US20100279924A1 - Peptides and peptide derivatives as well as pharmaceutical compositions containing the same - Google Patents

Peptides and peptide derivatives as well as pharmaceutical compositions containing the same Download PDF

Info

Publication number
US20100279924A1
US20100279924A1 US12/820,804 US82080410A US2010279924A1 US 20100279924 A1 US20100279924 A1 US 20100279924A1 US 82080410 A US82080410 A US 82080410A US 2010279924 A1 US2010279924 A1 US 2010279924A1
Authority
US
United States
Prior art keywords
pro
arg
gly
peptide
individual
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.)
Abandoned
Application number
US12/820,804
Inventor
Peter Petzelbauer
Rainer Henning
Sonja Reingruber
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.)
Fibrex Medical Research and Development GmbH
Original Assignee
Fibrex Medical Research and Development GmbH
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 Fibrex Medical Research and Development GmbH filed Critical Fibrex Medical Research and Development GmbH
Priority to US12/820,804 priority Critical patent/US20100279924A1/en
Assigned to IKARIA DEVELOPMENT SUBSIDIARY TWO LLC reassignment IKARIA DEVELOPMENT SUBSIDIARY TWO LLC LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: FIBREX MEDICAL RESEARCH & DEVELOPMENT GESMBH, FIBREX MEDICAL, INC.
Publication of US20100279924A1 publication Critical patent/US20100279924A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to peptides and peptide derivatives, to the production thereof as well as to their use for preparing a therapeutically and/or preventively active drug and to such a pharmaceutical drug.
  • EP1586586 describes the use of peptides from the sequence of fibrin possessing anti-inflammatory effects.
  • Said effect may be based on the fact that the fibrin and fibrin fragments generated during the breakdown thereof bind to endothelial cells via its neo-N-terminus of the Bbeta-chain and to cells in the bloodstream via the sequence of the Aalpha-chain, thereby leading to the adhesion and transmigration of these cells into the tissue.
  • the binding partner of the fibrin and fibrin fragments to the endothelial cells is the protein vascular endothelial (VE) cadherin, which is expressed exclusively in the adherens junction between neighboring endothelial cells.
  • VE vascular endothelial
  • the peptides according to the invention block this interaction and thereby counteract the transmigration of blood cells.
  • the natural defense against infections by the leukocytes in the blood is not adversely effected.
  • the composition of the same such as granulocytes, lymphocytes and monocytes, remains unaffected so that the natural defense process is maintained.
  • Fibrinogen is produced in the liver and, in this form, is biologically inactive and normally is provided in the blood at concentrations of around 3 g/l. Proteolytic cleavage of the proenzyme prothrombin results in the formation of thrombin, which cleaves off the fibrinopeptides A and B from the fibrinogen. In this way, fibrinogen is transformed into its biologically active form. Fibrin and fibrin cleavage products are generated.
  • Thrombin is formed whenever blood coagulation is activated, i.e., with damage to the tissue, be it of inflammatory, traumatic or degenerative genesis.
  • the formation of fibrin as mediated by thrombin is basically a protective process aimed at quickly sealing any defects caused to the vascular system.
  • the formation of fibrin also is a pathogenic process.
  • the appearance of a fibrin thrombus as the triggering cause of cardiac infarction is one of the most prominent problems in human medicine.
  • Fibrin binds to endothelial cells via its neo-N-terminus of Bbeta by means of the sequence to Bbeta and to cells in the bloodstream by means of the sequence Aalpha, thereby leading to the adhesion and transmigration of cells into the tissue.
  • the peptides or proteins according to the invention may prevent the adhesion of cells from the bloodstream to endothelial cells of the vascular wall and/or their subsequent transmigration from the blood into the tissue.
  • WO92/16221 describes polypeptides which are covalently linked to long-chain polymers, as for instance methoxy-polyethylene glycol (PEG).
  • PEG methoxy-polyethylene glycol
  • the binding of polypeptides to such polymers frequently results in a prolongation of the biological half-life of these polypeptides and delays their renal excretion.
  • PEG-groups exerts this effect in a way proportional to the molecular weight of the PEGylated peptide, as, up to a certain size of the molecule, the glomular filtration rate is inversely proportional to the molecular weight.
  • WO2004/101600 also describes new poly(ethylene glycol)-modified compounds and their use, in particular with emphasis on modified peptides activating the erythropoietin receptor.
  • a prolonged biological half-life is advantageous for various therapeutic uses of peptides. This is in particular true in cases of chronic diseases where the administration of the active agent over a prolonged period of time is indicated. With such indications this may improve the patient's compliance, as applying the active agent once a day will for instance be accepted more easily than continuous infusion.
  • a prolongation of the persistency of polypeptides may be obtained by modifying them in such a way that their degradation by proteolytic enzymes (e.g., exo- or endoproteases or peptidases) is prevented.
  • Calcitonin Lee et al., Pharm. Res. 1999, 16, 813
  • Growth Hormone Releasing Hormone Esposito et al., Advanced Drug Delivery Reviews, 2003, 55, 1279
  • Glucagon like peptide 1 Lee et al., Bioconjugate Res. 2005, 16, 377
  • Pegvisomant Ross et al., J. Clin. Endocrin. Metab.
  • peptides derived from the chain of the Bbeta(15-42)-fibrin fragment wherein one or several amino acids of the natural fibrin sequence have been substituted by other amino acids, as well as derivatives modified at the C-terminal end of the peptide sequence also have strong anti-inflammatory effects.
  • peptides and peptide derivatives the modification of which prevents their destruction by proteases or peptidases, as well as to peptide-PEG-conjugates derived from the basic sequence of the Bbeta(15-42)-fibrin fragment.
  • the invention relates to modified peptides which are derived from the chain of the Bbeta(15-42)-fibrin fragment and wherein one or several of the amino acids of the sequence have been substituted by genetically encoded or not genetically encoded amino acids or peptidomimetics. They may exist as free peptides or as C-terminal derivative and/or being linked to a polyethylene glycol (PEG)-polymer, and have anti-inflammatory and/or endothelium stabilizing effects. Esters or amides may for instance be taken into consideration as C-terminal derivatives.
  • the inventive compounds may have conservative substitutions of amino acids as compared to the natural sequence of fibrin of the warm blooded animals to be treated in one or several positions.
  • a conservative substitution is defined as the side chain of the respective amino acid being replaced by a side chain of similar chemical structure and polarity, the side chain being derived from a genetically coded or not genetically coded amino acid. Families of amino acids of this kind having similar side chains are known in the art.
  • amino acids having basic side chains lysines, arginines, histidine
  • acidic side chains aspartic acid, glutamic acid
  • uncharged polar side chains glycine, aspartamic acid, glutamine, serine, threonine, tyrosine, cysteine
  • non-polar side chains alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains threonine, valine, isoleucine
  • aromatic side chains tyrosine, phenylalanine, tryptophane, histidine.
  • amino acids having basic side chains lysines, arginines, histidine
  • acidic side chains aspartic acid, glutamic acid
  • uncharged polar side chains glycine, aspartamic acid, glutamine, serine, threonine, tyrosine, cysteine
  • non-polar side chains
  • the invention provides peptides and peptide derivatives of the following general Formula I:
  • the invention provides peptides and peptide derivatives of the general Formula I, wherein:
  • the invention provides peptides and peptide derivatives of Formula II,
  • the present invention provides compounds of Formula II, wherein
  • the invention provides peptide derivatives of Formula III,
  • residues X 19 , X 20 and X 21 each are a glycine residue and the remaining one is a residue C—(S-succinimido)-(PEG 5-40K ), the succinimido residue being linked to the sulfur atom of the cysteine residue via C-atom 3, and wherein X 17 denotes NR 2 R 3 , R 2 and R 3 being identical or different and being hydrogen or (C 1 -C 10 )-alkyl (SEQ ID NO: 10), as well as the physiologically acceptable salts thereof.
  • the invention provides peptide derivatives of Formula III,
  • residues X 19 , X 20 and X 21 each are a glycine residue and the remaining one is a residue K-(PEG 5-40K ), the PEG-residue being linked via the nitrogen atom in the side chain of the lysine residue, and wherein X 17 denotes NR 2 R 3 , R 2 and R 3 being identical or different and being hydrogen or (C 1 -C 10 )-alkyl (SEQ ID NO: 11), as well as the physiologically acceptable salts thereof.
  • phenylalanine is F
  • leucine is L
  • isoleucine is I
  • methionine is M
  • valine V
  • serine is S
  • proline is P
  • threonine is T
  • alanine is A
  • tyrosine Y
  • histidine H
  • glutamine Q
  • asparagine is N
  • lysine is K
  • aspartic acid is D
  • glutamic acid E
  • cysteine is C
  • tryptophan W
  • arginine is R
  • glycine G.
  • amino acid residues in the compounds of Formula I may either be present in their D or their L configuration.
  • peptide refers to a polymer of these amino acids, which are linked via an amide linkage.
  • FIG. 1 is a bar graph illustrating the number of neutrophils, specifically polymorphonuclear leukocytes (PMN), present in bronchioalveolar lavage liquid of mice having LPS-induced pneumonitis treated with either Control (100 ⁇ l NaCl) or 4.8 mg/kg of the agent of Example 1 (dissolved in 100 ⁇ l NaCl).
  • PMN polymorphonuclear leukocytes
  • “Physiologically acceptable” means that salts are formed with acids or bases the addition of which does not have undesirable effects when used for humans. Preferable are salts with acids or bases the use of which is listed for use with warm blooded animals, in particular humans, in the US Pharmacopoeia or any other generally recognized pharmacopoeia.
  • PEG stands for a polyethylene glycol residue having a molecular weight of between 5,000 and 60,000 Dalton, this molecular weight being the maximum of a molecular weight distribution, so that individual components of the mixture may have a higher or lower molecular weight.
  • the invention furthermore concerns processes for the production of the peptides and peptide derivatives of general Formula (I), characterized in that, either
  • Embodiments of the respective processing steps are not new per se and will be clear to an experienced specialist in the field of organic synthesis.
  • a cysteine (C)-residue may be reacted with PEG-maleimide, resulting in a succinimide residue as spacer for residue Z.
  • a further possibility is reacting an optionally activated C-terminal carboxy residue with an aminoalkyl-substituted PEG residue.
  • a further possibility is the introduction of a PEG residue by reacting an aldehyde-substituted PEG residue with the ⁇ -amino function of a lysine residue.
  • Activated PEG reagents having suitable spacers and reactive groups may for instance be obtained from NOF Corporation (Tokyo, Japan).
  • the substances according to the invention and the use of the substances according to the invention for the production of a pharmaceutical drug are of particular significance for the production of a pharmaceutical drug for the therapy of diseases resulting from the tissue-damaging effect of white blood cells, or wherein the integrity and full physiological integrity of the layer of endothelial cells lining the blood vessels is impaired.
  • Diseases belonging to this group are those in context with autoimmunity, as for instance collagenases, rheumatic diseases, inflammatory bowel diseases like Morbus Crohn or Colitis ulcerosa, psoriasis and psoriatic rheumatoid arthritis, and post/parainfectious diseases as well as diseases caused by a graft-versus-host reaction.
  • a healing effect takes place as this medical drug blocks the migration of the white blood cells into the tissue.
  • the white blood cells remain in the blood stream and cannot cause an autoreactive effect harmful to the tissue.
  • This effect of the inventive substances is furthermore important for the treatment of shock conditions, in particular in the case of septic shock triggered by infection with gram-positive or gram-negative bacterial pathogens as well as viral infections and haemorrhagic shock caused by heavy loss of blood because of severe injuries or bacterial or viral infections.
  • inventive substances may generally be used in situations that can be described with the terms “Systemic Inflammatory Response Syndrome (SIRS)”, “Acute Respiratory Distress Syndrome (ARDS)” and organ- or multiorgan failure, respectively.
  • SIRS Systemic Inflammatory Response Syndrome
  • ARDS acute Respiratory Distress Syndrome
  • organ- or multiorgan failure organ- or multiorgan failure
  • a pharmaceutical drug for the therapy and/or prevention of reperfusion trauma following surgically or pharmaceutically induced re-supply with blood e.g., following percutaneous coronary intervention, stroke, vessel surgery, cardiac bypass surgery and organ transplants
  • this pharmaceutical drug inhibits the migration of lymphocytes, neutrophils and monocytes into the wall of the vessel.
  • Reperfusion trauma is caused by a lack of oxygen/acidosis of the cells of the vessel during its re-supply with blood, leading to their activation and/or damage. Because of this, lymphocytes, neutrophils and monocytes adhere to the vessel wall and migrate into it.
  • Blocking the adherence and migration of lymphocytes, neutrophils and monocytes in the vessel wall causes the hypoxy/acidosis-induced damage to abate, without the subsequent inflammatory reaction causing permanent damage to the vessel.
  • the endothelium-stabilizing effect of the inventive compounds furthermore prevents the formation of oedemas as well as any further damage to the organs supplied via the respective blood vessels.
  • the pharmaceutical drug according to the invention may also be used for the transportation of another drug.
  • the inventive drug specifically binds a surface molecule on endothelial cells.
  • drugs linked thereto may be delivered to endothelial cells in high concentrations without any danger of them having side effects at other sites.
  • An example that may be cited here is the use of substances inhibiting the division of cells, which, specifically brought to endothelial cells, may have an antiangiogenetic effect. This brings about a healing effect in tumor patients, as tumor growth is blocked by preventing the proliferation of endothelial cells and thus by preventing neoangiogenesis.
  • inventive compounds themselves may also develop an antiangiogenetic effect, as they, because of their endothelium-stabilizing effect, prevent the endothelial cells from changing into a proliferative phenotype and thus prevent the formation of new capillary blood vessels. Therefore, they are themselves suitable for the treatment of all kinds of tumor diseases as well as the prevention and/or treatment of tumor metastases.
  • inventive compounds of Formula (I) together with pharmaceutical adjuvants and additives may be formulated into pharmaceutical preparations which also are a subject matter of the present invention.
  • a therapeutically effective dose of the peptide or peptide derivative is mixed with pharmaceutically acceptable diluents, stabilizers, solubilizers, emulsifying aids, adjuvants or carriers and brought into a suitable therapeutic form.
  • Such preparations for instance, contain a dilution of various buffers (e.g., Tris-HCl, acetate, phosphate) of different pH and ionic strength, detergents and solubilizers (e.g., Tween 80, Polysorbate 80), antioxidants (e.g., ascorbic acid), and fillers (e.g., lactose, mannitol).
  • buffers e.g., Tris-HCl, acetate, phosphate
  • detergents and solubilizers e.g., Tween 80, Polysorbate 80
  • antioxidants e.g., ascorbic acid
  • fillers e.g., lactose, mannitol
  • compositions according to the invention may be administered orally, parenterally (intramuscularly, intraperitoneally, intravenously or subcutaneously), transdermally or in an erodable implant of a suitable biologically degradable polymer (e.g., polylactate or polyglycolate).
  • a suitable biologically degradable polymer e.g., polylactate or polyglycolate
  • the biological effect and applicability for the claimed use of the inventive compounds may for instance be determined in an assay in which a culture of human umbilical cord endothelial cells is examined microscopically after stimulation with the “N-terminal disulfide knot protein II” (NDSK-II) or with thrombin.
  • the stimulation of endothelial cells causes the formation of gaps between the cells in a densely packed cell layer.
  • Treatment with the inventive compounds may prevent the formation of these gaps, and is successful in closing gaps that have already been formed.
  • This effect is predicative for the protective effect on the endothelium the inventive compounds have throughout the organism.
  • the inventive compounds have an effect in the range of concentrations from 0.01 nM to 1 mM, preferably in the range from 1 nM to 0.1 mM in the bath solution of cells.
  • the effectiveness in vivo may for instance be established using a model of acute pulmonitis in a rodent.
  • the treatment of the animal and the administration of the substance are carried out as described in Example 7 below.
  • the inventive compounds show an effect at a dose ranging from 0.001 mg/kg body weight to 500 mg/kg body weight, preferably at a dose ranging from 0.1 mg/kg to 50 mg/kg.
  • mice are, as described in Example 8, for instance infected with a dose of Dengue viruses, wherein 50% of the animals die within a period of 5-20 days after infection.
  • the inventive compounds bring about a reduction of this mortality at a dose ranging from 0.001 to 500 mg/kg body weight, preferably at a dose ranging from 0.1 to 50 mg/g body weight.
  • Tentagel-S-RAM (Rapp-Polymere) at a load of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • the FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 ⁇ l DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3 ⁇ 900 ⁇ l 30% piperidine in DMF and thorough mixing for 4 minutes.
  • DIC di-isopropy-carbodiimide
  • DIPEA di-isopropy-ethylamine
  • HBt hydroxybenzotriazole
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom fit of the reaction vessel.
  • the amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • the peptide resin When synthesis is completed the peptide resin is dried.
  • the peptide amide is subsequently cleaved off by treatment with trifluoracetic acid/TIS/EDT/water (95:2:2:1 vol) for 2 hours at room temperature.
  • concentration of the solution and precipitation by the addition of ice-cold diethyl ether the crude product (75 mg) is obtained as a solid.
  • the peptide is purified by RP-HPLC on Kromasil RP-18 250-20, 10 ⁇ m in 0.1% TFA with a gradient of 5 on 60% acetonitrile in 40 minutes at a flow rate of 12 ml/min and evaluation of the eluate by means of a UV detector at 215 nm.
  • the purity of the individual fractions is determined by analyt.
  • RP-HPLC and mass spectrometry Following combination of the purified fractions and lyophilisation, 48 mg of pure product are obtained Maldi-TOF, 3036.6 m/z (m.i.).
  • the monomeric peptide is synthesized as in Example 1, Tentagel (Rapp Polymere) being used as resin support here with FMOC-Cys(Trt) as the first amino acid.
  • reaction is carried out with a 2- to 8-fold molar excess of maleinimido-PEG 20K .
  • purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by way of analytical RP-HPLC and MALDI-MS.
  • Tentagel-S-RAM (Rapp-Polymere) at a load of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • the FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 ⁇ l DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3 ⁇ 900 ⁇ l 30% piperidine in DMF and thorough mixing for 4 minutes.
  • DIC di-isopropy-carbodiimide
  • DIPEA di-isopropy-ethylamine
  • HBt hydroxybenzotriazole
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom frit of the reaction vessel.
  • the amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • reaction is carried out with a 2- to 8-fold molar excess of maleinimido-PEG 20K .
  • purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by way of analytical RP-HPLC and MALDI-MS.
  • Tentagel-S-RAM (Rapp-Polymere) having a charge of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • the FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 ⁇ l DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3 ⁇ 900 ⁇ l 30% piperidine in DMF and thorough mixing for 4 minutes.
  • DIC di-isopropy-carbodiimide
  • DIPEA di-isopropy-ethylamine
  • HBt hydroxybenzotriazole
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom fit of the reaction vessel.
  • the amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • the peptide resin When synthesis is completed, the peptide resin is dried.
  • the peptide amide is subsequently cleaved by treatment with trifluoracetic acid/TIS/EDT/water (95:2:2:1 vol) for 2 hours at room temperature.
  • concentration of the solution and precipitation by the addition of ice-cold diethyl ether the crude product (75 mg) is obtained as a solid.
  • the peptide is purified by RP-HPLC on Kromasil RP-18 250-20.
  • the peptide thus obtained is reacted with maleinimido-PEG 20k .
  • purification by means of gel chromatography and lyophilisation, a pure product is obtained, the identity of which is confirmed by way of RP-HPLC and MALDI-MS.
  • Example 1 The biological effect of the compound of Example 1 was established in a model of LPS-induced pneumonitis. C57 Black mice were randomized in two groups of 6 animals each and treated as follows:
  • Group 1 received 100 ng/kg LPS intranasally, immediately after the administration of LPS the mice received 4.8 mg/kg of the agent of Example 1 (dissolved in 100 ⁇ l NaCl) i.p., a second dose followed 60 min after the administration of LPS.
  • Group 2 received 100 ng/kg LPS intranasally, immediately after the administration of LPS the mice received 100 ⁇ l NaCl i.p., 60 min after the LPS administration the mice again received 100 ⁇ l NaCl i.p. Furthermore, 6 hours after the application of LPS, all groups were submitted to a bronchioalveolar lavage and the lungs were removed. From the lavage liquids the number of neutrophils (PMN) was determined.
  • PMN neutrophils
  • mice The biological effect of the compound of Example 3 was established in a model of Dengue virus infection in mice.
  • 5-week-old male BALB/c mice were divided into 2 groups. All animals were infected subcutaneously with a mouse-adapted Dengue virus (DEN-2, strain P23085 at a dose of 1-2 LD 50 .
  • 15 mice received 0.1 ml of 0.8% saline as intramuscular injection (control).
  • the treated animals received 4.8 mg/kg/day of the agent of Example 3 as an intramuscular injection (diluted in 0.1 ml of 0.8% saline) once a day for 5 days, starting on day 3 after the virus infection.
  • reaction is carried out with a suitable excess of Br—CH 2 —CO—NH-PEG 20K .
  • purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by MALDI-MS.
  • Tentagel-S-RAM (Rapp-Polymere) at a load of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • the FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 ⁇ l DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3 ⁇ 900 ⁇ l 30% piperidine in DMF and thorough mixing for 4 minutes.
  • DIC di-isopropy-carbodiimide
  • DIPEA di-isopropy-ethylamine
  • HBt hydroxybenzotriazole
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom frit of the reaction vessel.
  • the amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • reaction is carried out with a suitable excess of Br—CH 2 —CO—NH-PEG 20K .
  • purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by MALDI-MS.
  • Tentagel-S-RAM (Rapp-Polymere) at a charge of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • the FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 ⁇ l DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3 ⁇ 900 ⁇ l 30% piperidine in DMF and thorough mixing for 4 minutes.
  • DIC di-isopropy-carbodiimide
  • DIPEA di-isopropy-ethylamine
  • HBt hydroxybenzotriazole
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom frit of the reaction vessel.
  • the amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • the peptide resin When synthesis is completed the peptide resin is dried.
  • the peptide amide is subsequently cleaved by treatment with trifluoracetic acid/TIS/EDT/water (95:2:2:1 vol) for 2 hours at room temperature.
  • concentration of the solution and precipitation by the addition of ice-cold diethyl ether the crude product (75 mg) is obtained as a solid.
  • the peptide is purified by RP-HPLC on Kromasil RP-18 250-20.
  • the peptide thus obtained is reacted with O-(iodoacetyl)-N-hydroxysuccinimide, followed by amino-ethyl-oxi-PEG 20k .

