US20120129774A1 - Kinin b1 receptor peptide agonists and uses thereof - Google Patents

Kinin b1 receptor peptide agonists and uses thereof Download PDF

Info

Publication number
US20120129774A1
US20120129774A1 US13/294,371 US201113294371A US2012129774A1 US 20120129774 A1 US20120129774 A1 US 20120129774A1 US 201113294371 A US201113294371 A US 201113294371A US 2012129774 A1 US2012129774 A1 US 2012129774A1
Authority
US
United States
Prior art keywords
phe
carboxylic acid
pro
sard
arg
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
US13/294,371
Inventor
Fernand Gobeil
Witold A. Neugebauer
Domenico Regoli
David Fortin
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.)
SOCPRA Sciences Sante et Humaines sec
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US13/294,371 priority Critical patent/US20120129774A1/en
Assigned to UNIVERSITE DE SHERBROOKE reassignment UNIVERSITE DE SHERBROOKE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FORTIN, DAVID, GOBEIL, FERNAND, JR., NEUGEBAUER, WITOLD A., REGOLI, DOMENICO
Assigned to SOCIETE DE COMMERCIALISATION DES PRODUITS DE LA RECHERCHE APPLIQUEE - SOCPRA-SCIENCES SANTE ET HUMAINES S.E.C. reassignment SOCIETE DE COMMERCIALISATION DES PRODUITS DE LA RECHERCHE APPLIQUEE - SOCPRA-SCIENCES SANTE ET HUMAINES S.E.C. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITE DE SHERBROOKE
Publication of US20120129774A1 publication Critical patent/US20120129774A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/18Kallidins; Bradykinins; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/08Antibacterial agents for leprosy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives

Definitions

  • the present invention relates to peptides derivatives of desArg 9 -bradykinin (desArg 9 -BK) which act as agonists at kinin B 1 receptors and their use as therapeutic agents.
  • kallikrein-kinin system may play key position in blood pressure regulation and organ protection (Hagiwara et al., 2004, Hypertens Res 27(6), 399-408; Couture and Girolami, 2004, Eur J Pharmacol 500(1-3), 467-485).
  • Kinins are a group of bioactive peptides formed by numerous tissues and within the blood.
  • Kinins can be divided in two major subgroups of naturally occurring peptides namely bradykinin (BK), kallidin (LysBK), [Hyp 3 ]-BK and Lys[Hyp 3 ]-BK and their respective bioactive metabolites produced by carboxypeptidases M and N (alias kininases I), desArg 9 BK, LysdesArg 9 BK, [Hyp 3 ]-desArg 9 BK and Lys[Hyp 3 ]-desArg 9 BK.
  • B 2 and B 1 specific receptors referred to as B 2 and B 1 , respectively, which are ubiquitously expressed on cell membranes in the affected tissues.
  • Kinin B 2 and B 1 receptors are found in a variety of cells as endothelia, smooth muscles, epithelia, white blood cells. They mediate vascular smooth muscle relaxation via release of autacoids, particularly from the endothelium. This function provides the basic mechanism of peripheral vasodilatation which is responsible for a large part of their in vivo hypotensive effect (Duka et al., 2006, Am J Physiol Endocrinol Metab [Epub ahead of print]).
  • B 1 receptors are usually not found in physiological conditions but is induced by various stimuli (i.e. cytokines) in several cell types including vascular endothelial and smooth muscle cells, fibroblasts, neurons. Inducible B 1 receptors are involved in various types of vascular inflammation associated with diabetic, hypertensive conditions, and angiogenesis.
  • cytokines i.e. cytokines
  • Inducible B 1 receptors are involved in various types of vascular inflammation associated with diabetic, hypertensive conditions, and angiogenesis.
  • the beneficial and protecting roles of kinin B 1 receptors in cardiovascular-related physiopathology have been recently reviewed (Couture and Girolami, vide supra).
  • the induction of B 1 receptor and subsequent activation may be seen as self-defense mechanism elicited by the vasculature against recurrent deleterious ischemic/hypoxic episodes.
  • Compounds of the present invention may be pharmacologically exploited for post-ischemic healing (Couture and Girolami, vide supra; Emanueli et al, vide supra; Hagiwara et at vide supra; Duguay et at, 2004, Brit J Pharmacol 141(4), 728-736; Emanueli and Madeddu, 2001, Trends Pharmacol. Sci. 22(9), 478-484; Parenti et al., 2001, FASEB J 15(8), 1487-1489).
  • the present invention relates to new biologically active peptides derivatives of the general formula (1) which act as agonists at the kinin B 1 receptors:
  • X is OH or NH 2
  • aa y is Sar, acetyl or other acyl group
  • aa x is Arg, Lys, Orn (L- or D-configuration) or another basic amino acid of the L- or D-configuration, L- or D-Cit or not substituted at this position;
  • aa 0 is Arg, Lys, Orn (L or D-configuration) or another basic amino acid of the L- or D-configuration, L- or D-Cit or not substituted at this position;
  • aa 1 is Arg or D-Arg
  • aa 2 is Pro, Oic, or another Pro-mimic amino acid, Hyp, ⁇ -(Me)Pro or another Pro-mimic amino acid derivative;
  • aa 4 is Gly, ethyl amine, or Aib;
  • aa 5 is Phe, Acc, Cha, Chg, Cpg, Igl, Pen, 4Bip, Phg or Thi;
  • D-Phe 8 is D-Phe or ⁇ -(Me)Phe.
  • residues -aa 2 -aa 3 -aa 4 - may be replaced by an aliphatic ⁇ -amino carboxyl (8 carbons chain length) linkers
  • residues -aa 2 -aa 3 -aa 4 -Phe 5 - may be replaced by an aliphatic ⁇ -amino carboxyl (11 carbons chain length) linkers.
  • the present invention relates to peptide derivatives acting as selective agonists toward B 1 receptor, that at least
  • Preferred B 1 receptor peptide agonists of the present invention may be illustrated by the following Tables 2 and 3.
  • Therapeutic applications of desArg 9 BK-related B 1 receptor agonists are based upon recognized beneficial actions mediated by kinin B 1 receptors such as vasodilatation/hypotension, neo-vascularization, angiogenesis, anti-ischemia, increase of vascular permeability specifically in surrounding brain tumors, and include treatment of pathological conditions where amount of exogenous B 1 receptor agonists is needed. These conditions may include treatment of life-threatening diseases specifically hypertension and diabetes associated with vasculopathies.
  • Compounds of the present invention may be used to prevent and/or reverse end-organ failure, owing to lack of tissue perfusion, in hypertensive and diabetic patients.
  • Such considerations may be extended to other diseases including cerebral and coronary artery diseases, to reduce incidence of stroke and myocardial infarction, respectively, and peripheral arterial disease to improve mobility of afflicted individuals.
  • Another therapeutic application of the present invention covers atherosclerotic coronary artery diseases where balloon angioplasty is to be performed to restore blood flow to blood-deprived heart tissue. Restenosis (repeat narrowing or blockage) of injured heart coronary vessels is frequently observed, usually one-third of the time, following angioplasty procedure.
  • Kinin B 1 receptors stimulants owing to their angiogenic properties, may assist to lower the rate of recurrence or restenosis.
  • Compounds of the present invention may be administered topically, subcutaneously, or by injection or infusion or delivered using suited biodegradable microsphere-based carrier systems or as an oral suspension in an appropriate vehicle or as tablets, pills, capsules, caplets or the like.
  • the dosage regimen and manner of administration will be defined by the application of the B 1 receptor peptide agonists and as per standard clinical testing to find the optimal dose.
  • SPPS solid phase peptide synthesis
  • DCC coupling reaction A 4-fold excess of Boc-amino acids over resin substitution rate is used in the Applied Bioscience 430A synthesizer. Boc-amino acids are activated for coupling with an equimolar amount of DCC and 2 equivalents of DIEA.
  • the solvent may be DCM, DMF, or NMP.
  • the resin is washed with the same solvent before and after coupling. Completeness of coupling is determined with a Kaiser test.
  • the deprotection reagent is 40% TFA in DCM, containing 1 mg/mL N-acetyl-LD-tryptophan. It is used for 30 min, following a prewash.
  • the neutralization reagent is 20% DIEA in DCM.
  • N-Terminal acylation A 5-fold excess of acyl chlorides and 10-fold excess of DIEA over peptide-resin are used in DCM for 30 min. The resin is washed with the same solvent after completion of the reaction.
  • Procedure D HF cleavage: A batch of 0.5 mmole of peptide-resin is mixed with 1.0 mL anisole and chilled in the reaction vessel (resistant to HF) to ⁇ 78° C., and 10 ml of anhydrous HF is distilled into the vessel under vacuum. The mixture is stirred at 0° C. for 1 h, and the HF is evaporated first under a nitrogen flow, then under vacuum. The peptide and resin mixture is washed three times with dry ether, and the peptide is extracted into 50% acetic acid. The peptide solution is concentrated under vacuum, diluted in water, and lyophilized.
  • Procedure E Purification: Preparative medium pressure chromatography may be carried out on a reversed phase C18 silica column in a gradient of 0.1% TFA in water to 0.05% TFA in acetonitrile. Eluted peptide is detected by UV at 254 nm. Analytical HPLC may be carried out in the same system to identified pure fractions.
  • the approach is an alternative approach of peptides synthesis and standard with Mitsunobu reaction ⁇ -amino acids residues. Synthesis may be carried out by use of PioneerTM continuous flow peptide synthesis system.
  • Procedure G The resin is placed in the column and a 2 to 4-fold excess of Fmoc-protected amino acids over resin substitution rate is placed in the sampler tray. Synthesis is performed using amine free DMF. All solutions needed for the solid phase continuous flow synthesis are prepared and loaded in the synthesizer. The synthesis protocol is prepared, loaded into the synthesizer, and run in normal or extended cycle mode. Fmoc deprotection is performed in 20% piperidine in DMF and monitored through UV detector at 364 nm. Fmoc-protected amino acids are activated for coupling with an equimolar amount of HATU or TBTU, and 2 equivalents of DIEA.
  • Procedure H N-terminal caping (acetylation): This step is optional and can be included in the synthesis protocol.
  • the acetylation reagents are 5% acetic anhydride and 6% 2,4-lutidine in DMF.
  • the resin is washed with the same solvent and isopropanol after completion of the reaction.
  • the resin is removed from the column synthesizer and dried under vacuum 12 hours.
  • TFA cleavage The cleavage solution, TFA: water: TIPS (95%:2.5%:2.5%), is mixed with peptide-resin, and stirred at room temperature for 2 h. The resin is filtrated and the peptide is precipitated in dry ether. The suspension is centrifuged. The ether solution is decanted and the precipitated peptide is dissolved in water and lyophilized. The peptide is purified and characterized as described in procedures E and F.
  • the peptide chain is assembled by Fmoc strategy.
  • O-NBS group is introduced after Fmoc deprotection at the N-terminal position as described in procedure J followed by 0.1 mmol of o-NBS-aa n -resin (0.2-0.6 mmol/g) is suspended in 1 mL of DME, and 1 mmol of appropriate alcohol is added to the suspension. Mistunobu reaction and deprotection of the o-NBS group are performed as described in procedure J.
  • the resin is treated with 10 equivalent of 1 M solution of mercaptoethanol/DBU in DMF for 1 h, and washed thoroughly with DMF and DCM.
  • the peptide is then cleaved with an appropriate TFA cocktail, see procedure I.
  • the peptide is purified and characterized as described in procedures E and F.
  • the rbA and the hUV without endothelium (which contains B 1 receptors) were used to determine the agonistic activities of each compound expressed in term of pEC 50 values (-log of the molar concentration of agonist required to produce 50% of the maximal response).
  • the rabbit jugular vein (which contains only B 2 receptor) was used to exclude any action of the new compounds at B 2 receptors and thus establish their selectivity.
  • Mean arterial blood pressure was continuously monitored by inserting a polyethylene catheter (filled with heparinized saline solution) into the right carotid artery attached to a transducer (model TDX-300; Micro-Med Inc., KY, USA) connected to a blood pressure analyzer (model BPA-400a; Micro-Med Inc., KY, USA).
  • a second arterectomy was performed on the left carotid artery for the administration of graded doses of compounds into the aorta.
  • the anesthetic sodium pentobarbital (30 mg/kg intravenously (i.v.) through the auricular vein) was used instead of the ketamine/xylasine solution.
  • LPS lipopolysaccharide
  • Enzymatic resistance of peptide compounds were measured in harsh conditions using rabbit lung and kidney extracts prepared as reported (Tramontana et al., 2001, J Pharmacol Exp Ther 296(3), 1051-1057) with minor modifications. Briefly, animals were euthanized, organs harvested and cleaned from connective and adipose tissues. Tissues were then homogenized with a Polytron homogenizer in 5 volumes (w/v) of cold buffer consisting of Tris HCl 50 mM pH 7.5, NaCl 300 mM and ZnCl 2 10 ⁇ M, and centrifuged at 1500 g for 15 min at 4° C. Concentration of proteins from the resulting supernatant was determined using Bradford method with bovine serum as standard.
  • Peptide compounds 150 ⁇ M were incubated at 37° C. in the presence of tissue enzymatic extracts (200 ⁇ g proteins) for different times (0, 15, 60 min) in a total reaction medium of 250 ⁇ l.
  • tissue enzymatic extracts 200 ⁇ g proteins
  • the naturally-occurring B 1 receptor agonist LysdesArg 9 -BK served as reference peptide. Hydrolysis was stopped by immersing samples in boiling water.
  • the delivery potency of peptide compounds was studied in syngeneic F98-Fischer glioma rat model based on previous reports (Barth, 1998, J Neurooncol 36, 91-102; Mathieu et al., 2005, The J Appl Res 5(1), 17-25). Briefly, the F98 cell line was cultured in monolayer and stereotactically implanted (1 ⁇ 10 4 cells in a volume of 5 ⁇ l; 1 ⁇ l/min) in the right frontal lobe of Fischer rats using standardized and validated coordinates. In this model, the tumor take has been shown to be 100%. Moreover, this model has been shown to adequately emulate the situation of primary malignant brain tumors in the human (Mathieu at al., vide supra).
  • rats were anesthetized with ketamine/xylazine solution.
  • Blood brain barrier disruption was performed in rats by administering specific B 1 interacting peptides or vehicle (isotonic saline) into the internal carotid artery in a retrograde fashion from the external carotid after catheter implantation.
  • the infusion rates of peptide or vehicle were set at 10 nmol/kg/min for 5 min followed by a 20 min resting period.

Abstract

The present invention provides for novel kinin B1 receptors peptide agonists of formula (1) having very good to excellent affinities and selectivity for the B1 receptor, in vitro and in vivo increased resistance to enzymatic degradation, superior pharmacokinetic properties to those of naturally occurring agents, capacity to significantly enhance delivery of chemotherapeutic substances across the blood brain barrier and within peripheral tissues for the treatment of tumors, capacity to protect and restore kidney, heart, brain and other organ functions, when given alone or in combination with other therapies in the treatment of hypertension, diabetes and other cardiovascular diseases particularly, but not limited to, atherosclerosis and arteriosclerosis.
aay-aax-aa0-aa1-aa2-aa3-aa4-aa5-Ser6-Pro7-D-Phe8-X (1)

Description

    BACKGROUND OF THE INVENTION
  • (a) Field of the Invention
  • The present invention relates to peptides derivatives of desArg9-bradykinin (desArg9-BK) which act as agonists at kinin B1 receptors and their use as therapeutic agents.
  • (b) Description of Prior Art
  • Emerging findings suggest that the kallikrein-kinin system may play key position in blood pressure regulation and organ protection (Hagiwara et al., 2004, Hypertens Res 27(6), 399-408; Couture and Girolami, 2004, Eur J Pharmacol 500(1-3), 467-485). Kinins are a group of bioactive peptides formed by numerous tissues and within the blood. Kinins can be divided in two major subgroups of naturally occurring peptides namely bradykinin (BK), kallidin (LysBK), [Hyp3]-BK and Lys[Hyp3]-BK and their respective bioactive metabolites produced by carboxypeptidases M and N (alias kininases I), desArg9BK, LysdesArg9BK, [Hyp3]-desArg9BK and Lys[Hyp3]-desArg9BK. Actions of bradykinin and congeners and desArg9-related peptides are relayed via specific receptors referred to as B2 and B1, respectively, which are ubiquitously expressed on cell membranes in the affected tissues. Kinin B2 and B1 receptors are found in a variety of cells as endothelia, smooth muscles, epithelia, white blood cells. They mediate vascular smooth muscle relaxation via release of autacoids, particularly from the endothelium. This function provides the basic mechanism of peripheral vasodilatation which is responsible for a large part of their in vivo hypotensive effect (Duka et al., 2006, Am J Physiol Endocrinol Metab [Epub ahead of print]).
  • Contrary to B2 receptors which are constitutively expressed, the B1 receptors are usually not found in physiological conditions but is induced by various stimuli (i.e. cytokines) in several cell types including vascular endothelial and smooth muscle cells, fibroblasts, neurons. Inducible B1 receptors are involved in various types of vascular inflammation associated with diabetic, hypertensive conditions, and angiogenesis. The beneficial and protecting roles of kinin B1 receptors in cardiovascular-related physiopathology have been recently reviewed (Couture and Girolami, vide supra). The induction of B1 receptor and subsequent activation may be seen as self-defense mechanism elicited by the vasculature against recurrent deleterious ischemic/hypoxic episodes. This view is supported by several lines of evidence. Exogenous perfusion of naturally-occurring B1 receptor agonist desArg9BK prior ischemic conditions, in a Langerdorff setup, decreased arrhythmias (Chahine et al., 1993, Brit J Pharmacol 108, 318-322) and protected endothelial functions in coronary arteries in the follow-up reperfusion process (Bouchard et al., 1998, Brit J Pharmacol 123, 413-420). In addition, systemic supplementation of a stable desArg9BK-related analogue caused a reparative angiogenesis in a murine of limb ischemic (Emanueli et al., 2002, Circulation 105, 360-366). The involvement of newly expressed B1 receptors in reparative angiogenesis of wounded arteries was equally seen in an animal model of balloon angioplasty (Pruneau et al., 1994, Brit J Pharmacol 111, 1029-1034; Agata et al., 2000, Hypertension 36,364-370). Also, the important role of B1 receptors in preservation of cardiac function after myocardial infarction has recently been demonstrated using kinin B1 receptor gene knockout mice (Xu et al., 2005, Hypertension 45, 747-753)
  • These salutary actions, within macro- and micro-vascular networks, lies in part on capacity of kinins including desArg9-BK derivatives to promote the release of nitric oxide and prostaglandins, which may serve as cytoprotective, angiogenic and dilatory factors thereby preserving functions and oxygenation of vital organs (Kichuk et al., 1996, Circulation 94, 44-51; Emanueli et al. vide supra; Sharma and Thani, 2004, IDrugs 7, 926-934). However, naturally-occurring B1 receptor agonists LysdesArg9BK and desArg9BK as many other endogenous peptide hormones, are subjected to rapid proteolysis by tissue and plasma enzymes, which limit their therapeutic potential (Rhaleb et al., 1990, Brit Pharmacol 99, 445-448; Drapeau et al., 1991, J Pharmacol Exp Ther 259, 997-1003). There is therefore a need for metabolically stable and potent compounds with long duration of action capable of selectively activating B1 receptors which interestingly, are not prone to desensitization after exposure with their cognate ligands. Compounds of the present invention may be pharmacologically exploited for post-ischemic healing (Couture and Girolami, vide supra; Emanueli et al, vide supra; Hagiwara et at vide supra; Duguay et at, 2004, Brit J Pharmacol 141(4), 728-736; Emanueli and Madeddu, 2001, Trends Pharmacol. Sci. 22(9), 478-484; Parenti et al., 2001, FASEB J 15(8), 1487-1489).
  • SUMMARY OF THE INVENTION
  • The present invention relates to new biologically active peptides derivatives of the general formula (1) which act as agonists at the kinin B1 receptors:
  • aay-aax-aa0-aa1-aa2-aa3-aa4-aa5-Ser6-Pro7-D-Phe8-X (1)
  • wherein:
  • X is OH or NH2
  • aay is Sar, acetyl or other acyl group;
  • aax is Arg, Lys, Orn (L- or D-configuration) or another basic amino acid of the L- or D-configuration, L- or D-Cit or not substituted at this position;
  • aa0 is Arg, Lys, Orn (L or D-configuration) or another basic amino acid of the L- or D-configuration, L- or D-Cit or not substituted at this position;
  • aa1 is Arg or D-Arg;
  • aa2 is Pro, Oic, or another Pro-mimic amino acid, Hyp, α-(Me)Pro or another Pro-mimic amino acid derivative;
  • aa3 is Pro, or another Pro-mimic amino acid, Hyp, Oic, α-(Me)Pro or another Pro-mimic amino acid derivative;
  • aa4 is Gly, ethyl amine, or Aib;
  • aa5 is Phe, Acc, Cha, Chg, Cpg, Igl, Pen, 4Bip, Phg or Thi; and
  • D-Phe8 is D-Phe or α-(Me)Phe.
  • According to the above formula, residues -aa2-aa3-aa4- may be replaced by an aliphatic ω-amino carboxyl (8 carbons chain length) linkers, and residues -aa2-aa3-aa4-Phe5- may be replaced by an aliphatic ω-amino carboxyl (11 carbons chain length) linkers.
  • Thus the present invention relates to peptide derivatives acting as selective agonists toward B1 receptor, that at least
  • 1) have good to high affinity and selectivity for the B1 receptor, and represent long lasting B1 receptor agonists.
  • 2) are more resistant to in vitro and in vivo enzymatic degradation.
  • 3) have pharmacokinetic properties superior to those of naturally occurring agents.
  • All references referred herein are hereby incorporated by reference.
  • For the purpose of the present invention the following abbreviations are defined in Table 1 below.
  • As used herein, abbreviations of natural α-amino acids are those accepted in the art (IUPAC-IUB Commission on Biochemical Nomenclature: Symbols for amino acids derivatives and peptides 1972. Biochem J 126, 773-780), and unless prefix with D are all L-configuration.
  • TABLE 1
    Ac Acetyl
    acyl CnH2n+1—CO—, where: n = 2-15
    Aib α-aminoisobutyric acid
    Acc 1-amino-1-cyclopentane carboxylic acid
    Boc tert-butyloxy carbonyl
    Cit Citrulline
    Cha β-cyclohexyl-alanine
    Chg α-cyclohexyl-glycine
    Cpg α-cyclopentyl-glycine
    4Bip 4-Phenyl-phenylalanine
    DBU diazabicyclo[5.4.0] undec-7-ene
    DCC Dicyclohexylcarbodiimide
    DCM dichloromethane
    DIAD diisopropyl azodicarbonate
    DIEA N,N-diisopropylethyl amine
    DME 1,2-dimethoxyethane
    DMF N,N-dimethylformamide
    Fmoc 9-fluorenylmethoxycarbonyl
    HATU O-(7-Azabenzotriazol-1-yl)-1,1,3,3-tetramethyluronium
    hexafluorophosphate
    Hyp trans-4-hydroxy-Pro
    Igl α-indanylglycine
    Me Methyl
    α-(Me)Phe α-methyl-phenylalanine
    α-(Me)Pro α-methyl-proline
    o-NBS ortho-nitrobenzenesulfonyl
    NMO N-methylmorpholine oxide
    Pen Penicillamine
    Phg Phenylglycine
    Pro-mimic 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-
    proline; azetidine-2-carboxylic acid; trans-4-cyano-
    proline; cis-4-cyano-proline; 2-ethylthiazolidine-4-
    carboxylic acid; thiazolidine-2-carboxylic acid;
    2-methylthiazolidine-4-carboxylic acid;
    3-phenylpyrrolidine-2-carboxylic acid
    Oic (2S,3aS,7aS)-1-octahydro-1H-indole-2-carboxylic acid
    Orn Ornithine
    Sar Sarcosine
    TBTU O-(benzotriazol-1-yl)-1,1,3,3-tetramethyluronium
    tetrafluoroborate
    Thi β-(2-thienyl)-alanine
    TIPS triisopropyl silane
    TFA trifluoroacetic acid
    TPP Triphenylphosphine
  • DETAILED DESCRIPTION OF THE INVENTION
  • Preferred B1 receptor peptide agonists of the present invention may be illustrated by the following Tables 2 and 3.
  • TABLE 2
     1 aay aax aa0 Arg1 Aa2 aa3 aa4 aa5 Ser6 Pro7 D-Phe8
     2 Ac Arg Pro Pro Gly Phe Ser Pro D-Phe
     3 acyl Arg Pro Pro Gly Phe Ser Pro D-Phe
     4 Sar Arg (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
     5 Ac Arg (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
     6 acyl Arg (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
     7 Sar Arg (L or D) Arg Pro Hyp Gly Phe Ser Pro D-Phe
     8 Ac Arg (L or D) Arg Pro Hyp Gly Phe Ser Pro D-Phe
     9 acyl Arg (L or D) Arg Pro Hyp Gly Phe Ser Pro D-Phe
    10 Sar Lys (L or D) Arg Pro Hyp Gly Phe Ser Pro D-Phe
    11 Ac Lys (L or D) Arg Pro Hyp Gly Phe Ser Pro D-Phe
    12 acyl Lys (L or D) Arg Pro Hyp Gly Phe Ser Pro D-Phe
    13 Sar Orn (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
    14a acyl Arg (L or D) Arg (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
    14b Sar Arg (L or D) Arg (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
    15a Ac Lys (L or D) Lys (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
    15b Sar Lys (L or D) Lys (L or D) Arg Pro Pro Gly Phe Ser Pro D-Phe
    16 Sar Lys or Arg Arg Pro Pro or Hyp Gly Acc Ser Pro D-Phe
    17 Sar Lys or Arg Arg Pro Pro or Hyp Gly 4Bip Ser Pro D-Phe
    18 Sar Lys or Arg Arg Pro Pro or Hyp Gly Cha Ser Pro D-Phe
    19 Sar Lys or Arg Arg Pro Pro or Hyp Gly Pen Ser Pro D-Phe
    20 Sar Lys or Arg Arg Pro Pro or Hyp Gly Thi Ser Pro D-Phe
    21 Sar Lys or Arg Arg Pro Pro or Hyp Gly Cpg Ser Pro D-Phe
    22 Sar Lys or Arg Arg Pro Pro or Hyp Gly Chg Ser Pro D-Phe
    23 Sar Lys or Arg Arg Pro Pro or Hyp Gly Phg Ser Pro D-Phe
    24 Sar Lys or Arg Arg NH—(CH2)7—CO Phe Ser Pro D-Phe
    25 Ac Lys or Arg Arg NH—(CH2)7—CO Phe Ser Pro D-Phe
    26a Sar Orn Arg NH—(CH2)7—CO Phe Ser Pro D-Phe
    26b Ac Orn Arg NH—(CH2)7—CO Phe Ser Pro D-Phe
    27 Sar Lys or Arg Arg NH—(CH2)10—CO Ser Pro D-Phe
    28 Ac Lys or Arg Arg NH—(CH2)10—CO Ser Pro D-Phe
    29 Sar Orn Arg NH—(CH2)10—CO Ser Pro D-Phe
    30 Ac Orn Arg NH—(CH2)10—CO Ser Pro D-Phe
  • TABLE 3
     2 AcArgProProGlyPheSerProD-Phe-X
     3 acylArgProProGlyPheSerProD-Phe-X
     4a SarArgArgProProGlyPheSerProD-Phe-X
     4b SarD-ArgArgProProGlyPheSerProD-Phe-X
     5a AcArgArgProProGlyPheSerProD-Phe-X
     5b AcD-ArgArgProProGlyPheSerProD-Phe-X
     6a acylArgArgProProGlyPheSerProD-Phe-X
     6b acylD-ArgArgProProGlyPheSerProD-Phe-X
     7a SarArgArgProHypGlyPheSerProD-Phe-X
     7b SarD-ArgArgProHypGlyPheSerProD-Phe-X
     8a AcArgArgProHypGlyPheSerProD-Phe-X
     8b AcD-ArgArgProHypGlyPheSerProD-Phe-X
     9a acylArgArgProHypGlyPheSerProD-Phe-X
     9b acylD-ArgArgProHypGlyPheSerProD-Phe-X
    10a SarLysArgProHypGlyPheSerProD-Phe-X
    10b SarD-LysArgProHypGlyPheSerProD-Phe-X
    11a AcLysArgProHypGlyPheSerProD-Phe-X
    llb AcD-LysArgProHypGlyPheSerProD-Phe-X
    12a acylLysArgProHypGlyPheSerProD-Phe-X
    12b acylD-LysArgProHypGlyPheSerProD-Phe-X
    13a SarOrnArgProProGlyPheSerProD-Phe-X
    13b SarD-OrnArgProProGlyPheSerProD-Phe-X
    14a acylArgArgArgProProGlyPheSerProD-Phe-X
    14b acyID-ArgArgArgProProGlyPheSerProD-Phe-X
    14c acylArgD-ArgArgProProGlyPheSerProD-Phe-X
    14d acylD-ArgD-ArgArgProProGlyPheSerProD-Phe-X
    14ba SarArgArgArgProProGlyPheSerProD-Phe-X
    14bb SarD-ArgArgArgProProGlyPheSerProD-Phe-X
    14bc SarArgD-ArgArgProProGlyPheSerProD-Phe-X
    14bd SarD-ArgD-ArgArgProProGlyPheSerProD-Phe-X
    15a AcLysLysArgProProGlyPheSerProD-Phe-X
    15b AcD-LysLysArgProProGlyPheSerProD-Phe-X
    15c AcLysD-LysArgProProGlyPheSerProD-Phe-X
    15d AcD-LysD-LysArgProProGlyPheSerProD-Phe-X
    15ba SarLysLysArgProProGlyPheSerProD-Phe-X
    15bb SarD-LysLysArgProProGlyPheSerProD-Phe-X
    15bc SarLysD-LysArgProProGlyPheSerProD-Phe-X
    15bd SarD-LysD-LysArgProProGlyPheSerProD-Phe-X
    16a SarLysArgProHypGlyAccSerProD-Phe-X
    16b SarArgArgProHypGlyAccSerProD-Phe-X
    17a SarLysArgProHypGly4BipSerProD-Phe-X
    17b SarArgArgProHypGly4BipSerProD-Phe-X
    18a SarLysArgProHypGlyChaSerProD-Phe-X
    18b SarArgArgProHypGlyChaSerProD-Phe-X
    19a SarLysArgProHypGlyPenSerProD-Phe-X
    19b SarArgArgProHypGlyPenSerProD-Phe-X
    20a SarLysArgProHypGlyThiSerProD-Phe-X
    20b SarArgArgProHypGlyThiSerProD-Phe-X
    21a SarLysArgProHypGlyCpgSerProD-Phe-X
    21b SarArgArgProHypGlyCpgSerProD-Phe-X
    22a SarLysArgProHypGlyChgSerProD-Phe-X
    22b SarArgArgProHypGlyChgSerProD-Phe-X
    23a SarLysArgProHypGlyPhgSerProD-Phe-X
    23b SarArgArgProHypGlyPhgSerProD-Phe-X
    24a SarLysArgNH-(CH2)7-COPheSerProD-Phe-X
    24b SarArgArgNH-(CH2)7-COPheSerProD-Phe-X
    25a AcLysArgNH-(CH2)7-COPheSerProD-Phe-X
    25b AcArgArgNH-(CH2)7-COPheSerProD-Phe-X
    26a SarOrnArgNH-(CH2)7-COPheSerProD-Phe-X
    26b AcOrnArgNH-(CH2)7-COPheSerProD-Phe-X
    27a SarLysArgNH-(CH2)10-COSerProD-Phe-X
    27b SarArgArgNH-(CH2)10-COSerProD-Phe-X
    28a AcLysArgNH-(CH2)10-COSerProD-Phe-X
    28b AcArgArgNH-(CH2)10-COSerProD-Phe-X
    29 SarOrnArgNH-(CH2)10-COSerProD-Phe-X
    39 AcOrnArgNH-(CH2)10-COSerProD-Phe-X
  • The synthesis of peptides described herein, including the preparation of appropriate amino acid derivatives, their activation and coupling to form peptides and methods for purification of peptides and determination of their purity are included in the general body of knowledge of peptide chemistry, as generally described in “Solid phase peptide synthesis” by Stewart and Young (1984, Solid phase peptide synthesis. Pierce Chemical Company, 2nd Edition) for the solution-phase synthesis and solid phase method.
  • Therapeutic applications of desArg9BK-related B1 receptor agonists are based upon recognized beneficial actions mediated by kinin B1 receptors such as vasodilatation/hypotension, neo-vascularization, angiogenesis, anti-ischemia, increase of vascular permeability specifically in surrounding brain tumors, and include treatment of pathological conditions where amount of exogenous B1 receptor agonists is needed. These conditions may include treatment of life-threatening diseases specifically hypertension and diabetes associated with vasculopathies. Compounds of the present invention may be used to prevent and/or reverse end-organ failure, owing to lack of tissue perfusion, in hypertensive and diabetic patients. Such considerations may be extended to other diseases including cerebral and coronary artery diseases, to reduce incidence of stroke and myocardial infarction, respectively, and peripheral arterial disease to improve mobility of afflicted individuals. Another therapeutic application of the present invention covers atherosclerotic coronary artery diseases where balloon angioplasty is to be performed to restore blood flow to blood-deprived heart tissue. Restenosis (repeat narrowing or blockage) of injured heart coronary vessels is frequently observed, usually one-third of the time, following angioplasty procedure. Kinin B1 receptors stimulants, owing to their angiogenic properties, may assist to lower the rate of recurrence or restenosis.
  • Limited therapeutic success in the treatment of central nervous system neoplasia with chemotherapy is attributed partly by delivery impediment related to blood brain barrier. Different approaches have been advocated to improve delivery across the blood brain barrier (Black, 1995, Adv Drug Delivery Rev 15, 37-52). Amongst these approaches, the infusion of proteolytically resistant bradykinin and desArg9BK surrogates to modulate permeability of the neoplastic blood vessels, have been studied (Emerich et al., 2000, Pharm Res 17, 1212-1219; Cardoso et al., 2004, BMC Neurosci 5, 38). Pharmacological approach, using kinin B2 receptor synthetic peptide agonist, has also prove successful for delivery of chemotherapeutics to solid peripheral tumors thereby increasing their efficacy (Emerich et al., 2001, J Pharmacol Exp Ther 296, 623-631). Altogether, these findings underscore the kinin B1 (and B2) receptors as potential targets to cure and/or to improve the quality of life of cancer patients.
  • Compounds of the present invention may be administered topically, subcutaneously, or by injection or infusion or delivered using suited biodegradable microsphere-based carrier systems or as an oral suspension in an appropriate vehicle or as tablets, pills, capsules, caplets or the like. The dosage regimen and manner of administration will be defined by the application of the B1 receptor peptide agonists and as per standard clinical testing to find the optimal dose.
  • The present invention will be more readily understood by referring to the following examples which are given to illustrate the invention rather than to limit its scope.
  • Example 1 Peptide Synthesis
  • Synthesis of the kinin B1 receptor agonists of the present invention by solid phase peptide synthesis (SPPS) may be carried out manually (see Stewart & Young and K. Wisniewski) or by use of the Applied Bioscience 430A for Boc-amino acids or by use of Pioneer™ continuous flow peptide synthesis system for Fmoc-amino acids. SPPS involves use of standard procedures, defined as follows:
  • General Method Involving Boc-Strategy
  • Procedure A, DCC coupling reaction: A 4-fold excess of Boc-amino acids over resin substitution rate is used in the Applied Bioscience 430A synthesizer. Boc-amino acids are activated for coupling with an equimolar amount of DCC and 2 equivalents of DIEA. The solvent may be DCM, DMF, or NMP. The resin is washed with the same solvent before and after coupling. Completeness of coupling is determined with a Kaiser test.
  • Procedure B, TFA deprotection and neutralization: The deprotection reagent is 40% TFA in DCM, containing 1 mg/mL N-acetyl-LD-tryptophan. It is used for 30 min, following a prewash. The neutralization reagent is 20% DIEA in DCM.
  • Procedure C, N-Terminal acylation: A 5-fold excess of acyl chlorides and 10-fold excess of DIEA over peptide-resin are used in DCM for 30 min. The resin is washed with the same solvent after completion of the reaction.
  • Procedure D, HF cleavage: A batch of 0.5 mmole of peptide-resin is mixed with 1.0 mL anisole and chilled in the reaction vessel (resistant to HF) to −78° C., and 10 ml of anhydrous HF is distilled into the vessel under vacuum. The mixture is stirred at 0° C. for 1 h, and the HF is evaporated first under a nitrogen flow, then under vacuum. The peptide and resin mixture is washed three times with dry ether, and the peptide is extracted into 50% acetic acid. The peptide solution is concentrated under vacuum, diluted in water, and lyophilized.
  • Procedure E, Purification: Preparative medium pressure chromatography may be carried out on a reversed phase C18 silica column in a gradient of 0.1% TFA in water to 0.05% TFA in acetonitrile. Eluted peptide is detected by UV at 254 nm. Analytical HPLC may be carried out in the same system to identified pure fractions.
  • Procedure F, Characterization: Final products are identified by analytical HPLC and by mass spectroscopy. MALDI spectra are recorded on a Tofspec™ 2E (micromass, UK).
  • General Method Involving Fmoc-Strategy
  • The approach is an alternative approach of peptides synthesis and standard with Mitsunobu reaction ω-amino acids residues. Synthesis may be carried out by use of Pioneer™ continuous flow peptide synthesis system.
  • Procedure G: The resin is placed in the column and a 2 to 4-fold excess of Fmoc-protected amino acids over resin substitution rate is placed in the sampler tray. Synthesis is performed using amine free DMF. All solutions needed for the solid phase continuous flow synthesis are prepared and loaded in the synthesizer. The synthesis protocol is prepared, loaded into the synthesizer, and run in normal or extended cycle mode. Fmoc deprotection is performed in 20% piperidine in DMF and monitored through UV detector at 364 nm. Fmoc-protected amino acids are activated for coupling with an equimolar amount of HATU or TBTU, and 2 equivalents of DIEA.
  • Procedure H, N-terminal caping (acetylation): This step is optional and can be included in the synthesis protocol. The acetylation reagents are 5% acetic anhydride and 6% 2,4-lutidine in DMF. The resin is washed with the same solvent and isopropanol after completion of the reaction. The resin is removed from the column synthesizer and dried under vacuum 12 hours.
  • Procedure I, TFA cleavage: The cleavage solution, TFA: water: TIPS (95%:2.5%:2.5%), is mixed with peptide-resin, and stirred at room temperature for 2 h. The resin is filtrated and the peptide is precipitated in dry ether. The suspension is centrifuged. The ether solution is decanted and the precipitated peptide is dissolved in water and lyophilized. The peptide is purified and characterized as described in procedures E and F.
  • Example 2 Synthesis of N-Terminal Alkylated Analogues
  • The peptide chain is assembled by Fmoc strategy. O-NBS group is introduced after Fmoc deprotection at the N-terminal position as described in procedure J followed by 0.1 mmol of o-NBS-aan-resin (0.2-0.6 mmol/g) is suspended in 1 mL of DME, and 1 mmol of appropriate alcohol is added to the suspension. Mistunobu reaction and deprotection of the o-NBS group are performed as described in procedure J. After the desired peptide is assembled, the resin is treated with 10 equivalent of 1 M solution of mercaptoethanol/DBU in DMF for 1 h, and washed thoroughly with DMF and DCM. The peptide is then cleaved with an appropriate TFA cocktail, see procedure I. The peptide is purified and characterized as described in procedures E and F.
  • Example 3 Functional Assays
  • In vitro and in vivo bioassays were used to assess the potency and selectivity indexes of peptide compounds at the inducible kinin B1 receptor subtype.
  • In Vitro Functional Assays (Isolated Preparations in Organ Baths)
  • Selected compounds were tested for activities in three isolated vessels: the rabbit aorta (rbA) and jugular vein (rbJV) and the human umbilical vein (hUV). All details regarding the collection and handling of human umbilical cords and rabbit vessels as well as, the procedures for preparing the isolated organs and the experimental protocols are described in these publications: (Gobeil et al., 1996, Br J Pharmacol 118, 289-294; Gobeil et al., 1999, Hypertension 33(3), 823-829). The rbA and the hUV without endothelium (which contains B1 receptors) were used to determine the agonistic activities of each compound expressed in term of pEC50 values (-log of the molar concentration of agonist required to produce 50% of the maximal response). The rabbit jugular vein (which contains only B2 receptor) was used to exclude any action of the new compounds at B2 receptors and thus establish their selectivity.
  • In Vivo Functional Assays (Rabbit Blood Pressure Model)
  • Selected compounds were tested as hypotensive agents in anesthetized rabbits. Surgical procedures and experimental methodologies used herein are based upon previous detailed reports (Gobeil et al., 1999, Immunopharmacol 43: 179-185; Gobeil et al., vide supra). Pathogen-free rabbits (which do not express functional B1 receptors) were anesthetized with a mixed solution of ketamine/xylazine injected intramuscularly in experiments designed to study possible interaction of the compounds with B2 receptors The trachea was intubated to facilitate breathing. Mean arterial blood pressure was continuously monitored by inserting a polyethylene catheter (filled with heparinized saline solution) into the right carotid artery attached to a transducer (model TDX-300; Micro-Med Inc., KY, USA) connected to a blood pressure analyzer (model BPA-400a; Micro-Med Inc., KY, USA). A second arterectomy was performed on the left carotid artery for the administration of graded doses of compounds into the aorta. In experiments designed to study potency of compounds at B1 receptors in rabbits, the anesthetic sodium pentobarbital (30 mg/kg intravenously (i.v.) through the auricular vein) was used instead of the ketamine/xylasine solution. For this purpose, rabbits were immunostimulated with lipopolysaccharide (LPS) (50 μg/kg i.v.) 5 hr before inducing the anesthesia; this endotoxin is a well known potent inducer of B1 receptor expression both in vitro and in vivo experiments. Hypotensive activities and duration of action of peptide compounds were measured following their intra-aortic administration as described in Gobeil et al. (1999, Hypertension 33(3), 823-829).
  • Example 4 Enzymatic Stability Studies
  • Enzymatic resistance of peptide compounds were measured in harsh conditions using rabbit lung and kidney extracts prepared as reported (Tramontana et al., 2001, J Pharmacol Exp Ther 296(3), 1051-1057) with minor modifications. Briefly, animals were euthanized, organs harvested and cleaned from connective and adipose tissues. Tissues were then homogenized with a Polytron homogenizer in 5 volumes (w/v) of cold buffer consisting of Tris HCl 50 mM pH 7.5, NaCl 300 mM and ZnCl2 10 μM, and centrifuged at 1500 g for 15 min at 4° C. Concentration of proteins from the resulting supernatant was determined using Bradford method with bovine serum as standard. Peptide compounds (150 μM) were incubated at 37° C. in the presence of tissue enzymatic extracts (200 μg proteins) for different times (0, 15, 60 min) in a total reaction medium of 250 μl. The naturally-occurring B1 receptor agonist LysdesArg9-BK served as reference peptide. Hydrolysis was stopped by immersing samples in boiling water.
  • Separation of peptide substrates and their metabolites was achieved by reverse-phase HPLC on a C18 μBondpak column (Waters Associates) with a linear gradient of 5% to 65% of water/acetonitrile (both containing 0.05% TFA) at 1 mL/min over a period of 20 min, as described (Gobeil et al., 1999, vide supra). A 50 μL aliquot of each assay was injected into the column. Peptide metabolism was calculated from the decrease in peptide substrate concentration. The elution positions of the peptides were determined following the absorbance at 214 nm and peak area integration was calculated using a computer software program (Baseline 810, Waters).
  • Example 5 Blood Brain Barrier Opening
  • The delivery potency of peptide compounds was studied in syngeneic F98-Fischer glioma rat model based on previous reports (Barth, 1998, J Neurooncol 36, 91-102; Mathieu et al., 2005, The J Appl Res 5(1), 17-25). Briefly, the F98 cell line was cultured in monolayer and stereotactically implanted (1×104 cells in a volume of 5 μl; 1 μl/min) in the right frontal lobe of Fischer rats using standardized and validated coordinates. In this model, the tumor take has been shown to be 100%. Moreover, this model has been shown to adequately emulate the situation of primary malignant brain tumors in the human (Mathieu at al., vide supra). At 14 days post-implantation, rats were anesthetized with ketamine/xylazine solution. Blood brain barrier disruption was performed in rats by administering specific B1 interacting peptides or vehicle (isotonic saline) into the internal carotid artery in a retrograde fashion from the external carotid after catheter implantation. The infusion rates of peptide or vehicle (total volume injected: 500 μl) were set at 10 nmol/kg/min for 5 min followed by a 20 min resting period. Animals were perfused via the same catheter using paraformaldehyde/glutaraldehyde solution, Brain tissues were than collected, embedded in paraffin, sectioned (3 μm) using a dedicated brain matrix and prepared for immunohistochemistry for albumin quantification (as marker of the extent of barrier opening) (Fortin, 2003, Prokai L, Prokai-Tatrai K eds. Peptide transport and delivery to the CNS. Progress in drug research, Birkhauser, Swittzerland, 61, 127-154). This quantification was expressed as the ratio of immunostaining regions against the treated cerebral hemisphere (Fortin, 2003, vide supra).
  • While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth, and as follows in the scope of the appended claims.
  • REFERENCES
    • IUPAC-IUB Commission on Biochemical Nomenclature: Symbols for amino acids derivatives and peptides 1972. Biochem J 126, 773-780
    • Agata at al., 2000, Hypertension 36,364-370
    • Barth, 1998, J Neurooncol 36, 91-102
    • Black, 1995, Adv Drug Delivery Rev 15, 37-52
    • Bouchard et al., 1998, Brit J Pharmacol 123, 413-420
    • Cardoso et al., 2004, BMC Neurosci 5, 38
    • Chahine et al., 1993, Brit J Pharmacol 108, 318-322
    • Couture and Girolami, 2004, Eur J Pharmacol 500(1-3), 467-485
    • Drapeau et al., 1991, J Pharmacol Exp Ther 259, 997-1003
    • Duguay et al., 2004, Brit J Pharmacol 141(4), 728-736
    • Duka et al., 2001, Circ Res 88, 275-281
    • Duka et al., 2006, Am J Physiol Endocrinol Metab [Epub ahead of print]
    • Emanueli and Madeddu, 2001, Trends Pharmacol. Sci. 22(9), 478-484
    • Emanueli et al., 2002, Circulation 105, 360-366
    • Emerich et al., 2000, Pharm Res 17, 1212-1219
    • Emerich et al., 2001, J Pharmacol Exp Ther 296, 623-631
    • Fortin, 2003, Prokai L, Prokai-Tatrai K eds. Peptide transport and delivery to the CNS, Progress in drug research, Birkhauser, Swittzerland, 61, 127-154
    • Gobeil et al., 1996, Br J Pharmacol 118, 289-294
    • Gobeil et al., 1999, Immunopharmacol 43, 179-185
    • Gobeil et al., 1999, Hypertension 33(3), 823-829
    • Hagiwara et al., 2004, Hypertens Res 27(6), 399-408
    • Kichuk et al., 1996, Circulation 94, 44-51
    • Mathieu et al., 2005, The J Appl Res 5(1), 17-25
    • Parenti et al., 2001, FASEB J 15(8), 1487-1489
    • Pruneau et al., 1994, Brit J Pharmacol 111, 1029-1034
    • Rhaleb et al., 1990, Brit J Pharmacol 99, 445-448
    • Sharma and Thani, 2004, Drug 7(10): 926-934
    • Stewart and Young, 1984, Solid phase peptide synthesis. Pierce Chemical Company, 2nd Edition
    • Tramontana et al., 2001, J Pharmacol Exp Ther 296(3), 1051-1057
    • Xu et al., 2005, Hypertension 45(4): 747-753

Claims (10)

1-20. (canceled)
21. A method for reducing the risks of kidney failure due to a lack of tissue perfusion in a patient suffering from diabetes, said method comprising the step of administering to said patient a compound of formula (1)
aay-aax-aa0-aa1-aa2-aa3-aa4-aa5-Ser6-Pro7-D-Phe8-X (1)
or a salt thereof;
wherein:
X is OH or NH2;
aay is Sar, acetyl or other acyl group;
aax is L-Arg, D-Arg, L-Lys, D-Lys, L-Orn, D-Orn, another basic amino acid of the L or D-configuration, L-Cit, D-Cit or is absent;
aa0 is L-Arg, D-Arg, L-Lys, D-Lys, L-Orn, D-Orn, another basic amino acid of the L or D-configuration, or L-Cit, D-Cit;
aa1 is L-Arg or D-Arg;
aa2 is L-Pro, L-Oic, another L-Pro mimic amino acid, L-Hyp, L-α-(Me)Pro or another Pro-mimic amino acid derivative chosen from 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-proline; azetidine-2-carboxylic acid; trans-4-cyano-proline; cis-4-cyano-proline; 2-ethylthiazolidine-4-carboxylic acid; thiazolidine-2-carboxylic acid; 2-methylthiazolidine-4-carboxylic acid; and 3-phenylpyrrolidine-2-carboxylic acid;
aa3 is L-Pro, or another L-Pro-mimic amino acid, L-Hyp, L-Oic, L-α-(Me)Pro or another Pro-mimic amino acid derivative chosen from 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-proline; azetidine-2-carboxylic acid; trans-4-cyano-proline, cis-4-cyano-proline; 2-ethylthiazolidine-4-carboxylic acid; thiazolidine-2-carboxylic acid; 2-methylthiazolidine-4-carboxylic acid; and 3-phenylpyrrolidine-2-carboxylic acid;
aa4 is Gly, ethyl amine, or Aib,
aa5 is L-Phe, D-Phe, Acc, L-Cha, D-Cha, L-Chg, D-Chg, L-Cpg, D-Cpg, L-Igl, D-Igl, L-Pen, D-Pen, L-4Bip, D-4Bip, L-Phg, D-Phg, L-Thi, or D-Thi; and
D-Phe8 is D-Phe. or
-aa2-aa3-aa4- is an aliphatic ω-amino carboxyl residue with a chain length of 8 carbon atoms; and X, aay, aax, aa0, aa1, aa5, and D-Phe8 are as previously defined; or
-aa2--aa3-aa4-aa5- is an aliphatic ω-amino carboxyl residue with a chain length of 11 carbon atoms; and X, aay, aax, aa0, aa1, and D-Phe8 are as previously defined.
22. The method of claim 21, wherein said compound is chosen from:
SarArgArgProProGlyPheSerProD-Phe-X SarD-ArgArgProProGlyPheSerProD-Phe-X AcArgArgProProGlyPheSerProD-Phe-X AcD-ArgArgProProGlyPheSerProD-Phe-X acylArgArgProProGlyPheSerProD-Phe-X acylD-ArgArgProProGlyPheSerProD-Phe-X SarArgArgProHypGlyPheSerProD-Phe-X SarD-ArgArgProHypGlyPheSerProD-Phe-X AcArgArgProHypGlyPheSerProD-Phe-X AcD-ArgArgProHypGlyPheSerProD-Phe-X acylArgArgProHypGlyPheSerProD-Phe-X acyLD-ArgArgProHypGlyPheSerProD-Phe-X SarLysArgProHypGlyPheSerProD-Phe-X SarD-LysArgProHypGlyPheSerProD-Phe-X AcLysArgProHypGlyPheSerProD-Phe-X AcD-LysArgProHypGlyPheSerProD-Phe-X acylLysArgProHypGlyPheSerProD-Phe-X acylD-LysArgProHypGlyPheSerProD-Phe-X SarOrnAraProProGlyPheSerProD-Phe-X SarD-OrnArgProProGlyPheSerProD-Phe-X acylArgArgArgProProGlyPheSerProD-Phe-X acyLD-ArgArgArgProProGlyPheSerProD-Phe-X acylArgD-ArgArgProProGlyPheSerProD-Phe-X acylD-ArgD-ArgArgProProGlyPheSerProD-Phe-X SarArgArgArgProProGlyPheSerProD-Phe-X SarD-ArgArgArgProProGlyPheSerProD-Phe-X SarArgD-ArgArgProProGlyPheSerProD-Phe-X SarD-ArgD-ArgArgProProGlyPheSerProD-Phe-X AcLysLysArgProProGlyPheSerProD-Phe-X AcD-LysLysArgProProGlyPheSerProD-Phe-X AcLysD-LysArgProProGlyPheSerProD-Phe-X AcD-LysD-LysArgProProGlyPheSerProD-Phe-X SarLysLysArgProProGlyPheSerProD-Phe-X SarD-LysLysArgProProGlyPheSerProD-Phe-X SarLysD-LysArgProProGlyPheSerProD-Phe-X SarD-LysD-LysArgProProGlyPheSerProD-Phe-X SarLysArgProHypayAccSerProD-Phe-X SarArgArgProHypGlyAccSerProD-Phe-X SarLysArgProHypGly4BipSerProDPhe-X SarArgArgProHypGly4BipSerProD-Phe-X SarLysArgProHypGlyChaSerProD-Phe-X SarArgArgProHypGlyChaSerProD-Phe-X SarLysArgProHypGlyPenSerProD-Phe-X SarArgArgProHypGlyPenSerProD-Phe-X SarLysArgProHypGlyThiSerProD-Phe-X SarArgArgProHypGlyThiSerProD-Phe-X SarLysArgProHypGlyCpgSerProD-Phe-X SarArgArgProHypGlyCogSerProD-Phe-X SarLysArgProHypGlyChgSerProD-Phe-X SarArgArgProHypGlyChgSerProD-Phe-X SarLysArgProHypGlyPhgSerProD-Phe-X SarArgArgProHypGlyPh9SerProD-Phe-X SarLysArGNH-(CH2)7-COPheSerProD-Phe-X SarArgArgNH-(CH2)7-COPheSerProD-Phe-X AcLysArgNH-(CH2)7-COPheSerProD-Phe-X AcArgArgNH-(CH2)7-COPheSerProD-Phe-X SarOrnArgNH-(CH2)7-COPheSerProD-Phe-X AcOrnArgNH-(CH2)7-COPheSerProD-Phe-X SarLysArgNH-(CH2)10-COSerProD-Phe-X SarArgArgNH-(CH2)10-COSerProD-Phe-X AcLysArgNH-(CH2)10-COSerProD-Phe-X AcArgArgNH-(CH2)10-COSerProD-Phe-X SarOrnArgNH-(CH2)10-COSerProD-Phe-X AcOrnArgNH-(CH2)10-COSerProD-Phe-X SarLysArgProHypGlyIglSerProD-Phe-X and SarArgArgProHypGlyIglSerProD-Phe-X.
23. The method of claim 21, wherein said compound is chosen from:
SarLysArgProHypGlyChaSerProD-Phe-X SarLysArgProHypGlyPhgSerProD-Phe-X SarLysArgProHypGlyIglSerProD-Phe-X SarLysArgProHypGlyPenSerProD-Phe-X and SarLysArgProHypGlyCpgSerProD-Phe-X.
24. A method for treating peripheral and coronary arterial diseases in a patient in need thereof, said method comprising the step of administering to said patient a compound of formula (1)
aay-aax-aa0-aa1-aa2-aa3-aa4-aa5-Ser6-Pro7-D-Phe8-X (1)
or a salt thereof;
wherein:
X is OH or NH2;
aay is Sar, acetyl or other acyl group;
aax is L-Arg, D-Arg, L-Lys, D-Lys, L-Orn, D-Orn, another basic amino acid of the L or D-configuration, L-Cit, D-Cit or is absent;
aa0 is L-Arg, D-Arg, L-Lys, D-Lys, L-Orn, D-Orn, another basic amino acid of the L or D-configuration, or L-Cit, D-Cit;
aa1 is L-Arg or D-Arg;
aa2 is L-Pro, L-Oic, another L-Pro mimic amino acid, L-Hyp, L-α-(Me)Pro or another Pro-mimic amino acid derivative chosen from 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-proline; azetidine-2-carboxylic acid; trans-4-cyano-proline; cis-4-cyano-proline; 2-ethylthiazolidine-4-carboxylic acid; thiazolidine-2-carboxylic acid; 2-methylthiazolidine-4-carboxylic acid; and 3-phenylpyrrolidine-2-carboxylic acid;
aa3 is L-Pro, or another L-Pro-mimic amino acid, L-Hyp, L-Oic, L-α-(Me)Pro or another Pro-mimic amino acid derivative chosen from 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-proline; azetidine-2-carboxylic acid; trans-4-cyano-proline; cis-4-cyano-proline; 2-ethylthiazolidine-4-carboxylic acid; thiazolidine-2-carboxylic acid; 2-methylthiazolidine-4-carboxylic acid; and 3-phenylpyrrolidine-2-carboxylic acid;
aa4 is Gly, ethyl amine, or Aib;
aa5 is L-Phe, D-Phe, Acc, L-Cha, D-Cha, L-Chg, D-Chg, L-Cpg, D-Cpg, L-Igl, D-Igl, L-Pen, D-Pen, L-4Bip, D-4Bip, L-Phg, D-Phg, L-Thi, or D-Thi; and
D-Phe5 is D-Phe. or
-aa2-aa3-aa4- is an aliphatic ω-amino carboxyl residue with a chain length of 8 carbon atoms; and X, aay, aax, aa0, aa1, aa5, and D-Phe8 are as previously defined; or
-aa2-aa3-aa4-aa5- is an aliphatic ω-amino carboxyl residue with a chain length of 11 carbon atoms; and X, aay, aax, aa0, aa1, and D-Phe8 are as previously defined.
25. The method of claim 24, wherein said compound is chosen from:
SarArgArgProProGlyPheSerProD-Phe-X SarD-ArgArgProProGlyPheSerProD-Phe-X AcArgArgProProGlyPheSerProD-Phe-X AcD-ArgArgProProGlyPheSerProD-Phe-X acylArgArgProProGlyPheSerProD-Phe-X aloyID-ArgArgProProGlyPheSerProD-Phe-X SarArgArgProHypGlyPheSerProD-Phe-X SarD-ArgArgProHypGlyPheSerProD-Phe-X AcArgArgProHypGlyPheSerProD-Phe-X AcD-ArgArgProHypGlyPheSerProD-Phe-X acylArgArgProHypGlyPheSerProD-Phe-X acylD-ArgArgProHypGlyPheSerProD-Phe-X SarLysArgProHypGlyPheSerProD-Phe-X SarD-LysArgProHypGlyPheSerProD-Phe-X AcLysArgProHypGlyPheSerProD-Phe-X AclD-LysArgProHypGlyPheSerProD-Phe-X acylLysArgProHypGlyPheSerProD-Phe-X acyID-LysArgProHypGlyPheSerProD-Phe-X SarOrnArgProProGlyPheSerProD-Phe-X SarD-OrnArgProProGlyPheSerProD-Phe-X acylArgArgArgProProGlyPheSerProD-Phe-X acy1D-ArgArgArgProProGlyPheSerProD-Phe-X acylArgD-ArgArgProProGlyPheSerProD-Phe-X acylD-ArgD-ArgArgProProGlyPheSerProD-Phe-X SarArgArgArgProProGlyPheSerProD-Phe-X SarD-ArgArgArgProProGlyPheSerProD-Phe-X SayArgD-ArgArgProProGlyPheSerProD-Phe-X SarD-ArgD-ArgArgProProGlyPheSerProD-Phe-X AcLysLysArgProProGlyPheSerProD-Phe-X AcD-LysLysArgProProGlyPheSerProD-Phe-X AcLysD-LysArgProProGlyPheSerProD-Phe-X AcD-LysD-LysArgProProGlyPheSerProD-Phe-X SarLysLysArgProProGlyPheSerPreD-Phe-X SarD-LysLysArgProProGlyPheSerProD-Phe-X SarLysD-LysArgProProGlyPheSerProD-Phe-X SarD-LysD-LysArgProProGlyPheSerProD-Phe-X SarLysArgProHypGlyAccSerProD-Phe-X SarArgArgProHypGlyAccSerProD-Phe-X SarLysArgProHypGly4BipSerProD-Phe-X SerArgArgProHypGly4BipSerProD-Phe-X SarLysArgProHypGlyChaSerProD-Phe-X SarArgArgProHypayChaSerProD-Phe-X SarLysArgProHypGlyPenSerProD-Phe-X SarArgArgProHypGlyPenSerProD-Phe-X SarLyeArgProHypGlyThiSerProD-Phe-X SerArgArgProHypGlyThiSerProD-Phe-X SarLysArgProHypGlyCpgSerProD-Phe-X SarArgArgProHypGlyCpgSerProD-Phe-X SarLysArgProHypGlyChgSerPreD-Phe-X SarArgArgProHypGlyChgSerProD-Phe-X SarLysArgProHypGlyPhgSerProD-Phe-X SarArgArgProHypGlyPhgSerProD-Phe-X SarLysArgNH-(CH2)7-COPheSerProD-Phe-X SarArgArgNH-(CH2)7-COPheSerProD-Phe-X AcLysArgNH-(CH2)7-COPheSerProD-Phe-X AcArgArgNH-(CH2)7-COPheSerProD-Phe-X SarOrnArgNH-(CH2)7-COPheSerProD-Phe-X AcOrnArgNH(CH2)7-COPheSerProD-Phe-X SarLysArgNH-(CH2)10-COSerProD-Phe-X SarArgArgNH-(CH2)10-COSerProD-Phe-X AcLysArgNH-(CH2)10-COSerProD-Phe-X AcArgArgNH-(CH2)10-COSerProD-Phe-X SarOrnArgNH-(CH2)10-COSerProD-Phe-X AcOrnArgNH-(CH2)10-COSerProD-Phe-X SarLysArgProHypGlyIglSerProD-Phe-X and SarArgArgProllyoGlyIglSerProD-Phe-X.
26. The method of claim 24, wherein said compound is chosen from:
SarLysArgProHypGlyChaSerProD-Phe-X SarLysArgProHypGlyPhgSerProD-Phe-X SarLysArgProHypGlyIglSerProD-Phe-X SarLysArgProHypGlyPenSerProD-Phe-X and SarLysArgProHypGlyCpgSerProD-Phe-X.
27. A method for adjunct treatment of coronary diseases with balloon catheter angioplasty, said method comprising the step of administering to a patient in need thereof a compound of formula (1);
aaY-aax-aa0-aa1-aa2-aa3-aa4-aa5-Ser6-Pro7-D-Phe8-X (1)
or a salt thereof;
wherein:
X is OH or NH2;
aay is Sar, acetyl or other acyl group;
aax is L-Arg, D-Arg, L-Lys, D-Lys, L-Orn, D-Orn, another basic amino acid of the L or D-configuration, L-Cit, D-Cit or is absent;
aa0 is L-Arg, D-Arg, L-Lys, D-Lys, L-Orn, D-Orn, another basic amino acid of the L or D-configuration, or L-Cit, D-Cit;
aa1 is L-Arg or D-Arg;
aa2 is L-Pro, L-Oic, another L-Pro mimic amino acid, L-Hyp, L-α-(Me)Pro or another Pro-mimic amino acid derivative chosen from 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-proline, azetidine-2-carboxylic acid; trans-4-cyano-proline; cis-4-cyano-proline; 2-ethylthiazolidine-4-carboxylic acid; thiazolidine-2-carboxylic acid; 2-methylthiazolidine-4-carboxylic acid; and 3-phenylpyrrolidine-2-carboxylic acid;
aa3 is L-Pro, or another L-Pro-mimic amino acid, L-Hyp, L-Oic, L-α-(Me)Pro or another Pro-mimic amino acid derivative chosen from 5,5-dimethylthiazolidine-4-carboxylic acid; 3,4-dehydro-proline; azetidine-2-carboxylic acid; trans-4-cyano-proline; cis-4-cyano-proline; 2-ethylthiazolidine-4-carboxylic acid; thiazolidine-2-carboxylic acid; 2-methylthiazolidine-4-carboxylic acid; and 3-phenylpyrrolidine-2-carboxylic acid;
aa4 is Gly, ethyl amine, or Aib;
aa5 is L-Phe, D-Phe, Acc, L-Cha, D-Cha, L-Chg, D-Chg, L-Cpg, D-Cpg, L-Igl, D-Igl, L-Pen, D-Pen, L-4Bip, D-4Bip, L-Phg, D-Phg, L-Thi, or D-Thi; and
D-Phe8 is D-Phe. or
-aa2-aa3-aa4- is an aliphatic ω-amino carboxyl residue with a chain length of 8 carbon atoms; and X, aay, aax, aa0, aa1, aa5, and D-Phe8 are as previously defined; or
-aa2-aa3-aa4-aa5- is an aliphatic ω-amino carboxyl residue with a chain length of 11 carbon atoms; and X, aay, aax, aa0, aa1, and D-Phe8 are as previously defined.
28. The method of claim 27, wherein said compound is chosen from:
SarArgArgProProGlyPheSerProD-Phe-X SarD-ArgArgProProGlyPheSerProD-Phe-X AcArgArgProProGlyPheSerProD-Phe-X AcD-ArgArgProProGlyPheSerProD-Phe-X acylArgArgProProGlyPheSerProD-Phe-X acylD-ArgArgProProGlyPheSerProD-Phe-X SarArgArgProHlypGlyPheSerProD-Phe-X SarD-ArgArgProHypGlyPheSerProD-Phe-X AcArgArgProHypGlyPheSerProD-Phe-X AcD-ArgArgProHypGlyPheSerProD-Phe-X acylArgArgProHypGlyPheSerProD-Phe-X acylD-ArgArgProHypGlyPheSerProD-Phe-X SarLysArgProHypGlyPheSerProD-Phe-X SaralysArgProHypGlyPheSerProD-Phe-X AcLysArgProHypGlyPheSerProD-Phe-X AcD-LysArgProHypayPheSerProD-Phe-X acylLysArgProHypayPheSerProD-Phe-X acylD-LysArgProHypGlyPheSerProD-Phe-X SarOrnArgProProGlyPheSerProD-Phe-X SarD-OrnArgProProGlyPheSerProD-Phe-X acylArgArgArgProProGlyPheSerProD-Phe-X acylD-ArgArgArgProProGlyPheSerProD-Phe-X acylArgD-ArgArgProProGlyPheSerProD-Phe-X acylD-Arga-ArgArgProProGlyPheSerProD-Phe-X SarArgArgArgProProGlyPheSerProD-Phe-X SarD-ArgArgArgProProGlyPheSerProD-Phe-X SarArgD-ArgArgProProGlyPheSerProD-Phe-X SarD-Arga-ArgArgProProGlyPheSerProD-Phe-X AcLysLysArgProProGlyPheSerProD-Phe-X AcD-LysLysArgProProGlyPheSerProD-Phe-X AcLysD-LysArgProProGlyPheSerProD-Phe-X AcD-LysD-LysArgProProGlyPheSerProD-Phe-X SarLysLysArgProProGlyPheSerProD-Phe-X SarD-LysLysArgProProGisePheSerProD-Phe-X SarLysD-LysArgProProGlyPheSerProD-Phe-X SarD-LysD-LysArgProProGlyPheSerProDPhe-X SarLysArgProHypGlyAccSerProD-Phe-X SarArgArgProHypGlyAccSerProD-Phe-X SarLysArgProHypGly4BipSerProD-Phe-X SarArgArgProHypGly4BipSerProD-Phe-X SarLysArgProHypGlyChaSerProD-Phe-X SarArgArgProHypGlyChaSerProD-Phe-X SarLysArgProHypGlyPenSerProD-Phe-X SarArgArgProHypGlyPenSerProD-Phe-X SarLysArgProHypGlyThiSerProD-Phe-X SarArgArgProHypGlyThiSerProD-Phe-X SarLysArgProHypGlyCpgSerProD-Phe-X SarArgArgProHypGlyCpgSerProD-Phe-X SarLysArgProHypGlyChgSerProD-Phe-X SarArgArgProHypGlyChgSerProD-Phe-X SarLysArgProHypGlyPhgSerProD-Phe-X SarArgArgProHypGlyPhgSerProD-Phe-X SarLysArgNH-(CH2)7-COPheSerProD-Phe-X SarArgArgNH-(CH2)7-COPheSerProD-Phe-X AcLysArgNH-(CH2)7-COPheSerProD-Phe-X AcArgArgNH-(CH2)7-COPheSerProD-Phe-X SarOrnArgNH-(CH2)7-COPheSerProD-Phe-X AcOrnArgNH-(CH2)7-COPheSerProD-Phe-X SarLysArgNH-(CH2)10-COSerProD-Phe-X SarArgArgNH-(CH2)10-COSerProD-Phe-X ActysArgNH-(CH2)10-COSerProD-Phe-X AcArgArgNH-(CH2)10-COSerProD-Phe-X SarOrnArgNH-(CH2)10COSerProD-Phe-X AcOrnArgNH-(CH2)10-COSerProD-Phe-X SarLysArgProHypGlyIgiSerProD-Phe-X and SarArgArgProHypGlyIglSerProD-Phe-X
29. The method of claim 27, wherein said compound is chosen from:
SarLysArgProHypGlyChaSerProD-Phe-X SarLysArgProHypGlyPhgSerProD-Phe-X SarLysArgProHypGlyIglSerProD-Phe-X SarLysArgProHypGlyPenSerProD-Phe-X and SarLysArgProHypGlyCpgSerProD-Phe-X,
US13/294,371 2005-06-01 2011-11-11 Kinin b1 receptor peptide agonists and uses thereof Abandoned US20120129774A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/294,371 US20120129774A1 (en) 2005-06-01 2011-11-11 Kinin b1 receptor peptide agonists and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US68588605P 2005-06-01 2005-06-01
PCT/CA2006/000890 WO2006128293A1 (en) 2005-06-01 2006-05-31 Kinin b1 receptor peptide agonists and uses thereof
US91613608A 2008-08-08 2008-08-08
US13/294,371 US20120129774A1 (en) 2005-06-01 2011-11-11 Kinin b1 receptor peptide agonists and uses thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/CA2006/000890 Continuation WO2006128293A1 (en) 2005-06-01 2006-05-31 Kinin b1 receptor peptide agonists and uses thereof
US91613608A Continuation 2005-06-01 2008-08-08

Publications (1)

Publication Number Publication Date
US20120129774A1 true US20120129774A1 (en) 2012-05-24

Family

ID=37481184

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/916,136 Expired - Fee Related US8076453B2 (en) 2005-06-01 2006-05-31 Kinin B1 receptor peptide agonists and uses thereof
US13/294,371 Abandoned US20120129774A1 (en) 2005-06-01 2011-11-11 Kinin b1 receptor peptide agonists and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/916,136 Expired - Fee Related US8076453B2 (en) 2005-06-01 2006-05-31 Kinin B1 receptor peptide agonists and uses thereof

Country Status (3)

Country Link
US (2) US8076453B2 (en)
EP (1) EP1937710B1 (en)
WO (1) WO2006128293A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2990053A1 (en) * 2015-06-25 2016-12-29 Societe De Commercialisation Des Produits De La Recherche Appliquee Socpra Sciences Sante Et Humaines, S.E.C. Novel kinin-based theranostic probes for solid cancers and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5686565A (en) * 1992-10-08 1997-11-11 Scios Inc. Bradykinin antagonist pseudopeptide derivatives of aminoalkanoic acids and related olefins
US5541286A (en) * 1992-10-08 1996-07-30 Scios Nova Inc. Bradykinin antagonist pseudopeptide derivatives of olefinic aminoalkanoic acids
US5521158A (en) * 1992-10-08 1996-05-28 Scios Nova Inc. Pseudopeptide bradykinin receptor antagonists
US5817756A (en) * 1993-09-09 1998-10-06 Scios Inc. Pseudo- and non-peptide bradykinin receptor antagonists

Also Published As

Publication number Publication date
US20090054347A1 (en) 2009-02-26
WO2006128293A1 (en) 2006-12-07
EP1937710A1 (en) 2008-07-02
EP1937710A4 (en) 2010-01-06
US8076453B2 (en) 2011-12-13
EP1937710B1 (en) 2012-10-17

Similar Documents

Publication Publication Date Title
US6046167A (en) Peptide YY analogs
EP1776133B1 (en) Peptidic vasopressin receptor agonists
JP3838656B2 (en) Polypeptide Bombesin Antagonist
EP2473518B1 (en) Stabilized melanocortin ligands
US8222202B2 (en) Use of peptidic vasopressin receptor agonists
HU208027B (en) Process for producing peptide compounds and pharmaceutical composition containing them as active components
CA2610496A1 (en) Synthetic peptide inhibitors of thrombin and thrombin activation of protease activated receptors 1 and 4
US8076453B2 (en) Kinin B1 receptor peptide agonists and uses thereof
US6083915A (en) Method for treating liver cancer
CA2405704C (en) Bombesin analogs for treatment of cancer
US10745443B2 (en) Vasopressin-2 receptor agonists
EP2198878A1 (en) Polypeptide bombesin antagonists
JPH02501224A (en) Bradykinin antagonist peptide
Derdowska et al. New analogues of bradykinin containing a conformationally restricted dipeptide fragment in their molecules: Authors' affiliations
US4587233A (en) Use of Arg-Phe-amide derivatives
AU2005202153B2 (en) Urotensin-II agonists
JPH0232098A (en) D-cys 6 vasopressin antagonist
JPS63303999A (en) Vasopressin antagonist

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITE DE SHERBROOKE, QUEBEC

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GOBEIL, FERNAND, JR.;NEUGEBAUER, WITOLD A.;REGOLI, DOMENICO;AND OTHERS;SIGNING DATES FROM 20070124 TO 20070129;REEL/FRAME:027656/0848

Owner name: SOCIETE DE COMMERCIALISATION DES PRODUITS DE LA RE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UNIVERSITE DE SHERBROOKE;REEL/FRAME:027657/0187

Effective date: 20071122

STCB Information on status: application discontinuation

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