US20020032214A1 - Pharmaceutical composition comprising a compound having anti-xa activity and a platelet aggregation antagonist compound - Google Patents

Pharmaceutical composition comprising a compound having anti-xa activity and a platelet aggregation antagonist compound Download PDF

Info

Publication number
US20020032214A1
US20020032214A1 US09/516,981 US51698100A US2002032214A1 US 20020032214 A1 US20020032214 A1 US 20020032214A1 US 51698100 A US51698100 A US 51698100A US 2002032214 A1 US2002032214 A1 US 2002032214A1
Authority
US
United States
Prior art keywords
compound
platelet aggregation
activity
pharmaceutical composition
aggregation antagonist
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
US09/516,981
Inventor
Andre Uzan
Alain Curaudeau
Robert Leadley
Christopher Dunwiddie
Mark Perrone
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.)
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=21861879&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20020032214(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US09/516,981 priority Critical patent/US20020032214A1/en
Publication of US20020032214A1 publication Critical patent/US20020032214A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • 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
    • 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
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • This invention is directed to a pharmaceutical composition comprising an anti-Xa activity compound and a platelet aggregation antagonist compound which exhibits unexpectedly efficacious activity for treating or preventing a physiological condition associated with a thrombosis related ischemic disorder in a patient.
  • the invention is also directed to a method of treating or preventing a thrombogenic condition associated with a thrombosis related ischemic disorder in a patient comprising administering pharmaceutically effective amounts of an anti-Xa activity compound and a platelet aggregation antagonist compound.
  • Platelet fibrinogen receptor antagonists have been shown to be effective agents for inhibiting platelet-dependent thrombus formation in animal models of coronary thrombosis. Likewise, results from clinical studies have shown that platelet fibrinogen receptor antagonists reduce the composite incidence of major ischemic events when administered to high-risk patients undergoing percutaneous transluminal coronary angioplasty. However, the therapeutic window is very narrow for this class of compounds, in part, because the high degree of inhibition of ex vivo platelet aggregation required for antithrombotic efficacy is often associated with a marked increase in template bleeding time, which is a marker of an undesirable bleeding complications.
  • LMWHs Low molecular weight heparins
  • HCs heparinoid compounds
  • LMWHs and HCs are gradually pervading into the treatment repertoire for arterial thrombotic indications.
  • Preliminary results favoring the use of LMWH or HCs over unfractionated heparin in arterial thrombotic indications are supported by several pharmacodynamic, pharmacokinetic, and mechanistic differences between these two classes of compounds. For example, reliable and safe anticoagulation can be achieved with LMWHs or HCs by subcutaneous dosing without monitoring.
  • LMWHs and HCs have a higher bioavailability, a relatively long half-life, and appear to have a safer profile.
  • LMWHs are more resistant than heparin to neutralization by platelet factor 4, which is released from activated platelets, presumably at the site of arterial thrombosis.
  • Haemostasis the biochemistry of blood coagulation, is an extremely complex phenomena whereby normal whole blood and body tissue spontaneously arrest bleeding from injured blood vessels. Effective haemostasis requires the combined activity of vascular, platelet and plasma factors as well as a controlling mechanism to prevent excessive clotting. Defects, deficiencies, or excesses of any of these components can lead to hemorrhagic or thrombotic consequences.
  • Platelet adhesion, spreading and aggregation on extracellular matrices are central events in thrombus formation. These events are mediated by a family of adhesive glycoproteins, i.e., fibrinogen, fibronectin, and von Willebrand factor.
  • Fibrinogen is a co-factor for platelet aggregation while fibronectin supports platelet attachments and spreading reactions, and von Willebrand factor is important in platelet attachment to and spreading on subendothelial matrices.
  • the binding sites for fibrinogen, fibronectin and von Willebrand factor have been located on the platelet membrane protein complex known as glycoprotein IIb/IIIa.
  • Adhesive glycoproteins like fibrinogen, do not bind with normal resting platelets. However, when a platelet is activated with an agonist such as thrombin or adenosine diphosphate, the platelet changes its shape, perhaps making the GPIIb/IIIa binding site accessible to fibrinogen. Blocking the fibrinogen receptor, thus inhibits platelet aggregation and subsequent thrombus formation, and is useful for the prevention and treatment of pathological thrombogenic conditions, such as stroke, peripheral arterial occlusive disease, disseminated intravascular coagulation, and acute coronary syndromes such as unstable angina and myocardial infarction.
  • pathological thrombogenic conditions such as stroke, peripheral arterial occlusive disease, disseminated intravascular coagulation, and acute coronary syndromes such as unstable angina and myocardial infarction.
  • the invention is also directed to pharmaceutical composition comprising a compound having anti-Xa activity, a platelet aggregation antagonist compound and a pharmaceutically acceptable carrier.
  • the invention is also directed to a method of treating or preventing a thrombogenic condition associated with a thrombosis related ischemic disorder in a patient comprising administering to said patient pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound.
  • FIG. 1 represents information regarding assorted blood sampling/hemodynamic measurements during administration of different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA), and composition thereof over time.
  • the anti-Xa activity compound Enoxaparin
  • the platelet aggregation antagonist compound the platelet aggregation antagonist compound
  • N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA) N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA)
  • FIG. 2 represents a graph of the number of cyclic flow reductions (CFRs) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • CFRs cyclic flow reductions
  • FIG. 3 represents a graph of the activated partial thromboplastin time (APTT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • APTT activated partial thromboplastin time
  • FIG. 4 represents a graph of the prothrombin time (PT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • PT prothrombin time
  • FIG. 5 represents a graph of the Anti-Xa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 6 represents a graph of the Anti-IIa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 7 represents a graph of the template bleeding time for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 8 represents a graph of the platelet count for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 9 represents a graph of collagen-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 10 represents a graph of ADP-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 11 represents a graph of arachidonic acid-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 12 represents a graph of thrombin-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time.
  • FIG. 13 represents information regarding assorted blood sampling/hemodynamic measurements during administration of different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L- ⁇ -cyclohexyl alanine amide (PBGACA) and composition thereof over time.
  • the anti-Xa activity compound Enoxaparin
  • the platelet aggregation antagonist compound the platelet aggregation antagonist compound
  • PBGACA N-[N-[N-(4-(piperidin-4-yl)butanoy
  • FIG. 14 represents a graph of the number of cyclic flow reductions (CFRs) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • CFRs cyclic flow reductions
  • FIG. 15 represents a graph of the activated partial thromboplastin time (APTT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • APTT activated partial thromboplastin time
  • FIG. 16 represents a graph of the prothrombin time (PT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • PT prothrombin time
  • FIG. 17 represents a graph of the Anti-Xa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • FIG. 18 represents a graph of the Anti-IIa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • FIG. 19 represents a graph of the template bleeding time for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound. PBGACA, and composition thereof over time.
  • FIG. 20 represents a graph of the platelet count for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • FIG. 21 represents a graph of collagen-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • FIG. 22 represents a graph of ADP-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • FIG. 23 represents a graph of arachidonic acid-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • FIG. 24 represents a graph of thrombin-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.
  • Patient includes both human and other mammals.
  • Effective amount is meant to describe an amount of composition according to the present invention effective in producing the desired therapeutic effect.
  • Plate aggregation antagonist compound means a compound that binds to the platelet GPIIb/IIIa receptor (GPIIb/IIIa receptor antagonist) and competitively inhibits binding of fibrinogen, fibronectin and von Willebrand factor as well as inhibits aggregation of activated platelets.
  • Anti-Xa activity compound means a heparinoid compound or low molecular weight heparin (LMWH), or synthetic derivatives thereof.
  • PAACs are those disclosed in International Publication No. WO 89/11538, International Publication No. WO 95/10295 or U.S. Pat. No. 5,292,756; further preferred are Reopro® (abciximab).
  • an anti-Xa activity compound may be produced as follows: enrichment by fractionation by ethanol and/or molecular sieving. e.g., gel filtration or membrane filtration of the LMWH present in standard heparin and controlled chemical (by nitrous acid, ⁇ -elimination or periodate oxidation) or enzymatic (by heparinases) depolymerization. The conditions for depolymerization can be carefully controlled to yield products of desired molecular weights. Nitrous acid depolymerization is commonly used.
  • LMWH with low anticoagulant activity and retaining basic chemical structure are prepared by depolymerization using periodate oxidation or by removing the antithrombin-binding fraction of LMWH, or prepared by other methods, using immobilized antithrombin for adsorption.
  • LMWH having an average molecular weight of about 3000 to about 6500.
  • Commercially available LMWHs useful according to the invention include the following: Clexane®/Klexane®/Lovenox® (Enoxaparin (ENOX)) having an average molecular mass of 4500 ⁇ 1000 Dalton (Da), molecular mass distribution comprising components of ⁇ 2000 Da (16.0 ⁇ 4.0%) and 2000 to 8000 Da (78.0 ⁇ 10.0%).
  • Anti-Xa activity (IU/mg on dry basis) of 90 to 125, and an Anti-Xa/Anti-IIa ratio of 3.3 to 5.3;
  • Fraxiparin® (Nardroparin) having an average molecular mass of 4300 ⁇ 700 Da, molecular mass distribution comprising components of ⁇ 2000 Da ( ⁇ 15%), 2000 to 4000 Da (45 ⁇ 10%) and 2000 to 8000 Da (85 ⁇ 10%), Anti-Xa activity (IU/mg on dry basis) of 95 to 130, and an Anti-Xa/Anti-IIa ratio of 2.5 to 4.0; Fragmin® (Dalteparin) having an average molecular mass of 6000 ⁇ 400 Da.
  • an anti-Xa activity compound is a heparinoid compound.
  • a preferred commercially available heparinoid compound useful according to the invention is Orgaran® (Danaparoid) having an average molecular mass of about 6500 Da, Anti-Xa activity (IU/mg on dry basis) of about 10, and an Anti-Xa/Anti-IIa ratio of about 28.
  • Preferred thrombogenic conditions treatable or preventable according to the invention include stroke, atherosclerosis, angiogenesis, thrombosis, thromboembolic conditions such as deep venous thrombosis.
  • pulmonary embolism or thrombophlebitis disseminated intravascular coagulation or thromboembolic syndromes associated with cancer, sepsis or obstetrical complications, peripheral arterial occlusive disease, and acute coronary syndromes such as unstable angina and myocardial infarction.
  • hemodialysis, or extra-corporeal circulation requirement associated with a surgical procedure or tissue damage caused by phospholipases A2 (PLA 2 ) more preferred is unstable angina and myocardial infarction.
  • Another preferred embodiment according to the invention useful in the course of a medical procedure wherein there is a potential for an occurrence pathological thrombogenic condition may occur, such as during coronary artery bypass surgery or percutaneous transluminal coronary angioplasty with or without placement of an intracoronary stent; more preferred is percutaneous transluminal coronary angioplasty with or without placement of an intracoronary stent.
  • Another preferred embodiment according to the invention is the use of pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound in the preparation of a medicament for treating or preventing a physiological condition associated with thrombosis related ischemic disorder.
  • the anti-Xa activity compound and platelet aggregation antagonist compound may be administered in different ways, such as in combination therapies optionally employing medical procedures.
  • the anti-Xa activity compound and platelet aggregation antagonist compound may be administered to a patient concomitantly or at different times provided that they are administered such that at some period of time there are pharmaceutically effective amounts of both compounds present in the patient such that a therapeutic effect according to the invention results.
  • kits for treating or preventing a physiological condition associated with thrombosis related ischemic disorder comprising a plurality of separate containers, wherein at least one of said containers contains a compound having anti-Xa activity and at least another of said containers contains a platelet aggregation antagonist compound, and said containers optionally contain a pharmaceutical carrier which kit may be effectively utilized for carrying out combination therapies according to the invention.
  • a further embodiment for a kit would be wherein of said containers at least one of said containers should contain the compound having anti-Xa activity without the presence of the platelet aggregation antagonist compound, and at least another of said containers should contain the platelet aggregation antagonist compound without the presence of the compound having anti-Xa activity.
  • the anti-Xa activity compound and platelet aggregation antagonist compound may be administered parenterally, topically, rectally, transdermally, intrapulmonary or orally, but they are preferably administered parenterally and/or orally.
  • Suitable compositions containing the compounds used according to the invention may be prepared by conventional means.
  • the compounds used according to the invention may be dissolved or suspended in a suitable carrier.
  • compositions containing the compounds used according to the invention which are suitable for use in human or veterinary medicine.
  • compositions may be prepared according to the customary methods, using one or more pharmaceutically acceptable carrier, which comprise adjuvants or excipients.
  • the adjuvants comprise, inter alia, diluents, sterile aqueous media and the various non-toxic organic solvents.
  • compositions may be presented in the form of tablets, pills, capsules, lozenges, troches, hard candies, granules, powders, aqueous solutions or suspensions, injectable solutions, elixirs or syrups, powders, solution or suspension for intrapulmonary administration and can contain one or more agents chosen from the group comprising sweeteners, flavorings, colorings, or stabilizers in order to obtain pharmaceutically acceptable preparations.
  • excipients such as sterile water, Ringer's solution, lactose, sodium citrate, isotonic saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride, or mixtures of such salts), calcium carbonate and disintegrating agents such as starch, alginic acids and certain complex silicates combined with lubricants such as magnesium stearate, sodium lauryl sulfate and talc may be used for preparing tablets.
  • excipients such as sterile water, Ringer's solution, lactose, sodium citrate, isotonic saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride, or mixtures of such salts), calcium carbonate and disintegrating agents such as starch, alginic acids and certain complex silicates combined with lubricants such as magnesium stearate, sodium lauryl sulfate and talc may be used for preparing tablets.
  • lactose and high molecular weight polyethylene glycols When aqueous suspensions are used they can contain emulsifying agents or agents which facilitate suspension. Diluents such as sucrose, ethanol, polyethylene glycol, propylene glycol, glycerol and chloroform or mixtures thereof may also be used.
  • emulsions, suspensions or solutions of the compounds used according to the invention in vegetable oil for example sesame oil, groundnut oil or olive oil, or aqueous-organic solutions such as water and propylene glycol, injectable organic esters such as ethyl oleate, as well as sterile aqueous solutions of the pharmaceutically acceptable salts, are useful.
  • the solutions of the salts of the compounds used according to the invention are especially useful for administration by intramuscular, intravenous, intraarterial or subcutaneous injection or infusion techniques.
  • aqueous solutions also comprising solutions of the salts in pure distilled water, may be used for intravenous administration with the proviso that their pH is suitably adjusted, that they are judiciously buffered and rendered isotonic with a sufficient quantity of glucose or sodium chloride and that they are sterilized by heating, irradiation or microfiltration.
  • the anti-Xa activity compound and platelet aggregation antagonist compound according to the invention may also be formulated in a manner which resists rapid clearance from the vascular (arterial or venous) wall by convection and/or diffusion, thereby increasing the residence time of the composition at the desired site of action.
  • Depot useful according to the invention may be in a copolymer matrix, such as ethylene-vinyl acetate, or a polyvinyl alcohol gel surrounded by a Silastic shell.
  • the anti-Xa activity compound and platelet aggregation antagonist compound may be delivered locally from a silicone polymer implanted in the adventitia.
  • microparticles may be comprised of a variety of synthetic polymers, such as polylactide for example, or natural substances, including proteins or polysaccharides. Such microparticles enable strategic manipulation of variables including total dose of a drug and kinetics of its release. Microparticles can be injected efficiently into the arterial or venous wall through a porous balloon catheter or a balloon over stent, and are retained in the vascular wall and the periadventitial tissue for at least about two weeks. Formulations and methodologies for local, intravascular site-specific delivery of therapeutic agents are discussed in Reissen et al. (J. Am. Coll. Cardiol. 1994; 23: 1234-1244), the entire contents of which are hereby incorporated by reference.
  • the medium for the anti-Xa activity compound and platelet aggregation antagonist compound can also be a hydrogel which is prepared from any biocompatible or non-cytotoxic (homo or hetero) polymer, such as a hydrophilic polyacrylic acid polymer that can act as a drug absorbing sponge.
  • a biocompatible or non-cytotoxic (homo or hetero) polymer such as a hydrophilic polyacrylic acid polymer that can act as a drug absorbing sponge.
  • Such polymers have been described, for example, in application WO93/08845. the entire contents of which are hereby incorporated by reference. Certain of them, such as, in particular, those obtained from ethylene and/or propylene oxide are commercially available.
  • the anti-Xa activity compound and platelet aggregation antagonist compound may be administered directly to the blood vessel wall by means of an angioplasty balloon which is coated with a hydrophilic film (for example a hydrogel), or by means of any other catheter containing an infusion chamber for the compounds, which can thus be applied in a precise manner to the site to be treated.
  • a hydrophilic film for example a hydrogel
  • the percentage of the anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention may be varied.
  • the compounds should constitute a proportion such that a suitable dosage shall be obtained.
  • several unit dosage forms may be administered.
  • the dose employed will be determined by the physician, and depends upon the desired therapeutic effect, the route of administration and the duration of the treatment, and the condition of the patient. In each particular case, the doses will be determined in accordance with the factors distinctive to the subject to be treated, such as age, weight, general state of health and other characteristics which can influence the efficacy of the medicinal product.
  • the dosages of the PAAC are generally from about 0.0001 to about 50, preferably about 0.0001 to about 5, mg/kg body weight per day by inhalation, from about 0.001 to about 100, preferably 0.01 to 70, more especially 0.05 to 10, mg/kg body weight per day by oral administration, and from about 0.0001 to about 10, preferably 0.001 to 1, mg/kg body weight per day by intravenous administration.
  • the dosages of the anti-Xa activity compound according to the invention is particularly useful in doses of about 10 to about 25,000 international units of anti-Xa activity.
  • the anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention may be administered as frequently as necessary in order to obtain the desired therapeutic effect.
  • the dosage regimen in carrying out the method of this invention is that which insures maximum therapeutic response until improvement is obtained and thereafter the minimum effective level which gives relief. Some patients may respond rapidly to a higher or lower dose and may find much weaker maintenance doses adequate. Both short- and long-term treatments regimens are contemplated for the invention.
  • Treatments at the rate of about 1 to about 4 doses per day are also contemplated, in accordance with the physiological requirements of each particular patient, bearing in mind of course, that in selecting the appropriate dosages in any specific case, consideration must be given to the patient's weight, general health, age, and other factors which may influence response to the drug. Thus, for other patients, it will be necessary to prescribe not more than one or two doses per day.
  • the compounds of the present invention may also be formulated for use in conjunction with other therapeutic agents such as agents or in connection with the application of therapeutic techniques to address pharmacological conditions which may be ameliorated through the application of a compound of formula I, such as in the following:
  • the compounds of the present invention may be used in the treatment of restenosis post angioplasty using any device such as balloon, ablation or laser techniques.
  • the compounds of the present invention may be used in the treatment of restenosis following stent placement in the vasculature either as 1) primary treatment for vascular blockage, or 2) in the instance where angioplasty using any device fails to give a patent artery.
  • the compounds of the present invention may be used either orally, by parenteral administration or the compound could be applied topically through the intervention of a specific device or as a properly formulated coating on a stent device.
  • the compounds of the present invention may be used in combination with any antihypertensive agent or cholesterol or lipid regulating agent in the treatment of restenosis or atherosclerosis concurrently with the treatment of high blood pressure or atherosclerosis.
  • agents that are useful in the treatment of high blood pressure include compounds of the following classes; beta-blockers, ACE inhibitors, calcium channel antagonists and alpha-receptor antagonists.
  • agents that are useful in the treatment of elevated cholesterol levels or disregulated lipid levels include compounds known to be HMGCoA reductase inhibitors, compounds of the fibrate class,
  • the anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention exhibit marked pharmacological activities according to tests described in the literature which tests results are believed to correlate to pharmacological activity in humans and other mammals.
  • the following pharmacological test results are typical characteristics of the anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention.
  • the following pharmacological tests evaluate the activity of anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention. Those tests include the hemodynamics measurements during administration of different concentrations of an anti-Xa activity compound, a platelet aggregation antagonist compound and composition thereof over time (FIGS. 1 and 13). More specifically measurements were taken of cyclic flow reduction (FIGS. 2 and 14), activated partial thromboplastin time (FIGS. 3 and 15), prothrombin time (FIGS. 4 and 16), Anti-Xa activity (FIGS. 5 and 17), Anti-IIa activity (FIGS. 6 and 18), template bleeding time (FIGS. 7 and 19). platelet count (FIGS.
  • compositions of the present invention exhibit marked activity in the foregoing tests and are considered useful in the prevention and treatment of thrombosis associated with certain disease states.
  • Antithrombotic activity in the ex vivo canine platelet aggregation assay is predictive of such activity in humans (see, for example Catalfamo, J. L., and Dodds, W. Jean, “Isolation of Platelets from Laboratory Animals”, Methods Enzymol. 169, Part A, 27 (1989)).
  • test protocol as follows is an experimental model of unstable angina.
  • Mongrel dogs of either sex (15-21 kg) are anesthetized with sodium pentobarbital (30 mg/kg, i.v., with supplements given as needed), intubated, and ventilated using a Harvard respirator (Harvard Apparatus. S. Natick, Mass.).
  • a tri-lumen catheter (SAFEDWELLplus, Becton Dickinson, Sandy, Utah) is placed in the right femoral vein for the administration of test agents and supplemental anesthesia.
  • the right femoral artery is cannulated for measurement of arterial blood pressure and for obtaining blood samples.
  • a left thoracotomy is performed at the 5th intercostal space and the heart is suspended in a pericardial cradle.
  • the left circumflex coronary artery (LCX) is isolated and dissected for a distance of 2 cm, ligating side branches when necessary.
  • An electromagnetic flow probe (Carolina Medical Electronics, 501D) is placed on the vessel to monitor coronary blood flow and a snare ligature is placed on the distal portion of the vessel to produce a temporary mechanical occlusion which is used to aid in adjusting the degree of stenosis and to aid in validating zero flow measurements.
  • a Lexan® occluder is positioned for the purpose of creating a critical stenosis, which is confirmed by abolishment of the hyperemic response to a 10 sec mechanical occlusion of the vessel.
  • the endothelium and vascular smooth muscle cells are damaged by compressing the vessel with a vascular clamp. These conditions result in platelet adhesion and aggregation at the damaged area, thus producing a gradual decrease in coronary blood flow.
  • the occluder is moved back and forth over the damaged area to mechanically dislodge the platelet-rich thrombus, thus restoring blood flow.
  • CFRs cyclic flow reductions
  • the compounds are administered as an intravenous bolus only (for N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA) or saline vehicle) or as an intravenous bolus plus a constant intravenous infusion (for heparin, enoxaparin (ENOX), or saline vehicle.
  • BSPBPA N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine
  • ENOX enoxaparin
  • the treatment groups are: I) BSPBPA (30 ⁇ g/kg), II) BSPBPA (300 ⁇ g/kg), III) ENOX (0.5 mg/kg+5 ⁇ g/kg/min) IV) ENOX (0.5 mg/kg+5 ⁇ g/kg/min) plus BSPBPA (30 ⁇ g/kg), V) heparin (60 U/kg+0.7 U/kg/min), and VI) heparin (60 U/kg+0.7 U/kg/min) plus BSPBPA (30 ⁇ g/kg). All compounds are diluted in saline and bolus injections are made using a volume of 5 mL and constant infusions are made using a volume of 22 mL.
  • the compounds for these experiments involves the administration of the following agents as a bolus and a constant infusion (for N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L- ⁇ -cyclohexyl alanine amide (PBGACA) or saline vehicle) or as an intravenous bolus plus a constant intravenous infusion (for heparin, ENOX, or saline vehicle.
  • the treatment groups were: I) PBGACA (10 ⁇ g/kg+0.15 ⁇ g/kg/min).
  • Blood samples (4.5 mL) obtained for analysis of anti-Xa and anti-IIa levels are collected in chilled syringes containing 0.5 mL trisodium citrate and immediately placed on ice. Mean arterial blood pressure, heart rate and EKG are recorded for the duration of the protocol (Grass polygraph, Model 7D, Grass Instruments, Mass.).
  • Activated partial thromboplastin time (APTT) and prothrombin time (PT) are measured using a Microsample Coagulation Analyzer (MCA210, Bio Data Corp, Horsham, Pa.) and Dade® reagents (Thromboplastin-C Plus and Actin® FS Activated PTT reagent, Baxter Diagnostics, Inc., Deerfield. Ill.).
  • the APTT is the most widely used method for monitoring intravenous heparin anticoagulation therapy. It is also a fundamental screening test for deficiencies or abnormalities of the intrinsic coagulation factors: VIII, IX, XI, XII, and factors common to both the intrinsic and extrinsic pathways: I (fibrinogen), II, V, X. When used in conjunction with deficient substrate plasma, the APTT provides the basis for the quantitation of specific coagulation factors.
  • the partial thromboplastin time becomes the “activated” partial thromboplastin time (APTT). Because coagulation endpoints are shorter and sharper than with the PTT, the APTT has proven to be a simple and highly reliable measurement of the intrinsic coagulation mechanism.
  • the prothrombin time is the method for monitoring oral anticoagulation therapy. It is also a fundamental screening test for a deficiency or abnormality of extrinsic coagulation factor VII, and the factors common to both the intrinsic and extrinsic hemostatic pathways: fibrinogen II, V and X. When used in conjunction with deficient substrate plasma, the PT provides the basis for the quantification of specific coagulation factors.
  • tissue thromboplastin The capability of blood to form a fibrin clot by way of the extrinsic hemostatic pathway requires tissue thromboplastin, calcium, factor VII, factor V, factor X, factor II (prothrombin) and factor I (fibrinogen).
  • tissue thromboplastin and calcium are added to a sample of citrated plasma the actions of the intrinsic factors are bypassed and the reaction becomes specific for the coagulation factors involved in the extrinsic and common pathways.
  • Template bleeding time measurements are obtained at the same time points as the blood samples, as mentioned previously. Template bleeding time is measured after a uniform incision is made on the mucous membrane of the inner upper lip with a Surgicutt® automated incision device (ITC, Edison, N.J.). Blood is blotted with Surgicutt® bleeding time blotting paper every 30 seconds, being careful not to disturb the incision site. Template bleeding time is measured from the moment of the incision until the blood no longer stained the blotting paper. Bleeding times of 10 minutes are taken to be maximal.
  • the 1st International LMWH Standard National Institute for Biological Standards and Control, London; anti-Xa activity 168 IU/mg and anti-IIa activity 66.5 IU/mg is used to construct standard curves for measuring heparin and ENOX anti-Xa and anti-IIa activity.
  • the curves are constructed using a four parameter curve-fitting model (Softmax Pro, Molecular Devices, Sunnyvale, Calif.). Values for anti-Xa and anti-IIa activity of heparin and ENOX are reported in International Units.
  • Platelet rich plasma is prepared by centrifugation of the blood samples at 150 x g for 10 min. After removal of the supernatant containing PRP, platelet-poor plasma (PPP) is prepared by centrifugation of the remaining sample at 1000 x g for 10 min. Platelet count is determined with a Coulter ZM or Coulter ZBI particle counter (Coulter Instruments, Hialeah, Fla.). When necessary, platelet count is adjusted to 3 ⁇ 10 8 platelets/mL using autologous PPP. PRP (250 ⁇ L) is incubated at 37° C. while being stirred at 1200 rpm. After preincubation with epinephrine for 1 min (1 ⁇ M.
  • platelet aggregation is induced by adenosine diphosphate (ADP, 10 ⁇ M, Chrono-par 384, Chrono-log Corp. Havertown, Pa.), collagen (equine tendon, 10 ⁇ g/mL, Chrono-par 385, Chrono-log Corp., Havertown, Pa.), arachidonic acid (1 mM, Biodata Corp, Horsham, Pa.) or thrombin (4 Units/mL, Enzyme Research Institute, South Bend, Ind.; plus Gly-Pro-Arg-Pro, a fibrin polymerization inhibitor, 2 mM, Sigma Chemical Co., St.
  • Platelet aggregation is monitored spectrophotometrically with a PAP-4C platelet aggregator (Bio Data Corp, Horsham. Pa.). Results are expressed as a percent inhibition of the rate of aggregation as compared to the pre-drug aggregation response.
  • an anti-Xa activity compound and a platelet aggregation antagonist compound used according to the invention provides for use of those compounds at doses that would be subefficacious if used individually, while effecting the inhibition of repetitive platelet thrombus formation to the same extent as high doses of the platelet aggregation antagonist compound without significantly increasing template bleeding time (as compared to: a >5-fold increase in template bleeding time caused by high-dose of the platelet aggregation antagonist compound, BSPBPA; and a ⁇ 3-fold increase in template bleeding time caused by high-dose of the platelet aggregation antagonist compound, PBGACA).

Abstract

The invention is also directed to pharmaceutical composition comprising a compound having anti-Xa activity, a platelet aggregation antagonist compound and a pharmaceutically acceptable carrier. The invention is also directed to a method of treating or preventing a thrombogenic condition associated with a thrombosis related ischemic disorder in a patient comprising administering to said patient pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound. In addition, this invention is directed to the use of pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound in the preparation of a medicament for treating or preventing a physiological condition associated with thrombosis related ischemic disorder. Furthermore, this invention is directed to a kit for treating or preventing a physiological condition associated with thrombosis related ischemic disorder, said kit comprising a plurality of separate containers, wherein at least one of said containers contains a compound having anti-Xa activity and at least another of said containers contains a platelet aggregation antagonist compound, and said containers optionally contain a pharmaceutical carrier.

Description

  • This application is a continuation application of copending PCT US97/21440, filed Nov. 24, 1997. which designates the United States, which, in turn, is a continuation-in-part application of U.S. patent application Ser. No. 60/031,878, filed Nov. 27, 1996, now abandoned.[0001]
  • FIELD OF THE INVENTION
  • This invention is directed to a pharmaceutical composition comprising an anti-Xa activity compound and a platelet aggregation antagonist compound which exhibits unexpectedly efficacious activity for treating or preventing a physiological condition associated with a thrombosis related ischemic disorder in a patient. The invention is also directed to a method of treating or preventing a thrombogenic condition associated with a thrombosis related ischemic disorder in a patient comprising administering pharmaceutically effective amounts of an anti-Xa activity compound and a platelet aggregation antagonist compound. [0002]
  • Platelet fibrinogen receptor antagonists have been shown to be effective agents for inhibiting platelet-dependent thrombus formation in animal models of coronary thrombosis. Likewise, results from clinical studies have shown that platelet fibrinogen receptor antagonists reduce the composite incidence of major ischemic events when administered to high-risk patients undergoing percutaneous transluminal coronary angioplasty. However, the therapeutic window is very narrow for this class of compounds, in part, because the high degree of inhibition of ex vivo platelet aggregation required for antithrombotic efficacy is often associated with a marked increase in template bleeding time, which is a marker of an undesirable bleeding complications. [0003]
  • Low molecular weight heparins (LMWHs) and heparinoid compounds (HCs) have been used effectively over the past few years for the prevention and treatment of venous thrombosis and the associated thromboembolism. However, LMWHs and HCs are gradually pervading into the treatment repertoire for arterial thrombotic indications. Preliminary results favoring the use of LMWH or HCs over unfractionated heparin in arterial thrombotic indications are supported by several pharmacodynamic, pharmacokinetic, and mechanistic differences between these two classes of compounds. For example, reliable and safe anticoagulation can be achieved with LMWHs or HCs by subcutaneous dosing without monitoring. Compared to heparin, LMWHs and HCs have a higher bioavailability, a relatively long half-life, and appear to have a safer profile. In addition, LMWHs are more resistant than heparin to neutralization by platelet factor 4, which is released from activated platelets, presumably at the site of arterial thrombosis. [0004]
  • Haemostasis the biochemistry of blood coagulation, is an extremely complex phenomena whereby normal whole blood and body tissue spontaneously arrest bleeding from injured blood vessels. Effective haemostasis requires the combined activity of vascular, platelet and plasma factors as well as a controlling mechanism to prevent excessive clotting. Defects, deficiencies, or excesses of any of these components can lead to hemorrhagic or thrombotic consequences. [0005]
  • Platelet adhesion, spreading and aggregation on extracellular matrices are central events in thrombus formation. These events are mediated by a family of adhesive glycoproteins, i.e., fibrinogen, fibronectin, and von Willebrand factor. Fibrinogen is a co-factor for platelet aggregation while fibronectin supports platelet attachments and spreading reactions, and von Willebrand factor is important in platelet attachment to and spreading on subendothelial matrices. The binding sites for fibrinogen, fibronectin and von Willebrand factor have been located on the platelet membrane protein complex known as glycoprotein IIb/IIIa. [0006]
  • Adhesive glycoproteins, like fibrinogen, do not bind with normal resting platelets. However, when a platelet is activated with an agonist such as thrombin or adenosine diphosphate, the platelet changes its shape, perhaps making the GPIIb/IIIa binding site accessible to fibrinogen. Blocking the fibrinogen receptor, thus inhibits platelet aggregation and subsequent thrombus formation, and is useful for the prevention and treatment of pathological thrombogenic conditions, such as stroke, peripheral arterial occlusive disease, disseminated intravascular coagulation, and acute coronary syndromes such as unstable angina and myocardial infarction. [0007]
  • SUMMARY OF THE INVENTION
  • The invention is also directed to pharmaceutical composition comprising a compound having anti-Xa activity, a platelet aggregation antagonist compound and a pharmaceutically acceptable carrier. The invention is also directed to a method of treating or preventing a thrombogenic condition associated with a thrombosis related ischemic disorder in a patient comprising administering to said patient pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound.[0008]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 represents information regarding assorted blood sampling/hemodynamic measurements during administration of different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA), and composition thereof over time. [0009]
  • FIG. 2 represents a graph of the number of cyclic flow reductions (CFRs) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0010]
  • FIG. 3 represents a graph of the activated partial thromboplastin time (APTT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0011]
  • FIG. 4 represents a graph of the prothrombin time (PT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0012]
  • FIG. 5 represents a graph of the Anti-Xa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0013]
  • FIG. 6 represents a graph of the Anti-IIa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0014]
  • FIG. 7 represents a graph of the template bleeding time for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0015]
  • FIG. 8 represents a graph of the platelet count for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0016]
  • FIG. 9 represents a graph of collagen-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0017]
  • FIG. 10 represents a graph of ADP-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0018]
  • FIG. 11 represents a graph of arachidonic acid-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0019]
  • FIG. 12 represents a graph of thrombin-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, BSPBPA, and composition thereof over time. [0020]
  • FIG. 13 represents information regarding assorted blood sampling/hemodynamic measurements during administration of different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L-β-cyclohexyl alanine amide (PBGACA) and composition thereof over time. [0021]
  • FIG. 14 represents a graph of the number of cyclic flow reductions (CFRs) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0022]
  • FIG. 15 represents a graph of the activated partial thromboplastin time (APTT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0023]
  • FIG. 16 represents a graph of the prothrombin time (PT) for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0024]
  • FIG. 17 represents a graph of the Anti-Xa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0025]
  • FIG. 18 represents a graph of the Anti-IIa activity for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0026]
  • FIG. 19 represents a graph of the template bleeding time for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound. PBGACA, and composition thereof over time. [0027]
  • FIG. 20 represents a graph of the platelet count for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0028]
  • FIG. 21 represents a graph of collagen-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0029]
  • FIG. 22 represents a graph of ADP-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0030]
  • FIG. 23 represents a graph of arachidonic acid-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time. [0031]
  • FIG. 24 represents a graph of thrombin-induced ex-vivo platelet aggregation for different concentrations of the anti-Xa activity compound, Enoxaparin, the platelet aggregation antagonist compound, PBGACA, and composition thereof over time.[0032]
  • DETAILED DESCRIPTION OF THE INVENTION
  • As used above, and throughout the description of the invention, the following terms, unless otherwise indicated, shall be understood to have the following meanings: [0033]
  • Definitions [0034]
  • “Patient” includes both human and other mammals. [0035]
  • “Effective amount” is meant to describe an amount of composition according to the present invention effective in producing the desired therapeutic effect. [0036]
  • “Platelet aggregation antagonist compound” (PAAC) means a compound that binds to the platelet GPIIb/IIIa receptor (GPIIb/IIIa receptor antagonist) and competitively inhibits binding of fibrinogen, fibronectin and von Willebrand factor as well as inhibits aggregation of activated platelets. [0037]
  • “Anti-Xa activity compound” means a heparinoid compound or low molecular weight heparin (LMWH), or synthetic derivatives thereof. [0038]
  • Preferred Embodiments [0039]
  • According to a preferred embodiment of the invention, the following references, incorporated herein by reference, describes useful PAACs: Lynch et al. J. Pharm. Expt. Thera. 272(1) 20 (1995); Kereiakes et al. JACC 27(3), 536 (1996); Peerlinck et al. Circulation 88(4), 1512 (1993); Barrett et al. Clin. Pharmacol. Ther. 56(4) 377 (1994); Cook et al. Thromb. Haemostas. 70(5), 838 (1993): Plow, et al.. Proc. Natl. Acad. Sci. USA 82, 8057-61 (1985); Ruggeri, et al., Proc. Natl. Acad. Sci. USA 5708-12 (1986); Ginsberg, et al., J. Biol. Chem. 260, 3931-36 (1985); and Gartner, et al., J. Biol. Chem. 260, 11,891-94 (1987); Plow, E. F., et al., Proc. Natl. Acad. Sci. USA 79, 3711-3715 (1982); Tjoeng. et al., U.S. Pat. Nos. 5,037,808, 4,879,313 and 4,992,463; Adams, et al., U.S. Pat. No. 4,857,508; Haverstick, D. M., et al., Blood 66(4), 946-952 (1985): Topol et al.. The Lancet, 343, 881 (1994), French Application No. 86/17507; Zimmerman, et al., U.S. Pat. No. 4,683,291; European Application Publication No. 0 319 506; U.S. Pat. No. 5,023,233; U.S. Pat. No. 4,952.562; International Publication No. WO 91/04746; U.S. application Ser. No. 5,086,069; International Publication No. WO 92/13117; U.S. Pat. No. 5,053,392; U.S. Pat. No. 5,064,814; U.S. Pat. No. 5,051,405; European Patent Application 0479,481; European Patent Application 0478,362; U.S. Pat. No. 5,292,756; International Publication No. WO 95/10295; and International Publication No. WO 89/11538. More preferred PAACs are those disclosed in International Publication No. WO 89/11538, International Publication No. WO 95/10295 or U.S. Pat. No. 5,292,756; further preferred are Reopro® (abciximab). N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]asparty]-L-β-cyclohexyl alanine, N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]-L-β-cyclohexyl alanine amide (PBGACA) or N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA). [0040]
  • According to a preferred embodiment of the invention, the following references, incorporated herein by reference, describes useful LMWHs according to the invention and methods for preparing the LMWHs: Eur. Pat. No. 0014184; Medicinal Research Reviews 12(4), 373 (1992); Drugs of the Future 12(1), 45 (1987); International Publication No. WO 92/19249; U.S. Pat. No. 4,692,435; Barrowcliffe, Thromb. Res. 12, 27-36 (1977); Eur. Pat. Appln. No. 37319; Eur. Pat. Appln. No. 76279; U.S. Pat. No. 4,804,652; WO81/3276; Eur. Pat. Appln. No. 244235, Eur. Pat. Appln. No. 244236; U.S. Pat. No. 4,486,420; U.S. Pat. No. 4,692,435; U.S. Pat. No. 4,826,827; U.S. Pat. No. 3,766,167; Eur. Pat. Appln. No. 40144; Eur. Pat. Appln. No.347588, Eur. Pat. Appln. No. 380943; U.S. Pat. No. 4,533,549; U.S. Pat. No. 4,629,699; Eur. Pat. Appln. No. 269981. For example, an anti-Xa activity compound may be produced as follows: enrichment by fractionation by ethanol and/or molecular sieving. e.g., gel filtration or membrane filtration of the LMWH present in standard heparin and controlled chemical (by nitrous acid, β-elimination or periodate oxidation) or enzymatic (by heparinases) depolymerization. The conditions for depolymerization can be carefully controlled to yield products of desired molecular weights. Nitrous acid depolymerization is commonly used. Also employed is depolymerization of the benzylic ester of heparin by β-elimination, which yields the same type of fragments as enzymatic depolymerization using heparinases. LMWH with low anticoagulant activity and retaining basic chemical structure are prepared by depolymerization using periodate oxidation or by removing the antithrombin-binding fraction of LMWH, or prepared by other methods, using immobilized antithrombin for adsorption. [0041]
  • Further preferred is a LMWH having an average molecular weight of about 3000 to about 6500. Commercially available LMWHs useful according to the invention include the following: Clexane®/Klexane®/Lovenox® (Enoxaparin (ENOX)) having an average molecular mass of 4500±1000 Dalton (Da), molecular mass distribution comprising components of <2000 Da (16.0±4.0%) and 2000 to 8000 Da (78.0±10.0%). Anti-Xa activity (IU/mg on dry basis) of 90 to 125, and an Anti-Xa/Anti-IIa ratio of 3.3 to 5.3; Fraxiparin® (Nardroparin) having an average molecular mass of 4300±700 Da, molecular mass distribution comprising components of <2000 Da (<15%), 2000 to 4000 Da (45±10%) and 2000 to 8000 Da (85±10%), Anti-Xa activity (IU/mg on dry basis) of 95 to 130, and an Anti-Xa/Anti-IIa ratio of 2.5 to 4.0; Fragmin® (Dalteparin) having an average molecular mass of 6000±400 Da. molecular mass distribution comprising components of <3000 Da (5.0 to 13.0%) and >8000 Da (15.0 to 25.0%). Anti-Xa activity (IU/mg on dry basis) of 110 to 210, and an Anti-Xa/Anti-IIa ratio of 1.9 to 3.2; Embolex®/Monoembolex® (Certroparin) having an average molecular mass of 5200±1000 Da, molecular mass distribution comprising components of <2000 Da (10 to 25%) and <8000 Da (75 to 90%), Anti-Xa activity (IU/mg on dry basis) of 80 to 120, and an Anti-Xa/Anti-IIa ratio of 1.5 to 2.5; Fluxum®/Minidalton®/Lowhepa® (Parnaparin) having an average molecular mass of 5000±1000 Da, molecular mass distribution comprising components of <3000 Da (20 to 30%) and 3000 to 8000 Da (50 to 60%), Anti-Xa activity (IU/mg on dry basis) of 75 to 110, and an Anti-Xa/Anti-IIa ratio of 2.0 to 3.0; Logiparin® (Tinzaparin) having an average molecular mass of 3400 to 5600 Da, molecular mass distribution comprising components of <2000 Da (2.0 to 16.0%), 2000 to 4000 Da (66.0±6.0%) and >8000 Da (12.0 to 38.0%), Anti-Xa activity (IU/mg on dry basis) of>70, and an Anti-Xa/Anti-IIa ratio of 1.5 to 2.5; Clivarine® (Reviparin) and Normiflo® (ardeparin/RD heparin/RDH). A preferred LMWH according to the invention is prepared according to the procedures disclosed in U.S. Pat. No. 4,486,420 or U.S. Pat. No. 4,692,435; more preferred enoxaparin. [0042]
  • According to another preferred embodiment of the invention, an anti-Xa activity compound is a heparinoid compound. The following references, incorporated herein by reference, describe a useful heparinoid compound according to the invention and methods for preparing the heparinoid compound: U.S. Pat. No. 4,438,108, Eur. Pat. No. EP 66908, Zammit et al. Thromb. Haemostas. 71(6), 759 (1994), and Gent et al. Circulation 93, 80 (1996). A preferred commercially available heparinoid compound useful according to the invention is Orgaran® (Danaparoid) having an average molecular mass of about 6500 Da, Anti-Xa activity (IU/mg on dry basis) of about 10, and an Anti-Xa/Anti-IIa ratio of about 28. [0043]
  • Preferred thrombogenic conditions treatable or preventable according to the invention include stroke, atherosclerosis, angiogenesis, thrombosis, thromboembolic conditions such as deep venous thrombosis. pulmonary embolism or thrombophlebitis, disseminated intravascular coagulation or thromboembolic syndromes associated with cancer, sepsis or obstetrical complications, peripheral arterial occlusive disease, and acute coronary syndromes such as unstable angina and myocardial infarction. hemodialysis, or extra-corporeal circulation requirement associated with a surgical procedure or tissue damage caused by phospholipases A2 (PLA[0044] 2), more preferred is unstable angina and myocardial infarction.
  • Another preferred embodiment according to the invention useful in the course of a medical procedure wherein there is a potential for an occurrence pathological thrombogenic condition may occur, such as during coronary artery bypass surgery or percutaneous transluminal coronary angioplasty with or without placement of an intracoronary stent; more preferred is percutaneous transluminal coronary angioplasty with or without placement of an intracoronary stent. [0045]
  • Another preferred embodiment according to the invention is the use of pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound in the preparation of a medicament for treating or preventing a physiological condition associated with thrombosis related ischemic disorder. [0046]
  • In the treatment or prevention method according to the invention the anti-Xa activity compound and platelet aggregation antagonist compound may be administered in different ways, such as in combination therapies optionally employing medical procedures. For example, the anti-Xa activity compound and platelet aggregation antagonist compound may be administered to a patient concomitantly or at different times provided that they are administered such that at some period of time there are pharmaceutically effective amounts of both compounds present in the patient such that a therapeutic effect according to the invention results. [0047]
  • Thus, it is a further object of the invention to provide a kit for treating or preventing a physiological condition associated with thrombosis related ischemic disorder, said kit comprising a plurality of separate containers, wherein at least one of said containers contains a compound having anti-Xa activity and at least another of said containers contains a platelet aggregation antagonist compound, and said containers optionally contain a pharmaceutical carrier which kit may be effectively utilized for carrying out combination therapies according to the invention. A further embodiment for a kit would be wherein of said containers at least one of said containers should contain the compound having anti-Xa activity without the presence of the platelet aggregation antagonist compound, and at least another of said containers should contain the platelet aggregation antagonist compound without the presence of the compound having anti-Xa activity. [0048]
  • In practice, the anti-Xa activity compound and platelet aggregation antagonist compound may be administered parenterally, topically, rectally, transdermally, intrapulmonary or orally, but they are preferably administered parenterally and/or orally. [0049]
  • Suitable compositions containing the compounds used according to the invention may be prepared by conventional means. For example, the compounds used according to the invention may be dissolved or suspended in a suitable carrier. [0050]
  • The compounds used according to the invention should be presented in forms permitting administration by the most suitable route, and the invention also relates to a pharmaceutical composition containing the compounds used according to the invention which are suitable for use in human or veterinary medicine. These compositions may be prepared according to the customary methods, using one or more pharmaceutically acceptable carrier, which comprise adjuvants or excipients. The adjuvants comprise, inter alia, diluents, sterile aqueous media and the various non-toxic organic solvents. The compositions may be presented in the form of tablets, pills, capsules, lozenges, troches, hard candies, granules, powders, aqueous solutions or suspensions, injectable solutions, elixirs or syrups, powders, solution or suspension for intrapulmonary administration and can contain one or more agents chosen from the group comprising sweeteners, flavorings, colorings, or stabilizers in order to obtain pharmaceutically acceptable preparations. [0051]
  • The choice of vehicle and the content of compounds used according to the invention in the vehicle are generally determined in accordance with the solubility and chemical properties of the compounds, the particular mode of administration and the provisions to be observed in pharmaceutical practice. For example, excipients such as sterile water, Ringer's solution, lactose, sodium citrate, isotonic saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride, or mixtures of such salts), calcium carbonate and disintegrating agents such as starch, alginic acids and certain complex silicates combined with lubricants such as magnesium stearate, sodium lauryl sulfate and talc may be used for preparing tablets. To prepare a capsule, it is advantageous to use lactose and high molecular weight polyethylene glycols. When aqueous suspensions are used they can contain emulsifying agents or agents which facilitate suspension. Diluents such as sucrose, ethanol, polyethylene glycol, propylene glycol, glycerol and chloroform or mixtures thereof may also be used. [0052]
  • For parenteral administration, emulsions, suspensions or solutions of the compounds used according to the invention in vegetable oil, for example sesame oil, groundnut oil or olive oil, or aqueous-organic solutions such as water and propylene glycol, injectable organic esters such as ethyl oleate, as well as sterile aqueous solutions of the pharmaceutically acceptable salts, are useful. The solutions of the salts of the compounds used according to the invention are especially useful for administration by intramuscular, intravenous, intraarterial or subcutaneous injection or infusion techniques. The aqueous solutions, also comprising solutions of the salts in pure distilled water, may be used for intravenous administration with the proviso that their pH is suitably adjusted, that they are judiciously buffered and rendered isotonic with a sufficient quantity of glucose or sodium chloride and that they are sterilized by heating, irradiation or microfiltration. [0053]
  • The anti-Xa activity compound and platelet aggregation antagonist compound according to the invention may also be formulated in a manner which resists rapid clearance from the vascular (arterial or venous) wall by convection and/or diffusion, thereby increasing the residence time of the composition at the desired site of action. Depot useful according to the invention may be in a copolymer matrix, such as ethylene-vinyl acetate, or a polyvinyl alcohol gel surrounded by a Silastic shell. Alternatively, the anti-Xa activity compound and platelet aggregation antagonist compound may be delivered locally from a silicone polymer implanted in the adventitia. [0054]
  • An alternative approach for minimizing washout of the anti-Xa activity compound and platelet aggregation antagonist compound during percutaneous, transvascular delivery comprises the use of nondiffusible, drug-eluting microparticles. The microparticles may be comprised of a variety of synthetic polymers, such as polylactide for example, or natural substances, including proteins or polysaccharides. Such microparticles enable strategic manipulation of variables including total dose of a drug and kinetics of its release. Microparticles can be injected efficiently into the arterial or venous wall through a porous balloon catheter or a balloon over stent, and are retained in the vascular wall and the periadventitial tissue for at least about two weeks. Formulations and methodologies for local, intravascular site-specific delivery of therapeutic agents are discussed in Reissen et al. (J. Am. Coll. Cardiol. 1994; 23: 1234-1244), the entire contents of which are hereby incorporated by reference. [0055]
  • The medium for the anti-Xa activity compound and platelet aggregation antagonist compound can also be a hydrogel which is prepared from any biocompatible or non-cytotoxic (homo or hetero) polymer, such as a hydrophilic polyacrylic acid polymer that can act as a drug absorbing sponge. Such polymers have been described, for example, in application WO93/08845. the entire contents of which are hereby incorporated by reference. Certain of them, such as, in particular, those obtained from ethylene and/or propylene oxide are commercially available. [0056]
  • In addition, the anti-Xa activity compound and platelet aggregation antagonist compound may be administered directly to the blood vessel wall by means of an angioplasty balloon which is coated with a hydrophilic film (for example a hydrogel), or by means of any other catheter containing an infusion chamber for the compounds, which can thus be applied in a precise manner to the site to be treated. [0057]
  • The percentage of the anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention may be varied. The compounds should constitute a proportion such that a suitable dosage shall be obtained. Obviously, several unit dosage forms may be administered. The dose employed will be determined by the physician, and depends upon the desired therapeutic effect, the route of administration and the duration of the treatment, and the condition of the patient. In each particular case, the doses will be determined in accordance with the factors distinctive to the subject to be treated, such as age, weight, general state of health and other characteristics which can influence the efficacy of the medicinal product. [0058]
  • In the adult, the dosages of the PAAC are generally from about 0.0001 to about 50, preferably about 0.0001 to about 5, mg/kg body weight per day by inhalation, from about 0.001 to about 100, preferably 0.01 to 70, more especially 0.05 to 10, mg/kg body weight per day by oral administration, and from about 0.0001 to about 10, preferably 0.001 to 1, mg/kg body weight per day by intravenous administration. [0059]
  • In the adult, the dosages of the anti-Xa activity compound according to the invention is particularly useful in doses of about 10 to about 25,000 international units of anti-Xa activity. [0060]
  • The anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention may be administered as frequently as necessary in order to obtain the desired therapeutic effect. The dosage regimen in carrying out the method of this invention is that which insures maximum therapeutic response until improvement is obtained and thereafter the minimum effective level which gives relief. Some patients may respond rapidly to a higher or lower dose and may find much weaker maintenance doses adequate. Both short- and long-term treatments regimens are contemplated for the invention. Treatments at the rate of about 1 to about 4 doses per day are also contemplated, in accordance with the physiological requirements of each particular patient, bearing in mind of course, that in selecting the appropriate dosages in any specific case, consideration must be given to the patient's weight, general health, age, and other factors which may influence response to the drug. Thus, for other patients, it will be necessary to prescribe not more than one or two doses per day. [0061]
  • The compounds of the present invention may also be formulated for use in conjunction with other therapeutic agents such as agents or in connection with the application of therapeutic techniques to address pharmacological conditions which may be ameliorated through the application of a compound of formula I, such as in the following: [0062]
  • The compounds of the present invention may be used in the treatment of restenosis post angioplasty using any device such as balloon, ablation or laser techniques. The compounds of the present invention may be used in the treatment of restenosis following stent placement in the vasculature either as 1) primary treatment for vascular blockage, or 2) in the instance where angioplasty using any device fails to give a patent artery. The compounds of the present invention may be used either orally, by parenteral administration or the compound could be applied topically through the intervention of a specific device or as a properly formulated coating on a stent device. [0063]
  • The compounds of the present invention may be used in the treatment of restenosis in combination with any anticoagulant, antiplatelet, antithrombotic or profibrinolytic agent. Often patients are concurrently treated prior, during and after interventional procedures with agents of these classes either in order to safely perform the interventional procedure or to prevent deleterious effects of thrombus formation. Some examples of classes of agents known to be anticoagulant, antiplatelet, antithrombotic or profibrinolytic agents include any formulation of thrombin inhibitors or Factor VIIa inhibitors. Some examples of classes of agents known to be anticoagulant, antiplatelet, antithrombotic or profibrinolytic agents include any formulation of aspirin, direct thrombin inhibitors, direct Factor Xa inhibitors, or Factor VIIa inhibitors. [0064]
  • The compounds of the present invention may be used in combination with any antihypertensive agent or cholesterol or lipid regulating agent in the treatment of restenosis or atherosclerosis concurrently with the treatment of high blood pressure or atherosclerosis. Some examples of agents that are useful in the treatment of high blood pressure include compounds of the following classes; beta-blockers, ACE inhibitors, calcium channel antagonists and alpha-receptor antagonists. Some examples of agents that are useful in the treatment of elevated cholesterol levels or disregulated lipid levels include compounds known to be HMGCoA reductase inhibitors, compounds of the fibrate class, [0065]
  • The anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention exhibit marked pharmacological activities according to tests described in the literature which tests results are believed to correlate to pharmacological activity in humans and other mammals. The following pharmacological test results are typical characteristics of the anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention. [0066]
  • The following pharmacological tests evaluate the activity of anti-Xa activity compound and platelet aggregation antagonist compound used according to the invention. Those tests include the hemodynamics measurements during administration of different concentrations of an anti-Xa activity compound, a platelet aggregation antagonist compound and composition thereof over time (FIGS. 1 and 13). More specifically measurements were taken of cyclic flow reduction (FIGS. 2 and 14), activated partial thromboplastin time (FIGS. 3 and 15), prothrombin time (FIGS. 4 and 16), Anti-Xa activity (FIGS. 5 and 17), Anti-IIa activity (FIGS. 6 and 18), template bleeding time (FIGS. 7 and 19). platelet count (FIGS. 8 and 20), collagen-induced ex-vivo platelet aggregation (FIGS. 9 and 21), ADP-induced ex-vivo platelet aggregation (FIGS. 10 and 22), arachidonic acid-induced ex-vivo platelet aggregation (FIGS. 11 and 23), and thrombin-induced ex-vivo platelet aggregation (FIGS. 12 and 24). [0067]
  • Compositions of the present invention exhibit marked activity in the foregoing tests and are considered useful in the prevention and treatment of thrombosis associated with certain disease states. Antithrombotic activity in the ex vivo canine platelet aggregation assay is predictive of such activity in humans (see, for example Catalfamo, J. L., and Dodds, W. Jean, “Isolation of Platelets from Laboratory Animals”, Methods Enzymol. 169, Part A, 27 (1989)). [0068]
  • Materials and Methods
  • All procedures in this study are performed in compliance with the Animal Welfare Act Regulations and with the Guide for the Care and Use of Laboratory Animals (DHEW Publication No. NIH 85-23, 1985). [0069]
  • The test protocol as follows is an experimental model of unstable angina. [0070]
  • Mongrel dogs of either sex (15-21 kg) are anesthetized with sodium pentobarbital (30 mg/kg, i.v., with supplements given as needed), intubated, and ventilated using a Harvard respirator (Harvard Apparatus. S. Natick, Mass.). A tri-lumen catheter (SAFEDWELLplus, Becton Dickinson, Sandy, Utah) is placed in the right femoral vein for the administration of test agents and supplemental anesthesia. The right femoral artery is cannulated for measurement of arterial blood pressure and for obtaining blood samples. [0071]
  • A left thoracotomy is performed at the 5th intercostal space and the heart is suspended in a pericardial cradle. The left circumflex coronary artery (LCX) is isolated and dissected for a distance of 2 cm, ligating side branches when necessary. An electromagnetic flow probe (Carolina Medical Electronics, 501D) is placed on the vessel to monitor coronary blood flow and a snare ligature is placed on the distal portion of the vessel to produce a temporary mechanical occlusion which is used to aid in adjusting the degree of stenosis and to aid in validating zero flow measurements. [0072]
  • Distal to the flow probe, a Lexan® occluder is positioned for the purpose of creating a critical stenosis, which is confirmed by abolishment of the hyperemic response to a 10 sec mechanical occlusion of the vessel. The endothelium and vascular smooth muscle cells are damaged by compressing the vessel with a vascular clamp. These conditions result in platelet adhesion and aggregation at the damaged area, thus producing a gradual decrease in coronary blood flow. When flow reaches zero, the occluder is moved back and forth over the damaged area to mechanically dislodge the platelet-rich thrombus, thus restoring blood flow. This repetitive pattern of decreasing blood flow that is restored by mechanical disruption of the platelet thrombus is referred to as cyclic flow reductions (CFRs). The antithrombotic effect of the test agents is quantitated by comparing the number of CFRs that occurred during a 20 min control period with the number of CFRs per 20 min for three consecutive 20 min periods after drug administration. A significant reduction in the number of CFRs is taken to represent an antithrombotic effect. [0073]
  • Experimental Protocol [0074]
  • a. Protocol using BSPBPA [0075]
  • Thirty dogs are assigned to one of six adjunctive treatment groups. The compounds are administered as an intravenous bolus only (for N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine (BSPBPA) or saline vehicle) or as an intravenous bolus plus a constant intravenous infusion (for heparin, enoxaparin (ENOX), or saline vehicle. The treatment groups are: I) BSPBPA (30 μg/kg), II) BSPBPA (300 μg/kg), III) ENOX (0.5 mg/kg+5 μg/kg/min) IV) ENOX (0.5 mg/kg+5 μg/kg/min) plus BSPBPA (30 μg/kg), V) heparin (60 U/kg+0.7 U/kg/min), and VI) heparin (60 U/kg+0.7 U/kg/min) plus BSPBPA (30 μg/kg). All compounds are diluted in saline and bolus injections are made using a volume of 5 mL and constant infusions are made using a volume of 22 mL. [0076]
  • a. Protocol using PBGACA [0077]
  • Thirty dogs are assigned to one of six adjunctive treatment groups. The compounds for these experiments involves the administration of the following agents as a bolus and a constant infusion (for N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L-β-cyclohexyl alanine amide (PBGACA) or saline vehicle) or as an intravenous bolus plus a constant intravenous infusion (for heparin, ENOX, or saline vehicle. The treatment groups were: I) PBGACA (10 μg/kg+0.15 μg/kg/min). II) PBGACA (30 μg/kg+0.15 μg/kg/min), III) ENOX (0.5 mg/kg+5 μg/kg/min) IV) ENOX (0.5 mg/kg+5 μg/kg/min) plus PBGACA (10 μg/kg+0.15 μg/kg/min), V) heparin (60 U/kg+0.7 U/kg/min), and VI) heparin (60 U/kg+PBGACA (10 μg/kg+0.15 μg/kg/min),. All compounds are diluted in saline and bolus injections are made using a volume of 5 mL and constant infusions were made using a volume of 22 mL. [0078]
  • After consistent CFRs are established for at least 20 minutes, compounds are administered as described above and blood flow is monitored during the one hour infusion period. Arterial blood samples are drawn before administration of test agents (control sample) and at 5, 10, 30, and 60 min after administration of compounds. Blood samples are drawn on {fraction (1/10)} volume of 3.8% trisodium citrate and are used for ex vivo platelet aggregation, platelet count, anti-Xa levels, anti-IIa levels, and coagulation time assays (activated partial thromboplastin time, APTT, and prothrombin time, PT). Blood samples (4.5 mL) obtained for analysis of anti-Xa and anti-IIa levels are collected in chilled syringes containing 0.5 mL trisodium citrate and immediately placed on ice. Mean arterial blood pressure, heart rate and EKG are recorded for the duration of the protocol (Grass polygraph, Model 7D, Grass Instruments, Mass.). [0079]
  • Coagulation times and template bleeding time. Activated partial thromboplastin time (APTT) and prothrombin time (PT) are measured using a Microsample Coagulation Analyzer (MCA210, Bio Data Corp, Horsham, Pa.) and Dade® reagents (Thromboplastin-C Plus and Actin® FS Activated PTT reagent, Baxter Diagnostics, Inc., Deerfield. Ill.). [0080]
  • The APTT is the most widely used method for monitoring intravenous heparin anticoagulation therapy. It is also a fundamental screening test for deficiencies or abnormalities of the intrinsic coagulation factors: VIII, IX, XI, XII, and factors common to both the intrinsic and extrinsic pathways: I (fibrinogen), II, V, X. When used in conjunction with deficient substrate plasma, the APTT provides the basis for the quantitation of specific coagulation factors. [0081]
  • The intrinsic capability of blood to form a fibrin clot requires coagulation factors XII, XI, IX, VIII, X, V, II (prothrombin), fibrinogen, platelet lipid, and calcium. [0082]
  • By adding a substance to activate factors XII and XI, the contact factors, the partial thromboplastin time becomes the “activated” partial thromboplastin time (APTT). Because coagulation endpoints are shorter and sharper than with the PTT, the APTT has proven to be a simple and highly reliable measurement of the intrinsic coagulation mechanism. [0083]
  • TEST PROCEDURE
  • 1. Preincubate 0.025 M Calcium Chloride to 37° C. [0084]
  • 2. Pipette 0.1 mL of reconstituted APTT reagent into a test cuvette. [0085]
  • 3. Add 0.1 mL test or control plasma. [0086]
  • 4. Incubate at 37° C. for 5 minutes exactly. [0087]
  • 5. Add 0.1 mL preincubated Calcium Chloride, simultaneously starting the timer. [0088]
  • 6. Record the clotting time. [0089]
  • The prothrombin time is the method for monitoring oral anticoagulation therapy. It is also a fundamental screening test for a deficiency or abnormality of extrinsic coagulation factor VII, and the factors common to both the intrinsic and extrinsic hemostatic pathways: fibrinogen II, V and X. When used in conjunction with deficient substrate plasma, the PT provides the basis for the quantification of specific coagulation factors. [0090]
  • The capability of blood to form a fibrin clot by way of the extrinsic hemostatic pathway requires tissue thromboplastin, calcium, factor VII, factor V, factor X, factor II (prothrombin) and factor I (fibrinogen). When tissue thromboplastin and calcium are added to a sample of citrated plasma the actions of the intrinsic factors are bypassed and the reaction becomes specific for the coagulation factors involved in the extrinsic and common pathways. [0091]
  • TEST PROCEDURE [0092]
  • 1. Preincubate reconstituted PT reagent to 37° C. [0093]
  • 2. Pipette 0.1 mL of test or control plasma into a test cuvette. [0094]
  • 3. Incubate at 37° C. for at least 2 minutes, but not more than 10 minutes. [0095]
  • 4. Inject 0.2 mL of the preincubated reagent, simultaneously starting the timer. [0096]
  • 5. Record the clotting time. [0097]
  • Template bleeding time measurements are obtained at the same time points as the blood samples, as mentioned previously. Template bleeding time is measured after a uniform incision is made on the mucous membrane of the inner upper lip with a Surgicutt® automated incision device (ITC, Edison, N.J.). Blood is blotted with Surgicutt® bleeding time blotting paper every 30 seconds, being careful not to disturb the incision site. Template bleeding time is measured from the moment of the incision until the blood no longer stained the blotting paper. Bleeding times of 10 minutes are taken to be maximal. [0098]
  • Anti-Xa and anti-IIa activity [0099]
  • These samples are centrifuiged at 1500 x g for 10 min at 4° C. The plasma is removed and stored at −70° C. until assayed. Anti-Xa and Anti-IIa activity are analyzed by chromogenic methods using kits supplied by American Diagnostica (actichrome® Heparin and actichrome® Heparin anti-IIa, Greenwich, Conn.), with minor modifications. Incubations and reactions are performed at 37° C. Amidolytic activity (milli-optical units, or mOD) is determined using a SPECTRAmax microtiter plate spectrophotometer and Softmax Pro software (Molecular Devices Corp., Sunnyvale, Calif.). The 1st International LMWH Standard (National Institute for Biological Standards and Control, London; anti-Xa activity 168 IU/mg and anti-IIa activity 66.5 IU/mg) is used to construct standard curves for measuring heparin and ENOX anti-Xa and anti-IIa activity. The curves are constructed using a four parameter curve-fitting model (Softmax Pro, Molecular Devices, Sunnyvale, Calif.). Values for anti-Xa and anti-IIa activity of heparin and ENOX are reported in International Units. [0100]
  • Platelet aggregation [0101]
  • The Inhibition of ex-vivo Platelet Aggregation assay is based on that of Zucker, “Platelet Aggregation Measured by the Photoelectric Method”, Methods in Enzymology 169, 117-133 (1989). [0102]
  • Platelet rich plasma (PRP) is prepared by centrifugation of the blood samples at 150 x g for 10 min. After removal of the supernatant containing PRP, platelet-poor plasma (PPP) is prepared by centrifugation of the remaining sample at 1000 x g for 10 min. Platelet count is determined with a Coulter ZM or Coulter ZBI particle counter (Coulter Instruments, Hialeah, Fla.). When necessary, platelet count is adjusted to 3×10[0103] 8 platelets/mL using autologous PPP. PRP (250 μL) is incubated at 37° C. while being stirred at 1200 rpm. After preincubation with epinephrine for 1 min (1 μM. Chrono-par 393, Chrono-log Corp., Havertown, Pa.), platelet aggregation is induced by adenosine diphosphate (ADP, 10 μM, Chrono-par 384, Chrono-log Corp. Havertown, Pa.), collagen (equine tendon, 10 μg/mL, Chrono-par 385, Chrono-log Corp., Havertown, Pa.), arachidonic acid (1 mM, Biodata Corp, Horsham, Pa.) or thrombin (4 Units/mL, Enzyme Research Institute, South Bend, Ind.; plus Gly-Pro-Arg-Pro, a fibrin polymerization inhibitor, 2 mM, Sigma Chemical Co., St. Louis, Mo.). Platelet aggregation is monitored spectrophotometrically with a PAP-4C platelet aggregator (Bio Data Corp, Horsham. Pa.). Results are expressed as a percent inhibition of the rate of aggregation as compared to the pre-drug aggregation response.
  • Statistics [0104]
  • Data obtained by multiple sampling during the experiment are analyzed by two-way repeated measures analysis of variance. Post-hoc multiple comparisons of means to control values within treatment groups, and comparison of ENOX data to other treatment groups, are performed using the least significant difference test. A p-value less than 0.05 is considered significant. [0105]
  • Results [0106]
  • The combined use of an anti-Xa activity compound and a platelet aggregation antagonist compound used according to the invention, provides for use of those compounds at doses that would be subefficacious if used individually, while effecting the inhibition of repetitive platelet thrombus formation to the same extent as high doses of the platelet aggregation antagonist compound without significantly increasing template bleeding time (as compared to: a >5-fold increase in template bleeding time caused by high-dose of the platelet aggregation antagonist compound, BSPBPA; and a ˜3-fold increase in template bleeding time caused by high-dose of the platelet aggregation antagonist compound, PBGACA). [0107]
  • Combination of a low-dose of a platelet aggregation antagonist compound with heparin, at a dose which increased APTT 2.0- to 2.5- fold over baseline, did not inhibit repetitive platelet thrombus formation. [0108]
  • The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. [0109]

Claims (17)

What is claimed is:
1. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound.
2. The pharmaceutical composition according to claim 1 wherein the compound having anti-Xa activity is a low molecular weight heparin.
3. The pharmaceutical composition according to claim 1 wherein the low molecular weight heparin is selected from the group consisting of enoxaparin, nardroparin, dalteparin, certroparin, parnaparin, reviparin, ardeparin/RD heparin/RDH or tinzaparin.
4. The pharmaceutical composition according to claim 1 wherein the low moleclular weight heparin is enoxaparin.
5. The pharmaceutical composition according to claim 1 wherein the compound having anti-Xa activity is a heparinoid compound.
6. The pharmaceutical composition according to claim 1 wherein the heparinoid compound is danaparoid.
7. The pharmaceutical composition according to claim 1 wherein the platelet aggregation antagonist compound is abciximab, N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L-β-cyclohexyl alanine, N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L-β-cyclohexyl alanine amide or N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine
8. The pharmaceutical composition according to claim 7 wherein the platelet aggregation antagonist compound antagonist is abciximab.
9. The pharmaceutical composition according to claim 7 wherein the platelet aggregation antagonist compound is N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L-β-cyclohexyl alanine.
10. The pharmaceutical composition according to claim 7 wherein the platelet aggregation antagonist compound is N-[N-[N-(4-(piperidin-4-yl)butanoyl)-N-ethylglycyl]aspartyl]-L-β-cyclohexyl alanine amide.
11. The pharmaceutical composition according to claim 7 wherein the platelet aggregation antagonist compound is N-(n-butylsulfonyl)-4-(piperidin-4-ylbutyloxy)-L-phenylalanine.
12. A method of treating or preventing a physiological condition associated with a thrombosis related ischemic disorder in a patient comprising administering to said patient pharmaceutically effective amounts of a compound having anti-Xa activity and a platelet aggregation antagonist compound.
13. The method of claim 12 wherein the physiological condition is selected from stroke, atherosclerosis, anaiogenesis, thrombosis, a thromboembolic condition, disseminated intravascular coagulation, peripheral arterial occlusive disease, hemodialysis, an extra-corporeal circulation requirement associated with a surgical procedure, tissue damage caused by phospholipases A2, an acute coronary syndrome, or a thromboembolic syndrome associated with cancer, sepsis or obstetrical complications.
14. The method of claim 13 wherein the physiological condition is the acute coronary syndrome.
15. The method of claim 14 wherein the acute coronary syndrome is unstable angina or myocardial infarction.
16. The method of claim 12 wherein the treating or preventing occurs in the course of coronary artery bypass surgery or percutaneous transluminal coronary angioplasty.
17. The method of claim 12 wherein the treating or preventing occurs in the course of percutaneous transluminal coronary angioplasty. carrier.
US09/516,981 1996-11-27 2000-03-01 Pharmaceutical composition comprising a compound having anti-xa activity and a platelet aggregation antagonist compound Abandoned US20020032214A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/516,981 US20020032214A1 (en) 1996-11-27 2000-03-01 Pharmaceutical composition comprising a compound having anti-xa activity and a platelet aggregation antagonist compound

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US3187896P 1996-11-27 1996-11-27
PCT/US1997/021440 WO1998023279A1 (en) 1996-11-27 1997-11-24 PHARMACEUTICAL COMPOSITION COMPRISING A COMPOUND HAVING ANTI-Xa ACTIVITY AND A PLATELET AGGREGATION ANTAGONIST COMPOUND
US09/192,710 US6103705A (en) 1996-11-27 1998-11-16 Pharmaceutical composition comprising a compound having anti-Xa activity and a platelet aggregation antagonist compound
US09/516,981 US20020032214A1 (en) 1996-11-27 2000-03-01 Pharmaceutical composition comprising a compound having anti-xa activity and a platelet aggregation antagonist compound

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/192,710 Continuation US6103705A (en) 1996-11-27 1998-11-16 Pharmaceutical composition comprising a compound having anti-Xa activity and a platelet aggregation antagonist compound

Publications (1)

Publication Number Publication Date
US20020032214A1 true US20020032214A1 (en) 2002-03-14

Family

ID=21861879

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/192,710 Expired - Lifetime US6103705A (en) 1996-11-27 1998-11-16 Pharmaceutical composition comprising a compound having anti-Xa activity and a platelet aggregation antagonist compound
US09/516,981 Abandoned US20020032214A1 (en) 1996-11-27 2000-03-01 Pharmaceutical composition comprising a compound having anti-xa activity and a platelet aggregation antagonist compound

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/192,710 Expired - Lifetime US6103705A (en) 1996-11-27 1998-11-16 Pharmaceutical composition comprising a compound having anti-Xa activity and a platelet aggregation antagonist compound

Country Status (28)

Country Link
US (2) US6103705A (en)
EP (1) EP0946185B2 (en)
JP (1) JP4498471B2 (en)
KR (1) KR100512671B1 (en)
CN (1) CN1121224C (en)
AP (1) AP1390A (en)
AT (1) ATE283062T1 (en)
AU (1) AU754936B2 (en)
BG (1) BG64542B1 (en)
BR (1) BR9713141A (en)
CA (1) CA2272317C (en)
CZ (1) CZ298552B6 (en)
DE (1) DE69731763T3 (en)
DK (1) DK0946185T4 (en)
EA (1) EA002475B1 (en)
ES (1) ES2230624T5 (en)
HK (1) HK1023504A1 (en)
HU (1) HUP9903496A3 (en)
IL (1) IL129877A (en)
NO (1) NO327740B1 (en)
OA (1) OA11049A (en)
PL (1) PL189116B1 (en)
PT (1) PT946185E (en)
SI (1) SI0946185T2 (en)
SK (1) SK286024B6 (en)
UA (1) UA63929C2 (en)
WO (1) WO1998023279A1 (en)
ZA (1) ZA9710691B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1537871A1 (en) * 2003-12-04 2005-06-08 Aventis Pharma S.A. Enoxaparin for the treatment of cancer

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SK286024B6 (en) * 1996-11-27 2008-01-07 Aventis Pharmaceuticals Inc. Pharmaceutical composition and use of the composition for medicine preparation
FR2764511B1 (en) 1997-06-13 2000-09-08 Sanofi Sa COMPOSITIONS FOR THE TREATMENT AND PREVENTION OF ARTERIAL THROMBOSIS AND USE OF A FACTOR XA INHIBITOR ALONE AND / OR IN COMBINATION WITH A PLAQUET ANTIAGGREGANT
US6136794A (en) * 1998-02-02 2000-10-24 Merck & Co., Inc. Platelet aggregation inhibition using low molecular weight heparin in combination with a GP IIb/IIIa antagonist
AU1712700A (en) * 1998-11-18 2000-06-05 G.D. Searle & Co. Restoration of platelet aggregation by antibody administration after gpiib/iiia antagonist treatment
BR0012202A (en) * 1999-06-30 2002-04-02 Hamilton Civic Hospitals Res Compositions of heparin of average molecular weight (mmwh), method of treating a thrombotic condition, method of preventing the formation of a thrombus, method of inhibiting the formation of thrombi, pharmaceutical composition, method of treating deep vein thrombosis, method of prevention of pulmonary embolism, method of preparation and uses of a composition of mmwh
ES2161615B1 (en) * 1999-07-23 2003-03-16 Rovi Lab Farmaceut Sa COMPOSITIONS OF HEPARINS OF VERY LOW MOLECULAR WEIGHT.
US6969705B2 (en) * 2000-07-21 2005-11-29 Aventis Pharma S.A. Compositions of polysaccharides derived from heparin, their preparation and pharmaceutical compositions containing them
AU2001291549A1 (en) * 2000-09-08 2002-03-22 Hamilton Civic Hospitals Research Development Inc. Antithrombotic compositions
US6982251B2 (en) * 2000-12-20 2006-01-03 Schering Corporation Substituted 2-azetidinones useful as hypocholesterolemic agents
DE10141106A1 (en) * 2001-08-22 2003-03-13 Aventis Pharma Gmbh Use of heparinoid derivatives for the treatment and diagnosis of heparinoid-treatable diseases
US20040171819A1 (en) * 2002-10-10 2004-09-02 Aventis Pharma S.A. Mixtures of polysaccharides derived from heparin, their preparation and pharmaceutical compositions containing them
ATE418551T1 (en) 2003-03-07 2009-01-15 Schering Corp SUBSTITUTED AZETIDINONE DERIVATIVES, THEIR PHARMACEUTICAL FORMULATIONS AND THEIR USE IN THE TREATMENT OF HYPERCHOLESTEROLEMIA
JP5137228B2 (en) * 2003-03-07 2013-02-06 メルク・シャープ・アンド・ドーム・コーポレーション Substituted azetidinone compounds, substituted azetidinone formulations and their use for the treatment of hypercholesterolemia
US7956046B2 (en) * 2003-07-24 2011-06-07 Aventis Pharma S.A. Oligosaccharide mixtures derived from heparin, preparation thereof and pharmaceutical compositions containing them
TWI396686B (en) 2004-05-21 2013-05-21 Takeda Pharmaceutical Cyclic amide derivative, and its production and use
KR100601429B1 (en) * 2004-07-09 2006-07-14 주식회사 대우일렉트로닉스 Damper driving circuit of mechanical microwave range
EP2014296A1 (en) * 2007-07-10 2009-01-14 PAION Deutschland GmbH Novel strategies for increasing the reperfusion in obstructed blood vessel
AR067345A1 (en) * 2007-07-10 2009-10-07 Paion Deutschland Gmbh USE OF THROMBOMODULIN FOR THE PREPARATION OF A THROMBOLITIC MEDICINAL PRODUCT
CN101783857B (en) * 2009-05-12 2011-11-30 上海海事大学 Image matrixing pretreatment method based on FPGA in high-resolution imaging system
IT1400232B1 (en) * 2010-05-07 2013-05-24 Advance Holdings Ltd PHARMACEUTICAL COMPOSITION TOPICAL COMPARING EPARIN
WO2011149110A1 (en) 2010-05-28 2011-12-01 Daiichi Sankyo Company, Limited Novel composition for the prevention and/or treatment of thromboembolism
EP2508892A1 (en) * 2011-04-04 2012-10-10 Siemens Healthcare Diagnostics Products GmbH Controls and kit for thrombocyte activity tests
RU2554803C1 (en) * 2014-06-26 2015-06-27 Федеральное государственное бюджетное учреждение "Ростовский научно-исследовательский онкологический институт" Министерства здравоохранения Российской Федерации Method of preventing development of post-operative venous thromboembolic complications in patients with colorectal cancer

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3766167A (en) * 1971-03-26 1973-10-16 Research Corp Orally active anticoagulant
FR2440376A1 (en) * 1978-11-06 1980-05-30 Choay Sa MUCOPOLYSACCHARIDIC COMPOSITION HAVING REGULATORY COAGULATION ACTIVITY, MEDICAMENT CONTAINING SAME AND METHOD FOR OBTAINING SAME
SE449753B (en) * 1978-11-06 1987-05-18 Choay Sa MUCOPOLYSACCARIDE COMPOSITION WITH REGULATORY EFFECTS ON COAGULATION, MEDICINAL CONTAINING ITS SAME AND PROCEDURE FOR PREPARING THEREOF
FR2478646A2 (en) * 1980-03-20 1981-09-25 Choay Sa MUCOPOLYSACCHARIDIC COMPOSITION HAVING A COAGULATION REGULATING ACTIVITY, MEDICAMENT CONTAINING THE SAME, AND METHOD FOR OBTAINING THE SAME
US4692435A (en) * 1978-11-06 1987-09-08 Choay, S.A. Mucopolysaccharide composition having a regulatory action on coagulation, medicament containing same and process of preparation
CA1136620A (en) * 1979-01-08 1982-11-30 Ulf P.F. Lindahl Heparin fragments having selective anticoagulation activity
US4826827A (en) * 1979-10-05 1989-05-02 Choay S.A. Short chained oligosaccharides having biological properties, a process for making the same and the use thereof as drugs
FR2482611B1 (en) * 1980-05-14 1986-03-07 Pharmindustrie NOVEL SULFATED POLYSACCHARIDES, METHODS FOR THEIR PREPARATION AND THEIR USE AS MEDICAMENTS
US4351938A (en) * 1980-05-19 1982-09-28 Riker Laboratories, Inc. Anticoagulant substance
FR2503714B1 (en) * 1981-04-10 1986-11-21 Choay Sa PROCESS FOR OBTAINING BIOLOGICALLY ACTIVE MUCOPOLYSACCHARIDES, OF HIGH PURITY, BY DEPOLYMERIZATION OF HEPARIN
ATE15142T1 (en) * 1981-05-21 1985-09-15 Akzo Nv ANTITHROMBOTICAL BASED ON POLYSACCHARIDS, PROCESS FOR ITS PRODUCTION AND MEDICINAL COMPOSITIONS.
US4533549A (en) * 1983-01-04 1985-08-06 Lasker Sigmund E Antithrombotic agent
IT1195497B (en) * 1983-03-08 1988-10-19 Opocrin Spa PROCEDURE FOR THE PREPARATION OF OLIGOSACCHARIDIC FRACTIONS EQUIPPED WITH PHARMACOLOGICAL PROPERTIES FOR CHEMICAL DEGRADATION OF HEPARIN
US4683291A (en) * 1985-10-28 1987-07-28 Scripps Clinic And Research Foundation Platelet binding inhibitors
DK196986D0 (en) * 1986-04-30 1986-04-30 Novo Industri As PREPARATION OF POLYSACCHARIDES
DK196886D0 (en) * 1986-04-30 1986-04-30 Novo Industri As PREPARATION OF POLYSACCHARIDES
US4833155A (en) * 1986-11-25 1989-05-23 Syntex (U.S.A.) Inc. 3-(ω-(3,5,-di-t-butyl-4-hydroxyphenyl)-alkanoyl)pyrroles, and anti-inflammatory uses thereof
EP0275748B1 (en) * 1986-12-15 1992-08-19 Institut National De La Sante Et De La Recherche Medicale (Inserm) Peptide derivatives and their therapeutical use
US4857508A (en) * 1987-12-03 1989-08-15 Monsanto Company Novel platelet-aggregation inhibitor peptide derivatives
KR940009084B1 (en) * 1988-05-18 1994-09-29 체크 포인트 시스템스, 인코오퍼레이티드 Antenna system for magnetic and resonant circuit detection
IT1234508B (en) * 1988-06-10 1992-05-19 Alfa Wassermann Spa HEPARIN DERIVATIVES AND PROCEDURE FOR THEIR PREPARATION
US5037808A (en) * 1988-07-20 1991-08-06 Monsanto Co. Indolyl platelet-aggregation inhibitors
US4879313A (en) * 1988-07-20 1989-11-07 Mosanto Company Novel platelet-aggregation inhibitors
US4992463A (en) * 1988-07-20 1991-02-12 Monsanto Company Thienyl peptide mimetic compounds which are useful in inhibiting platelet aggregation
IT1234826B (en) * 1989-01-30 1992-05-29 Alfa Wassermann Spa HEPARIN DERIVATIVES AND PROCEDURE FOR THEIR PREPARATION
US5023233A (en) * 1989-07-28 1991-06-11 Merck & Co., Inc. Fibrinogen receptor antagonists
US4952562A (en) * 1989-09-29 1990-08-28 Rorer Pharmaceutical Corporation Anti-thrombotic peptides and pseudopeptides
CA2066047A1 (en) * 1989-09-29 1991-03-30 Scott I. Klein Anti-thrombotic peptides and pseudopeptides
US5051405A (en) * 1989-10-10 1991-09-24 Rhone-Poulenc Rorer Pharmaceuticals Inc. Anti-thrombotic peptides and pseudopeptides
US5053392A (en) * 1989-12-01 1991-10-01 Rhone-Poulenc Rorer Pharmaceuticals Inc. Novel arginine, glycine, aspartic acid derivatives as platelet-aggregation inhibitors
US5086069A (en) * 1990-02-05 1992-02-04 Rorer Pharmaceutical Corporation Anti-thrombotic peptide and pseudopeptide derivatives
US5064814A (en) * 1990-04-05 1991-11-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Anti-thrombotic peptide and pseudopeptide derivatives
NZ239846A (en) * 1990-09-27 1994-11-25 Merck & Co Inc Sulphonamide derivatives and pharmaceutical compositions thereof
IL99539A0 (en) * 1990-09-27 1992-08-18 Merck & Co Inc Fibrinogen receptor antagonists and pharmaceutical compositions containing them
IL99538A0 (en) * 1990-09-27 1992-08-18 Merck & Co Inc Fibrinogen receptor antagonists and pharmaceutical compositions containing them
UA26910C2 (en) * 1991-01-24 1999-12-29 Аероджет-Джеhерал Корпорейшh METHOD OF PROCESSING A COMPOSITION CONTAINING ENERGY COMPOUNDS (VARIATES)
HUT67136A (en) * 1991-05-02 1995-02-28 Yeda Res & Dev Compositions for the prevention and/or treatment of pathological processes
WO1993008845A1 (en) * 1991-11-08 1993-05-13 Massachusetts Institute Of Technology Localized oligonucleotide therapy
EP0691987A1 (en) * 1993-03-29 1996-01-17 The Du Pont Merck Pharmaceutical Company CYCLIC COMPOUNDS USEFUL AS INHIBITORS OF PLATELET GLYCOPROTEIN IIb/IIIa
PL181749B1 (en) * 1993-10-15 2001-09-28 Aventis Pharm Prod Inc Antihrombotic azacycloalkanoyl peptides and pseudopeptides
US5639469A (en) * 1994-06-15 1997-06-17 Minnesota Mining And Manufacturing Company Transmucosal delivery system
US5763427A (en) * 1995-03-31 1998-06-09 Hamilton Civic Hospitals Research Development Inc. Compositions and methods for inhibiting thrombogenesis
US5721214A (en) * 1995-06-07 1998-02-24 Cor Therapeutics, Inc. Inhibitors of factor Xa
AU2202397A (en) * 1996-03-28 1997-10-17 G.D. Searle & Co. Iib/iiia antagonists co-administered with aspirin and/or heparin
SK286024B6 (en) * 1996-11-27 2008-01-07 Aventis Pharmaceuticals Inc. Pharmaceutical composition and use of the composition for medicine preparation

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1537871A1 (en) * 2003-12-04 2005-06-08 Aventis Pharma S.A. Enoxaparin for the treatment of cancer
WO2005053713A1 (en) * 2003-12-04 2005-06-16 Aventis Pharma S.A. Enoxaparin for the treatment of cancer
US20050164985A1 (en) * 2003-12-04 2005-07-28 Aventis Pharma S.A. Therapeutic application of enoxaparin

Also Published As

Publication number Publication date
SK67399A3 (en) 2000-07-11
EP0946185A1 (en) 1999-10-06
BG64542B1 (en) 2005-07-29
CN1121224C (en) 2003-09-17
EP0946185B1 (en) 2004-11-24
DE69731763T3 (en) 2010-12-30
DE69731763D1 (en) 2004-12-30
US6103705A (en) 2000-08-15
ZA9710691B (en) 1998-06-12
PL189116B1 (en) 2005-06-30
ATE283062T1 (en) 2004-12-15
KR20000057265A (en) 2000-09-15
IL129877A0 (en) 2000-02-29
NO327740B1 (en) 2009-09-14
AP1390A (en) 2005-04-15
NO992555L (en) 1999-05-27
ES2230624T5 (en) 2010-10-25
CZ298552B6 (en) 2007-10-31
BR9713141A (en) 2000-02-08
SI0946185T1 (en) 2005-06-30
DK0946185T3 (en) 2005-03-21
BG103372A (en) 2000-01-31
EP0946185B2 (en) 2010-05-05
AU5454698A (en) 1998-06-22
NO992555D0 (en) 1999-05-27
HUP9903496A2 (en) 2000-10-28
SI0946185T2 (en) 2010-08-31
AP9901517A0 (en) 1999-06-30
EA199900499A1 (en) 2000-04-24
DE69731763T2 (en) 2005-06-30
AU754936B2 (en) 2002-11-28
UA63929C2 (en) 2004-02-16
CA2272317C (en) 2001-01-30
SK286024B6 (en) 2008-01-07
ES2230624T3 (en) 2005-05-01
EP0946185A4 (en) 2001-06-27
HUP9903496A3 (en) 2001-03-28
HK1023504A1 (en) 2000-09-15
KR100512671B1 (en) 2005-09-07
DK0946185T4 (en) 2010-08-30
JP2001506603A (en) 2001-05-22
WO1998023279A1 (en) 1998-06-04
PT946185E (en) 2005-02-28
CN1238693A (en) 1999-12-15
IL129877A (en) 2004-08-31
JP4498471B2 (en) 2010-07-07
EA002475B1 (en) 2002-06-27
CZ174299A3 (en) 1999-09-15
OA11049A (en) 2002-02-18

Similar Documents

Publication Publication Date Title
US6103705A (en) Pharmaceutical composition comprising a compound having anti-Xa activity and a platelet aggregation antagonist compound
Hanke et al. Inhibition of cellular proliferation after experimental balloon angioplasty by low-molecular-weight heparin.
Staudinger et al. Influence of prothrombin complex concentrates on plasma coagulation in critically ill patients
JPH04504710A (en) antithrombotic composition
Kawasaki et al. Pharmacological properties of YM-254890, a specific Gαq/11 inhibitor, on thrombosis and neointima formation in mice
Buchanan et al. Prevention of thrombus formation and growth by antithrombin III and heparin cofactor II-dependent thrombin inhibitors: importance of heparin cofactor II
Rebello et al. Antithrombotic efficacy of a novel factor Xa inhibitor, FXV673, in a canine model of coronary artery thrombolysis
Köhler Aerosolized heparin
JP3838680B2 (en) Medicaments suitable for use as antidote to anticoagulants and their use
CN1128168A (en) The use of vWF-containing concentrates as a therapy which is employed in combination with antithrombotic and fibrinolytic therapy
IML Queiroz et al. An evaluation of the efficacy of local hemostatic measures in dental patients taking oral anticoagulants: a critical review of the literature over the past two decades
Rogers et al. Effects of a factor Xa inhibitor, DX-9065a, in a novel rabbit model of venous thrombosis
Stow et al. Continuing Medical Education Article Anticoagulants in anaesthesia
MXPA99005008A (en) PHARMACEUTICAL COMPOSITION COMPRISING A COMPOUND HAVING ANTI-Xa ACTIVITY AND A PLATELET AGGREGATION ANTAGONIST COMPOUND
Rebello et al. Superiority of enoxaparin over heparin in combination with a GPIIb/IIIa receptor antagonist during coronary thrombolysis in dogs
Mattsson et al. Mechanism of action of the oral direct thrombin inhibitor ximelagatran
Bunce et al. Platelet function in patients receiving enprostil
PIZZO et al. in Vivo Regulation of Thrombin Generation and Activitya
JPH11100322A (en) New ischemic/reperfusion disorder inhibitor

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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