Abstract

Therapeutic compositions can include modified peptides which are derived from the chain of the Bbeta(15-42)-fibrin fragment and wherein one or several of the amino acids of the sequence have been substituted by genetically encoded or not genetically encoded amino acids or peptidomimetics. They may exist as free peptides or as C-terminal derivative and/or being linked to a polyethylene glycol (PEG)-polymer, and have anti-inflammatory and/or endothelium stabilizing effects. Esters or amides may for instance be taken into consideration as C-terminal derivatives. Processes for production of the peptides and derivatives thereof are also described.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation of U.S. patent application Ser. No. 11/678,535, filed Feb. 23, 2007, which claims priority to Austrian Application No. AT 301/2006, filed Feb. 23, 2006, the entire contents of which applications are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to peptides and peptide derivatives, to the production thereof as well as to their use for preparing a therapeutically and/or preventively active drug and to such a pharmaceutical drug.
  • BACKGROUND OF THE INVENTION
  • EP1586586 describes the use of peptides from the sequence of fibrin possessing anti-inflammatory effects.
  • Said effect may be based on the fact that the fibrin and fibrin fragments generated during the breakdown thereof bind to endothelial cells via its neo-N-terminus of the Bbeta-chain and to cells in the bloodstream via the sequence of the Aalpha-chain, thereby leading to the adhesion and transmigration of these cells into the tissue. The binding partner of the fibrin and fibrin fragments to the endothelial cells is the protein vascular endothelial (VE) cadherin, which is expressed exclusively in the adherens junction between neighboring endothelial cells. The peptides according to the invention block this interaction and thereby counteract the transmigration of blood cells. However, the natural defense against infections by the leukocytes in the blood is not adversely effected. Thus, the composition of the same, such as granulocytes, lymphocytes and monocytes, remains unaffected so that the natural defense process is maintained.
  • Fibrinogen is produced in the liver and, in this form, is biologically inactive and normally is provided in the blood at concentrations of around 3 g/l. Proteolytic cleavage of the proenzyme prothrombin results in the formation of thrombin, which cleaves off the fibrinopeptides A and B from the fibrinogen. In this way, fibrinogen is transformed into its biologically active form. Fibrin and fibrin cleavage products are generated.
  • Thrombin is formed whenever blood coagulation is activated, i.e., with damage to the tissue, be it of inflammatory, traumatic or degenerative genesis. The formation of fibrin as mediated by thrombin is basically a protective process aimed at quickly sealing any defects caused to the vascular system. However, the formation of fibrin also is a pathogenic process. The appearance of a fibrin thrombus as the triggering cause of cardiac infarction is one of the most prominent problems in human medicine.
  • The role which fibrin plays during the extravasation of inflammatory cells from the bloodstream into the tissue, which, on the one hand, is a desired process for the defense against pathogenic microorganisms or tumor cells in the tissue, but, on the other hand, is a process which, by itself, induces or prolongs damage done to the tissue, has so far not been examined at all or not to a sufficient extent. Fibrin binds to endothelial cells via its neo-N-terminus of Bbeta by means of the sequence to Bbeta and to cells in the bloodstream by means of the sequence Aalpha, thereby leading to the adhesion and transmigration of cells into the tissue.
  • By way of the mechanism described above the peptides or proteins according to the invention may prevent the adhesion of cells from the bloodstream to endothelial cells of the vascular wall and/or their subsequent transmigration from the blood into the tissue.
  • WO92/16221 describes polypeptides which are covalently linked to long-chain polymers, as for instance methoxy-polyethylene glycol (PEG). The binding of polypeptides to such polymers frequently results in a prolongation of the biological half-life of these polypeptides and delays their renal excretion. A summary of these properties may be found in Davis et al., Polymeric Materials Pharmaceuticals for Biomedical Use, pp. 441-451 (1980). The addition of PEG-groups exerts this effect in a way proportional to the molecular weight of the PEGylated peptide, as, up to a certain size of the molecule, the glomular filtration rate is inversely proportional to the molecular weight.
  • WO2004/101600 also describes new poly(ethylene glycol)-modified compounds and their use, in particular with emphasis on modified peptides activating the erythropoietin receptor.
  • Further examples for the covalent modification of peptides and proteins with PEG residues are interleukins (Knauf et al., J. Biol. Chem. 1988, 263, 15064; Tsutumi et al., J. Controlled Release 1995, 33, 447), Interferons (Kita et al., Drug Delivery Res. 1990, 6, 157), Catalase (Abuchowski et al., J. Biol. Chem. 1997, 252, 3582). A review of the prior art may be found in Reddy, Ann. of Pharmacotherapy, 2000, 34, 915.
  • A prolonged biological half-life is advantageous for various therapeutic uses of peptides. This is in particular true in cases of chronic diseases where the administration of the active agent over a prolonged period of time is indicated. With such indications this may improve the patient's compliance, as applying the active agent once a day will for instance be accepted more easily than continuous infusion. Apart from increasing the molecular mass by covalent modification, a prolongation of the persistency of polypeptides may be obtained by modifying them in such a way that their degradation by proteolytic enzymes (e.g., exo- or endoproteases or peptidases) is prevented.
  • Using various examples it has been shown that it is necessary to customize the appropriate modification for each peptide so as to prevent a significant influence on the pharmacodynamic effect as compared to the unmodified peptide. In this context the following may be referred to: Calcitonin (Lee et al., Pharm. Res. 1999, 16, 813), Growth Hormone Releasing Hormone (Esposito et al., Advanced Drug Delivery Reviews, 2003, 55, 1279), Glucagon like peptide 1 (Lee et al., Bioconjugate Res. 2005, 16, 377), as well as the growth hormone-receptor antagonist Pegvisomant (Ross et al., J. Clin. Endocrin. Metab. 2001, 86, 1716). The reviews by Caliceti and Veronese (Adv. Drug Deliv. Rev. 2003, 55 1261) and by Harris and Chess (Nature Rev. Drug Discovery 2003, 2, 214) discuss that in case of designing peptide- or protein-PEG-conjugates it is necessary to take into consideration the structure of the original substance, the molecular weight of the peptide and the polymer, the number of conjugated polymer chains as well as the linker chemistry, so as to obtain an effective peptide-PEG-conjugate.
  • SUMMARY OF THE INVENTION
  • Surprisingly, it has now been found that peptides derived from the chain of the Bbeta(15-42)-fibrin fragment, wherein one or several amino acids of the natural fibrin sequence have been substituted by other amino acids, as well as derivatives modified at the C-terminal end of the peptide sequence also have strong anti-inflammatory effects. The same applies to peptides and peptide derivatives the modification of which prevents their destruction by proteases or peptidases, as well as to peptide-PEG-conjugates derived from the basic sequence of the Bbeta(15-42)-fibrin fragment.
  • Thus, the invention relates to modified peptides which are derived from the chain of the Bbeta(15-42)-fibrin fragment and wherein one or several of the amino acids of the sequence have been substituted by genetically encoded or not genetically encoded amino acids or peptidomimetics. They may exist as free peptides or as C-terminal derivative and/or being linked to a polyethylene glycol (PEG)-polymer, and have anti-inflammatory and/or endothelium stabilizing effects. Esters or amides may for instance be taken into consideration as C-terminal derivatives.
  • The inventive compounds may have conservative substitutions of amino acids as compared to the natural sequence of fibrin of the warm blooded animals to be treated in one or several positions. A conservative substitution is defined as the side chain of the respective amino acid being replaced by a side chain of similar chemical structure and polarity, the side chain being derived from a genetically coded or not genetically coded amino acid. Families of amino acids of this kind having similar side chains are known in the art. They comprise for instance amino acids having basic side chains (lysines, arginines, histidine), acidic side chains (aspartic acid, glutamic acid), uncharged polar side chains (glycine, aspartamic acid, glutamine, serine, threonine, tyrosine, cysteine), non-polar side chains (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (threonine, valine, isoleucine) and aromatic side chains (tyrosine, phenylalanine, tryptophane, histidine). Such conservative substitutions of side chains may preferably be carried out in non-essential positions. In this context, an essential position in the sequence is one wherein the side chain of the relevant amino acid is of significance for its biological effect.
  • In one embodiment, the invention provides peptides and peptide derivatives of the following general Formula I:

  • H2N-GHRPX1X2X3X4X5X6X7X8PX9X10X11PX12PPPX13X14X15X16GYR—X17  (I),
  • wherein:
    • X1-X16 denote one of the 20 genetically encoded amino acids,
    • X17 denotes OR1 with R1=hydrogen or (C1-C10)-alkyl, or
      • NR2R3, R2 and R3 being identical or different and denoting hydrogen, (C1-C10)-alkyl, or a residue-PEG5-60K, wherein the PEG-residue is linked to the N atom via a spacer (SEQ ID NO: 1), or
      • a residue NH—Y—Z-PEG5-60K, wherein Y denotes a chemical bond or a genetically coded amino acid from among the group of S, C, K or R, and Z denotes a spacer by way of which a polyethylene glycol (PEG)-residue may be linked (SEQ ID NO: 2), as well as the physiologically acceptable salts thereof,
    • or wherein
    • X15 or X16 denote an amino acid from the group of C or K, which is linked to a residue Z-PEG5-60K via the heteroatom in the side chain, and wherein
    • X17 denotes OR1, with R1=hydrogen or (C1-C10)-alkyl, or
      • NR2R3, R2 and R3 being identical or different and denoting hydrogen or (C1-C10)-alkyl (SEQ ID NO: 3), as well as the physiologically acceptable salts thereof.
  • In one embodiment, the invention provides peptides and peptide derivatives of the general Formula I, wherein:
    • X1, X9, X10, X14 denote L, I, S, M or A,
    • X2, X6, X7 denote E or D,
    • X3, X4, X5, X11 denote R or K
    • X8, X12 denote A, G, S, or L
    • X13 denotes I, L or V and wherein
    • X15, X16 and X17 have the same meaning as given above, namely,
    • X15-X16 denote one of the 20 genetically encoded amino acids,
    • X17 denotes OR1 with R1=hydrogen or (C1-C10)-alkyl, or
      • NR2R3, R2 and R3 being identical or different and denoting hydrogen, (C1-C10)-alkyl, or a residue-PEG5-60K, wherein the PEG-residue is linked to the N atom via a spacer (SEQ ID NO: 4), or
      • a residue NH—Y—Z-PEG5-60K, wherein Y denotes a chemical bond or a genetically coded amino acid from among the group of S, C, K or R, and Z denotes a spacer by way of which a polyethylene glycol (PEG)-residue may be linked (SEQ ID NO: 5), as well as the physiologically acceptable salts thereof,
    • or wherein
    • X15 or X16 denote an amino acid from the group of C or K, which is linked to a residue Z-PEG5-60K via the heteroatom in the side chain, and wherein
    • X17 denotes OR1, with R1=hydrogen or (C1-C10)-alkyl, or
      • NR2R3, R2 and R3 being identical or different and denoting hydrogen or (C1-C10)-alkyl (SEQ ID NO: 6),
        as well as the physiologically acceptable salts thereof.
  • In one embodiment, the invention provides peptides and peptide derivatives of Formula II,

  • H2N-GHRPLDKKREEAPSLRPAPPPISGGGYR—X17  (II),
  • wherein X17 has the same meaning as given above for Formula I, namely,
    • X17 denotes OR1 with R1=hydrogen or (C1-C10)-alkyl, or
      • NR2R3, R2 and R3 being identical or different and denoting hydrogen, (C1-C10)-alkyl, or a residue-PEG5-60K, wherein the PEG-residue is linked to the N atom via a spacer (SEQ ID NO: 7), or
      • a residue NH—Y—Z-PEG5-60K, wherein Y denotes a chemical bond or a genetically coded amino acid from among the group of S, C, K or R, and Z denotes a spacer by way of which a polyethylene glycol (PEG)-residue may be linked (SEQ ID NO: 8),
        as well as the physiologically acceptable salts thereof.
  • In one embodiment, the present invention provides compounds of Formula II, wherein
    • X17 denotes NR2R3, R2 and R3 being identical or different and being hydrogen or (C1-C10)-alkyl, or a residue
      • C(NR2R3)—(S-succinimido)-(PEG5-40K), the succinimide residue being linked via C-atom 3 to the sulfur atom of the cysteine residue (SEQ ID NO: 9)
        as well as the physiologically acceptable salts thereof.
  • In one embodiment, the invention provides peptide derivatives of Formula III,

  • H2N-GHRPLDKKREEAPSLRPAPPPIS—X19—X20—X21—YR—X17  (III)
  • wherein two of the residues X19, X20 and X21 each are a glycine residue and the remaining one is a residue C—(S-succinimido)-(PEG5-40K), the succinimido residue being linked to the sulfur atom of the cysteine residue via C-atom 3,
    and wherein X17 denotes NR2R3, R2 and R3 being identical or different and being hydrogen or (C1-C10)-alkyl (SEQ ID NO: 10),
    as well as the physiologically acceptable salts thereof.
  • In one embodiment, the invention provides peptide derivatives of Formula III,

  • H2N-GHRPLDKKREEAPSLRPAPPPIS—X19—X20—X21—YR—X17  (III)
  • wherein two of the residues X19, X20 and X21 each are a glycine residue and the remaining one is a residue K-(PEG5-40K), the PEG-residue being linked via the nitrogen atom in the side chain of the lysine residue, and wherein
    X17 denotes NR2R3, R2 and R3 being identical or different and being hydrogen or (C1-C10)-alkyl (SEQ ID NO: 11), as well as the physiologically acceptable salts thereof.
  • In the above formulas I and II the following letters represent amino acid residues in accordance with the general annotation for proteins and peptides: phenylalanine is F, leucine is L, isoleucine is I, methionine is M, valine is V, serine is S, proline is P, threonine is T, alanine is A, tyrosine is Y, histidine is H, glutamine is Q, asparagine is N, lysine is K, aspartic acid is D, glutamic acid is E, cysteine is C, tryptophan is W, arginine is R, glycine is G.
  • The amino acid residues in the compounds of Formula I may either be present in their D or their L configuration.
  • The term peptide refers to a polymer of these amino acids, which are linked via an amide linkage.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 is a bar graph illustrating the number of neutrophils, specifically polymorphonuclear leukocytes (PMN), present in bronchioalveolar lavage liquid of mice having LPS-induced pneumonitis treated with either Control (100 μl NaCl) or 4.8 mg/kg of the agent of Example 1 (dissolved in 100 μl NaCl).
  • DETAILED DESCRIPTION OF THE INVENTION
  • “Physiologically acceptable” means that salts are formed with acids or bases the addition of which does not have undesirable effects when used for humans. Preferable are salts with acids or bases the use of which is listed for use with warm blooded animals, in particular humans, in the US Pharmacopoeia or any other generally recognized pharmacopoeia.
  • PEG stands for a polyethylene glycol residue having a molecular weight of between 5,000 and 60,000 Dalton, this molecular weight being the maximum of a molecular weight distribution, so that individual components of the mixture may have a higher or lower molecular weight.
  • The invention furthermore concerns processes for the production of the peptides and peptide derivatives of general Formula (I), characterized in that, either
      • (A) the first amino acid at the C-terminal end of the respective sequence is linked to a polymeric resin via a suitable cleavable spacer, the subsequent amino acids, optionally containing suitable protective groups for functional groups, are linked step by step according to methods known in the art, the finished peptide is cleaved off the polymeric resin according to suitable methods known in the art, the protective groups, if present, are cleaved off by suitable methods and the peptide or peptide derivative is purified according to suitable methods, or
      • (B) a PEG-group having a desired molecular weight is linked to a polymeric resin via a suitable spacer, the first amino acid at the N-terminal end of the peptide is linked using suitable methods, the remaining steps being the same as described in (A), or
      • (C) a lysine residue, containing a suitable protective group at the ε-amino group is linked to a suitable polymeric resin via a suitable spacer using suitable methods, the peptide chain is synthesized as described in (A), following cleavage from the polymeric resin and purification, if necessary, the protective group at the ε-amino group is cleaved off using suitable methods, a PEG group having a desired molecular weight is linked to the ε-amino group using a suitable activated reagent, the optionally remaining protective groups are cleaved off and the final product is purified using suitable methods, or
      • (D) a peptide containing a cysteine residue is reacted with a PEG-maleimide to form compounds of Formula (III).
  • Suitable processing steps following (A), (B) or (C) as well as suitable reagents are for instance described in document WO 2004/101600.
  • Embodiments of the respective processing steps are not new per se and will be clear to an experienced specialist in the field of organic synthesis.
  • Processes for linking a PEG-residue to a peptide chain will be known to the skilled artisan. For instance, a cysteine (C)-residue may be reacted with PEG-maleimide, resulting in a succinimide residue as spacer for residue Z. A further possibility is reacting an optionally activated C-terminal carboxy residue with an aminoalkyl-substituted PEG residue. A further possibility is the introduction of a PEG residue by reacting an aldehyde-substituted PEG residue with the ε-amino function of a lysine residue. Activated PEG reagents having suitable spacers and reactive groups may for instance be obtained from NOF Corporation (Tokyo, Japan).
  • The substances according to the invention and the use of the substances according to the invention for the production of a pharmaceutical drug are of particular significance for the production of a pharmaceutical drug for the therapy of diseases resulting from the tissue-damaging effect of white blood cells, or wherein the integrity and full physiological integrity of the layer of endothelial cells lining the blood vessels is impaired.
  • Diseases belonging to this group are those in context with autoimmunity, as for instance collagenases, rheumatic diseases, inflammatory bowel diseases like Morbus Crohn or Colitis ulcerosa, psoriasis and psoriatic rheumatoid arthritis, and post/parainfectious diseases as well as diseases caused by a graft-versus-host reaction. A healing effect takes place as this medical drug blocks the migration of the white blood cells into the tissue. Thus, the white blood cells remain in the blood stream and cannot cause an autoreactive effect harmful to the tissue. This effect of the inventive substances is furthermore important for the treatment of shock conditions, in particular in the case of septic shock triggered by infection with gram-positive or gram-negative bacterial pathogens as well as viral infections and haemorrhagic shock caused by heavy loss of blood because of severe injuries or bacterial or viral infections.
  • The inventive substances may generally be used in situations that can be described with the terms “Systemic Inflammatory Response Syndrome (SIRS)”, “Acute Respiratory Distress Syndrome (ARDS)” and organ- or multiorgan failure, respectively.
  • With a pharmaceutical drug for the therapy and/or prevention of rejection reactions of organ transplants there is a healing effect as this pharmaceutical drug prevents the migration of white blood cells from the blood stream into the donor organ, and the donor organ can therefore not be destroyed for instance by autoreactive lymphocytes.
  • With a pharmaceutical drug for the therapy and/or prevention of arteriosclerosis there is a healing and/or preventive effect as this pharmaceutical drug blocks the migration of lymphocytes and monocytes into the wall of the tissue and thus the activation of the cells of the tissue wall. Thus, the progress of arteriosclerosis is minimized or stopped, the progredience of arteriosclerotic plaque resulting therefrom is inhibited, causing the arteriosclerosis to recede.
  • With a pharmaceutical drug for the therapy and/or prevention of reperfusion trauma following surgically or pharmaceutically induced re-supply with blood, e.g., following percutaneous coronary intervention, stroke, vessel surgery, cardiac bypass surgery and organ transplants, there is a healing and/or preventive effect as this pharmaceutical drug inhibits the migration of lymphocytes, neutrophils and monocytes into the wall of the vessel. Reperfusion trauma is caused by a lack of oxygen/acidosis of the cells of the vessel during its re-supply with blood, leading to their activation and/or damage. Because of this, lymphocytes, neutrophils and monocytes adhere to the vessel wall and migrate into it. Blocking the adherence and migration of lymphocytes, neutrophils and monocytes in the vessel wall causes the hypoxy/acidosis-induced damage to abate, without the subsequent inflammatory reaction causing permanent damage to the vessel. The endothelium-stabilizing effect of the inventive compounds furthermore prevents the formation of oedemas as well as any further damage to the organs supplied via the respective blood vessels.
  • With a pharmaceutical drug for the therapy and/or prevention of arteriosclerosis as a consequence of metabolic diseases or the process of aging, there is a healing and/or preventive effect as this pharmaceutical drug inhibits the migration of lymphocytes, neutrophils and monocytes into the vessel wall, thus inhibiting the progredience of arteriosclerotic plaque resulting therefrom.
  • The pharmaceutical drug according to the invention may also be used for the transportation of another drug. The inventive drug specifically binds a surface molecule on endothelial cells. Thus, drugs linked thereto may be delivered to endothelial cells in high concentrations without any danger of them having side effects at other sites. An example that may be cited here is the use of substances inhibiting the division of cells, which, specifically brought to endothelial cells, may have an antiangiogenetic effect. This brings about a healing effect in tumor patients, as tumor growth is blocked by preventing the proliferation of endothelial cells and thus by preventing neoangiogenesis. The inventive compounds themselves may also develop an antiangiogenetic effect, as they, because of their endothelium-stabilizing effect, prevent the endothelial cells from changing into a proliferative phenotype and thus prevent the formation of new capillary blood vessels. Therefore, they are themselves suitable for the treatment of all kinds of tumor diseases as well as the prevention and/or treatment of tumor metastases.
  • The inventive compounds of Formula (I) together with pharmaceutical adjuvants and additives may be formulated into pharmaceutical preparations which also are a subject matter of the present invention. In order to prepare such formulations a therapeutically effective dose of the peptide or peptide derivative is mixed with pharmaceutically acceptable diluents, stabilizers, solubilizers, emulsifying aids, adjuvants or carriers and brought into a suitable therapeutic form. Such preparations, for instance, contain a dilution of various buffers (e.g., Tris-HCl, acetate, phosphate) of different pH and ionic strength, detergents and solubilizers (e.g., Tween 80, Polysorbate 80), antioxidants (e.g., ascorbic acid), and fillers (e.g., lactose, mannitol). These formulations may influence the biological availability and the metabolic behavior of the active agents.
  • The pharmaceutical preparations according to the invention may be administered orally, parenterally (intramuscularly, intraperitoneally, intravenously or subcutaneously), transdermally or in an erodable implant of a suitable biologically degradable polymer (e.g., polylactate or polyglycolate).
  • The biological effect and applicability for the claimed use of the inventive compounds may for instance be determined in an assay in which a culture of human umbilical cord endothelial cells is examined microscopically after stimulation with the “N-terminal disulfide knot protein II” (NDSK-II) or with thrombin. The stimulation of endothelial cells causes the formation of gaps between the cells in a densely packed cell layer. Treatment with the inventive compounds may prevent the formation of these gaps, and is successful in closing gaps that have already been formed. This effect is predicative for the protective effect on the endothelium the inventive compounds have throughout the organism. The inventive compounds have an effect in the range of concentrations from 0.01 nM to 1 mM, preferably in the range from 1 nM to 0.1 mM in the bath solution of cells.
  • The effectiveness in vivo may for instance be established using a model of acute pulmonitis in a rodent. For this the treatment of the animal and the administration of the substance are carried out as described in Example 7 below. The inventive compounds show an effect at a dose ranging from 0.001 mg/kg body weight to 500 mg/kg body weight, preferably at a dose ranging from 0.1 mg/kg to 50 mg/kg.
  • A further possibility for establishing the biological effect in vivo is the reduction or complete suppression of mortality because of an infection with haemolytic viruses or bacteria. For this purpose, mice are, as described in Example 8, for instance infected with a dose of Dengue viruses, wherein 50% of the animals die within a period of 5-20 days after infection. The inventive compounds bring about a reduction of this mortality at a dose ranging from 0.001 to 500 mg/kg body weight, preferably at a dose ranging from 0.1 to 50 mg/g body weight.
  • The following examples serve to illustrate the invention without limiting it to the examples.
  • General Preparation and Purification of Peptides According to the Invention
  • The preparation and purification of the above peptide derivatives generally takes place by way of FMOC-strategy on acid-labile resin supports using a commercially available batch peptide synthesizer as also described in the literature (e.g., “solid phase peptide synthesis—A practical approach” by E. Atherton, R. C. Sheppard, Oxford University press 1989). N-alpha-FMOC-protected derivatives, the functional side-chains of which are protected by acid-sensitive protective groups, are used as amino acid components. Unless otherwise stated, purification is carried out by means of RP-chromatography using a water/acetonitrile gradient and 0.1% TFA as ion pair reagent.
  • Example 1
  • (SEQ ID NO: 12)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Gly-Gly-Tyr-Arg-NH2
  • 100 mg Tentagel-S-RAM (Rapp-Polymere) at a load of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • The FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 μl DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3×900 μl 30% piperidine in DMF and thorough mixing for 4 minutes.
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom fit of the reaction vessel.
  • The amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • When synthesis is completed the peptide resin is dried. The peptide amide is subsequently cleaved off by treatment with trifluoracetic acid/TIS/EDT/water (95:2:2:1 vol) for 2 hours at room temperature. By way of filtration, concentration of the solution and precipitation by the addition of ice-cold diethyl ether the crude product (75 mg) is obtained as a solid.
  • The peptide is purified by RP-HPLC on Kromasil RP-18 250-20, 10 μm in 0.1% TFA with a gradient of 5 on 60% acetonitrile in 40 minutes at a flow rate of 12 ml/min and evaluation of the eluate by means of a UV detector at 215 nm. The purity of the individual fractions is determined by analyt. RP-HPLC and mass spectrometry. Following combination of the purified fractions and lyophilisation, 48 mg of pure product are obtained Maldi-TOF, 3036.6 m/z (m.i.).
  • Example 2
  • (SEQ ID NO: 13)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)-OH
  • The monomeric peptide is synthesized as in Example 1, Tentagel (Rapp Polymere) being used as resin support here with FMOC-Cys(Trt) as the first amino acid.
  • After cleavage and purification of the peptide, reaction is carried out with a 2- to 8-fold molar excess of maleinimido-PEG20K. Following recovery, purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by way of analytical RP-HPLC and MALDI-MS.
  • Example 3
  • (SEQ ID NO: 14)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)-amide
  • 100 mg Tentagel-S-RAM (Rapp-Polymere) at a load of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • The FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 μl DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3×900 μl 30% piperidine in DMF and thorough mixing for 4 minutes.
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom frit of the reaction vessel.
  • The amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • After cleavage and purification of the peptide, reaction is carried out with a 2- to 8-fold molar excess of maleinimido-PEG20K. Following recovery, purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by way of analytical RP-HPLC and MALDI-MS.
  • Example 4
  • (SEQ ID NO: 15)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Cys-(S-succinimido-PEG20K)-Gly-Tyr-Arg-amide
  • 100 mg Tentagel-S-RAM (Rapp-Polymere) having a charge of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • The FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 μl DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3×900 μl 30% piperidine in DMF and thorough mixing for 4 minutes.
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom fit of the reaction vessel.
  • The amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • When synthesis is completed, the peptide resin is dried. The peptide amide is subsequently cleaved by treatment with trifluoracetic acid/TIS/EDT/water (95:2:2:1 vol) for 2 hours at room temperature. By way of filtration, concentration of the solution and precipitation by the addition of ice-cold diethyl ether, the crude product (75 mg) is obtained as a solid.
  • The peptide is purified by RP-HPLC on Kromasil RP-18 250-20. The peptide thus obtained is reacted with maleinimido-PEG20k. Following recovery, purification by means of gel chromatography and lyophilisation, a pure product is obtained, the identity of which is confirmed by way of RP-HPLC and MALDI-MS.
  • Example 5
  • Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Cys-(S-succinimido-PEG20K)-Gly-Gly-Tyr-Arg-amide (SEQ ID NO: 16) is obtained as in Example 4, the sequence of protected amino acids being appropriately altered.
  • Example 6
  • Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-Gly-Cys-(S-succinimido-PEG20K)-Tyr-Arg-amide (SEQ ID NO: 17) is obtained as in Example 4, the sequence of protected amino acids being appropriately altered.
  • The following were prepared as in Example 1:
  • (SEQ ID NO: 18)
    Gly-His-Arg-Pro-Ile-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 19)
    Gly-His-Arg-Pro-Ala-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 20)
    Gly-His-Arg-Pro-Leu-Asp-Arg-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 21)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Arg-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 22)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Asp-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 23)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Asp-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 24)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Lys-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 25)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Gly-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 26)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Leu-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 27)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Lys-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 28)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ala-Gly-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 29)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ala-Ala-Gly-Gly-Tyr-Arg-NH2
    (SEQ ID NO: 30)
    Gly-His-Arg-Pro-Ile-Asp-Lys-Arg-Arg-Glu-Glu-Ala-Pro-Ser-Ile-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ala-Gly-Gly-Gly-Tyr-Arg-NH2
  • The following were prepared as in Example 2:
  • (SEQ ID NO: 31)
    Gly-His-Arg-Pro-Ile-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)OH
    (SEQ ID NO: 32)
    Gly-His-Arg-Pro-Leu-Asp-Arg-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG10K)OH
    (SEQ ID NO: 33)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Asp-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)OH
    (SEQ ID NO: 34)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)OH
    (SEQ ID NO: 35)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ala-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)OH
    (SEQ ID NO: 36)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG10K)OH
    (SEQ ID NO: 37)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)OH
    (SEQ ID NO: 38)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Ile-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)OH
  • The following were prepared as in Example 3:
  • (SEQ ID NO: 39)
    Gly-His-Arg-Pro-Ile-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)amide
    (SEQ ID NO: 40)
    Gly-His-Arg-Pro-Leu-Asp-Arg-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG10K)amide
    (SEQ ID NO: 41)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Asp-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)amide
    (SEQ ID NO: 42)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)amide
    (SEQ ID NO: 43)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ala-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)amide
    (SEQ ID NO: 44)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG10K)amide
    (SEQ ID NO: 45)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)amide
    (SEQ ID NO: 46)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Ile-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S-succinimido-PEG20K)amide
  • Example 7
  • The biological effect of the compound of Example 1 was established in a model of LPS-induced pneumonitis. C57 Black mice were randomized in two groups of 6 animals each and treated as follows:
  • Group 1 received 100 ng/kg LPS intranasally, immediately after the administration of LPS the mice received 4.8 mg/kg of the agent of Example 1 (dissolved in 100 μl NaCl) i.p., a second dose followed 60 min after the administration of LPS.
  • Group 2 received 100 ng/kg LPS intranasally, immediately after the administration of LPS the mice received 100 μl NaCl i.p., 60 min after the LPS administration the mice again received 100 μl NaCl i.p. Furthermore, 6 hours after the application of LPS, all groups were submitted to a bronchioalveolar lavage and the lungs were removed. From the lavage liquids the number of neutrophils (PMN) was determined.
  • Example 8
  • The biological effect of the compound of Example 3 was established in a model of Dengue virus infection in mice. 5-week-old male BALB/c mice were divided into 2 groups. All animals were infected subcutaneously with a mouse-adapted Dengue virus (DEN-2, strain P23085 at a dose of 1-2 LD50. 15 mice received 0.1 ml of 0.8% saline as intramuscular injection (control). The treated animals received 4.8 mg/kg/day of the agent of Example 3 as an intramuscular injection (diluted in 0.1 ml of 0.8% saline) once a day for 5 days, starting on day 3 after the virus infection.
  • At the end of the treatment period (day 10) the survival rates were compared.
  • The following results were obtained:
  • Mortality Percentage
    Control 8/15 47%
    Example 3 0/10  0%
    p < 0.05
  • Example 9
  • (SEQ ID NO: 47)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)-OH
  • The synthesis of the monomeric peptide is carried analogically to Example 1, Tentagel of (Rapp Polymere) being used as resin support here with FMOC-Cys(Trt) as the first amino acid.
  • Following cleavage and purification of the peptide, reaction is carried out with a suitable excess of Br—CH2—CO—NH-PEG20K. Following recovery, purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by MALDI-MS.
  • Example 10
  • (SEQ ID NO: 48)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)-amide
  • 100 mg Tentagel-S-RAM (Rapp-Polymere) at a load of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • The FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 μl DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3×900 μl 30% piperidine in DMF and thorough mixing for 4 minutes.
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom frit of the reaction vessel.
  • The amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • Following cleavage and purification of the peptide, reaction is carried out with a suitable excess of Br—CH2—CO—NH-PEG20K. Following recovery, purification is carried out on Kromasil RP-18, and the identity of the product is confirmed by MALDI-MS.
  • Example 11
  • (SEQ ID NO: 49)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-
    Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-
    Cys-(S—CH2—CO—NH-PEG20K)-Gly-Tyr-Arg-amide
  • 100 mg Tentagel-S-RAM (Rapp-Polymere) at a charge of 0.24 mmol/g are transferred to a commercially available peptide synthesis device (PSMM(Shimadzu)), wherein the peptide sequence is constructed step-by-step according to the carbodiimide/HOBt method.
  • The FMOC-amino acid derivatives are pre-activated by adding a 5-fold equimolar excess of di-isopropy-carbodiimide (DIC), di-isopropy-ethylamine (DIPEA) and hydroxybenzotriazole (HOBt) and, following their transfer into the reaction vessel, mixed with the resin support for 30 minutes. Washing steps are carried out by 5 additions of 900 μl DMF and thorough mixing for 1 minute. Cleavage steps are carried out by the addition of 3×900 μl 30% piperidine in DMF and thorough mixing for 4 minutes.
  • Removal of the individual reaction and wash solutions is effected by forcing the solutions through the bottom frit of the reaction vessel.
  • The amino acid derivatives FMOC-Ala, FMOC-Arg(Pbf), FMOC-Asp, FMOC-Gly, FMOC-His(Trt), FMOC-Ile, FMOC-Leu, FMOC-Lys(BOC), FMOC-Pro, FMOC-Ser(tBu), FMOC-Cys(Trt) and FMOC-Tyr(tBu) (Orpegen) are employed.
  • When synthesis is completed the peptide resin is dried. The peptide amide is subsequently cleaved by treatment with trifluoracetic acid/TIS/EDT/water (95:2:2:1 vol) for 2 hours at room temperature. By way of filtration, concentration of the solution and precipitation by the addition of ice-cold diethyl ether, the crude product (75 mg) is obtained as a solid.
  • The peptide is purified by RP-HPLC on Kromasil RP-18 250-20. The peptide thus obtained is reacted with O-(iodoacetyl)-N-hydroxysuccinimide, followed by amino-ethyl-oxi-PEG20k. After recovery, purification by means of gel chromatography and lyophilisation, a pure product is obtained, the identity of which is confirmed by MALDI-MS.
  • Example 12
  • Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-Ile-Ser-Gly-Cys-(S—CH2—CO—NH-PEG20K)-Gly-Tyr-Arg-amide (SEQ ID NO: 50) is obtained as in Example 11, the sequence of protected amino acids being appropriately altered.
  • The following were produced as in Example 9:
  • (SEQ ID NO: 51)
    Gly-His-Arg-Pro-Ile-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)OH
    (SEQ ID NO: 52)
    Gly-His-Arg-Pro-Leu-Asp-Arg-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG10K)OH
    (SEQ ID NO: 53)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Asp-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)OH
    (SEQ ID NO: 54)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)OH
    (SEQ ID NO: 55)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ala-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)OH
    (SEQ ID NO: 56)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG10K)OH
    (SEQ ID NO: 57)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)OH
    (SEQ ID NO: 58)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Ile-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)OH
  • The following were produced as in Example 10:
  • (SEQ ID NO: 59)
    Gly-His-Arg-Pro-Ile-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)amide
    (SEQ ID NO: 60)
    Gly-His-Arg-Pro-Leu-Asp-Arg-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG10K)amide
    (SEQ ID NO: 61)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Asp-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)amide
    (SEQ ID NO: 62)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)amide
    (SEQ ID NO: 63)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ala-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ala-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)amide
    (SEQ ID NO: 64)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG10K)amide
    (SEQ ID NO: 65)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Leu-Arg-Pro-Ala-Pro-Pro-
    Pro-Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)amide
    (SEQ ID NO: 66)
    Gly-His-Arg-Pro-Leu-Asp-Lys-Lys-Arg-Glu-Glu-Ala-Pro-Ser-Ile-Arg-Pro-Ala-Pro-Pro-Pro-
    Ile-Ser-Gly-Gly-Gly-Tyr-Arg-Cys-(S—CH2—CO—NH-PEG20K)amide

Claims (19)

1-7. (canceled)
8. A peptide of Formula II:
(SEQ ID NO: 9) H2N-GHRPLDKKREEAPSLRPAPPPISGGGYR-X17
or a physiologically acceptable salt thereof, wherein X17 denotes NR2R3, R2 and R3 being identical or different and denoting hydrogen, (C1-C10)-alkyl, or a residue-PEG5-40K, wherein the PEG-residue is linked to the N atom via a spacer.
9. The peptide of claim 8, wherein X17 denotes NR2R3, R2 and R3 being identical and denoting hydrogen.
10. A pharmaceutical composition, comprising the peptide of claim 8 and a pharmaceutically acceptable additive.
11. A peptide of Formula II:
(SEQ ID NO: 67) H2N-GHRPLDKKREEAPSLRPAPPPISGGGYR-X17
or a physiologically acceptable salt thereof, wherein X17 denotes a residue C(NR2R3)—(S-succinimido)-(PEG5-60K), R2 and R3 being identical and denoting hydrogen, and the succinimido residue being linked to the sulfur atom of the cysteine residue via C-atom 3, or a physiologically acceptable salts thereof.
12. The peptide of claim 12, wherein PEG is 20K.
13. A pharmaceutical composition, comprising the peptide of claim 11 and a pharmaceutically acceptable additive.
14. A method for preventing or treating Acute Respiratory Distress Syndrome in an individual comprising administering to the individual an effective amount of the peptide of claim 8.
15. A method for preventing or treating Acute Respiratory Distress Syndrome in an individual comprising administering to the individual an effective amount of the peptide of claim 11.
16. A method for preventing or treating Systemic Inflammatory Response Syndrome in an individual comprising administering to the individual an effective amount of the peptide of claim 8.
17. A method for preventing or treating Systemic Inflammatory Response Syndrome in an individual comprising administering to the individual an effective amount of the peptide of claim 11.
18. A method for preventing or treating shock in an individual comprising administering to the individual an effective amount of the peptide of claim 8.
19. A method for preventing or treating shock in an individual comprising administering to the individual an effective amount of the peptide of claim 11.
20. A method for preventing or treating reperfusion trauma in an individual comprising administering to the individual an effective amount of the peptide of claim 8.
21. A method for preventing or treating reperfusion trauma in an individual comprising administering to the individual an effective amount of the peptide of claim 11.
22. A method for preventing or treating Dengue virus in an individual comprising administering to the individual an effective amount of the peptide of claim 8.
23. A method for preventing or treating Dengue virus in an individual comprising administering to the individual an effective amount of the peptide of claim 11.
24. A method for preventing or treating organ failure in an individual comprising administering to the individual an effective amount of the peptide of claim 8.
25. A method for preventing or treating organ failure in an individual comprising administering to the individual an effective amount of the peptide of claim 11.
US12/820,804 2006-02-23 2010-06-22 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same Abandoned US20100279924A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/820,804 US20100279924A1 (en) 2006-02-23 2010-06-22 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
ATA301/2006 2006-02-23
AT3012006 2006-02-23
US11/678,535 US7799758B2 (en) 2006-02-23 2007-02-23 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same
US12/820,804 US20100279924A1 (en) 2006-02-23 2010-06-22 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/678,535 Continuation US7799758B2 (en) 2006-02-23 2007-02-23 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same

Publications (1)

Publication Number Publication Date
US20100279924A1 true US20100279924A1 (en) 2010-11-04

Family

ID=38042580

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/280,543 Abandoned US20110098442A1 (en) 2006-02-23 2007-02-23 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same
US11/678,535 Expired - Fee Related US7799758B2 (en) 2006-02-23 2007-02-23 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same
US12/820,804 Abandoned US20100279924A1 (en) 2006-02-23 2010-06-22 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US12/280,543 Abandoned US20110098442A1 (en) 2006-02-23 2007-02-23 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same
US11/678,535 Expired - Fee Related US7799758B2 (en) 2006-02-23 2007-02-23 Peptides and peptide derivatives as well as pharmaceutical compositions containing the same

Country Status (15)

Country Link
US (3) US20110098442A1 (en)
EP (2) EP2256131A1 (en)
JP (1) JP2009527502A (en)
KR (1) KR20090005300A (en)
CN (1) CN101389653A (en)
AT (1) ATE513853T1 (en)
AU (1) AU2007219031B2 (en)
CA (1) CA2641908A1 (en)
ES (1) ES2371038T3 (en)
IL (1) IL193435A0 (en)
MX (1) MX2008010930A (en)
NO (1) NO20084002L (en)
RU (1) RU2008137796A (en)
WO (1) WO2007095660A1 (en)
ZA (1) ZA200807028B (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2331958T3 (en) * 2004-06-25 2010-01-21 FIBREX MEDICAL RESEARCH &amp; DEVELOPMENT GMBH USE OF PEPTIDES DERIVED FROM CHAIN B BETA OF HUMAN FIBRINOGEN FOR THE TREATMENT OF SHOCK.
WO2009038729A2 (en) * 2007-09-17 2009-03-26 Regenerx Biopharmaceuticals, Inc. Compositions and methods utilizing fibrin beta chain fragments of the bbeta chain of fibrinogen
JP5410997B2 (en) * 2008-01-31 2014-02-05 則行 川村 Depression and depression markers and detection / diagnosis using them
US20090286740A1 (en) * 2008-05-15 2009-11-19 Fibrex Medical Research & Development Gmbh Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
US20090286725A1 (en) * 2008-05-15 2009-11-19 Fibrex Medical Research & Development Gmbh Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
US7884074B2 (en) * 2008-05-15 2011-02-08 Ikaria Development Subsidiary Two, LLC Compounds and methods for prevention and/or treatment of inflammation using the same
US8088890B2 (en) * 2008-09-26 2012-01-03 Fibrex Medical Research & Development Gmbh Peptides and peptidomimetic compounds, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
WO2010043444A2 (en) * 2008-10-15 2010-04-22 Fibrex Medical Research & Development Gmbh Pharmaceutical preparation for the treatment and/or prevention of ischemia/reperfusion injury and the sequels thereof
US20100152832A1 (en) * 2008-12-12 2010-06-17 Medtronic Vascular, Inc. Apparatus and Methods for Treatment of Aneurysms With Fibrin Derived Peptide B-Beta
US8841257B2 (en) * 2009-04-10 2014-09-23 Board Of Regents, The University Of Texas System Inhibitors of STAT3 and uses thereof
WO2019011879A1 (en) 2017-07-09 2019-01-17 Rainer Henning Therapeutic for treating capillary leak syndrome
JP7429032B2 (en) * 2019-12-24 2024-02-07 学校法人順天堂 Method for creating a non-human animal model of interstitial pneumonia
EP3912679A1 (en) 2020-05-19 2021-11-24 Johann Wolfgang Goethe-Universität Frankfurt am Main Bbeta-15-42 for the treatment of viral endothelitis

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4966848A (en) * 1988-02-08 1990-10-30 The General Hospital Corporation Isolation, purification, characterization, cloning and sequencing of N α-acetyltransferase
US5223421A (en) * 1989-10-25 1993-06-29 The General Hospital Corporation Identification of methionine Nα-acetyltransferase
US5837218A (en) * 1995-09-15 1998-11-17 Resolution Pharmaceuticals Inc. Non-receptor cell mediated imaging agents
US6086849A (en) * 1992-04-30 2000-07-11 Diatide, Inc. Technetium-99m labeled peptides for imaging
US6323311B1 (en) * 1999-09-22 2001-11-27 University Of Utah Research Foundation Synthesis of insulin derivatives
US20040192496A1 (en) * 2003-03-28 2004-09-30 Jurgen Legner Drive train for powering a mobile vehicle
US20070037749A1 (en) * 2000-12-12 2007-02-15 Peter Petzelbauer Therapeutic fibrin-derived peptides and uses thereof
US20080249006A1 (en) * 2004-06-25 2008-10-09 Fibrex Medical Research & Development Gesmbh Pharmaceutical Preparation for the Treatment of Shock
US20090005310A1 (en) * 2005-12-23 2009-01-01 Fibrex Medical Research & Development Gmbh (En) Pharmaceutical Composition for Treating Haemorrhagic Shock and its Consecutive Symptoms

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0575545B1 (en) * 1991-03-15 2003-05-21 Amgen Inc. Pegylation of polypeptides
DE602004028725D1 (en) 2003-05-12 2010-09-30 Affymax Inc NEW POLY (ETHYLENGLYCOL) MODIFIED ERYTHROPOIETINAGONISTS AND THEIR USES

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4966848A (en) * 1988-02-08 1990-10-30 The General Hospital Corporation Isolation, purification, characterization, cloning and sequencing of N α-acetyltransferase
US5223421A (en) * 1989-10-25 1993-06-29 The General Hospital Corporation Identification of methionine Nα-acetyltransferase
US6086849A (en) * 1992-04-30 2000-07-11 Diatide, Inc. Technetium-99m labeled peptides for imaging
US5837218A (en) * 1995-09-15 1998-11-17 Resolution Pharmaceuticals Inc. Non-receptor cell mediated imaging agents
US6323311B1 (en) * 1999-09-22 2001-11-27 University Of Utah Research Foundation Synthesis of insulin derivatives
US20070037749A1 (en) * 2000-12-12 2007-02-15 Peter Petzelbauer Therapeutic fibrin-derived peptides and uses thereof
US7271144B2 (en) * 2000-12-12 2007-09-18 Fibrex Medical Research & Development Gmbh Therapeutic fibrin-derived peptides and uses thereof
US20040192496A1 (en) * 2003-03-28 2004-09-30 Jurgen Legner Drive train for powering a mobile vehicle
US20080249006A1 (en) * 2004-06-25 2008-10-09 Fibrex Medical Research & Development Gesmbh Pharmaceutical Preparation for the Treatment of Shock
US20090005310A1 (en) * 2005-12-23 2009-01-01 Fibrex Medical Research & Development Gmbh (En) Pharmaceutical Composition for Treating Haemorrhagic Shock and its Consecutive Symptoms

Also Published As

Publication number Publication date
WO2007095660A1 (en) 2007-08-30
KR20090005300A (en) 2009-01-13
IL193435A0 (en) 2011-08-01
ES2371038T3 (en) 2011-12-26
RU2008137796A (en) 2010-03-27
ATE513853T1 (en) 2011-07-15
CN101389653A (en) 2009-03-18
ZA200807028B (en) 2009-11-25
EP2256131A1 (en) 2010-12-01
NO20084002L (en) 2008-11-18
US7799758B2 (en) 2010-09-21
JP2009527502A (en) 2009-07-30
EP1987062B1 (en) 2011-06-22
MX2008010930A (en) 2008-10-29
CA2641908A1 (en) 2007-08-30
AU2007219031A1 (en) 2007-08-30
US20080039388A1 (en) 2008-02-14
EP1987062A1 (en) 2008-11-05
AU2007219031B2 (en) 2011-11-03
US20110098442A1 (en) 2011-04-28

Similar Documents

Publication Publication Date Title
US7799758B2 (en) Peptides and peptide derivatives as well as pharmaceutical compositions containing the same
US8067533B2 (en) Peptides and peptide derivatives, the production thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
US7884074B2 (en) Compounds and methods for prevention and/or treatment of inflammation using the same
US8088890B2 (en) Peptides and peptidomimetic compounds, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
US20090286725A1 (en) Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
US20090286740A1 (en) Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition

Legal Events

Date Code Title Description
AS Assignment

Owner name: IKARIA DEVELOPMENT SUBSIDIARY TWO LLC, NEW JERSEY

Free format text: LICENSE;ASSIGNORS:FIBREX MEDICAL, INC.;FIBREX MEDICAL RESEARCH & DEVELOPMENT GESMBH;REEL/FRAME:024600/0420

Effective date: 20090717

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION