys 195 200
(2) INFORMATION FOR SEQ ID NO: 14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 202 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Lymnea stagnalis
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:
Met Ser Thr Met Asn Pro Asp Tyr Asp Tyr Leu Phe Lys Leu Leu Leu 1 5 10 15
He Gly Asp Ser Gly Val Gly Lys Ser Cys Leu Leu Leu Arg Phe Ala 20 25 30
Asp Asp Thr Tyr Thr Glu Ser Tyr He Ser Thr He Gly Val Asp Phe 35 40 45
Lys He Arg Thr He Glu Leu Asp Gly Lys Thr He Lys Leu Gin He 50 55 60
Trp Asp Thr Ala Gly Gin Glu Arg Phe Arg Thr He Thr Ser Ser Tyr 65 70 75 80
Tyr Arg Gly Ala His Gly He He Val Val Tyr Asp Val Thr Asp Gin 85 90 95
Glu Ser Phe Asn Asn Val Lys Gin Trp Leu Gin Glu He Asp Arg Tyr 100 105 110
Ala Ser Glu Asn Val Asn Lys Leu Leu Val Gly Asn Lys Ser Asp Leu 115 120 125
Thr Thr Lys Lys Val Asp Phe Thr Thr Ala Lys Glu Tyr Ala Asp Gin 130 135 140
Leu Gly He Pro Phe Leu Glu Thr Ser Ala Lys Asn Ala Thr Asn Val 145 150 155 160
Glu Gin Ala Phe Met Thr Met Ala Ala Glu He Lys Asn Arg Met Gly 165 170 175
Pro He Thr Ala Ser Asp Ser Lys Pro Ser Val Lys He Asn Ser Ser 180 185 190
Thr Pro Ser Ala Asn Lys Gly Gly Cys Cys 195 200
(2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 208 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:
Met Ala Tyr Ala Tyr Leu Phe Lys Tyr He He He Gly Asp Thr Gly 1 5 10 15
Val Gly Lys Ser Cys Leu Leu Leu Gin Phe Thr Asp Lys Arg Phe Gin 20 25 30
Pro Val His Asp Leu Thr He Gly Val Glu Phe Gly Ala Arg Met He 35 40 45
Thr He Asp Gly Lys Gin He Lys Leu Gin He Trp Asp Thr Ala Gly 50 55 60
Gin Glu Ser Phe Arg Ser He Thr Arg Ser Tyr Tyr Arg Gly Ala Ala 65 70 75 80
Gly Ala Leu Leu Val Tyr Asp He Thr Arg Arg Asp Thr Phe Asn His 85 90 95
Leu Thr Thr Trp Leu Glu Asp Ala Arg Gin His Ser Asn Ser Asn Met 100 105 110
Val He Met Leu He Gly Asn Lys Ser Asp Leu Glu Arg Arg Glu Val 115 120 125
Lys Lys Glu Glu Gly Glu Ala Phe Ala Glu His Gly Leu He Phe Met 130 135 140
Glu Thr Ala Lys Thr Ala Ser Val Glu Glu Ala Phe He Asn Thr Ala 145 150 155 160
Lys Glu He Tyr Glu Lys He Gin Glu Gly Val Phe Asp He Asn Asn 165 170 175
Glu Ala Asn Gly He Lys He Gly Pro Gin His Ala Ala Thr Asn Ala 180 185 190
Thr His Ala Gly Asn Gin Gly Gly Gin Gin Ala Gly Gly Gly Cys Cys 195 200 205
(2) INFORMATION FOR SEQ ID NO:16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 210 amino acids
(B) TYPE: ammo acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Lymnea stagnalis
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:16 :
Met Ser Tyr Ala Tyr Leu Phe Lys Tyr He He He Gly Asp Thr Gly 1 5 10 15
Val Gly Lys Ser Cys Leu Leu Leu Gin Phe Thr Asp Lys Arg Phe Gin 20 25 30
Pro Val His Asp Leu Thr He Gly Val Glu Phe Gly Ala Arg Met He 35 40 45
Thr He Asp Gly Lys Gin He Lys Leu Gin He Trp Asp Thr Ala Gly 50 55 60
Gin Glu Ser Phe Arg Ser He Thr Arg Ser Tyr Tyr Arg Gly Ala Ala 65 70 75 80
Gly Ala Leu Leu Val Tyr Asp He Thr Arg Arg Asp Thr Phe Asn His 85 90 95
Leu Thr Thr Trp Leu Glu Asp Ala Arg Gin His Ser Asn Ser Asn Met 100 105 110
Val He Met Leu He Gly Asn Lys Ser Asp Leu Glu Ala Arg Arg Val 115 120 125
Lys Lys Glu Glu Gly Glu Ala Phe Arg Glu His Gly Leu He Phe Met 130 135 140
Glu Thr Ser Ala Lys Thr Ala Ala Asn Val Glu Glu Ala Phe He Asn 145 150 155 160 Thr Ala Lys Glu He Tyr Gin Lys He Gin Asp Gly Val Phe Asp He 165 170 175
Asn Asn Glu Ala Asn Gly He Lys He Gly Pro Gin His Ser Pro Ala 180 185 190
Ser Gin Ser Leu Asn Val Gly Gly Ser Gly Gly Asn Gin Gly Gly Asn 195 200 205
Cys Cys 210 INFORMATION FOR SEQ ID NO: 17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 208 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Oryctolagus cuniculus
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
Met Ala Tyr Ala Tyr Leu Phe Lys Tyr He He He Gly Asp Thr Gly 1 5 10 15
Val Gly Lys Ser Cys Leu Leu Leu Gin Phe Thr Asp Lys Arg Phe Gin 20 25 30
Pro Val His Asp Leu Thr He Gly Val Glu Phe Gly Ala Arg Met He 35 40 45
Thr He Asp Gly Lys Gin He Lys Leu Gin He Trp Asp Thr Ala Gin 50 55 60
Glu Ser Phe Arg Ser He Arg Ser Tyr Tyr Arg Gly Ala Gly Ala Leu 65 70 75 80
Leu Val Tyr Asp He Thr Arg Arg Asp Thr Phe Asn His Leu Thr Thr 85 90 95
Trp Leu Glu Asp Ala Arg Gin His Ser Asn Ser Asn Met Val He Met 100 105 110
Leu He Gly Asn Lys Ser Asp Leu Glu Ser Arg Arg Glu Val Lys Lys 115 120 125
Glu Glu Gly Glu Ala Phe Ala Arg Glu His Gly Leu He Phe Met Glu 130 135 140
Thr Ser Ala Lys Thr Ala Ser Asn Val Glu Glu Ala Phe He Asn Thr 145 150 155 160
Ala Lys Glu He Tyr Glu Lys He Gin Glu Gly Val Phe Asp He Asn 165 170 175
Asn Glu Ala Asn Gly He Lys He Gly Pro Gin His Gly Ala Thr Asn 180 185 190 Ala His Ala Gly Asn Gin Gly Gly Gin Gin Ala Gly Gly Gly Cys Cys 195 200 205
(2) INFORMATION FOR SEQ ID NO:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 212 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Rattus norvegicus
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
Met Ala Tyr Ala Tyr Leu Phe Lys Tyr He He He Gly Asp Thr Gly 1 5 10 15
Val Gly Lys Ser Cys Leu Leu Leu Gin Phe Thr Asp Lys Arg Phe Gin 20 25 30
Pro Val His Asp Leu Thr Met Gly Val Glu Phe Gly Ala Arg Met He 35 40 45
Thr He Asp Gly Lys Gin He Lys Leu Gin He Trp Asp Thr Ala Gly 50 55 60
Gin Glu Ser Phe Arg Ser He Thr Arg Ser Tyr Tyr Arg Gly Ala Ala 65 70 75 80
Gly Ala Leu Leu Val Tyr Asp He Thr Arg Arg Asp Thr Phe Asn His 85 90 95
Leu Thr Thr Trp Leu Glu Asp Ala Arg Gin His Ser Asn Ser Asn Met 100 105 110
Val He Met Leu He Gly Asn Lys Ser Asp Leu Glu Ser Arg Arg Glu 115 120 125
Val Lys Lys Glu Glu Gly Glu Ala Phe Ala Arg Glu His Gly Leu He 130 135 140
Phe Met Glu Thr Ser Ala Lys Thr Ala Ser Asn Val Glu Glu Ala Phe 145 150 155 160
He Asn Thr Ala Lys Glu He Tyr Glu Lys He Gin Glu Gly Val Phe 165 170 175
Asp He Asn Asn Glu Ala Asn Gly He Lys He Gly Pro Gin His Ala 180 185 190
Ala Thr Asn Ala Ser His Gly Gly Asn Gin Gly Gly Gin Gin Ala Gly 195 200 205
Gly Gly Cys Cys 210
(2) INFORMATION FOR SEQ ID NO:19: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 218 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Drosophila melanogaster
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:
Met Ala Gly Gly Asp Pro Lys Trp Gin Lys Asp Ala Ala Asp Gin Asn 1 5 10 15
Phe Asp Tyr Met Phe Lys Leu Leu He He Gly Asn Ser Ser Val Gly 20 25 30
Lys Thr Ser Phe Leu Phe Arg Tyr Ala Asp Asp Ser Phe Thr Ser Ala 35 40 45
Phe Val Ser Thr Val Gly He Asp Phe Lys Val Lys Thr Val Phe Arg 50 55 60
His Asp Lys Arg Val Lys Leu Gin He Trp Asp Thr Ala Gly Gin Glu 65 70 75 80
Arg Tyr Arg Thr He Thr Thr Ala Tyr Tyr Arg Gly Ala Met Gly Phe 85 90 95
He Leu Met Tyr Asp Val Thr Asn Glu Asp Ser Phe Asn Ser Val Gin 100 105 110
Asp Trp Val Thr Gin He Lys Thr Tyr Ser Trp Asp Asn Ala Gin Val 115 120 125
He Leu Val Gly Asn Lys Cys Asp Met Glu Asp Gin Arg Val He Ser 130 135 140
Phe Glu Arg Gly Arg Gin Leu Ala Asp Gin Leu Gly Val Glu Phe Phe 145 150 155 160
Glu Thr Ser Ala Lys Glu Asn Val Asn Val Lys Ala Val Phe Glu Arg 165 170 175
Leu Val Asp He He Cys Lys Met Ser Glu Ser Leu Asp Ala Asp Pro 180 185 190
Thr Leu Val Gly Gly Gly Gin Lys Gly Gin Arg Leu Thr Asp Gin Pro 195 200 205
Gin Gly Thr Pro Asn Ala Asn Cys Asn Cys 210 215
(2) INFORMATION FOR SEQ ID NO:20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 208 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Rattus norvegicus
