EP0555328A1 - Blutplättchenaggregationshemmstoffe - Google Patents

Blutplättchenaggregationshemmstoffe

Info

Publication number
EP0555328A1
EP0555328A1 EP91919821A EP91919821A EP0555328A1 EP 0555328 A1 EP0555328 A1 EP 0555328A1 EP 91919821 A EP91919821 A EP 91919821A EP 91919821 A EP91919821 A EP 91919821A EP 0555328 A1 EP0555328 A1 EP 0555328A1
Authority
EP
European Patent Office
Prior art keywords
polypeptide
amino
alkyl
alkoxy
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP91919821A
Other languages
English (en)
French (fr)
Inventor
John P. Burnier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Publication of EP0555328A1 publication Critical patent/EP0555328A1/de
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/75Fibrinogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/78Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin or cold insoluble globulin [CIG]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/02Linear peptides containing at least one abnormal peptide link
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • C07K7/56Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring the cyclisation not occurring through 2,4-diamino-butanoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to inhibitors of platelet aggregation.
  • the invention is directed to peptides comprising the tripeptide sequence -Lys-Gly-Asp- capable of acting as antagonists of the final common pathway of platelet aggregation and that act as potent antithrombotics.
  • the invention further relates to therapeutic applications of these inhibitors in diseases for which blocking platelet aggregation and intracellular adhesion is indicated.
  • Platelets are particles found in whole blood known to participate in thrombus formation and blood coagulation.
  • a membrane spanning glycoprotein receptor, GP li Wa. is present on the surface of platelets and is known to be involved in the coagulation process.
  • GP lib I Ha is a non-covalent, calcium ion dependent heterodimer complex composed of alpha and beta subunits (Jennings, et al., J. Biol. Chem. (1982) 257, 10458) capable of binding protein ligands.
  • This glycoprotein receptor contributes to normal platelet function through interactions with protein ligands containing the tripeptide amino acid sequence Arg-Gly-Asp (RGD).
  • Fibrinogen contains two RGD sequences located at A ⁇ 95-97 and A ⁇ 572-574 (Doolittle, R. F., Watt, K. W. K., Co lltrell, B. A., Strong, D. D. and Riley, M. (1979) Nature 280, 464-468) that have been shown to interact with the GP H Hla receptor (Hawiger.e. al., Biochemistry, 28, 2909-2914 (1989).
  • GP li Ilia The interaction of GP li Ilia with fibrinogen is stimulated by certain factors released or exposed when a blood vessel is injured. Multiple factors, including a variety of physiologic stimuli and soluble mediators, initiate platelet activation via several pathways. These pathways have, as a common final step, the activation of the GP lib Efl a receptor on the platelet surface and its subsequent binding to fibrinogen followed by aggregation and thrombus formation. By virtue of these interactions GP lib Wa is an important component of the platelet aggregation system (Pytela et al., Science (1986) 231 , 1559).
  • integrin receptors may be discovered that also interact with RGD containing ligands.
  • a particularly useful antithrombotic would be one that specifically inhibited the interaction between RGD containing proteins and the platelet GP llbWa receptor while not effecting the interaction between the other integrins and their endogenous ligands.
  • Atherosclerotic plaques form niduses for platelet plugs and thrombii that lead to vascular narrowing and occlusion, resulting in myocardial and cerebral ischemic disease. This may happen spontaneously or following procedures such as angioplasty or endarteroectomy. Thrombii that break off and are released into the circulation cause infarction of different organs, especially the brain, extremities, heart and kidneys.
  • platelets may also play a role in venous thrombosis.
  • a large percentage of such patients have no antecedent risk factors and develop venous thrombophlebitis and subsequent pulmonary emboii without a known cause.
  • Other patients who form venous thrombi have underlying diseases known to predispose to these syndromes. Some of these patients may have genetic or acquired deficiencies of factors that normally prevent hypercoagulability, such as antithrombin-3. Others have mechanical obstructions to venous flow, such as tumor masses, that lead to low flow states and thrombosis. Patients with malignancy have a high incidence of thrombotic phenomena for unclear reasons. Antithrombotic therapy in this situation with currently available agents is dangerous and often ineffective.
  • the number of available therapeutic agents is limited and these, for the most part, act by inhibiting or reducing levels of circulating clotting factors. These agents are frequently not effective against the patient's underlying hematologic problem, which often concerns an increased propensity for platelet aggregation and adhesion. They also cause the patient to be susceptible to abnormal bleeding. Available antiplatelet agents, such as aspirin, inhibit only part of the platelet activation process and are therefore often inadequate for therapy.
  • An agent which effectively inhibits the final common pathway of platelet activation, namely fibrinogen binding to the GP lib Ilia receptor, should accordingly be useful in a large group of disorders characterized by a hyperthrombotic state as described above.
  • the present invention contemplates such an agent which is a new composition, namely a polypeptide that may consist in part of natural amino acids and in part of unnatural amino acids as well as non- peptidyl portions. This new composition is believed to interfere with the interaction of Arg-Gly- Asp containing peptides, particularly fibrinogen, with the GP lib Hla complex thereby preventing platelet aggregation.
  • Platelet aggregation has been identified as an early step in the formation of platelet plugs, emboii and thrombii in the circulatory system which in turn have been shown to play an active role in cardiovascular complications and disease. Inhibition of fibrinogen binding to the GP lib Hla complex has been shown to be an effective antithrombotic treatment in animals (H. K. Gold, et al., Circulation (1988) 77, 670-677; T. Yasuda, et al., J. Clin. Invest.
  • EPO 0368486 A2 discloses a Arg-Tyr-Asp-21 mer that is about 10-fold less active in a platelet aggregation assay than the corresponding Arg-GIy-Asp-21 mer.
  • a peptide having high platelet aggregation inhibition activity It is a further object of this invention to provide peptides having high specificity for the GP HbHIa receptor. It is still a further object to produce small cyclic peptides that are stable to ring opening having the above described properties. It is still a further object of this invention to provide a platelet aggregation inhibitor exhibiting diminished in vivo side effects such increased bleeding times and optionally to provide such inhibitors with increased lifetime
  • Xaa is Omithine (Orn) or Lysine (Lys).
  • the peptide contains the sequence Lys-Gly-Asp and contains fewer than about 345 amino acid residues.
  • the peptide is cyclic, having from 5-10 amino acids in the cycle. Most preferably, the cyclic peptide has 5 amino acids forming the ring of the cycle. More preferably, the ring of the cyclic peptide contains from about 17 to about 18 atoms, most preferably 18 atoms.
  • a particulariy preferred compound of the instant invention is a polypeptide having the structure
  • Xaa is a D or L ⁇ -amino acid linked to Z through the ⁇ amino group
  • Xaa2 is Orn or Lys
  • Xaa3 is a D or L ⁇ -amino acid linked through the side chain to Z; Z is an amide bond, disulfide, COCH2S, COCH2SO, or COCH(C6H5)S; and R is OH or NH2.
  • the ring will contain a D amino acid most preferably linked to the Lys of the tripeptide sequence.
  • the invention in its broad aspects relates to peptide derivatives which are useful as inhibitors of platelet function mediated by the GP lib Hla receptor and for the prevention of thrombus formation.
  • Preferred compounds of this invention are represented by Formula I:
  • and Rg are the same or different and are selected from hydroxy
  • R 8 are the same or different and are selected from hydrogen, C6-C12 aryl where the aryl group is unsubstituted or substituted by one or more of the groups nitro, hydroxy, halo (F, Cl, Br, I), Ci-C ⁇ alkyl, halo-C ⁇ - C ⁇ alkyl, C-t-C ⁇ -alkoxy, amino, phenyloxy, phenyl, acetamido, benzamido, di-C-
  • C6-C12 aryl wherein the aryl group is unsubstituted or substituted by one or more of the groups nitro, hydroxy, halo, C-
  • R2 or R3 may be optionally joined with R4 to form a piperidine, pyrrolidine or thiazolidi ⁇ e ring
  • R14 is selected from hydrogen, C1 -C ⁇ -alkyl, C3-C ⁇ -alkenyl, C6-C12-aryl, and C6-C12 aryl-Ci -C ⁇ - alkyl
  • X is selected from an O or S atom, an S atom bearing one or two O atoms
  • R13 is hydrogen, C-i-C ⁇ -alkyi, C3-C ⁇ -alkenyl, C ⁇ -Ci 2-aryl, C6-Ci2-aryl-Ci -C ⁇ -alkyl, Ct-C ⁇ alkanoyl, and C6-C12 aroyl, and C6-Ci2 aryl, C-
  • alkyl, alkenyl and alkynyl denote straight and branched hydrocarbon chains having single, double and triple bonds, respectively;
  • C6-C12 aryl groups denote unsubstituted aromatic ring or fused rings such as, for example, phenyl or naphthyl;
  • hetero denotes the heteroatoms O, N, or S;
  • aromatic heterocyclic groups have 5-10 ring atoms and contain up to four heteroatoms; halogen or halo denote F, Cl Br, or I atoms;
  • alkoxy denotes an alkyl group attached to O.
  • Ci-C ⁇ alkyl or C2-C6 alkenyl groups include methyl, ethyl, propyl, isopropyl, butyl, t-butyl, pentyl, isopentyl, hexyl, vinyl, ally I, butenyl and the like;
  • examples of C3-C ⁇ o-cycloalkyl groups include cyclopropyl, cyctopentyl, cyctohexyl, and the like; aromatic heterocyclic groups include but are not limited to pyridyl, thienyl, furyl, indolyl, benzthienyl, imidazolyl, thiazoiyl, quinolinyl and isoquinolinyl.
  • the present invention includes a method of making the compounds of Formula I.
  • the present invention also includes a method for reducing platelet aggregation in a mammal. This method involves administering a therapeutically effective amount of the compounds of the present invention alone or in combination with a pharmacologically acceptable carrier. This general method may also be applied to treat a mammal having an increased propensity for thrombus formation.
  • the present invention is directed to compositions of matter for reducing platelet aggregation in a mammal; treating a mammal having an increased propensity for thrombus formation; or inhibiting binding of a ligand to GP lib Hla in a mammal; wherein each of these compositions contains as an active ingredient one or more of the cyclic peptides defined in Formula I.
  • KGD containing polypeptide are potent inhibitors of platelet aggregation and in the Fg/GP HbHIa ELISA.
  • the most potent KGD-containing inhibitors are small cyclic peptides and thus these peptides are preferred.
  • the instant KGD peptides are weak inhibitors of the fibronectin (Fn)/fibronectin receptor (FnR) interaction and of the vitronectin ( Vn) vitronectin receptor VnR interaction.
  • inhibition potency as measured by IC50, is from 10-500 fold higher (i.e. a lower IC50) in the Fg/GPIIbHIa ELISA than in the Vn/VnR ELISA.
  • the instant polypeptides exhibit high specificity for the GPIIbHIa receptor. It is contemplated that this high specificity will reduce the number and severity of side effects for these antithrombotic compounds. It is believed these compounds will exhibit high platelet aggregation inhibition without substantially increasing bleeding time in a mammal.
  • Polypeptides of this invention can be made by chemical synthesis or by employing recombinant technology. These methods are known in the art. Chemical synthesis, especially solid phase synthesis, is prefered for short (eg. less than 50 residues) polypeptides or those containing unnatural or unusual amino acids such as; D-Tyr, Omithine, amino adipic acid, and the like. Recombinant procedures are prefered for longer polypeptides or for mutant or variant peptides containing the KGD sequence.
  • a synthetic gene When recombinant procedures are selected, a synthetic gene may be constructed de novo or a natural gene may be mutanigized by, for example, casette mutagenisis. Set forth below are exemplary general recombinant procedures.
  • a KGD-containing protein may be produced using recombinant DNA techniques. These techniques contemplate, in simplified form, taking the gene, either natural or synthetic, for the protein; inserting it into an appropriate vector; inserting the vector into an appropriate host cell; culturing the host cell to cause expression of the gene; and purifying the protein produced thereby.
  • DNA sequence encoding a KGD-containing protein is cloned and manipulated so that it may be expressed in a convenient host.
  • DNA encoding parent polypeptides can be obtained from a genomic library, from cDNA derived from mRNA from cells expressing the protein, or by synthetically constructing the DNA sequence (Sambrook, J., Fritsch, E.F., and Maniatis, T., (1989), Molecular Cloning (2d ed.), Cold Springs Harbor Laboratory, N.Y.). The parent DNA is then inserted into an appropriate plasmid or vector which is used to transform a host cell.
  • plasmid vectors containing replication and control sequences which are derived from species compatible with the host cell are used in connection with those hosts.
  • the vector ordinarily carries a replication site, as well as sequences which encode proteins that are capable of providing phenotypic selection in transformed cells.
  • E * cadi may be transformed using pBR322, a plasmid derived from an £ coli species (Mandel, M. et al. (1970) J. Mol. Biol.53, 154). Plasmid pBR322 contains genes for ampicillin and tetracycline resistance, and thus provides easy means for selection. Other vectors include different features such as different promoters, which are often important in expression. For example, plasmids pKK223-3, pDR?20, and pPL-lambda represent expression vectors with the tac, trp, or P
  • a preferred vector is pB0475.
  • This vector contains origins of replication for phage and £.fi ⁇ li which allow it to be shuttled between such hosts, thereby facilitating both mutagenesis and expression (Cunningham, B., et al. (1989), Science 243, 1330-1336; Wells, J. and
  • vectors are pR1T5 and pRlT2T (Pharmacia Biotechnology). These vectors contain appropriate promoters followed by the Z domain of protein A, altowing genes inserted into the vectors to be expressed as fusion proteins. Further discussion of these vectors may be found below.
  • compositions described above can be constructed using standard techniques by combining the relevant traits of the vectors described above.
  • Relevant traits include the promoter, the ribosome binding site, the decorsin or omatin gene or gene fusion (the Z domain of protein A and decorsin or omatin and its linker), the antibiotic resistance markers, and the appropriate origins of replication.
  • the host cell may be prokaryotic or eukaryotic.
  • Prokaryotes are preferred for cloning and expressing DNA sequences to produce parent polypeptides, segment substituted polypeptides, residue-substituted polypeptides and polypeptide variants.
  • £. K12 strain 294 ATCC No.31446
  • E. coli B £,221 X1776 (ATCC No. 31537)
  • £. C2li c600 and c ⁇ OOhf I, E ⁇ W3110 F-, gamma-, prototrophic /ATCC No.
  • prokaryote J ⁇ W3110 (ATCC 27325).
  • the polypeptides When expressed by prokaryotes the polypeptides typically contain an N-terminal methionine or a formyl methionine and are not glycosylated. In the case of fusion proteins, the N-terminal methionine or formyl methionine resides on the amino terminus of the fusion protein or the signal sequence of the fusion protein.
  • eukaryotic organisms such as yeast cultures, or cells derived from multicellular organisms may be used.
  • any such cell culture is workable.
  • interest has been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a reproducible procedure ( ⁇ ssue Culture, Academic Press, Kruse and Patterson, editors (1973)).
  • useful host cell lines are VERO and HeLa cells, Chinese Hamster Ovary (CHO) cell lines, W138, 293, BHK, COS-7 and MDCK cell lines.
  • Gene Fusions A variation on the above procedures contemplates the use of gene fusions, wherein the gene encoding the desired protein is associated, in the vector, with a gene encoding another protein or a gragment of another protein. This results in the desired protein - here, a KGD-containing protein - being produced by the host cell as a fusion with another protein.
  • the "other" protein is often a protein or peptide which can be secreted by the cell, making it possible to isolate and purify the desired protein from the culture medium and eliminating the necessity of destroying the host cells which arises when the desired protein remains inside the cell.
  • the fusion protein can be expressed intracellulariy. It is useful to use fusion proteins that are highly expressed.
  • a KGD-containing protein expressed a fusion protein may be properly folded or require folding to obtain the native structure.
  • the properly folded fusion protein may be active and useful as a GP lib Hla antagonist and inhibitor of platelet aggregation. More preferred would be the correctly folded" native" protein that is obtained from the fusion protein by methods known in the art.
  • Fusion proteins can be cleaved using chemicals, such as cyanogen bromide, which cleaves at a methionine, or hydroxylamine, which cleaves between an asn and giy. Using standard recombinant DNA methodology, the nucleotide base pairs encoding these amino acids may be inserted just prior to the 5' end of the KGD- containing protein gene.
  • proteolytic cleavage of fusion proteins which has been recently reviewed (Carter, P. (1990) in Protein Purification: From Molecular Mechanisms to Large-Scale Processes, Ladisch, M. R., Willson, R. C, Painton, C. C, and Builder, S. E., eds., American Chemical Society Symposium Series No. 427, Ch 13, 181-193).
  • Proteases such Factor Xa, thrombin, subtilisin and mutants, and a number of other have been successfully used to cleave fusion proteins.
  • a peptide linker that is amenable to cleavage by the protease used is inserted between the "other" protein (e.g., the Z domain of protein A) and the KGD-contaning protein of interest.
  • the nucleotide base pairs encoding the linker are inserted between the genes or gene fragments coding for the other proteins.
  • Proteolytic cleavage of the partially purified fusion protein containing the correct linker can then be earned out on either the native fusion protein, or the reduced or denatured fusion protein.
  • the protein may or may not be properly folded when expressed as a fusion protein.
  • the specific peptide linker containing the cleavage site may or may not be accessible to the protease. These factors determine whether the fusion protein must be denatured and refolded, and if so, whether these procedures are employed before or after cleavage.
  • a chaotrope such as a guanidine HCI
  • a redox buffer containing, for example, reduced and oxidized dithiothreitol or glutathione at the appropriate ratios, pH, and temperature, such that the protein of interest is refolded to its native structure.
  • peptides When peptides are not prepared using recombinant DNA technology, they are preferably prepared using solid-phase synthesis, such as that generally described by Merrifield, J. Am. Chem. Soc. (1963) 85, 2149, although other equivalent chemical syntheses known in the art are employable as previously mentioned.
  • Solid-phase synthesis is initiated from the C-terminus of the peptide by coupling a protected ⁇ -amino acid to a suitable resin.
  • a starting material can be prepared by attaching an ⁇ -amino-protected amino acid by an ester linkage to a chloromethylated resin or a hydroxymethyl resin, or by an amide bond to a BHA resin or MBHA resin.
  • the amino acids are coupled to the peptide chain using techniques well known in the art for the formation of peptide bonds.
  • One method involves converting the amino acid to a derivative that will render the carboxyl group more susceptible to reaction with the free N- terminal amino group of the peptide fragment.
  • the amino acid can be converted to a mixed anhydride by reaction of a protected amino acid with ethylchloroformate, phenyl chloroformate, sec-butyl chloroformate, isobutyl chloroformate, pivaloyl chloride or like acid chlorides.
  • the amino acid can be converted to an active ester such as a 2,4,5- trichlorophenyl ester, a pentachlorophenyl ester, a pentafluorophenyl ester, a p-nitrophenyl ester, a N-hydroxysuccinimide ester, or an ester formed from 1-hydroxybenzotriazole.
  • an active ester such as a 2,4,5- trichlorophenyl ester, a pentachlorophenyl ester, a pentafluorophenyl ester, a p-nitrophenyl ester, a N-hydroxysuccinimide ester, or an ester formed from 1-hydroxybenzotriazole.
  • Another coupling method involves use of a suitable coupling agent such as N,N'- dicyclohexylcarbodiimide or N,N'-diisopropyl-carbodiimide.
  • a suitable coupling agent such as N,N'- dicyclohexylcarbodiimide or N,N'-diisopropyl-carbodiimide.
  • Other appropriate coupling agents apparent to those skilled in the art, are disclosed in E. Gross & J. Meienhofer, The Peptides: Analysis, Structure, Biology, Vol. I: Major Methods of Peptide Bond Formation (Academic Press, New York, 1979).
  • ⁇ -amino group of each amino acid employed in the peptide synthesis must be protected during the coupling reaction to prevent side reactions involving there active ⁇ -amino function.
  • certain amino acids contain reactive side-chain functional groups (e.g. sulfhydryl, amino, carboxyl, and hydroxyl) and that such functional groups must also be protected with suitable protecting groups to prevent a chemical reactton from occurring at that site during both the initial and subsequent coupling steps.
  • suitable protecting groups known in the art, are described in E. Gross & J. Meienhofer, The Peptides: Analysis, Structure, Biology, Vol.3: Protection of Functional
  • An ⁇ -amino protecting group (a) must render the ⁇ -amino function inert under the conditions employed in the coupling reaction, (b) must be readily removable after the coupling reaction under conditions that will not remove side-chain protecting groups and will not alter the structure of the peptide fragment, and (c) must eliminate the possibility of racemization upon activation immediately prior to coupling.
  • a side-chain protecting group (a) must render the side chain functional group inert under the conditions employed in the coupling reaction, (b) must be stable under the conditions employed in removing the ⁇ -amino protecting group, and (c) must be readily removable upon completion of the desired amino acid peptide under reaction conditions that will not alter the structure of the peptide chain.
  • protecting groups known to be useful for peptide synthesis will vary in reactivity with the agents employed for their removal.
  • certain protecting groups such as triphenylmethyl and 2-(p- biphenylyl)isopropyioxycarbonyl are very labile and can be cleaved under mild acid conditions.
  • Other protecting groups such as t-butyloxycarbonyl (BOC), t-amyloxycarbonyl.
  • adamantyl- oxycarbonyl, and p-methoxybenzyloxycarbonyl are less labile and require moderately strong acids, such as trifluoroacetic, hydrochloric, or boron trifiuoride in acetic acid, for their removal.
  • Still other protecting groups such as benzytoxycarbonyl (CBZ or Z), hatobenzytoxycarbonyl, p- nitrobenzyloxycarbonyl cyctoalkytoxycarbonyl, and isopropyloxycarbonyl, are even less labile and require stronger acids, such as hydrogen fluoride, hydrogen bromide, or boron trifluoroacetate in trifiuor oacetic acid, for their removal.
  • useful amino acid protecting groups are included:
  • aromatic urethane-type protecting groups such as fluorenyimethyloxycarbonyl (FMOC) CBZ, and substituted CBZ, such as, e.g., p- chlorobenzyloxycarbonyl, p-6-nitrobenzytoxycarbonyl, p-bromobenzyloxycarbonyl, and p-methoxybenzytoxycarbonyl, o-chlorobenzytoxycarbonyl, 2,4- dichtorobenzytoxycarbonyl, 2,6-dichlorobenzytoxycarbonyl, and the like; (b) aliphatic urethane-type protecting groups, such as BOC, t-amytoxycarbonyl, isopropyloxycarbonyl, 2-(p-biphenylyl)-isopropyloxycarbonyl, allyloxycarbonyl and the like; (c) cycloalkyl urethane-type protecting groups,
  • protection may be by nitro, tosyl, CBZ, adamantytoxycarbonyl, 2,2,5,7,8-pentamethytohroman-6-sulfonyl or 2,3,6 -trimethyl-4- methoxyphenylsulfonyl, or BOC.
  • (4) for the hydroxyl group of Ser, Thr, or Tyr protection maybe, for example, by C1-C4 alkyl, such as t-butyi; benzyl (BZL); substituted BZL, such as p-methoxybenzyl, p- ⁇ itrobenzyi, p-chlorobenzyl, o-chtorobenzyl, and 2,6-dichtorobenzyl.
  • C1-C4 alkyl such as t-butyi; benzyl (BZL); substituted BZL, such as p-methoxybenzyl, p- ⁇ itrobenzyi, p-chlorobenzyl, o-chtorobenzyl, and 2,6-dichtorobenzyl.
  • protection may be, for example, by esterification using groups such as BZL, t- butyl, cyctohexyl, cyctopentyl, and the like.
  • a protecting group such as tetrahydropyranyl, tert- butyl, trityl, BZL, chlorobenzyl, 4-bromobenzyl, and 2,6- dichtorobenzyl are suitably employed.
  • the preferred protecting group is 2,6- dichlorobenzyl.
  • xanthyl (Xan) is preferably employed.
  • the amino acid is preferably left unprotected.
  • p-methoxybenzyl is typically employed.
  • the C-terminal amino acid, e.g., Lys is protected at the N-amino position by an appropriately selected protecting group, in the case of Lys, BOC.
  • the BOC-Lys-OH can be first coupled to the benzyhydrylamine or chloromethylated resin according to the procedure set forth in Horiki et al., Chemistry Letters, (1978)165-168 or using isopropylcarbodiimide at about 25°C for 2 hours with stirring.
  • the ⁇ -amino protecting group is removed, as by using trifluoroacetic acid (TFA) in methylene chloride or TFA alone.
  • TFA trifluoroacetic acid
  • the deprotection is carried out at a temperature between about 0°C and room temperature.
  • Other standard cleaving reagents, such as HCI in dioxane, and conditions for removal of specific ⁇ -amino protecting groups are described in Schroder & Lubke, supra, Chapter I, pp. 72-75.
  • the remaining ⁇ -amino and side-chain protected amino acids are coupled step within the desired order.
  • some may be coupled to one another prior to addition to the solid-phase synthesizer.
  • the selection of an appropriate coupling reagent is within the skill of the art. Particularly suitable as a coupling reagent is N,N'-dicyclohexyl carbodiimide or diisopropylcarbodiimide.
  • Each protected amino acid or amino acid sequence is introduced into the solid-phase reactor in excess, and the coupling is suitably carried out in a medium of dimethylformamide (DMF) or CH 2 CI 2 or mixtures thereof. If incomplete coupling occurs, the coupling procedure is repeated before removal of the N-amino protecting group prior to the coupling of the next amino acid.
  • the success of the coupling reaction at each stage of the synthesis may be monitored. A preferred method of monitoring the synthesis is by the ninhydrin reaction, as described by Kaiser etal., Anal. Biochem, (1970) 34, 595.
  • the coupling reactions can be performed automatically using well known methods, for example, a Biosearch 9500 Peptide Synthesizer.
  • the protected peptide Upon completion of the desired peptide sequence, the protected peptide must be cleaved from the resin support, and all protecting groups must be removed. The cleavage reaction and removal of the protecting groups is suitably accomplished simultaneously or stepwise.
  • the bond anchoring the peptide to the resin is an ester linkage formed between the free carboxyl group of the C-terminal residue and one of the many chloromethyl groups present on the resin matrix. It will be appreciated that the anchoring bond can be cleaved by reagents that are known to be capable of breaking an ester linkage and of penetrating the resin matrix.
  • One especially convenient method is by treatment with liquid anhydrous hydrogen fluoride.
  • This reagent not only will cleave the peptide from the resin but also will remove all protecting groups. Hence, use of this reagent will directly afford the fully deprotected peptide.
  • hydrogen fluoride treatment results in the formation of the free peptide acids.
  • benzhydrylami ⁇ e resin is used, hydrogen fluoride treatment results directly in the free peptide amines. Reaction with hydrogen fluoride in the presence of anisole and dimethylsulfide at O 0 C for one hour will simultaneously remove the side-chain protecting groups and release the peptide from the resin.
  • the protected peptide-resin can undergo methanolysis to yield the protected peptide in which the C-terminal carboxyl group is methylated.
  • the methyl ester is then hydrolyzed under mild alkaline conditions to give the free C-terminal carboxyl group.
  • the protecting groups on the peptide chain then are removed by treatment with a strong acid, such as liquid hydrogen fluoride.
  • a strong acid such as liquid hydrogen fluoride.
  • a particularly useful technique for methanolysis is that of Moore etal., Peptides, Proc. Fifth Amer. Pept. Symp., M. Goodman and J. Meienhofer, Eds., (John Wiley, N.Y., 1977), p. 518-521 , in which the protected peptide-resin is treated with methanol and potassium cyanide in the presence of crown ether.
  • Another method for cleaving the protected peptide from the resin when the chloromethylated resin is employed is by ammonolysis or by treatment with hydrazine. If desired, the resulting C-terminal amide or hydrazide can be hydrolyzed to the free C-terminal carboxyl moiety, and the protecting groups can be removed conventionally.
  • the protecting group present on the N-terminal ⁇ -amino group may be removed preferentially either before or after the protected peptide is cleaved from the support.
  • Purification of the polypeptides of the invention is typically achieved using conventional procedures such as preparative HPLC (including reversed phase HPLC) or other known chromatographic techniques such as gel permeation, ton exchange, partition chromatography, affinity chromotography (including monoclonal antibody columns) or countercurrent distribution.
  • preparative HPLC including reversed phase HPLC
  • other known chromatographic techniques such as gel permeation, ton exchange, partition chromatography, affinity chromotography (including monoclonal antibody columns) or countercurrent distribution.
  • Polypeptide chains are polymerized by crosslinking monomer chains with polyfunctional crosslinking agents, including compound 1 , either directly or indirectly through multifunctional polymers.
  • polyfunctional crosslinking agents including compound 1
  • two substantially identical polypeptides are crosslinked at their C or N termini using a bifunctional crosslinking agent.
  • the agent is used to crosslink the terminal amino and/or carboxyl groups.
  • both terminal carboxyl groups or both terminal amino groups are crosslinked to one another, although by selectton of the appropriate crosslinking agent the alpha amino of one polypeptide is crosslinked to the terminal carboxyl group of the other polypeptide.
  • the polypeptides are substituted at their C-termini with cysteine.
  • a disulfide bond can be formed between the terminal cysteines, thereby crosslinking the polypeptide chains.
  • disulfide bridges are conveniently formed by metal-catalyzed oxidation of the free cysteines or by nucleophilic substitution of a suitably modified cysteine residue. Selectton of the crosslinking agent will depend upon the identities of there active side chains of the amino acids present in the polypeptides. For example, disulfide crosslinking would not be preferred if cysteine was present in the polypeptide at additional sites other than the C-terminus. Also within the scope hereof are peptides crosslinked with methylene bridges.
  • Suitable crosslinking sites on the peptides aside from the N-terminal amino and C- terminal carboxyl groups, include epsilo ⁇ amino groups found on lysine residues, as well as amino, imino, carboxyl, sulf hydryi and hydroxyl groups located on the side chains of internal residues of the peptides or residues introduced into flanking sequences.
  • Crosslinking through externally added crosslinking agents is suitably achieved, e.g., using any of a number of reagents familiar to those skilled in the art, for example, via carbodiimide treatment of the polypeptide.
  • Suitable multifunctional (ordinarily bifunctional) crosslinking agents include 1 ,1-bis(diazoacetyl)-2-phenylethane; glutarakJehyde; N-hydroxysuccinimide esters (Bragg and Hou, Arch. Biochem. Biophvs. (1975) 167, 3II-32I; Anjaneyla and Staros, Int. J. Pep. Pro. Res.
  • esters with 4-azidosalicylic acid such as esters with 4-azidosalicylic acid; homobifunctional imidoesters including disuccinimidyl esters such as 3,3'-dithtobis (succinimidyl-propionate) and dimethyladipimidate dihydrochloride (Zahn, Agnew. Chem. (1955) 67, 561-572; Golden and Harrison, Biochemistry (1982) 21, 3862-3866); bifunctional maleimides such as bis-N-maleimido-l,8-octane; disuccinimidyl suberate (Novick etal., J. Biol. Chem.
  • SMCC 4-(N-maleimidomethyI) cyclohexane - 1 - carboxylate (SMCC) (Mahan etal. Anal. Biochem. (1987)162, 163-170); sulfo-SMCC (Hashida et al., J. Applied Biochem.
  • MBS m- maleimidobenzoyl-N-hydroxysuccinimide ester
  • Sulfo-MBS succinimidyl 4-(p- maleimidophenyl) butyrate (SMPB); sulfo-SMPB; N-succinimidyl(4-iodoacetyl)aminobenzoate (SIAB); sulfo-SIAB; 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochtoride (EDC); and
  • Crosslinking agents such as methyl-3-[(p-azido-phenyl)dithio] propioimidate yield photoactivatable intermediates which are capable of forming crosslinks in the presence of light. If necessary, sensitive residues such as the side chains of the diargininyl group are protected during crosslinking and the protecting groups removed thereafter.
  • Polymers capable of multiple crosslinking serve as indirect crosslinking agents. For example, cyanogen bromide activated carbohydrates and the systems described in U.S.
  • patents 3,959,080; 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; 4,055,635 and 4,330,440 are suitably modified for crosslinking the peptides herein.
  • Crosslinking to amino groups of the peptides is accomplished by known chemistries based upon cyanuric chloride, carbonyl diimidazole, aldehyde reactive groups (PEG alkoxide plus diethyl acetal of bromoacetaldehyde; PEG plus DMSO and acetic anhydride, or PEG chloride plus the phenoxide of 4-hydroxybenzaldehyde).
  • succinimidyl active esters activated dithiocarbonate PEG, and 2,4,5 -trichlorophenyl-chloroformate- or p-nitrophenyl- chloroformate-activated PEG.
  • Carboxyl groups are derivatized by coupling PEG-amine using carbodiimide.
  • the crosslinking agent is not a multifunctional polymer but instead is a small molecule being less than about 500 in MW.
  • the peptides of this invention also may be conformationally stabilized by cyclization.
  • the peptides ordinarily are cyclized by covalently bonding the N and C-terminal domains of one peptide to the corresponding domain of another peptide of this invention so as to form cyclooligomers containing two or more iterated peptide sequences, each internal peptide having substantially the same sequence.
  • cyclized peptides are crosslinked to form 1-3 cyclic structures having from 2 to 6 peptides comprised therein.
  • the peptides preferably are not covalently bonded through ⁇ -amino and main chain carboxyl groups (head to tail), but rather are cross-linked through the side chains of residues located in the N and C-terminal domains.
  • the linking sites thus generally will be between the side chains of the residues.
  • a and B represent the peptides of this invention and are the same or different.
  • a and B are single peptides or head-to-tail polymers of two or more of such peptides.
  • C represents one or more bonds or crosslinking moieties.
  • Lys/Asp cyclization has been accomplished using N ⁇ -Boc-amino acids on solid-phase support with Fmoc/9-fluorenylmethyl (OFm) side-chain protection for
  • Lys/Asp the process is completed by piperidine treatment followed by cyclization.
  • Glu and Lys side chains also have been crosslinked in preparing cyclic or bicyclic peptides: the peptide is synthesized by solid phase chemistry on a p-methylbenzhydrylamine resin. The peptide is cleaved from the resin and depr otected. The cyclic peptide is formed using diphenylphosphorylazide in diluted methylformamide. For an altemative procedure, see
  • thiomethylene bridges ⁇ Tetrahedron Letters (1984) 25, 2067-2068). See also Cody et al., J. Med. Chem. (1985) 28, 583.
  • the desired cyclic or polymeric peptides are purified by gel filtration followed by reversed-phase high pressure liquid chromatography or other conventional procedures.
  • the peptides are sterile filtered and formulated into conventional pharmacologically acceptable vehicles.
  • a peptide derivative bound to a polymer support, depicted by intermediate II, may be prepared by sequential coupling of individual amino acid derivatives by standard techniques.
  • Methods Merrifield, R. B., J. Am. Chem. Soc. (1963) 85, 2149-2154; Stewart, J. M. and Young, J. D., Solid Phase Peptide Synthesis (1984), Pierce Chemical Co., Rockford, IL and additional references cited in the above publications.
  • the terminal amino group is acylated with a suitable carboxylic acid derivative III.
  • the acylation to yield IV may be accomplished using a number of standard methods which require activation of the carboxylic acid group of III.
  • activation may be obtained by the addition of an equimolar amount of dicyctohexylca od iimfcte or related carbodiimide reagent. If desired an additive such as 1-hydroxybenztriazole or N-hydroxysuccinimide may be incorporated.
  • the carboxyl group may be activated by conversion to a halo derivative.
  • the chtoride may be obtained by treatment of the acid with thionyl chtoride or oxalyl chtoride in a compatible solvent such as dichtoromethane, toluene, or ethylene dichloride if desired.
  • the substituent W is chosen such that it is readily displaceable by the group X.
  • R-i ⁇ may be selectively cleaved from X using a very dilute solution of a strong acid such as trifluoroacetic acid in a solvent compatible with the polymer resin.
  • resin compatible solvents are dimethylacetamk e, dimethylformamide or dichtoromethane and the like.
  • the end result of the cleavage process is replacement of the R-f ⁇ group with a hydrogen atom.
  • a base such as N-methylmorpholine may be incorporated into the reactton.
  • Rg may be a group which affords an ester such as methoxy, ethoxy, benzyloxy, t-butyloxy and the like or an amide or substituted amide.
  • R15 may be a protecting group such as ten 1 -butyloxycarbonyl.
  • Final cleavage of the cyclized peptide product from the polymer resin may be accomplished in a variety of ways dependent upon the type of resin used and the chemical linkage between the cyclized peptide and the resin. If, for example, the resin is derived from a polymerized p-alkoxybenzyl alcohol derivative, then cleavage of the peptide-resin linkage may be carried out using a strong acid such as trifluoroacetic acid. If desired, additives such as phenol, anisoie and ethanedithtol may be added to the reactton.
  • the groups Rg and R15 may be chosen, if desired, to also be cleavable concurrently with cleavage of the cyclized peptide from the polymer resin.
  • the crude product thus obtained may be further purified using chromatographic or other methods of chemical purification to obtain I. Further derivatization of I may be earned out if desired.
  • the linear peptide derivative IV may be cleaved from the resin prior to cyclization to yield VI.
  • IV is synthesized on a polystyrene resin the cleavage can be accomplished using liquid hydrogen flouride.
  • the groups Rg, R15 and R16 may, if desired, be cleaved concurrently under these conditions.
  • Rg are t- butyloxy, benzyloxy or cyclohexyloxy
  • R15 is t-butyloxycarbonyl
  • R16 is triphenylmethyl or p-methylbenzyl if X is either O or S, or t-butoxycarbonyl if X is NR13. Cleavage of these groups would result in Rg being OH and R15 and 16 being hydrogen.
  • the peptide derivative VI may then be cyclized in solution in the presence of a weak base such as ammonium hydroxide.
  • the group W is as described in Method A.
  • the resulting crude I may then be purified as described above in Method A.
  • the purified I may be further transformed as described in Method A. Additionally and if desired, when X is NR13 and R13 is hydrogen, I may be acylated with, for example, acetyl chloride, acetic anhydride or benzoyl chtoride, methanesulfonyl chtoride or p-toluenesuKonyl chtoride and the like.
  • Intermediate VI may be prepared by the sequential coupling of amino acid derivatives in solution without the use of polymer resin or other solid supports.
  • the methods useful for solution phase peptide synthesis are well documented in the chemical literature and are known to those skilled in the art (Houben-Weyl, Methoden der Organischen Chemie, 4th Edn., Vol. 15, Georg Thieme Verlag, Stuttgart 1974).
  • the attached substituents R-j, Rg, R15 and Ri 6 may be chosen such that they are transformable concurrently or sequentially as described in Methods A and B above. Cyclization of of VI wherein R ⁇ is H under conditions described above in Method B will provide compounds of Formula I.
  • the carbon atoms bearing the (CH2)n sidechain and the (CH2)m sfctechain are generally preferred to have the S configuration.
  • the carbon atom bearing the substituents R2 and R3 is generally preferred to have a configuratton corresponding to that of a D amino acid.
  • the configuratton may be assigned R or S depending on the chemical compositton of R2 and R3.
  • the compounds described in this invention may be isolated as the free acid or base or converted to salts of various inorganic and organic acids and bases. Such salts are within the scope of this invention. .Examples of such salts include ammonium, metal salts like sodium, potassium, calcium and magnesium; salts with organic bases like dtoyctohexylamine, N-methyl-
  • Salts with inorganic and organic acids may be likewise prepared, for example, using hydrochloric, hydrobromic, sulfuric, phosphoric, trifluoroacetic, methanesuifonto, malic, maleic, fumaric and the like.
  • Non- toxic and physiologically compatible salts are particulariy useful although other less desirable salts may have use in the processes of isolation and purification.
  • a number of methods are useful for the preparation of the salts described above and are known to those skilled in the art. For example, reactton of the free acid or free base form of a compound of Formula I with one or more molar equivalents of the desired acid or base in a solvent or solvent mixture in which the salt is insoluble; or in a solvent like water after which the solvent is removed by evaporation, distillation or freeze drying.
  • the free acid or base form of the product may be passed over an ion exchange resin to form the desired salt or one salt form of the product may be converted to another using the same general process.
  • the compounds described in the present invention inhibit the binding of fibrinogen to its receptor on platelets, GP lib Hl a , and thus prevent the aggregation of platelets and the formation of platelet plugs, emboii and thrombii in the circulatory system in mammals.
  • Thromboembolic disorders have been shown to be directly related to the susceptibility of blood platelets to aggregate. Mammals exposed to medical procedures such as angioplasty and thrombolytic therapy are particularly susceptible to thrombus formation.
  • the compounds of the present invention can be used to inhibit thrombus formation following angioplasty.
  • thrombolytic agents such as tissue plasminogen activator and its derivatives (US patents 4,752,603; 4,766,075; 4,777,043; EP 199,574; EP 0238,304; EP 228,862; EP 297,860; PCT WO89/04368; PCT WO89/00197), streptokinase and its derivatives, or urokinase and its derivatives to prevent arterial reocclusion following thrombolytic therapy.
  • tissue plasminogen activator and its derivatives US patents 4,752,603; 4,766,075; 4,777,043; EP 199,574; EP 0238,304; EP 228,862; EP 297,860; PCT WO89/04368; PCT WO89/00197
  • streptokinase and its derivatives or urokinase and its derivatives to prevent arterial reocclusion following thrombolytic therapy.
  • the compounds of the present invention may be administered prior to,
  • Mammals exposed to renal dialysis, blood oxygenation, cardiac catheterization and similar medical procedures as well as mammals fitted with certain prosthetic devices are also susceptible to thromboembolic disorders.
  • Physiologic conditions, with or without known cause may also lead to thromboembolic disorders.
  • the compounds described herein are useful in treating thromboembolic disorders in mammals.
  • the compounds described herein may also be used as adjuncts to anticoagulant therapy, for example in combination with aspirin, heparin or warfarin and other anticoagulant agents.
  • the application of the compounds described herein for these and related disorders will be apparent to those skilled in the art.
  • inhibitors of the fibrinogen-platelet interaction is guided by in vitro receptor binding assays and in vitro platelet aggregation inhibition assays.
  • Microtiter plates are coated with fibrinogen (10 ⁇ g/ml) and then blocked with TACTS buffer containing 0.5% bovine serum albumin (BSA).
  • TACTS buffer contains 20mM Tris.HCI, pH 7.5, 0.02% sodium azide, 2 mM calcium chtoride, 0.05% Tween 20, 150 mM sodium chtoride.
  • PBS phosphate buffered saline
  • solubilized GP lib Hla receptor 40 ⁇ g/ml
  • platelet aggregation assays may be performed in human platelet rich plasma (PRP).
  • PRP human platelet rich plasma
  • Fifty millil ' rters of whole human blood (9 parts) is drawn on 3.6% sodium citrate (1 part) from a donor who has not taken aspirin or related medications for at least two weeks.
  • the blood is centrifuged at 160 x g for 10 min at 22° C and then altowed to stand for 5 min after which the PRP is decanted.
  • Platelet poor plasma (PPP) is isolated from the remaining blood after centrifugation at 2000 x g for 25 min. T e platelet count of the PRP was adjusted to ca.300,000 per microliter with PPP.
  • a 225 ⁇ L aliquot of PRP plus 25 ⁇ L of either a dilution of the test sample or a control (PBS) is incubated for 5 min in a Chrono-log Whole Blood Aggregometer at 25 * C.
  • An aggregating agent (collagen, 1 mg/ml; U46619, 100 ng/ml; or ADP, 8 ⁇ M) is added and the platelet aggregation recorded.
  • the compounds of this invention may be utilized in compositions such as tablets, capsules or elixers for oral administration; suppositories for rectal administration; sterile solutions or suspensions for injectable administration, and the like.
  • Animals in need of treatment using compounds of this invention can be administered dosages that will provide optimal efficacy.
  • the dose and method of administration will vary from animal to animal and be dependent upon such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.
  • Dosage Formulations of the cyclic polypeptides of the present invention are prepared for storage or administration by mixing the the cyclic polypeptide having the desired degree of purity with physiologically acceptable carriers, excipients, or stabilizers.
  • physiologically acceptable carriers excipients, or stabilizers.
  • Such materials are non-toxic to the recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, acetate and other organic acid salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) peptides such as polyarginine, proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as poiyvinylpyrrolidinone; amino acids such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, or dextrins; chelating agents such as EDTA
  • Dosage formulations of the cyclic polypeptides of the present invention to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes such as 0.2 microne membranes. Cyclic polypeptide formulations ordinarily will be stored in lyophilized form or as an aqueous solution.
  • the pH of the cyclic polypeptide preparations typically will be between 3 and 11 , more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of cyclic polypeptide salts. While the preferred route of administration is by hypodermic injection needle, other methods of administration are also anticipated such as suppositories, aerosols, oral dosage formulations and topical formulations such as ointments, drops and dermal patches.
  • Therapeutic cyclic polypeptide formulations generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by hypodermic injection needle.
  • Therapeutically effective dosages may be determined by either in vitro or in vivo methods.
  • One method of evaluating therapeutically effective dosages consists of taking the cyclic polypeptide cyclo-S-acetyl-Gly-Lys-Gly -Asp-Cys-OH and determining a 50% inhibitory concentration (IC50) of inhibiting fibrinogen binding to the GP lib Hla platelet receptor. Similarly, in a platelet aggregation assay using the same cyclic peptide, the IC50 is measured. Based upon such in vitro assay techniques, a therapeutically effective dosage range may be determined. For each particular cyclic polypeptide of the present invention, individual determinations may be made to determine the optimal dosage required.
  • the range of therapeutically effective dosages will naturally be influenced by the route of administration. For injection by hypodermic needle it may be assumed the dosage is delivered into the body's fluids. For other routes of administration, the absorption efficiency must be individually determined for each cyclic polypeptide by methods well known in pharmacology.
  • the range of therapeutic dosages is from about 0.001 nM to 1.0 mM, more preferably from 0.1 nM to 100 ⁇ M, and most preferably from 1.0 nM to 50 ⁇ M.
  • Typical formulation of compounds of Formula I as pharmaceutical compositions are discussed below.
  • a compound or mixture of compounds of Formula I as the free acid or base form or as a pharmaceutically acceptable salt, is compounded with a physiologically acceptable vehicle, carrier, excipient, binder, preservative, stabilizer, flavor, etc., as called for by accepted pharmaceutical practice.
  • a physiologically acceptable vehicle, carrier, excipient, binder, preservative, stabilizer, flavor, etc. as called for by accepted pharmaceutical practice.
  • the amount of active ingredient in these compositions is such that a suitable dosage in the range indicated is obtained.
  • Typical adjuvants which may be incorporated into tablets, capsules and the like are a binder such as acacia, com starch or gelatin; an excipient such as microcrystalline cellulose; a disintegrating agent like com starch or alginic acid; a lubricant such as magnesium stearate; a sweetening agent such as sucrose or lactose; a flavoring agent such as peppermint, wintergreen or cherry.
  • a binder such as acacia, com starch or gelatin
  • an excipient such as microcrystalline cellulose
  • a disintegrating agent like com starch or alginic acid
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose or lactose
  • a flavoring agent such as peppermint, wintergreen or cherry.
  • a liquid carrier such as a fatty oil.
  • Other materials of various types may be used as coatings or as modifiers of the physical form of the dosage unit.
  • a syrup or elixer may contain the active compound, a sweetener such as sucrose, preservatives like propyl paraben, a cotoring agent and a flavoring agent such as cherry.
  • a sweetener such as sucrose
  • preservatives like propyl paraben a cotoring agent
  • a flavoring agent such as cherry.
  • Sterile composittons for injectton can be formulated according to conventional pharmaceutical practice. For example, dissolution or suspension of the active compound in a vehicle such as water or naturally occurring vegetable oil like sesame, peanut, or cottonseed oil or a synthetic fatty vehicle like ethyl oleate or the like may be desired. Buffers, preservatives, antioxidants and the like can be incorporated according to accepted pharmaceutical practice.
  • common ⁇ -amino acids may be described by the standard three letter amino acid code when referring to intermediates and final products.
  • common ⁇ - amino acids is meant those amino acids incorporated into proteins under mRNA direction. Standard abbreviattons are listed in The Merck Index, 10th Edition, pp Misc-2 - Misc-3. Unless otherwise designated the common ⁇ -amino acids have the natural or "L"- configuratton at the alpha carbon atom. If the code is preceded by a "D" this signifies the opposite enantiomer of the common ⁇ -amino acid.
  • Example 2 The compound prepared in Example 1 is dissolved in deionized water (1mg/ml) and the pH of the solution is adjusted to 7.0-8.5 with ammonium hydroxide. After stirring for 4 hr at ambient temperature the reactton solution is acidified to pH 3.0 - 3.5 with trifluoroacetic acid and then lyophilized. The resulting crude product is purified by HPLC using the conditions described in Example 1. The desired title compound elutes after 11 minutes.
  • Bromoacetyl-Gly-Lys(t-butyloxycarbonyl)-Gly-Asp(beta-t-butyl)-Cys(S- triphenylmethyl)-O-(polymer resin) is prepared using standard solid phase peptide synthesis utilizing fiuorenylmethoxycarbonyl (FMOC) protecting group chemistry on a p-alkoxybenzyl alcohol resin.
  • FMOC fiuorenylmethoxycarbonyl
  • substituted bromoacetic acids listed below may be used in place of bromoacetic acid in Example 1 and in combination with the amino acid derivatives listed above to provide the compounds shown in Table 1 with variable substituents at R5, R&.
  • bromoacetic acid is used in combination with the amino acid derivatives listed above R5 and Q are hydrogen
  • L-Pennicillamine may be substituted for L-cysteine in Example 1 to produce compounds in Table 1 where R7 and Re are methyl.
  • Example 7 The purified product from Example 2 is dissolved in water at a concentration of 10 mg per mL. The pH of the solution is adjusted to 7. A 50% solution of hydrogen peroxide is added to make a final concentration of 3% hydrogen peroxide and the resulting reaction mixture is stirred overnight at room temperature. The solutton is toaded directly onto an octadecylsilyl reverse phase chromatography column. The sulfoxide isomers formed in the reactton are eluted with a linear gradient of acetonitrile in 1% trifluoroacetic acid in water. Example 7
  • Microtiter plates are coated with fibrinogen (10 ⁇ g/ml) and then blocked with TACTS buffer containing 0.5% BSA.
  • TACTS buffer contains 20mM Tris.HCI, pH 7.5, 0.02% sodium azkJe, 2 mM calcium chloride, 0.05% Tween 20, 150 mM sodium chtoride.
  • the plate is washed with phosphate buffered saline containing 0.01% Tween 20 and a dilution of the sample to be determined added, foltowed by addition of solubilized HbHIa receptor (40 ⁇ g/ml) in TACTS, 0.5% BSA.
  • Vitronectin Binding to Vitronectin Receptor a. Coat 96 -well microliter plates (Nunc Maxisorp) with human vitronectin (Telios) made up at 15 ⁇ g/ml in PBS. Use 50 ⁇ l/well. Incubate overnight 4 * C. b. Remove coat solutton. Wash plates one time with 200 ⁇ l of assay buffer (50 mM Tris, 100 mM NaCI, 1 mM CaCl2, 1 mM MgCl2, 1 mM MnCl2, pH 7.4) with the addition of 3.5% BSA in assay buffer. Add additional 150 ⁇ l assay buffer/well. Block plate for 1 hour at room temperature. c.
  • test compounds in assay buffer Prepare human vitronectin receptor, purified at 50 ⁇ g/ml concentration. d. Add 25 ⁇ l of test compounds or buffer control into the plates. Add 25 ⁇ l of the receptor solution into the plates. Incubate at room temperature with shaking for 1 hour, e . Prepare antibody solution during incubation. Combine 4B12 mab (a ⁇ 3 specific antibody) at 1 :1650 with a rabbit F(ab')2 anti-murine Fc-HRP conjugate (Pel-Freez)l :7500 in assay buffer. f. Decant plates. Wash 4 times (150 ⁇ l/well) with PBS, .05% Tween 20. Add in 50 ⁇ l of the antibody solution per well.
  • the present invention may have application in the treatment of a large group of disorders associated with, or characterized by, a hyperthrombotto state.
  • disorders are genetic or acquired deficiencies of factors which normally prevent a hyperthrombotto state; medical procedures such as angioplasty and thrombolytic therapy; mechanical obstructtons to blood flow, such as tumor masses, prosthetic synthetic cardiac valves, and extracorporeal pertuston devices; atherosclerosis; and coronary artery disease.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Diabetes (AREA)
  • Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
EP91919821A 1990-11-02 1991-10-24 Blutplättchenaggregationshemmstoffe Ceased EP0555328A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60841790A 1990-11-02 1990-11-02
US608417 1990-11-02

Publications (1)

Publication Number Publication Date
EP0555328A1 true EP0555328A1 (de) 1993-08-18

Family

ID=24436416

Family Applications (1)

Application Number Title Priority Date Filing Date
EP91919821A Ceased EP0555328A1 (de) 1990-11-02 1991-10-24 Blutplättchenaggregationshemmstoffe

Country Status (4)

Country Link
EP (1) EP0555328A1 (de)
JP (1) JPH06502407A (de)
CA (1) CA2092315A1 (de)
WO (1) WO1992007870A1 (de)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6017877A (en) * 1990-04-06 2000-01-25 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
US5780303A (en) * 1990-04-06 1998-07-14 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
US6521594B1 (en) 1990-04-06 2003-02-18 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
US5672585A (en) * 1990-04-06 1997-09-30 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
US5635477A (en) * 1991-09-30 1997-06-03 The Dupont Merck Pharmaceutical Company Cyclic compounds useful as inhibitors of platelet glycoprotein IIB/IIIA
CA2101599A1 (en) * 1992-08-31 1994-03-01 Wilhelm Bannwarth Tri- and tetracyclic compounds
WO1994011398A1 (en) * 1992-11-18 1994-05-26 The Du Pont Merck Pharmaceutical Company CYCLIC COMPOUNDS LINKED BY A HETEROCYCLIC RING USEFUL AS INHIBITORS OF PLATELET GLYCOPROTEIN IIb/IIIa
EP0691986B1 (de) * 1993-03-29 1998-12-02 The Du Pont Merck Pharmaceutical Company Prozess und zwischenverbindungen zur herstellung von blaettchen glycoprotein iib/iiia hemmern
US5879657A (en) * 1993-03-30 1999-03-09 The Dupont Merck Pharmaceutical Company Radiolabeled platelet GPIIb/IIIa receptor antagonists as imaging agents for the diagnosis of thromboembolic disorders
AU6663794A (en) * 1993-05-13 1994-12-12 Du Pont Merck Pharmaceutical Company, The Processes and intermediate compounds useful for the preparation of platelet glycoprotein iib/iiia inhibitors
JP2004528025A (ja) 2001-02-21 2004-09-16 サーロメッド・インコーポレーテッド 修飾されたアネキシン蛋白質及び血栓症を防ぐための方法
US7635676B2 (en) 2001-02-21 2009-12-22 Alavita Pharmaccuticals, Inc. Modified annexin proteins and methods for their use in organ transplantation
US7635680B2 (en) 2001-02-21 2009-12-22 Alavita Pharmaceuticals, Inc. Attenuation of reperfusion injury
US7645739B2 (en) 2001-02-21 2010-01-12 Alavita Pharmaceuticals, Inc. Modified annexin compositions and methods of using same
WO2011028915A2 (en) * 2009-09-03 2011-03-10 University Of Chicago Platelet aggregation inhibitors
CN103113456B (zh) * 2013-03-05 2014-07-16 中国药科大学 具有抗血小板聚集活性的僵蚕多肽及其制备方法和应用
CN108203457B (zh) * 2016-12-20 2022-09-06 山西医科大学 一种靶向抑制血小板聚集的抗栓小肽ωKWR

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZW6189A1 (en) * 1988-05-09 1990-05-09 Smithkline Beckman Corp Anti-aggregatory peptides
US5318899A (en) * 1989-06-16 1994-06-07 Cor Therapeutics, Inc. Platelet aggregation inhibitors
US5384309A (en) * 1989-07-17 1995-01-24 Genentech, Inc. Cyclized peptides and their use as platelet aggregation inhibitors
AU6470590A (en) * 1989-10-23 1991-04-26 Smithkline Beecham Corporation Cyclic anti-aggregatory peptides
EP0749979A3 (de) * 1990-04-06 1998-03-11 La Jolla Cancer Research Foundation Methode und Zusammensetzung zur Behandlung von Thrombose

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9207870A1 *

Also Published As

Publication number Publication date
CA2092315A1 (en) 1992-05-03
WO1992007870A1 (en) 1992-05-14
JPH06502407A (ja) 1994-03-17

Similar Documents

Publication Publication Date Title
EP0578728B1 (de) PLAETTCHENAGGREGATIONSINHIBITOREN MIT HOHER SPEZIFIZITAET ZUM GP IIbIIIa
JP2755351B2 (ja) 抗―凝集ペプチド
EP0482080B1 (de) Kleine zyklische inhibitoren der blutplättchenaggregation
JP2922281B2 (ja) 環状抗凝集性ペプチド類
JP3281370B2 (ja) 血小板凝集阻害剤
US6001961A (en) Cyclic adhesion inhibitors
US5643872A (en) Cyclic anti-aggregatory peptides
Barker et al. Cyclic RGD peptide analogs as antiplatelet antithrombotics
EP0555328A1 (de) Blutplättchenaggregationshemmstoffe
JPH03118330A (ja) フイブリノーゲンレセプター拮抗剤
US6127335A (en) Cyclic adhesion inhibitors
EP0570507A1 (de) Anti-aggregierende peptide, die einen aromatischen ester oder ein amid aufweisen
JP2669510B2 (ja) ヒルジンのアミノ酸配列に基づくトロンビン阻害剤
AU641215B2 (en) Stabilized sulfonate, sulfate, phosphonate and phosphate derivatives of hirudin
AU690923B2 (en) Linear adhesion inhibitors
JP4127325B2 (ja) ビオチン誘導体
JP3532920B2 (ja) 三機能性の抗血栓及び抗血小板ペプチド類
US5858972A (en) Antithrombotic agents and methods of use
US20030191278A1 (en) Cyclic pentapeptides and their preparation
JPH05504559A (ja) メルカプタンまたはスルフィド基含有の抗―凝集ペプチド
JPH06157588A (ja) 新規な細胞接着活性ペプチド誘導体

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LI LU NL SE

17P Request for examination filed

Effective date: 19930524

17Q First examination report despatched

Effective date: 19960322

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

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 19990722