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20:
Met Ser Glu Thr Tyr Asp Phe Leu Lys Phe Leu Val He Gly Asn Ala 1 5 10 15
Gly Thr Gly Lys Ser Cys Leu Leu His Gin Phe He Glu Lys Lys Phe 20 25 30
Lys Asp Asp Ser Asn His Thr He Gly Val Glu Phe Gly Gin Lys He
35 40 45
He Asn Val Gly Gly Lys Tyr Val Lys Leu Gin He Trp Asp Thr Ala 50 55 60
Gly Gin Glu Arg Phe Arg Val Thr Thr Ser Tyr Arg Gly Ala Ala Gly 65 70 75 80
Ala Leu Leu Val Tyr Asp He Thr Ser Arg Glu Thr Tyr Asn Ala Leu 85 90 95
Thr Asn Trp Leu Thr Asp Ala Arg Met Leu Ala Ser Gin Asn He Val 100 . 105 110
He Cys Gly Asn Lys Lys Asp Leu Asp Ala Asp Arg Glu Val Thr Phe 115 120 125
Leu Glu Ala Ser Arg Phe Ala Gin Glu Asn Glu Leu Met Phe Leu Glu 130 135 140
Thr Ser Ala Leu Thr Gly Glu Asn Val Glu Glu Ala Phe Met Gin Cys 145 150 155 160
Ala Arg Lys He Leu Asn Lys He Glu Ser Gly Glu Leu Asp Pro Glu 165 170 175
Arg Met Gly Ser Gly He Gin Tyr Gly Asp Ala Ala Leu Arg Gin Leu 180 185 190
Arg Ser Pro Arg Arg Thr Gin Ala Pro Ser Ala Gin Glu Cys Gly Cys 195 200 205
(2) INFORMATION FOR SEQ ID NO:21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 203 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Caenorhabditis elegans (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21:
Met Ala Asp Phe Thr Asn Asn Ala Leu Lys Lys Phe Lys Leu Val Phe 1 5 10 15
Leu Gly Glu Gin Ser Val Gly Lys Thr Ser He He Thr Arg Phe Met 20 25 30
Tyr Asp Ser Phe Asp Asn Thr Tyr Gin Ala Thr He Gly He Asp Phe 35 40 45
Leu Ser Lys Thr Met Tyr Leu Glu Asp Arg Thr He Arg Leu Gin Leu 50 55 60
Trp Asp Thr Ala Gly Gin Glu Arg Phe Arg Ser Leu He Pro Ser Tyr 65 70 75 80
He Arg Asp Ser Ser Val Ala Val Val Val Tyr Asp He Thr Asn Ala 85 90 95
Asn Ser Phe His Gin Thr Thr Lys Trp Val Asp Asp Val Arg Asn Glu 100 105 110
Arg Gly Cys Asp Val He He Val Leu Val Gly Asn Lys Thr Asp Leu 115 120 125
Ala Asp Lys Arg Gin Val Ser Thr Glu Asp Gly Glu Lys Lys Ala Arg 130 135 140
Asp Leu Asn Val Met Phe He Glu Thr Ser Ala Lys Ala Gly Tyr Asn 145 150 . 155 160
Val Lys Gin Leu Phe Arg Lys He Ala Leu Pro Gly He Val Gin Glu 165 170 175
Glu Thr Pro Glu Gin Pro Asn He Val He Met Asn Pro Pro Lys Asp 180 185 190
Ala Glu Glu Ser Gin Gly Arg Gin Cys Pro Cys 195 200
(2) INFORMATION FOR SEQ ID NO:22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 207 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:
Met Ser Thr Gly Gly Asp Phe Gly Asn Pro Leu Arg Lys Phe Lys Leu
1 5 10 15
Val Phe Leu Gly Glu Gin Ser Val Gly Lys Thr Ser Leu He Thr Arg 20 25 30
Phe Met Tyr Asp Ser Phe Asp Asn Thr Tyr Gin Ala Thr He Gly He 35 40 45
Asp Phe Leu Ser Lys Thr Met Tyr Leu Glu Asp Arg Thr Val Arg Leu 50 55 60
Gin Leu Trp Asp Thr Ala Gly Gin Glu Arg Phe Arg Ser Leu He Pro 65 70 75 80
Ser Tyr He Arg Asp Ser Thr Val Ala Val Val Val Tyr Asp He Thr 85 90 95
Asn Val Asn Ser Phe Gin Gin Thr Thr Lys Trp He Asp Asp Val Arg 100 105 110
Thr Glu Arg Gly Ser Asp Val He He Met Leu Val Gly Asn Lys Thr 115 120 125
Asp Leu Ala Asp Lys Arg Gin Val Ser He Glu Glu Gly Glu Arg Lys 130 135 140
Ala Lys Glu Leu Asn Val Met Phe He Glu Ser Ala Lys Ala Gly Tyr 145 150 155 160
Asn Val Lys Gin Leu Phe Arg Arg Val Ala Ala Ala Leu Pro Gly Met 165 170 175
Glu Ser Thr Gin Asp Arg Ser Arg Glu Asp Met He Asp He Lys Leu 180 185 190
Glu Lys Pro Gin Glu Gin Pro Val Ser Glu Gly Gly Cys Ser Cys 195 200 205
(2) INFORMATION FOR SEQ ID NO:23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 203 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Canis familiaris
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23:
Met Thr Ser Arg Lys Lys Val Leu Leu Lys Val He He Leu Gly Asp 1 5 10 15
Ser Gly Val Gly Lys Thr Ser Leu Met Asn Gin Tyr Val Asn Lys Lys 20 25 30
Phe Ser Asn Gin Tyr Lys Ala Thr He Gly Ala Asp Phe Leu Thr Lys 35 40 45
Glu Val Met Val Asp Asp Arg Leu Val Thr Met Gin He Trp Asp Thr 50 55 60
Ala Gly Gin Glu Arg Phe Gin Ser Leu Gly Val Phe Tyr Arg Gly Ala 65 70 75 80
Asp Cys Cys Val Leu Val Phe Asp Val Thr Ala Pro Asn Thr Phe Lys 85 90 95
Thr Leu Asp Ser Trp Arg Asp Glu Phe Leu He Gin Ala Ser Pro Arg 100 105 110
Asp Pro Glu Asn Phe Pro Phe Val Val Leu Gly Asn Lys He Asp Leu 115 120 125
Glu Asn Arg Gin Val Ala Thr Lys Arg Ala Gin Ala Trp Cys Tyr Ser 130 135 140
Lys Asn Asn He Pro Tyr Phe Glu Thr Ser Ala Lys Glu Ala He Asn 145 150 155 160
Val Glu Gin Ala Phe Gin Thr He Ala Arg Asn Ala Leu Lys Gin Glu 165 170 175
Thr Glu Val Glu Leu Tyr Asn Glu Phe Pro Glu Pro He Lys Leu Asp 180 185 190
Lys Asp Ala Lys Thr Ser Ala Glu Cys Ser Cys 195 200
(2) INFORMATION FOR SEQ ID NO:24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 202 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyosteliu discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:
Met Thr Lys Lys Lys Val Leu Leu Lys Val He He Leu Gly Asp Ser
1 5 10 15
Gly Val Gly Lys Thr Ser Leu Met Asn Gin Tyr Val Asn Lys Lys Phe 20 25 30
Ser Asn Gin Tyr Lys Ala Thr He Gly Ala Asp Phe Leu Thr Lys Glu 35 40 45
Leu Met Val Asp Asp Arg Val Val Thr Met Gin He Trp Asp Thr Ala 50 55 60
Gly Gin Glu Arg Phe Gin Ser Leu Gly Val Ala Phe Tyr Arg Gly Ala 65 70 75 80
Asp Cys Cys Val Leu Cys Tyr Asp Val Asn Val Ala Lys Thr Phe Glu
85 90 95
Asn Leu Asp Ser Trp Arg Asp Glu Phe Leu He Gin Ala Gly Pro Arg 100 105 no
Asp Pro Asp Asn Phe Pro Phe Val Val Leu Gly Asn Lys He Asp Leu 115 120 125
Glu Asn Gin Arg Val Val Ser Gin Lys Arg Ala Ala Ser Trp Cys Gin 130 135 140
Ser Lys Gly Asn He Pro Tyr Phe Glu Thr Ser Ala Lys Glu Ala He 145 150 155 160
Asn Val Glu Gin Ala Phe Gin Thr He Ala Arg Asn Ala He Lys Leu 165 170 175
Glu Asp Gly Leu Val Phe Pro He Pro Thr Asn He Gin Val He Pro 180 185 190
Glu Pro Gin Pro Ala Lys Ser Gly Cys Cys 195 200
(2) INFORMATION FOR SEQ ID NO:25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 205 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Canis familiaris
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:
Met Lys Thr Tyr Asp Tyr Leu Phe Lys Leu Leu Leu He Gly Asp Ser
1 5 10 15
Gly Val Gly Lys Thr Cys Val Leu Phe Arg Phe Ser Glu Asp Ala Phe 20 25 30
Asn Ser Thr Phe He Ser Thr He Gly He Asp Phe Lys He Arg Thr
35 40 45
He Glu Leu Asp Gly Lys Arg He Lys Leu Gin He Trp Asp Thr Ala 50 55 60
Gly Gin Glu Arg Phe Arg Thr He Thr Thr Ala Tyr Tyr Arg Ala Met 65 70 75 80
Gly He Met Leu Val Tyr Asp He Thr Asn Glu Lys Ser Phe Asp Asn 85 90 95
He Arg Asn Trp He Arg Asn He Glu Glu His Ala Ser Ala Asp Val 100 105 110
Glu Lys Met He Leu Gly Asn Lys Cys Asp Val Asn Asp Lys Arg Gin
115 120 125
Val Ser Lys Glu Arg Gly Glu Lys Leu Ala Leu Asp Tyr Gly He Lys 130 135 140
Phe Met Glu Thr Ser Ala Lys Ala Asn He Asn Val Glu Asn Ala Phe 145 150 155 160
Phe Thr Leu Ala Arg Asp He Lys Ala Lys Met Asp Lys Lys Leu Glu 165 170 175
Gly Asn Ser Pro Gin Gly Ser Asn Gin Gly Val Lys He Thr Pro Asp 180 185 190
Gin Gin Lys Arg Ser Ser Phe Phe Arg Cys Val Leu Leu 195 200 205
(2) INFORMATION FOR SEQ ID NO:26:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 191 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:26:
Met Glu Glu Glu He Leu Tyr Lys He He Leu Val Gly Glu Ser Gly 1 5 10 15
Val Gly Lys Ser Ser He Leu Val Arg Phe Thr Asp Asn Thr Phe Ser 20 25 30
Gin His Phe Ala Pro Thr Leu Gly Val Phe Val Lys Thr He Arg Asn 35 40 45
Lys Glu Thr Gly Gin Thr Val Lys Leu Gin Leu Trp Asp Thr Ala Gly 50 55 60
Gin Glu Arg Phe Lys Ser He Thr Gin Phe Tyr Arg Gly Ser His Gly 65 70 75 80
Val He Val Val Tyr Asp Val Thr Asp Pro Lys Ser Phe Glu Arg Leu 85 90 95
Lys Asn Trp Val Glu Asp He Asn Gin Tyr Thr Gin Asp Gly Met He 100 105 110
He He Leu Val Gly Asn Lys Ser Asp Met Val Ala Gin Arg Lys Val 115 120 125
Thr Phe Glu Gin Gly Gin Glu Met Ala Glu Gin Leu Lys Thr Lys Phe 130 135 140
Leu Glu Val Ser Ala Lys Glu Asn Asn Gly Val Thr Gin Val Phe Asp 145 150 155 160
Leu Leu Val Gin Asp He Glu Ala Thr Met Lys Asn Ser Lys Val Ala 165 170 175
Gin Asn Gin Leu Asn Leu Ser Val Gly Gin Glu Arg Gly Cys Cys 180 185 190
(2) INFORMATION FOR SEQ ID NO:27:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 189 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Caenorhabditis elegans
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:27:
Met Gin Ala He Lys Cys Val Val Val Gly Asp Gly Ala Val Gly Lys 1 5 10 15
Thr Cys Leu Leu He Ser Tyr Thr Thr Asn Ala Phe Pro Gly Glu Tyr 20 25 30
He Pro Thr Val Phe Asp Asn Tyr Ser Ala Asn Val Met Val Asp Gly 35 40 45
Arg Pro He Asn Leu Gly Leu Trp Asp Thr Ala Gly Gin Asp Tyr Asp 50 55 60
Arg Leu Arg Pro Leu Ser Tyr Pro Gin Thr Asp Val Phe Leu Val Cys 65 70 75 80
Phe Ala Leu Asn Asn Pro Ala Ser Phe Glu Asn Val Arg Ala Lys Trp 85 90 95
Tyr Pro Glu Val Ser His His Cys Pro Asn Thr Pro He He Leu Val 100 105 110
Gly Thr Lys Ala Asp Leu Arg Glu Asp Asp Thr Val Glu Arg Leu Arg 115 120 125
Glu Arg Arg Leu Gin Pro Val Ser Gin Thr Gin Gly Tyr Val Met Ala 130 135 140
Lys Glu He Lys Ala Val Lys Tyr Leu Glu Cys Ser Ala Leu Thr Gin 145 150 155 160
Arg Gly Leu Lys Gin Val Phe Asp Glu Ala He Arg Ala Val Val Thr 165 170 175
Pro Pro Gin Arg Ala Lys Lys Ser Lys Cys Thr Val Leu 180 185
(2) INFORMATION FOR SEQ ID NO:28:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 191 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:28: Met Gin Ala He Lys Cys Val Val Val Gly Asp Gly Ala Val Gly Lys 1 5 10 15
Thr Cys Leu Leu He Ser Tyr Thr Thr Asn Ala Phe Pro Gly Glu Tyr 20 25 30
He Pro Thr Val Phe Asp Asn Tyr Ser Ala Asn Val Met Val Asp Gly 35 40 45
Lys Pro He Asn Leu Gly Leu Trp Asp Thr Ala Gly Gin Glu Asp Tyr 50 55 60
Asp Arg Leu Arg Pro Leu Ser Tyr Pro Gin Thr Asp Val Phe Leu He 65 70 75 80
Cys Phe Ser He He Ser Pro Ser Ser Phe Glu Asn Val Asn Gly Lys 85 90 95
Trp His Pro Glu He Cys His His Pro Asn Val Pro He Leu Val Gly 100 105 110
Thr Lys Leu Asp Met Arg Asp Lys Glu Thr Gin Asp Arg Leu Lys Glu 115 120 125
Lys Lys Leu Tyr Pro He Ser Tyr Glu Gin Gly Leu Ala Lys Met Lys 130 135 140
Glu He Asn Ala Val Lys Tyr Leu Glu Cys Ser Ala Leu Thr Glu Lys 145 150 155 160
Gly Leu Lys Thr Val Phe Asp Glu Ala He Arg Ala Val He Asn Pro 165 . 170 175
Pro Leu Ser Lys Lys Lys Lys Ser Ser Gly Gly Cys Asn He Leu 180 185 190
(2) INFORMATION FOR SEQ ID NO:29:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 192 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:29:
Met Gin Ser He Lys Leu Val Val Val Gly Asp Gly Ala Val Gly Lys 1 5 10 15
Thr Cys Leu Leu He Ser Tyr Thr Ser Asn Ser Phe Pro Thr Glu Tyr 20 25 30
Val Pro Thr Val Phe Asp Asn Tyr Ser Ala Asn Val Met Val Asp Asn 35 40 45
Lys Thr Val Ser Leu Gly Leu Trp Asp Thr Ala Gly Gin Glu Asp Tyr 50 55 60
Asp Arg Leu Arg Pro Leu Ser Tyr Pro Gin Thr Asp Val Phe Leu He 65 70 75 80
Cys Phe Ala He He Ser Gin Ser Tyr Thr Asn Val Lys Ser Lys Trp 85 90 95
Trp Pro Glu Val Thr His His Cys Pro Asn Cys Thr He Leu Val Gly 100 105 110
Thr Lys Cys Asp Leu Arg Asp Lys Glu Ser Leu Glu Lys Leu Arg Glu 115 120 125
Lys His Gin Gin Pro Leu Thr Phe Gin Gin Gly Glu Gin Met Ala Lys 130 135 140
Glu He Lys Ala Phe Cys Tyr Met Glu Cys Ser Ala Leu Thr Gin Lys 145 150 155 160
Gly Leu Lys Gin Val Phe Asp Glu Ala He Lys Ala Val He Phe Pro 165 170 175
Asp Arg Asp Lys Ala Thr Asn Lys Lys Asn Ser Lys Cys Ser He Leu 180 185 190
(2) INFORMATION FOR SEQ ID NO:30:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 185 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:30:
Met Ser Ala Ala Glu Val He Lys Leu Val Val He Gly Gly Ala Val 1 5 10 15
Gly Lys Thr Cys Leu Leu He Tyr Ala Asn Asn Arg Phe Pro Glu Asp 20 25 30
Tyr He Pro Thr Val Phe Asp Asn Tyr Val Val Asn Leu Thr Ala Gly 35 40 45
Asp Arg Asn He Glu Leu Gly Leu Trp Asp Thr Ala Gly Glu Tyr Asp 50 55 60
Lys Leu Arg Pro Leu Ser Tyr Ala Asn Asn Val Phe Leu He Cys Phe 65 70 75 80
Ser He Asn Pro Val Ser Phe Glu Asn Val Tyr Thr Lys Trp Tyr Pro 85 90 95
Glu Val Met His Phe Cys Pro Glu Val Gin He Leu Val Gly Thr Lys 100 105 110
Leu Asp Thr Arg Asp Asp Arg Gly Val Leu Asp Lys Leu Gin Gin Thr 115 120 125 Gly His Lys Pro He Thr Thr Glu Gin Gly Asn Asp Leu Ala Arg Arg 130 135 140
He Lys Ala He Lys Tyr Met Glu Cys Ser Ala Lys Thr Ser Gin Asn 145 150 155 160
Leu Lys Gin Val Phe Asp Glu Ala He Lys Ser Val Leu Phe He Lys 165 170 175
Lys Lys Lys Ser Lys Cys He Val Met 180 185
(2) INFORMATION FOR SEQ ID NO: 31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 205 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
( i) SEQUENCE DESCRIPTION: SEQ ID NO:31:
Met Ala Ala Asn Lys Pro Lys Gly Gin Asn Ser Leu Ala Leu His Lys 1 5 10 15
Val He Met Val Gly Ser Gly Gly Val Gly Lys Ser Ala Leu Thr Leu 20 25 30
Gin Phe Met Tyr Asp Glu Phe Val Glu Asp Tyr Glu Pro Thr Lys Ala 35 40 45
Asp Ser Tyr Arg Lys Lys Val Val Leu Asp Gly Glu Glu Val Gin He 50 55 60
Asp He Leu Asp Thr Ala Gly Gin Glu Asp Tyr Ala Ala He Arg Asp 65 70 75 80
Asn Tyr Phe Arg Ser Gly Glu Gly Phe Leu Cys Val Phe Ser He Thr 85 90 95
Glu Met Glu Ser Phe Ala Ala Thr Ala Asp Phe Arg Glu Gin He Leu 100 105 110
Arg Val Lys Glu Asp Glu Asn Val Pro Phe Leu Leu Val Gly Asn Lys 115 120 125
Ser Asp Leu Glu Asp Lys Arg Gin Val Ser Val Glu Glu Ala Lys Asn 130 135 140
Arg Ala Glu Gin Trp Asn Val Asn Tyr Val Glu Thr Ser Ala Lys Thr 145 150 155 160
Arg Ala Asn Val Asp Lys Val Phe Phe Asp Leu Met Arg Glu He Arg 165 170 175
Ala Arg Lys Met Glu Asp Ser Lys Lys Asn Gly Lys Lys Lys Arg Lys 180 185 190 Ser Leu Ala Lys Arg He Arg Glu Arg Cys Cys He Leu 195 200 205
(2) INFORMATION FOR SEQ ID NO:32:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 204 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:32:
Met Ala Ala Asn Lys Ser Lys Gly Gin Ser Ser Leu Ala Leu His Lys 1 5 10 15
Val He Met Val Gly Ser Gly Gly Val Gly Lys Ser Ala Leu Thr Leu 20 25 30
Gin Phe Met Tyr Asp Glu Phe Val Glu Asp Tyr Glu Pro Thr Lys Ala 35 40 45
Asp Ser Tyr Arg Lys Lys Val Val Leu Asp Gly Glu Glu Val He Asp 50 55 60
He Leu Asp Thr Ala Gly Gin Glu Asp Tyr Ala He Arg Asp Asn Tyr 65 70 75 80
Phe Arg Ser Gly Glu Gly Phe Leu Leu Val Phe Ser He Thr Glu His 85 90 95
Glu Ser Phe Thr Ala Thr Ala Glu Phe Arg Glu Gin He Leu Arg Val 100 105 110
Lys Ala Glu Glu Asp Lys He Pro Leu Leu Val Val Gly Asn Lys Ser 115 120 125
Asp Leu Glu Glu Arg Arg Gin Val Pro Val Glu Glu Ala Arg Ser Lys 130 135 140
Ala Glu Glu Trp Gly Val Gin Tyr Val Glu Thr Ser Ala Lys Thr Arg 145 150 155 160
Ala Asn Val Asp Lys Val Phe Phe Asp Leu Met Arg Glu He Arg Thr 165 170 175
Lys Lys Met Ser Glu Asn Lys Asp Lys Asn Gly Lys Lys Ser Ser Lys 180 185 190
Asn Lys Lys Ser Phe Lys Glu Arg Cys Cys Leu Leu 195 200
(2) INFORMATION FOR SEQ ID NO:33:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 200 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Discopyge ommata
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:33:
Met Ala Ala Asn Lys Asn Lys Asn Gin Ser Ser Leu Leu Lys Val He 1 5 10 15
Met Val Gly Ser Gly Gly Val Gly Lys Ser Ala Leu Thr Leu Gin Phe 20 25 30
Met Tyr Asp Glu Phe Val Glu Asp Tyr Glu Pro Thr Lys Ala Asp Ser 35 40 45
Tyr Arg Lys Lys Val Val Leu Asp Gly Glu Val Gin He Asp He Leu 50 55 60
Asp Thr Ala Gly Gin Glu Asp Tyr Ala He Arg Asp Asn Tyr Phe Arg 65 70 75 80
Ser Gly Glu Gly Phe Leu Cys Val Phe Ser He Glu Gin Glu Ser Phe 85 90 95
Thr Ala Thr Val Glu Phe Arg Glu Gin He Leu Arg Val Lys Glu Glu 100 105 110
Asp Lys He Pro Leu Leu Leu Val Gly Asn Lys Ser Asp Leu Glu Asp 115 120 125
Arg Arg Gin Val Ser He Glu Glu Ala Arg Ser Lys Ala Glu Glu Trp 130 135 140
Gly Val Gin Tyr Val Glu Thr Ser Ala Lys Thr Arg Ala Asn Val Asp 145 150 155 160
Lys Val Phe Phe Asp Leu Met Arg Glu Val Arg Ala Lys Lys Met Ser 165 170 175
Glu Asn Lys Asp Lys Asn Gly Lys Lys Ser Ser Arg Asn Lys Lys Ser 180 185 190
Leu Arg Glu Arg Cys Cys He Leu 195 200
(2) INFORMATION FOR SEQ ID NO:34:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 194 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Discopyge ommata (xi) SEQUENCE DESCRIPTION: SEQ ID NO:34:
Met Ala Lys Lys Thr Tyr Asp Leu Leu Phe Lys Leu Leu Leu He Gly 1 5 10 15
Asp Ser Gly Val Gly Lys Thr Cys Val Leu Phe Arg Phe Ser Asp Asp 20 25 30
Ala Phe Asn Thr Thr Phe He Ser Thr He Gly He Asp Phe Lys He 35 40 45
Lys Thr Val Glu Leu His Gly Lys Lys He Lys Leu Gin He Trp Asp 50 55 60
Thr Ala Gly Gin Glu Arg Phe His Thr He Thr Ser Tyr Tyr Arg Gly 65 70 75 80
Ala Met Gly He Met Leu Val Tyr Asp He Thr Asn Ala Lys Ser Phe 85 90 95
Glu Asn He Ser Lys Trp Leu Arg Asn He Asp Glu His Ala Asn Glu 100 105 110
Asp Val Glu Arg Met Leu Leu Gly Asn Lys Asp Met Glu Asp Lys Arg 115 120 125
Val Val Leu Lys Ser Lys Gly Gin He Ala Glu His Ala He Arg Phe 130 135 140
Phe Glu Thr Ser Ala Lys Ala Asn He Asn He Glu Lys Ala Phe Leu 145 150 155 160
Thr Leu Ala Glu Asp He Leu Gin Lys Thr Pro Val Lys Glu Pro Asp 165 170 175
Arg Glu Asn Val Asp He Ser Thr Gly Gly Gly Gly Leu Lys Lys Cys 180 185 190
Cys Ser
(2) INFORMATION FOR SEQ ID NO:35:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 207 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Discopyge ommata
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 35:
Met Lys Thr Tyr Asp Tyr Leu Phe Lys Leu Leu Leu He Gly Asp Ser 1 5 10 15
Gly Val Gly Lys Thr Cys Leu Leu Phe Arg Phe Ser Glu Asp Ala Phe 20 25 30 Asn Thr Thr Phe He Ser Thr He Gly He Asp Phe Lys He Arg Thr 35 40 45
Val Glu Leu Asp Gly Lys Lys He Lys Leu Gin He Trp Asp Thr Ala 50 55 60
Gly Gin Glu Arg Phe Arg Thr He Thr Ala Tyr Tyr Arg Gly Ala Met 65 70 75 80
Gly He Met Lys Val Asp He Thr Asn Glu Lys Ser Phe Asp Asn He 85 90 95
Lys Asn Trp He Arg Asn He Glu Glu His Ala Ser Ser Asp Val Glu 100 105 no
Arg Met He Leu Gly Asn Lys Cys Asp Met Asn Glu Lys Arg Gin Val 115 120 125
Ser Lys Glu Arg Gly Glu Lys Leu Ala He Asp Tyr Gly He Lys Phe 130 135 140
Leu Glu Thr Ser Ala Lys Ser Ser He Asn Val Glu Glu Ala Phe He 145 150 155 160
Thr Leu Ala Arg Asp He Met Thr Lys Leu Asn Lys Lys Met Asn Glu 165 170 175
Asn Ser Leu Gin Glu Ala Val Asp Lys Leu Lys Ser Pro Pro Lys Lys 180 185 190
Pro Ser Gin Lys Lys Lys Gin Leu Ser Phe Arg Cys Ser Leu Leu 195 200 205
(2) INFORMATION FOR SEQ ID NO:36:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 213 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Discopyge ommata
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:36:
Met Gly Thr Arg Asp Asp Glu Tyr Asp Tyr Leu Phe Lys Val Val Leu 1 5 10 15
He Gly Asp Ser Gly Val Gly Lys Ser Asn Leu Leu Ser Arg Phe Thr 20 25 30
Arg Glu Phe Asn Leu Glu Ser Lys Ser Thr He Gly Val Glu Phe Ala 35 40 45
Thr Arg Ser He Gin Val Asp Gly Lys Thr He Lys Gin He Trp Asp 50 55 60
Thr Gly Gin Glu Arg Tyr Arg Ala He Thr Ser Ala Tyr Tyr Arg Gly 65 70 75 80 Ala Val Gly Ala Leu Leu Val Tyr Asp He Ala Lys His Leu Thr Tyr 85 90 95
Glu Asn Val Glu Arg Trp Leu Lys Glu Leu Arg Asp His Ala Asp Asn 100 105 110
Asn He Val He Met Leu Val Gly Asn Lys Ser Asp Leu Arg His Leu 115 120 125
Arg Val Pro Thr Asp Ala Arg Ala Phe Ala Glu Lys Asn Asn Leu Ser 130 135 140
Phe He Glu Thr Ser Ala Leu Asp Ser Thr Asn Val Glu Glu Ala Phe 145 150 155 160
Lys Asn He Leu Thr Glu He Tyr Arg He Val Ser Gin Lys Gin He 165 170 175
Ser Asp Arg Ser Ala His Asp Glu Ser Pro Gly Asn Asn Val Val Asp 180 185 190
He Ser Val Pro Pro Thr Thr Asp Gly Gin Lys Ser Asn Lys Leu Gin 195 200 205
Cys Cys Gin Asn Met 210
(2) INFORMATION FOR SEQ ID NO:37:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 184 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 37:
Met Pro Leu Arg Phe Lys He Val Val Leu Gly Ser Gly Gly Val Gly 1 5 10 15
Lys Ser Ala Leu Thr Val Gin Phe Val Gin Gly He Phe Val Glu Lys 20 25 30
Tyr Asp Pro Thr He Glu Asp Ser Tyr Arg Lys Gin Val Glu Val Asp 35 40 45
Ser Asn Gin Cys Met Leu Glu He Leu Asp Thr Ala Gly Thr Glu Gin 50 55 60
Phe Thr Met Arg Asp Leu Tyr Met Lys Asn Gly Gin Gly Phe Val Leu 65 70 75 80
Val Tyr Ser He He Ser Asn Ser Thr Phe Asn Glu Leu Pro Asp Leu 85 90 95
Arg Glu Gin He Leu Arg Val Lys Asp Cys Glu Asp Val Pro Met Val 100 105 110 Leu Val Gly Asn Lys Cys Asp Leu His Asp Gin Arg Val He Ser Thr 115 120 125
Glu Gin Gly Glu Glu Leu Ala Arg Lys Phe Gly Asp Cys Tyr Phe Leu 130 135 140
Glu Ala Ser Ala Lys Asn Lys Val Asn Val Glu Gin He Phe Tyr Asn 145 150 155 160
Leu He Arg Gin He Asn Arg Lys Asn Pro Val Gly Pro Pro Ser Lys 165 170 175
Ala Lys Ser Lys Cys Ala Leu Leu 180
(2) INFORMATION FOR SEQ ID NO:38:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 179 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:38:
Met Arg Glu Tyr Lys Val Val Val Leu Gly Ser Gly Gly Val Gly Lys 1 5 10 15
Ser Ala Leu Thr Val Gin Phe Val Thr Gly Thr Phe He Glu Lys Tyr 20 25 30
Asp Pro Thr He Glu Asp Phe Tyr Arg Lys Glu He Glu Val Asp Ser 35 40 45
Ser Pro Ser Val Leu Glu He Leu Asp Thr Ala Gly Thr Glu Gin Phe 50 55 60
Ala Ser Arg Asp Leu Tyr He Lys Asn Gly Gin Gly Phe He Leu Val 65 70 75 80
Tyr Ser Leu Val Asn Gin Gin Phe Gin Asp He Lys Pro Met Arg Asp 85 90 95
Gin He He Arg Val Lys Tyr Glu Lys Val Pro Val He Leu Val Gly 100 105 110
Asn Lys Val Asp Leu Glu Ser Glu Arg Glu Val Ser Ser Ser Glu Gly 115 120 125
Arg Ala Leu Ala Glu Glu Trp Gly Cys Pro Phe Met Glu Thr Ser Ala 130 135 140
Lys Ser Lys Thr Met Val Asp Glu Leu Phe Ala Glu He Val Arg Gin 145 150 155 160
Met Asn Tyr Ala Ala Gin Pro Asp Lys Asp Asp Pro Cys Cys Ser Ala 165 170 175 Cys Asn Gin
(2) INFORMATION FOR SEQ ID NO: 39:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 183 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:39:
Met Arg Glu Tyr Lys Val Val Val Leu Gly Ser Gly Gly Val Gly Lys 1 5 10 15
Ser Ala Leu Thr Val Gin Phe Val Thr Gly Ser Phe He Glu Lys Tyr 20 25 30
Asp Pro Thr He Glu Asp Phe Tyr Arg Lys Glu He Glu Val Asp Ser 35 40 45
Ser Pro Ser Val Leu Glu He Leu Asp Thr Ala Gly Thr Glu Gin Phe 50 55 60
Ala Ser Met Arg Asp Leu Tyr He Lys Asn Gly Gin Gly Phe He Leu 65 70 75 80
Val Tyr Ser Leu Val Asn Gin Gin Ser Phe Gin Asp He Lys Pro Met 85 90 95
Arg Asp Gin He He Arg Val Lys Arg Tyr Glu Arg Val Pro Met He 100 105 110
Leu Val Gly Asn Lys Val Asp Leu Glu Gly Glu Arg Glu Val Ser Tyr 115 120 125
Gly Glu Gly Lys Ala Leu Ala Glu Glu Trp Ser Cys Pro Phe Met Glu 130 135 140
Thr Ser Ala Lys Asn Lys Ala Ser Val Asp Glu Leu Phe Ala Glu He 145 150 155 160
Val Arg Gin Met Asn Tyr Ala Ala Gin Ser Asn Gly Asp Glu Gly Cys 165 170 175
Cys Ser Ala Cys Val He Leu 180
(2) INFORMATION FOR SEQ ID NO:40:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 184 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Discopyge ommata
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:40:
Met Arg Glu Tyr Lys Leu Val Val Leu Gly Ser Gly Gly Val Gly Lys 1 5 10 15
Ser Ala Leu Thr Val Gin Phe Val Gin Gly He Phe Val Glu Lys Tyr 20 25 30
Asp Pro Thr He Glu Asp Ser Tyr Arg Lys Gin Val Glu Val Asp Cys 35 40 45
Gin Pro Cys Met Leu Glu He Leu Asp Thr Ala Gly Thr Glu Gin Phe 50 55 60
Thr Ala Met Arg Asp Leu Tyr Met Lys Asn Gly Gin Gly Phe Ala Leu 65 70 75 80
Val Tyr Ser He Thr Ala Gin Ser Thr Phe Asn Asp Leu Gin Asp Leu 85 90 95
Arg Glu Gin He Leu Arg Val Lys Asp Thr Glu Asp Val Pro Met He 100 105 110
Leu Val Gly Asn Lys Cys Asp Leu Glu Asp Glu Arg Val Val Gly Lys 115 120 125
Glu Gin Gly Gin Asn Leu Ala Arg Gin Trp Asn Asn Cys Ala Phe Leu 130 135 140
Glu Ser Ser Ala Lys Ser Lys He Asn Val Asn Glu He Phe Tyr Asp 145 150 155 160
Leu Val Arg Gin He Asn Arg Lys Ala Pro Val Glu Lys Cys Lys Lys 165 170 175
Lys Lys Ser Gin Cys Thr Leu Leu 180
(2) INFORMATION FOR SEQ ID NO: 1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 180 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:41:
Met Arg Glu Tyr Lys Leu Val Val Gly Ser Gly Gly Val Gly Lys Ser 1 5 10 15 Ala Leu Thr Val Gin Phe Val Gin Gly Phe Val Glu Lys Tyr Asp Pro 20 25 30
Thr He Glu Asp Ser Tyr Arg Lys Gin Val Glu Val Asp Cys Gin Gin 35 40 45
Cys Met Leu Glu Asp Thr Ala Gly Thr Glu Gin Phe Thr Ala Met Arg 50 55 60
Asp Leu Tyr Met Lys Asn Gly Gin Gly Phe Ala Leu Val Tyr Ser He 65 70 75 80
Thr Ala Gin Ser Thr Phe Asn Asp Leu Gin Asp Leu Arg Glu Gin He 85 90 95
Leu Arg Val Lys Asp Thr Glu Asp Val Pro Met He Leu Val Gly Asn 100 105 110
Lys Cys Asp Leu Glu Asp Glu Arg Val Val Gly Lys Glu Gin Gly Gin 115 120 125
Asn Leu Ala Arg Gin Trp Cys Asn Cys Ala Phe Leu Glu Ser Ser Ala 130 135 140
Lys Ser Lys He Asn Val Asn Glu He Phe Tyr Asp Leu Val Arg Gin 145 150 155 160
He Asn Arg Lys Thr Pro Val Glu Lys Lys Lys Pro Lys Lys Lys Ser 165 170 175
Cys Leu Leu Leu 180
(2) INFORMATION FOR SEQ ID NO:42:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 179 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:42:
Met Arg Glu Tyr Lys Leu Val Val Leu Gly Ser Gly Gly Val Gly Lys 1 5 10 15
Ser Ala Leu Thr Val Gin Phe Val Gin Gly He Phe Val Glu Lys Tyr 20 25 30
Asp Pro Thr He Glu Asp Ser Tyr Arg Lys Gin Val Glu Val Asp Ala 35 40 45
Gin Gin Cys Met Leu Glu He Leu Asp Thr Ala Gly Thr Glu Gin Phe 50 55 60
Thr Ala Met Arg Asp Leu Tyr Met Lys Asn Gly Gin Gly Phe Ala Leu 65 70 75 80 Val Tyr Ser He Thr Ala Gin Ser Thr Phe Asn Asp Leu Gin Asp Leu 85 90 95
Arg Glu Gin He Leu Arg Val Lys Asp Thr Asp Asp Val Pro Met He 100 105 110
Leu Val Gly Asn Lys Cys Asp Leu Glu Asp Glu Arg Val Val Gly Lys 115 120 125
Glu Gin Gly Gin Asn Leu Ala Arg Gin Trp Asn Asn Cys Ala Phe Leu 130 135 140
Glu Ser Ser Ala Lys Ser Lys He Asn Val Glu He Phe Tyr Asp Leu 145 150 155 160
Val Arg Gin He Asn Arg Lys Thr Pro Val Pro Gly Lys Ala Arg Lys 165 170 175
Lys Ser Ser
(2) INFORMATION FOR SEQ ID NO:43:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 184 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Drosophila melanogaster
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:43:
Met Arg Glu Tyr Lys He Val Val Leu Gly Ser Gly Gly Val Gly Lys 1 5 10 15
Ser Ala Leu Thr Val Gin Phe Val Gin Cys He Phe Val Glu Lys Tyr 20 25 30
Asp Pro Thr He Glu Asp Ser Tyr Arg Lys Gin Val Glu Val Asp Gly 35 40 45
Gin Gin Cys Met Leu Glu He Leu Asp Thr Ala Gly Thr Glu Gin Phe 50 55 60
Thr Ala Met Arg Asp Leu Tyr Met Lys Asn Gly Gin Gly Phe Val Leu 65 70 75 80
Val Tyr Ser He Thr Ala Gin Ser Thr Phe Asn Asp Leu Gin Asp Leu 85 90 95
Arg Glu Gin He Leu Arg Val Lys Asp Thr Asp Asp Val Pro Met Val 100 105 110
Leu Val Gly Asn Lys Cys Asp Leu Glu Glu Glu Arg Val Val Gly Lys 115 120 125
Glu Leu Gly Lys Asn Leu Ala Thr Gin Phe Asn Cys Ala Phe Met Glu 130 135 140 Thr Ser Ala Lys Ala Lys Val Asn Val Asn Asp He Phe Tyr Asp Leu 145 150 155 160
Val Arg Gin He Asn Lys Lys Ser Pro Glu Lys Lys Gin Lys Lys Pro 165 170 175
Lys Lys Ser Leu Cys Val Leu Leu 180
(2) INFORMATION FOR SEQ ID NO:44:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 182 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:44:
Met Thr Glu Tyr Lys Leu Val He Val Gly Gly Gly Gly Val Gly Lys 1 5 10 15
Ser Leu Thr He Gin Leu He Gin Asn His Phe Asp Glu Tyr Asp Pro 20 25 30
Thr He Glu Asp Ser Tyr Arg Lys Gin Val Ser He Asp Asp Glu Thr 35 40 45
Cys Leu Leu He Leu Asp Thr Ala Gly Gin Glu Glu Ser Ala Met Arg 50 55 60
Asp Gin Tyr Met Arg Thr Gly Gin Gly Phe Leu Cys Val Tyr Ser He 65 70 75 80
Thr Ser Arg Ser Ser Tyr Asp Glu He Ala Ser Phe Arg Glu Gin He 85 90 95
Leu Arg Val Lys Asp Lys Asp Arg Val Pro Leu He Leu Val Gly Asn 100 105 110
Lys Ala Asp Leu Asp His Glu Arg Gin Val Ser Val Asn Glu Gly Gin 115 120 125
Glu Leu Ala Lys Asp Ser Leu Ser Phe His Glu Ser Ser Ala Lys Ser 130 135 140
Arg He Asn Val Glu Glu Ala Phe Tyr Ser Leu Val Arg Glu He Arg 145 150 155 160
Lys Glu Leu Lys Gly Asp Gin Ser Ser Gly Lys Ala Gin Lys Lys Lys 165 170 175
Lys Gin Cys Leu He Leu 180
(2) INFORMATION FOR SEQ ID NO:45:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 190 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:45:
Met Ser Val Ser Asn Glu Tyr Lys Leu Val Val Gly Gly Gly Gly Val 1 5 10 15
Gly Lys Ser Ala Leu Thr He Gin Phe Gin Asn His Phe He Glu Glu 20 25 30
Tyr Asp Pro Thr He Glu Asp Ser Tyr Arg Arg Gin Cys Gin Val Asp 35 40 45
Glu Asp Thr Cys Leu Leu Asp He Leu Asp Thr Ala Gly Gin Asp Asp 50 55 60
Tyr Ser Met Arg Asp Gin Tyr Met Arg Thr Gly Gin Gly Phe Leu Val 65 70 75 80
Tyr Asp Val Ser Arg Thr Ser Phe Glu Glu He Asn Val Val Glu Gin 85 90 95
He Arg Val Lys Asp Asn Asp Lys Val Pro He Val Leu Val Gly Asn 100 105 110
Lys Cys Asp Leu Glu Asn Leu Arg Glu Val Thr Glu Gly Glu Gly Ser 115 120 125
Glu Leu Ala Lys Ser Phe Ser Val Pro Phe Leu Glu Thr Ser Ala Lys 130 135 140
Lys Arg Leu Asn Val Asp Glu Cys Phe Phe Glu Val Val Arg Glu He 145 150 155 160
Lys Lys Ser Leu Lys Glu Pro Gly Arg Ser Lys Lys Asp Lys Lys Gly 165 170 175
Gly He Leu Lys Lys Phe Lys Gly Gly Asp Cys Leu He Leu 180 185 190
(2) INFORMATION FOR SEQ ID NO:46:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 188 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum (xi) SEQUENCE DESCRIPTION: SEQ ID NO:46:
Met Ser Lys Leu Leu Lys Leu Val He Val Gly Asp Gly Gly Val Gly 1 5 10 15
Lys Ser Ala Leu Thr He Gin Leu Thr Gin Asn Gin Phe He Ala Glu 20 25 30
Tyr Asp Pro Thr He Glu Asn Ser Tyr Arg Lys Gin Val Asn He Asp 35 40 45
Glu Glu Val Tyr Met Leu Asp He Leu Asp Thr Ala Gly Gin Glu Glu 50 55 60
Tyr Ser Ala Met Arg Asp Gin Tyr He Arg Ser Gly Arg Gly Phe Leu 65 70 75 80
He Val Tyr Ser He He Ser Arg Ala Ser Phe Glu Ala Val Thr Thr 85 90 95
Phe Arg Glu Gin He Leu Arg Val Lys Asp Leu Ser Thr Tyr Pro He 100 105 110
Val He He Gly Asn Lys Ala Asp Leu Pro Asp Lys Asp Arg Lys Val 115 120 125
Pro Pro Met Glu Gly Lys Glu Leu Ala Lys Phe Gly Ala Pro Phe Leu 130 135 140
Glu Thr Ser Ala Lys Ser Arg Val Asn Val Glu Glu Ala Phe Phe Thr 145 150 155 160
Leu Val Arg Glu He Lys Arg Trp Asn Gin Asn Pro Gin Asn Glu Glu 165 170 175
Met Leu Pro Pro Lys Lys Arg Gly Cys He He Leu 180 185
(2) INFORMATION FOR SEQ ID NO:47:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 188 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Dictyostelium discoideum
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:47:
Met Glu Tyr Lys Leu Val He Val Gly Gly Gly Gly Val Gly Lys Ser
1 5 10 15
Ala Leu Thr He Gin Leu He Gin Asn His Phe He Asp Glu Tyr Asp
20 25 30
Pro Thr He Glu Asp Ser Tyr Arg Lys Gin Val Thr He Asp Glu Glu
35 40 45 Thr Cys Leu Leu Asp He Leu Asp Thr Ala Gly Gin Glu Glu Tyr Ser 50 55 60
Ala Met Arg Asp Gin Tyr Met Arg Thr Gly Gin Gly Phe Leu Cys Val 65 70 75 80
Tyr Ser He Thr Ser Arg Ser Ser Phe Asp Glu He Ala Ser Phe Arg 85 90 95
Glu Gin He Leu Arg Val Lys Asp Lys Asp Arg Val Pro Met He Val 100 105 110
Val Gly Asn Lys Cys Asp Leu Glu Ser Asp Arg Gin Val Thr Thr Gly 115 120 125
Glu Gly Gin Asp Leu Ala Lys Ser Phe Gly Ser Pro Phe Leu Glu Thr 130 135 140
Ser Ala Lys He Arg Val Asn Val Glu Glu Ala Phe Tyr Ser Leu Val 145 150 155 160
Arg Glu He Arg Lys Asp Leu Lys Gly Asp Ser Lys Pro Glu Lys Gly 165 170 175
Lys Lys Lys Arg Pro Leu Lys Ala Cys Thr Leu Leu 180 185
(2) INFORMATION FOR SEQ ID NO:48:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 204 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Caenorhabditis elegans
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:48:
Met Ser Ser Ser Leu Gin Ser Asn Arg Gin Ser Leu Asn Arg Lys Val 1 5 10 15
Ala Val Met Gly Tyr Pro His Val Gly Lys Ser Ala Leu Val Leu Arg 20 25 30
Phe Thr Gin Asn He Phe Pro Glu Arg Tyr Glu Ser Thr He Glu Asp 35 40 45
Gin His Ser Lys His He Ala Ala Phe His Arg Asp Tyr His Leu Arg 50 55 60
Val Thr Asp Thr Ala Gly Gin Gin Glu Tyr Thr Val Phe Pro Arg Ser
65 70 75 80
Cys Ser Leu Asp He Asn Gly Phe He Leu Val Tyr Ala He Asp Asp 85 90 95
Arg Lys Ser Phe Glu Met Cys Ser Asn He Tyr Glu Lys He Val Arg 100 105 110 Thr Tyr Gly Asp Thr Ser He Pro He Val He Val Gly Lys Thr Asp 115 120 125
Leu Ser Thr Gin Val Val Arg Ala Glu Glu Gly Glu Glu Leu Ala Arg 130 135 140
Gin Trp Asp Ala Lys Phe Val Glu He Thr Ala Arg Glu Ser Asn Arg 145 150 155 160
Val His Glu Val Phe Glu Leu Leu Leu Arg Glu He Glu He Ser Arg 165 170 175
Gly Asn Leu Ser Pro Thr Glu Arg Pro Asn Gly Asn Ser Pro Lys Arg 180 185 190
Pro Phe Lys Asp Asp Gly Lys Pro Cys Ser He Ser 195 200
(2) INFORMATION FOR SEQ ID NO:49:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 215 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Coprinus cinereus
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:49:
Met Ala Ala Arg Ala Gin Phe Leu Arg Glu Tyr Lys Leu Val Val Val 1 5 10 15
Gly Gly Gly Gly Val Gly Lys Ser Ala Leu Thr He Gin Phe He Gin 20 25 30
Ser His Phe Val Asp Glu Tyr Asp Pro Thr He Glu Asp Ser Tyr Arg 35 40 45
Lys Gin Cys He He Asp Asp Glu Val Ala Leu Leu Asp Val Leu Asp 50 55 60
Thr Ala Gly Gin Glu Glu Tyr Gly Ala Met Arg Glu Gin Tyr Met Arg 65 70 75 80
Thr Gly Glu Gly Phe Leu Leu Val Tyr Ser He Thr Ser Arg Asn Ser 85 90 95
Phe Glu Glu He Ser He Phe His Gin Gin He Leu Arg Val Lys Asp 100 105 110
Gin Asp Ser Phe Pro Val He Val Val Ala Asn Lys Cys Asp Leu Glu 115 120 125
Tyr Glu Arg Gin Val Gly Met Asn Glu Gly Arg Asp Leu Ala Lys His 130 135 140
Phe Gly Cys Lys Phe He Glu Thr Ser Ala Lys Gin Arg He Asn Val 145 150 155 160 Asp Glu Ala Phe Ser Asn Leu Val Arg Glu He Arg Lys Tyr Asn Arg 165 170 175
Glu Gin Gin Thr Gly Arg Pro Ala He Ala Ala Gly Gly Gly Gly Pro 180 185 190
Ala Gly Ser Tyr Thr Gin Asp Arg His His Asp Glu Ala Pro Gly Cys 195 200 205
Cys Ala Gly Cys Val He Ala 210 215
(2) INFORMATION FOR SEQ ID NO:50:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 206 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Geodia cydonium
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 50:
Met Thr Glu Tyr Lys He Val Val Gly Gly Gly Leu Val Gly Lys Ser 1 5 10 15
Ala Leu Thr Leu Gin Leu Val Gin Val Cys He Lys Asp Gin Tyr Tyr 20 25 30
Leu He Glu Phe Gin Asn Asn Gin Phe Gin Phe Glu Asn Leu Gin Asn 35 40 45
His Tyr He Asp Tyr Asp Pro Thr Val Glu Asp Ser Arg Arg Glu Val 50 55 60
Ser He Asp Asp Gin Thr Cys Leu Asn He Leu Asp Thr Ala Gly Gin 65 70 75 80
Gin His Ser Asn Ala Gin Ser Met Asp Ala His Trp Ser Thr Val Phe 85 90 95
Val Cys Leu Phe Asn Tyr Phe Asn He Thr Ser Met Tyr Asp Glu He 100 105 110
Ala Ser Phe Arg Glu Gin He Leu Arg Val Lys Asp Gly Ala Lys Asp 115 120 125
Leu Val Pro Leu He Leu He He Asn Lys Ala Asp Leu Asp His Glu 130 135 140
Ser Gin Gly Ser Gly Asn Glu Gly Gin Leu Ala Lys Asp Ser Leu Ser 145 150 155 160
Phe His Gin Ser Ser Ala Lys Ser Arg He Asn Leu Glu Glu He Pro 165 170 175
Tyr Ser Leu Val Arg Glu Leu Arg Lys Glu Leu Lys Leu Asp Gin Ser 180 185 190 Ser Gly Lys Ala Gin Lys Lys Lys Lys Gin Cys Leu He He 195 200 205
(2) INFORMATION FOR SEQ ID NO:51:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 198 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Canis familiaris
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:51:
Met Lys Lys Thr Tyr Asp Leu Leu Phe Lys Leu Leu Leu He Gly Asp 1 5 10 15
Ser Gly Val Gly Lys Thr Cys Val Leu Phe Arg Phe Ser Asp Asp Ala 20 25 30
Phe Asn Thr Thr Phe He Ser He Gly He Asp Phe Lys He Lys Thr 35 40 45
Val Glu Leu Gin Gly Lys Lys He Lys Leu Gin He Trp Asp Thr Ala 50 55 60
Gly Gin Glu Arg Phe His Thr He Thr Thr Ser Tyr Tyr Arg Gly Ala 65 70 75 80
Met Gly He Met Leu Val Tyr Asp He Thr Asn Gly Lys Ser Phe Glu 85 90 95
Asn He Ser Lys Trp Leu Arg Asn He Asp Glu His Ala Asn Glu Asp 100 105 110
Val Glu Arg Met Leu Leu Gly Asn Lys Cys Asp Met Asp Asp Lys Arg 115 120 125
Val Val Pro Lys Gly Lys Gly Glu Gin He Ala Arg Glu His Gly He 130 135 140
Arg Phe Phe Glu Thr Ser Ala Lys Val Asn He Asn He Glu Lys Ala 145 150 155 160
Phe Leu Thr Leu Ala Glu Asp He Leu Arg Lys Thr Pro Val Lys Glu 165 170 175
Pro Asn Ser Glu Asn Val Asp He Ser Ser Gly Gly Gly Val Thr Gly 180 185 190
Trp Lys Ser Lys Cys Cys 195
(2) INFORMATION FOR SEQ ID NO:52:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 214 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:52:
Met Gly Thr Arg Asp Asp Glu Tyr Asp Tyr Leu Phe Lys Val Val Leu 1 5 10 15
He Gly Asp Ser Gly Val Gly Lys Ser Asn Leu Leu Ser Arg Phe Thr 20 25 30
Arg Asn Glu Phe Asn Leu Glu Ser Lys Ser Thr He Gly Val Glu Phe 35 40 45
Ala Thr Arg Ser He Gin Val Asp Gly Lys Thr He Lys Ala Gin He 50 55 60
Trp Asp Thr Ala Gly Gin Glu Arg Tyr Arg Ala He Thr Ser Ala Tyr 65 70 75 80
Tyr Arg Gly Ala Val Gly Ala Leu Leu Val Tyr Asp He Ala Lys His 85 90 95
Leu Thr Tyr Glu Asn Val Glu Arg Trp Leu Lys Glu Leu Arg Asp His 100 105 110
Ala Asp Ser Asn He Val He Met Leu Val Gly Asn Lys Ser Asp Leu 115 120 125
Arg His Leu Arg Ala Val Pro Thr Asp Glu Ala Arg Ala Phe Ala Glu 130 135 140
Lys Asn Gly Leu Ser Phe He Glu Thr Ser Ala Leu Asp Ser Thr Asn 145 150 155 160
Val Glu Ala Ala Phe Gin Thr He Leu Thr Glu He Tyr Arg He Val 165 170 175
Ser Gin Lys Gin Met Ser Asp Arg Glu Asn Asp Met Ser Pro Ser Asn 180 185 190
Asn Val Val Pro He His Val Pro Pro Thr Thr Glu Lys Pro Lys Val 195 200 205
Gin Cys Cys Gin Asn He 210

Claims

We claim: 1. A composition comprising at least one component selected from the group consisting of a peptide having an amino acid sequence selected from the group consisting of Val-Val-Ile, Ile-Lys-Arg-Val-Lys-Asp (SEQ ID NO : 1), Lys-Cys-Asp-Leu-Ala (SEQ ID NO : 2), Cys-Asp-Leu-Ala-Ala-Arg-Thr (SEQ ID NO : 3) and Asp-Leu-Ala-Ala (SEQ ID NO:4) or a physiologically acceptable salt of said peptide, a cyclized peptide having a formula selected from the group consisting of cyclo [- R(1) R(2) Thr-Ile-Glu-Asp-Ser-Tyr-Arg-Lys-Gln-Val-Val-Ile- Asp R(3) R(4)-], cyclo [-R(1) R(2) Val-Val-Ile R(3) R(4)-], cyclo [-R(1) R(2) Tyr-Arg-Glu-Gln-Ile-Lys-Arg-Val-Lys-Asp- Ser-Asp-Asp-Val-Pro R(3) R(4)-], cyclo [-R(1) R(2) Lys-Arg- Val R(3) R(4)-], cyclo [-R(1) R(2) Ile-Lys-Arg-Val-Lys-Asp R(3) R(4)-], cyclo [-R(1) R(2) Gly-Asn-Lys-Cys-Asp-Leu-A;a- Ala-Arg-Thr-Val-Glu R(3) R(4)-], cyclo [-R(1) R(2) Lys-Cys- Asp-Leu-Ala R(3) R(4)-], cyclo [-R(1) R(2) Cys-Asp-Leu-Ala- Ala-Arg-Thr R(3) R(4)-], cyclo [-R(1) R(2) Asp-Leu-A; a-Ala R(3) R(4)-], cyclo [- R(1) R(2) D-Thr-Ile-Glu-Asp-Ser-Tyr- Arg-Lys-Gln-Val-D-Val-Ile-Asp R(3) R(4)-], cyclo [-R(1) R(2) D-Val-D-Val-D-Ile R(3) R(4)-], cyclo [-R(1) R(2) D- Tyr-Arg-Glu-Gln-Ile-Lys-Arg-Val-Lys-Asp-D-Ser-Asp-D-Asp-Val Pro R(3) R(4)-], cyclo [-R(1) R(2) D-Lys-D-Arg-D-Val-R (3) R(4)-], cyclo [-R(1) R(2) D-Ile-Lys-Arg-Val-Lys-D-Asp-R (3) R(4)-], cyclo [-R(1) R(2) Gly-D-Asn-Lys-Cys-Asp-Leu-D-Ala- Ala-Arg-Thr-D-Val-Glu R(3) R(4)-], cyclo [-R(1) R(2) D-Lys- Cys-Asp-Leu-D-Ala R(3) R(4)-], cyclo [-R(1) R(2) Cys-Asp- Leu-Ala-Ala-Arg-D-Thr R(3) R(4)-], cyclo [-R(1) R(2) Asp-D- Leu-D-Ala-D-Ala R(3) R(4)-], and
Figure imgf000095_0001
wherein R(1) R(2), R(3) and R(4) represent independently alanine, ornithine, cysteine, lysine, glutamic and aspartic acid, and wherein there is a covalent bond between the carboxyl and amino termini by which R(1) and R(4) are interconnected to each other via a methylene bridge of type ╌(CH2)m ╌or ╌(CH2)m╌M╌ (CH2)m,╌, wherein m and m' are integers from 1, 2, 3, or 4, and M is NH, N[R(5)], O, or S, and wherein R(5) is methyl, ethyl, n-propyl, isopropyl, cyclopropyl, or cyclobutyl, or the sidechain of any naturally occurring amino acid, and a physiologically acceptable salt thereof and a peptidomimetic falling within the structure
Figure imgf000096_0001
wherein the sidechain R attached at the carbon atom numbered 6 on the sterol nucleus can be NH-CH2-CH2NH3 +, alkylamino, arylamino, or aralkylamino group, and wherein the sidechain attached at the carbon number 3 can be replaced with -O-C(=O)╌(CH2)m╌COOH, where m is an integer from 1 to 6, inclusive, preferably from 1 to 3, inclusive, and more preferably 2, and one of X and Y at each position independently, can be one H, a small alkyl group of C1 to C3, preferably C1; a halogen, preferably F, or an amino group where the other of one of X and Y is H. Preferably, each of X and Y is H and a pharmaceutically acceptable salt thereof.
2. The composition of claim 1 wherein said component is a peptide has a structure having the structure
Figure imgf000097_0002
3. The composition of claim 1 wherein said peptidomimetic capable of inhibiting the oncogenic activity of p21 Ras has the structure
Figure imgf000097_0001
wherein the sidechain R attached at the carbon atom numbered 6 on the sterol nucleus can be NH-CH2-CH2NH3% alkylamino, arylamino, or aralkylamino group, and wherein the sidechain attached at the carbon number 3 can be replaced with -O-C(=O)╌(CH2)m╌COOH, where m is an integer from 1 to 6, inclusive, preferably from 1 to 3, inclusive, and more preferably 2, and one of X and Y at each position independently, can be one H, a small alkyl group of C1 to C3, preferably C1; a halogen, preferably F, or an amino group where the other of one of X and Y is H. Preferably, each of X and Y is H and a pharmaceutically acceptable salt thereof.
4. The composition of claim 1 wherein said peptidomimetic has the structure
Figure imgf000098_0001
or a pharmaceutically acceptable salt thereof.
5. A method for inhibiting the oncogenic or transforming activity of p2l Ras, said method comprising the step of administering an amount of the composition of of claim 1 effective to achieve the result that the transforming and/or oncogenic activity of the p21 Ras protein is inhibited.
6. A method for inhibiting the oncogenic or transforming activity of p21 Ras, said method comprising the step of administering an amount of a composition comprising at least one cyclized peptide as set forth in claim 1, effective to achieve the result that the transforming and/or oncogenic activity of the p21 Ras protein is inhibited.
7. The method of claim 6 wherein the cyclized peptide has a structure
Figure imgf000099_0001
8. A method for inhibiting the oncogenic or transforming activity of p21 Ras, said method comprising the step of administering an effective amount of at least one peptidomimetic as set forth in claim 4, with the result that the transforming and/or oncogenic activity of the p2l Ras protein is inhibited. 9. The method of claim 8 wherein said peptidomimetic is
3-malonoxy-6-N-(2aminoethyl)
aminocyclopentanoperhydrophenanthrene.
PCT/US1996/015098 1995-09-21 1996-09-20 Peptides and peptidomimetics inhibiting the oncogenic action of p21 ras WO1997010836A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU73668/96A AU7366896A (en) 1995-09-21 1996-09-20 Peptides and peptidomimetics inhibiting the oncogenic action of p21 ras

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US409195P 1995-09-21 1995-09-21
US60/004,091 1995-09-21
US08/531,525 1995-09-21
US08/531,525 US5840683A (en) 1995-09-21 1995-09-21 Peptides inhibiting the oncogenic action of p21 ras

Publications (1)

Publication Number Publication Date
WO1997010836A1 true WO1997010836A1 (en) 1997-03-27

Family

ID=26672587

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/015098 WO1997010836A1 (en) 1995-09-21 1996-09-20 Peptides and peptidomimetics inhibiting the oncogenic action of p21 ras

Country Status (3)

Country Link
AU (1) AU7366896A (en)
CA (1) CA2232750A1 (en)
WO (1) WO1997010836A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6022326A (en) * 1998-10-30 2000-02-08 Lifepoint, Inc. Device and method for automatic collection of whole saliva
JP2005529882A (en) * 2002-04-19 2005-10-06 アンスティテュ ナショナル ドゥ ラ サン エ ドゥ ラ ルシュルシェ メディカル Aminoalkyl sterol compounds having antitumor and neuroprotective activity
US8080517B2 (en) 2005-09-12 2011-12-20 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8183339B1 (en) 1999-10-12 2012-05-22 Xigen S.A. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8236924B2 (en) 1999-10-12 2012-08-07 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8748395B2 (en) 2005-09-12 2014-06-10 Xigen Inflammation Ltd. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8981052B2 (en) 2010-06-21 2015-03-17 Xigen Inflammation Ltd. JNK inhibitor molecules
US9006185B2 (en) 2008-05-30 2015-04-14 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US9150618B2 (en) 2010-10-14 2015-10-06 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory eye diseases
US9180159B2 (en) 2008-05-30 2015-11-10 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory digestive diseases
US20160151456A1 (en) * 2011-05-25 2016-06-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv Lipopeptide inhibitors of ras oncoproteins
US10023615B2 (en) 2008-12-22 2018-07-17 Xigen Inflammation Ltd. Efficient transport into white blood cells
US10596223B2 (en) 2011-12-21 2020-03-24 Xigen Inflammation Ltd. JNK inhibitor molecules for treatment of various diseases
US10624948B2 (en) 2013-06-26 2020-04-21 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US11331364B2 (en) 2014-06-26 2022-05-17 Xigen Inflammation Ltd. Use for JNK inhibitor molecules for treatment of various diseases
CN114409737B (en) * 2022-02-24 2023-04-25 南阳师范学院 Novel antitumor peptide and application thereof
US11779628B2 (en) 2013-06-26 2023-10-10 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ANTICANCER RESEARCH, Volume 11, No. 4, issued 1991, CHUNG et al., "A Peptide from the GAP-Binding Domain of the Ras-p21 Protein and Azatyrosine Block Ras - Induced Maturation of Xenopus Oocytes", pages 1373-1378. *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6022326A (en) * 1998-10-30 2000-02-08 Lifepoint, Inc. Device and method for automatic collection of whole saliva
US8183339B1 (en) 1999-10-12 2012-05-22 Xigen S.A. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8236924B2 (en) 1999-10-12 2012-08-07 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8278413B2 (en) 1999-10-12 2012-10-02 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8569447B2 (en) 1999-10-12 2013-10-29 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
JP2005529882A (en) * 2002-04-19 2005-10-06 アンスティテュ ナショナル ドゥ ラ サン エ ドゥ ラ ルシュルシェ メディカル Aminoalkyl sterol compounds having antitumor and neuroprotective activity
US7947668B2 (en) 2002-04-19 2011-05-24 Institut National De La Sante Et De La Recherche Medicale Aminoalkyl sterol compounds having an anti-tumoral and neuroprotective activity
US8524694B2 (en) 2002-04-19 2013-09-03 Institut National De La Sante Et De La Recherche Medicale (Inserm) Aminoalkylsterol compounds with antitumoral and neuroprotective activity
US8957055B2 (en) 2002-04-19 2015-02-17 Institut National De La Sante Et De La Recherche Medicale (Inserm) Aminoalkylsterol compounds with antitumoral and neuroprotective activity
US8080517B2 (en) 2005-09-12 2011-12-20 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US8748395B2 (en) 2005-09-12 2014-06-10 Xigen Inflammation Ltd. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US9290538B2 (en) 2005-09-12 2016-03-22 Xigen Inflammation Ltd. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US9006185B2 (en) 2008-05-30 2015-04-14 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US9180159B2 (en) 2008-05-30 2015-11-10 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory digestive diseases
US9610330B2 (en) 2008-05-30 2017-04-04 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US10023615B2 (en) 2008-12-22 2018-07-17 Xigen Inflammation Ltd. Efficient transport into white blood cells
US8981052B2 (en) 2010-06-21 2015-03-17 Xigen Inflammation Ltd. JNK inhibitor molecules
US9624267B2 (en) 2010-06-21 2017-04-18 Xigen Inflammation Ltd. JNK inhibitor molecules
US9150618B2 (en) 2010-10-14 2015-10-06 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory eye diseases
US20160151456A1 (en) * 2011-05-25 2016-06-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv Lipopeptide inhibitors of ras oncoproteins
US10596223B2 (en) 2011-12-21 2020-03-24 Xigen Inflammation Ltd. JNK inhibitor molecules for treatment of various diseases
US10624948B2 (en) 2013-06-26 2020-04-21 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US11779628B2 (en) 2013-06-26 2023-10-10 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US11331364B2 (en) 2014-06-26 2022-05-17 Xigen Inflammation Ltd. Use for JNK inhibitor molecules for treatment of various diseases
CN114409737B (en) * 2022-02-24 2023-04-25 南阳师范学院 Novel antitumor peptide and application thereof

Also Published As

Publication number Publication date
AU7366896A (en) 1997-04-09
CA2232750A1 (en) 1997-03-27

Similar Documents

Publication Publication Date Title
AU2008260326B2 (en) Cyclic peptide CXCR4 antagonists
WO1997010836A1 (en) Peptides and peptidomimetics inhibiting the oncogenic action of p21 ras
AU746461B2 (en) Peptide parathyroid hormone analogs
RU2510399C2 (en) Synthetic peptide amides and their dimers
BRPI0619399A2 (en) metastine derivative or salt thereof, compound or salt thereof, prodrug, medicament, and use of an effective dose of the metastine derivative
BG65065B1 (en) Peptide antiangiogenic drugs
US20020183249A1 (en) Method of identifying inhibitors of CDC25
US9982015B2 (en) Cyclin based inhibitors of CDK2 and CDK4
JP2009108078A (en) Neuromedin b and somatostatin receptor agonist
US5910478A (en) Peptidomimetics inhibiting the oncogenic action of p21 ras
AU6382096A (en) Peptides and compounds that bind to the il-1 receptor
CN117279933A (en) Cyclic compounds having selective KRAS inhibition relative to HRAS and NRAS
AU4236393A (en) Peptides and compounds that bind to elam-1
TW200410986A (en) Pharamaceutical comiposition comprising cyclic somatostatin analogs II
US5840683A (en) Peptides inhibiting the oncogenic action of p21 ras
AU7077400A (en) Method of identifying inhibitors of cdc25
EP2004352B1 (en) Amino acid surrogates for peptidic constructs
JP2023537111A (en) Cyclic chemerin-9 derivatives
CN113727991A (en) Ras inhibitory peptide
Frączak et al. The Biological Consequences of Replacing d‐Ala in Biphalin with Amphiphilic α‐Alkylserines
WO2002070680A1 (en) Method of identifying inhibitors of cdc25
WO2001027077A2 (en) Hydroxysulfonylalkylene-, phosphonoalkylene- or difluoro(phosphononon)methyl- substituted benzene, or benzofuran derivatives as non-peptidic cdc25 inhibitors
WO2011088462A2 (en) Alpha-fetoprotein 'ring and tail' peptides
Ungwitayatorn Synthesis and biochemical properties of peptide mimetic analogues of enkephalin

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI

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

Ref document number: 2232750

Country of ref document: CA

Ref country code: CA

Ref document number: 2232750

Kind code of ref document: A

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase