WO2003057218A1 - Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof - Google Patents

Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof Download PDF

Info

Publication number
WO2003057218A1
WO2003057218A1 PCT/EP2003/000153 EP0300153W WO03057218A1 WO 2003057218 A1 WO2003057218 A1 WO 2003057218A1 EP 0300153 W EP0300153 W EP 0300153W WO 03057218 A1 WO03057218 A1 WO 03057218A1
Authority
WO
WIPO (PCT)
Prior art keywords
rapamycin
inhibitor
active
drug delivery
derivative
Prior art date
Application number
PCT/EP2003/000153
Other languages
French (fr)
Inventor
Margaret Forney Prescott
Walter Schuler
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MXPA04006731A priority Critical patent/MXPA04006731A/en
Application filed by Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Priority to JP2003557576A priority patent/JP2005514411A/en
Priority to AU2003205586A priority patent/AU2003205586A1/en
Priority to CA002472198A priority patent/CA2472198A1/en
Priority to US10/501,210 priority patent/US20050020614A1/en
Priority to EP03702413A priority patent/EP1465624A1/en
Priority to IL16271903A priority patent/IL162719A0/en
Priority to HU0402594A priority patent/HUP0402594A3/en
Priority to KR10-2004-7010710A priority patent/KR20040076278A/en
Priority to BR0306858-7A priority patent/BR0306858A/en
Publication of WO2003057218A1 publication Critical patent/WO2003057218A1/en
Priority to NO20043309A priority patent/NO20043309L/en
Priority to US12/242,980 priority patent/US20090036352A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/0092Hollow drug-filled fibres, tubes of the core-shell type, coated fibres, coated rods, microtubules or nanotubes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus

Definitions

  • the present invention relates to drug delivery systems for the prevention and treatment of proliferative diseases, particularly vascular diseases.
  • PCTA percutaneous transluminal coronary angioplasty
  • PTA percutaneous transluminal angioplasty
  • atherectomy bypass grafting or other types of vascular grafting procedures.
  • Re-narrowing (restenosis) of an artherosclerotic coronary artery after various revascularization procedures occurs in 10-80% of patients undergoing this treatment, depending on the procedure used and the aterial site.
  • revascularization also injures endothelial cells and smooth muscle cells within the vessel wall, thus initiating a thrombotic and inflammatory response.
  • Cell derived growth factors such as platelet derived growth factor, infiltrating macrophages, leukocytes or the smooth muscle cells themselves provoke proliferative and migratory responses in the smooth muscle cells.
  • inflammatory cells also invade the site of vascular injury and may migrate to the deeper layers of the vessel wall. Proliferation/migration usually begins within one to two days post-injury and, depending on the revascularization procedure used, continues for days and weeks.
  • the newly formed tissue is called neointima, intimal thickening or restenotic lesion and usually results in narrowing of the vessel lumen. Further lumen narrowing may take place due to constructive remodeling, e.g. vascular remodeling, leading to further intimal thickening or hyperplasia.
  • atherosclerotic lesions which do not limit or obstruct vessel blood flow but which form the so-called "vulnerable plaques”.
  • Atherosclerotic lesions or vulnerable plaques are prone to rupture or ulcerate, which results in thrombosis and thus produces unstable angina pectoris, myocardial infarction or sudden death. Inflamed atherosclerotic plaques can be detected by thermography.
  • vascular access dysfunction in hemodialysis patients is generally caused by outflow stenoses in the venous circulation (Schwam S. J., et al., Kidney Int. 36: 707-711 , 1989).
  • Vascular access related morbidity accounts for about 23 percent of all hospital stays for advanced renal disease patients and contributes to as much as half of all hospitalization costs for such patients (Feldman H. I., J. Am. Soc. Nephrol. 7: 523 -535,1996).
  • vascular access dysfunction in chemotherapy patients is generally caused by outflow stenoses in the venous circulation and results in a decreased ability to administer medications to cancer patients. Often the outflow stenoses is so severe as to require intervention.
  • TPN total parenteral nutrition
  • Vascular access dysfunction is the most important cause of morbidity and hospitalization in the hemodialysis population. Venous neointimal hyperplasia characterized by stenosis and subsequent thrombosis accounts for the overwhelming majority of pathology resulting in dialysis graft failure. The most common form of vascular access procedure performed in chronic hemodialysis patients in the United States is the arteriovenous PTFE graft, which accounts for approximately 70% of all hemodialysis access.
  • VNH venous neointimal hyperplasia
  • VNH in the setting of hemodialysis grafts appears to be a far more aggressive lesion as compared to the more common arterial neointimal hyperplasia that occurs in peripheral bypass grafts.
  • VNH in the setting of dialysis access grafts appears to be a far more aggressive lesion as compared to the more common arterial neointimal hyperplasia that occurs in peripheral bypass grafts.
  • compare the 50% one year primary patency in PTFE dialysis access grafts with an 88% five year patency for aortoiliac grafts and a 70 to 80% one year patency for femoro-popliteal grafts.
  • Venous stenoses in the setting of dialysis access grafts also have a poorer response to angioplasty (40% three month survival if thrombosed and a 50% six month survival if not thrombosed) as compared to arterial stenoses.
  • rapamycin and rapamycin derivatives having mTOR inhibiting properties have beneficial effects on above mentioned disorders, diseases or dysfunctions.
  • Rapamycin is a known macrolide antibiotic produced by Streptomyces hygroscopicus, which inhibits mTOR.
  • rapamycin derivative having mTOR inhibiting properties is meant a substituted rapamycin, e.g. a 40-substituted-rapamycin or a 16-substituted rapamycin, or a 32-hydrogenated rapamycin, for example a compound of formula I
  • rapamycin derivatives of formula I are e.g. 32-deoxorapamycin, 16-pent-2- ynyloxy-32-deoxorapamycin, 16-pent-2-ynyloxy-32(S or R)-dihydro-rapamycin, 16-pent-2- ynyloxy-32(S or R)-dihydro-40-O-(2-hydroxyethyl)-rapamycin, 40-[3-hydroxy-2- (hydroxymethyl)-2-methylpropanoate]-rapamycin (also called CCI779) or 40-epi-(tetrazolyl)- rapamycin (also called ABT578).
  • a preferred compound is e.g.
  • Rapamycin derivatives may also include the so-called rapalogs, e.g. as disclosed in WO 98/02441 and WO01/14387, e.g. AP23573.
  • rapamycin or a rapamycin derivative having mTOR inhibiting properties may be applied as the sole active ingredient or in conjunction with one or more active co-agents selected from a) an immunosuppressive agent, e.g. a calcineurin inhibitor, e.g. a cyclosporin, for example cyclosporin A, ISA tx 247 or FK506, b) an EDG-receptor agonist having lymphocyte depleting properties, e.g. FTY720 (2-amino- 2-[2-(4-octylphenyI) ethyl]propane-1 ,3-diol in free form or in a pharmaceutically acceptable salt form, e.g.
  • an immunosuppressive agent e.g. a calcineurin inhibitor, e.g. a cyclosporin, for example cyclosporin A, ISA tx 247 or FK506,
  • an anti-inflammatory agent e.g. a steroid, e.g. a corticosteroid, e.g. dexamethasone or prednisone, a NSAID, e.g.
  • a cyclooxygenase inhibitor e.g. a cox-2 inhibitor, e.g. celecoxib, rofecoxib, etoricoxib or valdecoxib, an ascomycin, e.g. ASM981 (or pimecrolimus), a cytokine inhibitor, e.g. a lymphokine inhibitor, e.g. an IL-1 , -2 or -6 inhibitor, for example pralnacasan or anakinra, or a TNF inhibitor, for instance Etanercept, or a chemokine inhibitor; d) an anti- thrombotic or anti-coagulant agent, e.g.
  • a cox-2 inhibitor e.g. celecoxib, rofecoxib, etoricoxib or valdecoxib
  • an ascomycin e.g. ASM981 (or pimecrolimus)
  • a cytokine inhibitor e.g. a lymph
  • heparin or a glycoprotein llb/llla inhibitor e.g. abciximab, eptifibatide or tirofibran
  • an antiproliferative agent e.g. a microtubule stabilizing or destabilizing agent including but not limited to taxanes, e.g. taxol, paclitaxel or docetaxel, vinca alkaloids, e.g. vinblastine, especially vinblastine sulfate, vincristine especially vincristine sulfate, and vinorelbine, discodermolides or epothilones or a derivative thereof.e.g.
  • epothilone B or a derivative thereof; a protein tyrosine kinase inhibitor, e.g. protein kinase C or Pl(3) kinase inhibitor, for example staurosporin and related small molecules, e.g.
  • EGF receptor tyrosine kinase a compound which binds to EGF or reduces expression of the EGF receptor e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the compound of ex.
  • trastuzumab (Herpetin R ), cetuximab, OSI-774, CI-1033, EKB-569, GW-2016, E1.1 , E2.4, E2.5, E6.2, E6.4, E2.11 , E6.3 or E7.6.3, retinoic acid, alpha-, gamma- or delta-tocopherol or alpha-, gamma- or delta-tocotrienol, or compounds affecting GRB2, IMC-C225; or a compound or antibody which inhibits the VEGF receptor tyrosine kinase or a VEGF receptor or a compound which binds to VEGF, e.g.
  • WO 98/35958 e.g. 1-(4- chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g. the succinate, or in WO 00/09495, WO 00/27820, WO 00/59509, WO 98/11223, WO 00/27819, WO 00/37502, WO 94/10202 and EP 0 769 947, those as described by M. Prewett et al in Cancer Research 59 (1999) 5209-5218, by F. Yuan et al in Proc. Natl. Acad. Sci. USA, vol.
  • RhuMab e.g. having HMG-CoA reductase inhibition activity, e.g. fluvastatin, lovastatin, simvastatin, pravastatin, atorvastatin, cerivastatin, pitavastatin, rosuvastatin or nivastatin; g) a compound, protein, growth factor or compound stimulating growth factor production that will enhance endothelial regrowth of the luminal endothelium, e.g. FGF, IGF; h) a matrix metalloproteinase inhibitor, e.g.
  • HMG-CoA reductase inhibition activity e.g. fluvastatin, lovastatin, simvastatin, pravastatin, atorvastatin, cerivastatin, pitavastatin, rosuvastatin or nivastatin
  • a compound, protein, growth factor or compound stimulating growth factor production that will enhance endothelial regrowth of the luminal end
  • a modulator i.e. antagonists or agonists of kinases, e.g. JNK, ERK1/2, MAPK or STAT;
  • a compound stimulating the release of (NO) or a NO donor e.g. diazeniumdiolates, S- nitrosothiols, mesoionic oxatriazoles, isosorbide or a combination thereof, e.g. mononitrate and/or dinitrate;
  • a somatostatin analogue e.g. octreotide, lanreotide, vapreotide or a cyclohexapeptide having somatostatin agonist properties, e.g.
  • a modified GH analogue chemically linked to PEG e.g. Pegvisomant
  • an altosterone synthetase inhibitor or aldosterone receptor blocker e.g. eplerenone
  • a compound inhibiting the renin-angiotensin system e.g. a renin inhibitor, e.g. SPP100, an ACE inhibitor, e.g.
  • captopril enalapril, lisinopril, fosinopril, benazepril, quinapril, ramipril, imidapril, perindopril erbumine, trandolapril or moexipril, or an ACE receptor blocker, e.g. losartan, irbesartan, candesartan cilexetil, valsartan or olmesartan medoxomil; o) mycophenolic acid or a salt thereof, e.g. sodium mycophenolate, or a prodrug thereof, e.g. mycophenolate mofetil.
  • ACE receptor blocker e.g. losartan, irbesartan, candesartan cilexetil, valsartan or olmesartan medoxomil
  • mycophenolic acid or a salt thereof e.g. sodium mycophenolate, or a pro
  • antibody By antibody is meant monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
  • a pharmaceutical combination comprising i) rapamycin or a rapamycin derivative having mTOR properties and ii) pimecrolimus, also form part of the present invention.
  • rapamycin is preferably locally administered or delivered in conjunction with one or more co-agents selected from b), e), f), g), h), k), m), n), o), a cox-2 inhibitor, a cytokine inhibitor or a chemokine inhibitor, as defined above.
  • a method for preventing or treating smooth muscle cell proliferation and migration in hollow tubes, or increased cell proliferation or decreased apoptosis or increased matrix deposition in a subject in need thereof comprising local administration of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • a method for the prevention or treatment of intimal thickening in vessel walls comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • the intimal thickening in vessel walls is stenosis, restenosis, e.g. following revascularization or neovascularization, and/or inflammation and/or thrombosis.
  • a method for stabilizing vulnerable plaques in blood vessels of a subject in need of such a stabilization comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • a method as defined in 1.1 to 1.4 associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of rapamycin or a derivative thereof having mTOR inhibiting properties, e.g. a compound of formula I.
  • rapamycin or the derivative thereof, e.g. of formula I is administered orally.
  • a method as defined in 1.1 to 1.4 may be associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of the co- agent.
  • a method for preventing or treating restenosis in diabetic patients comprising administering to said patients a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • a method for preventing or treating restenosis in diabetic patients comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • a method for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter, preferably a large bore catheter, into a vein or artery, or actual treatment, in a subject in need thereof which comprises administering to the subject rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above, or a controlled delivery from a drug delivery medical device or system of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • the invention relates to the prevention or reduction of vascular access dysfunction in hemodialysis.
  • a method for the stabilization or repair of arterial or venous aneurisms in a subject comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • 1.11 A method for the prevention or treatment of anastomic hyperplasia in a subject comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • 1.12 A method for the prevention or treatment of arterial, e.g.
  • aortic, by-pass anastomosis in a subject comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
  • a method as defined in 1.9 to 1.12 associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of rapamycin or a derivative thereof, e.g. a compound of formula I.
  • rapamycin or the derivative thereof, e.g. of formula I is administered orally.
  • a method as defined in 1.9 to 1.12 may be associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of the co- agent.
  • a drug delivery device or system comprising i) a medical device adapted for local application or administration in hollow tubes, e.g. a catheter-based delivery device or a medical device intraluminal or outside of hollow tubes such as an implant or a sheath placed within the adventitia, and ii) a therapeutic dosage of a rapamycin derivative having mTOR inhibiting properties or rapamycin, optionally in conjunction with a therapeutic dosage of one or more other active co-agents, e.g. as disclosed above, each being releasably affixed to the delivery device or system.
  • a medical device adapted for local application or administration in hollow tubes e.g. a catheter-based delivery device or a medical device intraluminal or outside of hollow tubes such as an implant or a sheath placed within the adventitia
  • a therapeutic dosage of a rapamycin derivative having mTOR inhibiting properties or rapamycin optionally in conjunction with a therapeutic dosage of one or more other active co-agents,
  • rapamycin or a rapamycin derivative having mTOR inhibiting properties in any of the method as defined under 1.4, 1.6 or 1.9 optionally in conjunction with one or more other active co-agent, or in the manufacture of a medicament for use in any of the method as defined under 1.4, 1.6 or 1.9 optionally in conjunction with one or more other active co-agent.
  • a pharmaceutical composition for use in any method as defined under 1.4, 1.6 or 1.9 comprising rapamycin or a derivative thereof having mTOR properties, e.g. CCI779, ABT578, a rapalog or a compound of formula I, together with one or more pharmaceutically acceptable diluents or carriers therefor.
  • a local delivery device or system according to the invention can be used to reduce stenosis or restenosis as an adjunct to revascularization, bypass or grafting procedures performed in any vascular location including coronary arteries, carotid arteries, renal arteries, peripheral arteries, cerebral arteries or any other arterial or venous location, to reduce anastomic stenosis or hyperplasia including in the case of arterial-venous dialysis access with or without PTFE or e.g. Gore-Tex grafting and with or without stenting, or in conjunction with any other heart or transplantation procedures, or congenital vascular interventions.
  • the present invention also provides a drug delivery system or device as disclosed above additionally comprising a source delivering a therapeutic dosage of a compound or antibody which inhibits the PDGF receptor tyrosine kinase or a compound which binds to PDGF or reduces expression of the PDGF receptor e.g. as disclosed above, a compound or antibody which inhibits the EGF receptor tyrosine kinase or a compound which binds to EGF or reduces expression of the EGF receptor e.g. as disclosed above, a compound or antibody which inhibits the VEGF receptor tyrosine kinase or a VEGF receptor or a compound which binds to VEGF, e.g. as disclosed above, each being releasably affixed to the catheter-based delivery device or medical device.
  • Rapamycin or rapamycin derivative having mTOR inhibiting properties will be referred to hereinafter as “active agent”.
  • drug(s) means active agent or the active agent and the active co-agent.
  • the local administration preferably takes place at or near the lesion sites, e.g. vascular lesion sites.
  • the local administration may be by one or more of the following routes: via catheter or other intravascular delivery system, intranasally, intrabronchially, interperitoneally or eosophagal, or via delivery balloons used in the musculature.
  • Hollow tubes include natural body vessels or ducts, e.g. circulatory system vessels such as blood vessels (arteries or veins), tissue lumen, lymphatic pathways, digestive tract including alimentary duct, e.g. esophagus or biliary ducts, respiratory tract, e.g. trachea, excretory system tubes, e.g. intestines, ureters or urethra-prostate, reproductive system tubes and ducts, body cavity tubes, etc.
  • circulatory system vessels such as blood vessels (arteries or veins), tissue lumen, lymphatic pathways, digestive tract including alimentary duct, e.g. esophagus or biliary ducts, respiratory tract, e.g. trachea, excretory system
  • Local administration or application of the drug(s) may afford concentrated delivery of said drug(s), achieving tissue levels in target tissues not otherwise obtainable through other administration route. Additionally local administration or application may reduce the risk of remote or systemic toxicity.
  • the smooth muscle cell proliferation or migration is inhibited or reduced according to the invention immediately proximal or distal to the locally treated or stented area.
  • Means for local delivery of the drug(s) to hollow tubes can be by physical delivery of the drug(s) either internally or externally to the hollow tube.
  • Local drug(s) delivery includes catheter delivery systems, local injection devices or systems or indwelling devices. Such devices or systems would include, but not be limited to, stents, coated stents, endolumenal sleeves, stent-grafts, sheathes, balloons, liposomes, controlled release matrices, polymeric endoluminal paving, or other endovascular devices, embolic delivery particles, cell targeting such as affinity based delivery, internal patches around the hollow tube, external patches around the hollow tube, hollow tube cuff, external paving, external stent sleeves, and the like. See, Eccleston et al.
  • Interventional Cardiology Monitor 1 33-40-41
  • Interventional Cardiol. 1 103-116
  • Regar E Sianos G
  • Serruys PW Stent development and local drug delivery
  • the delivery device or system fulfils pharmacological, pharmacokinetic and mechanical requirements.
  • it also is suitable for sterilisation.
  • the stent according to the invention can be any stent, including self-expanding stent, or a stent that is radially expandable by inflating a balloon or expanded by an expansion member, or a stent that is expanded by the use of radio frequency which provides heat to cause the stent to change its size.
  • a stent composed of or coated with a polymer or other biocompatible materials, e.g. porous ceramic, e.g. nanoporous ceramic, into which the drug(s) has been impregnated or incorporated can be used.
  • Stents can be biodegradable or can be made of metal or alloy, e.g. Ni and Ti, or another stable substance when intented for permanent use.
  • the drug(s) may also be entrapped into the metal of the stent or graft body which has been modified to contain micropores or channels. Also lumenal and/or ablumenal coating or external sleeve made of polymer or other biocompatible materials, e.g. as disclosed below, that contain the drug(s) can also be used for local delivery.
  • biocompatible a material which elicits no or minimal negative tissue reaction including e.g. thrombus formation and/or inflammation.
  • Stents may commonly be used as a tubular structure left inside the lumen of a duct to relieve an obstruction. They may be inserted into the duct lumen in a non-expanded form and are then expanded autonomously (self-expanding stents) or with the aid of a second device in situ, e.g. a catheter-mounted angioplasty balloon which is inflated within the stenosed vessel or body passageway in order to shear and disrupt the obstructions associated with the wall components of the vessel and to obtain an enlarged lumen.
  • a catheter-mounted angioplasty balloon which is inflated within the stenosed vessel or body passageway in order to shear and disrupt the obstructions associated with the wall components of the vessel and to obtain an enlarged lumen.
  • stents being easily deformed at lower temperature to be inserted in the hollow tubes may be used: after deployment at site, such stents recover their original shape and exert a retentive and gentle force on the internal wall of the hollow tubes, e.g. of the esophagus or trachea.
  • the drug(s) may be incorporated into or affixed to the stent in a number of ways and utilizing any biocompatible materials; it may be incorporated into e.g. a polymer or a polymeric matrix and sprayed onto the outer surface of the stent.
  • a mixture of the drug(s) and the polymeric material may be prepared in a solvent or a mixture of solvents and applied to the surfaces of the stents also by dip-coating, brush coating and/or dip/spin coating, the solvent (s) being allowed to evaporate to leave a film with entrapped drug(s).
  • a solution of a polymer may additionally be applied as an outlayer to control the drug(s) release; alternatively, the active agent may be comprised in the micropores, struts or channels and the active co-agent may be incorporated in the outlayer, or vice versa.
  • the active agent may also be affixed in an inner layer of the stent and the active co-agent in an outer layer, or vice versa.
  • the drug(s) may also be attached by a covalent bond, e.g. esters, amides or anhydrides, to the stent surface, involving chemical derivatization.
  • the drug(s) may also be incorporated into a biocompatible porous ceramic coating, e.g. a nanoporous ceramic coating.
  • the medical device of the invention is configured to release the active co-agent concurrent with or subsequent to the release of the active agent.
  • polymeric materials include hydrophilic, hydrophobic or biocompatible biodegradable materials, e.g. polycarboxylic acids; cellulosic polymers; starch; collagen; hyaluronic acid; gelatin; lactone-based polyesters or copolyesters, e.g.
  • polylactide polyglycolide; polylactide-glycolide; polycaprolactone; polycaprolactone-glycolide; poly(hydroxybutyrate); poly(hydroxyvalerate); polyhydroxy(butyrate-co-valerate); polyglycolide-co-trimethylene carbonate; poly(diaxanone); polyorthoesters; polyanhydrides; polyaminoacids; polysaccharides; polyphospoeters; polyphosphoester-urethane; polycyanoacrylates; polyphosphazenes; poly(ether-ester) copolymers, e.g.
  • PEO-PLLA fibrin; fibrinogen; or mixtures thereof; and biocompatible non-degrading materials, e.g. polyurethane; polyolefins; polyesters; polyamides; polycaprolactame; polyimide; polyvinyl chloride; polyvinyl methyl ether; polyvinyl alcohol or vinyl alcohol/olefin copolymers, e.g. vinyl alcohol/ethylene copolymers; polyacrylonitrile; polystyrene copolymers of vinyl monomers with olefins, e.g.
  • styrene acrylonitrile copolymers ethylene methyl methacrylate copolymers; polydimethylsiloxane; poly(ethylene-vinylacetate); acrylate based polymers or coplymers, e.g. polybutylmethacrylate, poly(hydroxyethyl methylmethacrylate); polyvinyl pyrrolidinone; fluorinated polymers such as polytetrafluoethylene; cellulose esters e.g. cellulose acetate, cellulose nitrate or cellulose propionate; or mixtures thereof.
  • a polymeric matrix When a polymeric matrix is used, it may comprise 2 layers, e.g. a base layer in which the drug(s) is/are incorporated, e.g. ethylene-co-vinylacetate and polybutylmethacrylate, and a top coat, e.g. polybutylmethacrylate, which is drug(s)-free and acts as a diffusion-control of the drug(s).
  • the active agent may be comprised in the base layer and the active co-agent may be incorporated in the outlayer, or vice versa. Total thickness of the polymeric matrix may be from about 1 to 20 ⁇ or greater.
  • the drug(s) may elute passively, actively or under activation, e.g. light-activation.
  • the drug(s) elutes from the polymeric material or the stent over time and enters the surrounding tissue, e.g. up to ca. 1 month to 1 year.
  • the local delivery according to the present invention allows for high concentration of the drug(s) at the disease site with low concentration of circulating compound.
  • the amount of drug(s) used for local delivery applications will vary depending on the compounds used, the condition to be treated and the desired effect.
  • a therapeutically effective amount will be administered; for example, the drug delivery device or system is configured to release the active agent and/or the active co-agent at a rate of 0.001 to 200 ⁇ g/day.
  • therapeutically effective amount is intended an amount sufficient to inhibit cellular proliferation and resulting in the prevention and treatment of the disease state.
  • a contemplated treatment period for use in the prevention or reduction of vascular access dysfunction of the present invention is about 85, e.g. 70, preferably 50, e.g. 28, more preferably 28 days in association with the insertion or repair of an indwelling shunt, fistula or catheter, or actual treatment.
  • a preferred method of use in the prevention or reduction of vascular access dysfunction is a method for preventing or reducing vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling access clotting shunt, fistula or catheter associated with insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, in dialysis patients.
  • a preferred method of use in the prevention or reduction of vascular access dysfunction is a method for preventing or reducing vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling vascular access shunt, fistula or catheter associated with insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, in cancer patients.
  • a preferred method of use in the prevention or reduction of vascular access dysfunction is a method for preventing or reducing vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling vascular access shunt, fistula or catheter associated with insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, in total parenteral nutrition (TPN) patients.
  • TPN total parenteral nutrition
  • prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter is meant that the incidence of vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling vascular access shunt, fistula or catheter in patients treated according to the invention collected over the observation period are prevented or reduced in comparison to untreated patients.
  • the treatment according to the invention can commence immediately, for example within 4 to 8 hours, after insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, such as dialysis treatment; within a few days, for example about 7 days, preferably about 1 or 2 days, after insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, such as dialysis treatment; or for a period of days, for example about 30 days, preferably about 14 days, preferably about 7 days, prior to insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, such as dialysis treatment.
  • in association with the insertion or repair of an indwelling shunt, fistula or catheter is a dosing protocol in which a dose or several doses, are skipped, for example in the morning of or on the day of insertion, repair or treatment.
  • in association with the insertion or repair of an indwelling shunt, fistula or catheter is a dosing protocol in which a day of drug treatment or several days of drug treatment, are skipped.
  • treatment when used herein to refer surgical procedures, are procedures selected from access surgery, placement of fistula or shunt, catheter insertion, actual disease treatment, such as dialysis treatment, and declotting of an access shunt, fistula or catheter. Further, treatment for insertion access also includes repair/revision of the access. For example, a patient experiencing a failure in a dialysis access shunt will have the access repaired, for instance, by angioplasty.
  • collected over the observation period means a period of up to or about 12 months, preferably 12 months.
  • rapamycin or a rapamycin derivative having mTOR inhibiting properties is administered systemically or is additionally administered by systemic application, e.g. in the prevention or reduction of vascular access dysfunction, according to the invention.daily dosages required in practicing the method of the present invention will vary depending upon, for example, the compound used, the host, the mode of administration and the severity of the condition to be treated.
  • a preferred daily dosage range is about from 0.1 to 25 mg as a single dose or in divided doses. Suitable daily dosages for patients are on the order of from e.g. 0.1 to 25 mg p.o.
  • the compound may be administered by any conventional route, in particular enterally, e.g. orally, e.g.
  • Suitable unit dosage forms for oral administration comprise from ca. 0.05 to 12.5 mg, usually 0.25 to 10 mg compound, together with one or more pharmaceutically acceptable diluents or carriers therefor.
  • Preferred combinations according to the invention are those comprising a compound of formula I, e.g. 40-0-(2-hydroxyethyl)-rapamycin or 32-deoxorapamycin, or CCI-779, ABT578 or a rapalog in conjunction or association with a compound having antiproliferative properties, e.g. taxol, paclitaxel, docetaxel, an epothilone, a tyrosine kinase inhibitor, e.g.
  • a protein kinase C or Pl(3) kinase inhibitor for example staurosporin or a related small molecule, a PDGF receptor tyrosine kinase inhibitor, a PDGF receptor inhibitor, a compound binding to PDGF, e.g. imatinib, a VEGF receptor tyrosine kinase inhibitor, a VEGF receptor inhibitor, a compound binding to VEGF, e.g. 1-(4-chloroanilino)-4-(4- pyridylmethyl)phtalazine, a cox-2 inhibitor, an ascomycin, e.g. pimecrolimus, or a calcineurin inhibitor, e.g.
  • a combination of rapamycin or a rapamycin derivative as mentioned above with a compound having anti-inflammatory properties, pimecrolimus, or an EDG-receptor agonist having lymphocyte depleting properties, has particularly beneficial effects when used in the treatment or prevention of restenosis in diabetic patients.
  • a combination of rapamycin or a rapamycin derivative as mentioned above with a statin or an aldosterone synthetase inhibitor or an aldosterone receptor blocker, or with a compound inhibiting the renin-angiotensin system has also beneficial properties; such a combination also forms part of the invention.
  • Rapamycin or the rapamycin derivative having mTOR inhibiting properties may also be applied to the drug delivery device or system in admixture with an antioxidant, e.g. 2,6-di- tert.-butyl-4-methylphenol, e.g. at an amount up to 0.5% by weight, preferably 0.2% by weight.
  • an antioxidant e.g. 2,6-di- tert.-butyl-4-methylphenol
  • Utility of the drug(s) may be demonstrated in animal test methods as well as in clinic, for example in accordance with the methods hereinafter described.
  • Rats are dosed orally with placebo or a compound of formula I. Daily dosing starts 3 days prior to surgery and continues for 31 days. Rat carotid arteries are balloon injured using a method described by Clowes et al. Lab. Invest. 1983;49;208-215. Following sacrifice at 28 days post-balloon injury, carotid arteries are removed and processed for histologic and morphometric evaluation. In this assay the compounds of formula I, e.g. 40-O-(2- hydroxyethyl)-rapamycin, significantly reduce neointimal lesion formation at 28 days following balloon injury when administered at a dose of from 0.5 to 2.0 mg/kg.
  • the compounds of formula I e.g. 40-O-(2- hydroxyethyl)-rapamycin
  • a combined angioplasty and stenting procedure is performed in New Zealand White rabbit iliac arteries.
  • Iliac artery balloon injury is performed by inflating a 3.0 x 9.0 mm angioplasty balloon in the mid-portion of the artery followed by "pull-back" of the catheter for 1 balloon length.
  • Balloon injury is repeated 2 times, and a 3.0 x 12 mm stent is deployed at 6 atm for 30 seconds in the iliac artery. Balloon injury and stent placement is then performed on the contralateral iliac artery in the same manner.
  • a post-stent deployment angiogram is performed. All animals receive oral aspirin 40 mg/day daily as anti-platelet therapy and are fed standard low-cholesterol rabbit chow.
  • mice Twenty-eight days after stenting, animals are anesthetized and euthanized and the arterial tree is perfused at 100 mmHg with lactated Ringer's for several minutes, then perfused with 10% formalin at 100 mmHg for 15 minutes.
  • the vascular section between the distal aorta and the proximal femoral arteries is excised and cleaned of periadventitial tissue.
  • the stented section of artery is embedded in plastic and sections are taken from the proximal, middle, and distal portions of each stent. All sections are stained with hematoxylin-eosin and Movat pentachrome stains.
  • Computerized planimetry is performed to determine the area of the internal elastic lamina (IEL), external elastic lamina (EEL) and lumen.
  • the neointima and neointimal thickness is measured both at and between the stent struts.
  • the vessel area is measured as the area within the EEL.
  • Data are expressed as mean ⁇ SEM.
  • Statistical analysis of the histologic data is accomplished using analysis of variance (ANOVA) due to the fact that two stented arteries are measured per animal with a mean generated per animal. A P ⁇ 0.05 is considered statistically significant.
  • a compound of formula I e.g. 40-O-(2-hydroxyethyl)-rapamycin
  • 40-O-(2-hydroxyethyl)-rapamycin is administered orally by gavage at a loading dose of 1.5 mg/kg one day prior to stenting, then dosed at 0.75 mg/kg/day from the day of stenting until day 27 post-stenting.
  • the treatment with the compounds of formula I results in a marked reduction in the extent of restenotic lesion formation: for example, the treatment with 40-O-(2-hydroxyethyl)-rapamycin produces a significant (P ⁇ 0.03) reduction in neointimal thickness (40% reduction), neointimal area (24% reduction), and percent arterial stenosis (26% reduction) with a significant 32% increase in lumen area.
  • mice Male Sprague Dawley rats weighing 250 to 500 mg are housed individually and allowed to acclimate prior to surgery. All animals receive standard rat chow and water ad libitum. Group size is 12 animals per group.
  • the drug(s) administration is perivascular.
  • a segment of ballooned carotid is encircled with a 1 cm length of silastic tubing (0.25 inch inside diameter, 0.47 inch outside diameter) to which is attached a catheter which feeds into an osmotic pump containing either compound or vehicle.
  • This delivery system provides continuous, local delivery to the adventitia of the wrapped portion of vessel.
  • Local drug(s) administration ranges between 5 ⁇ g and 10 mg, locally per day, depending on the solubility characteristics of the individual compounds.
  • the left common carotid arteries are denuded of endothelium using a 2F Fogarty catheter as previously described (Prescott Am. J. Pathol. (1991) 139:1291-1296, Clowes et al., (1983) Lab Invest. 49:327-333). Briefly, rats are anesthetized with ketamine (50 mg/ml) and rompun (10mg/ml) administered intraperitoneally at a dose of 1.5 ml/kg. A midline incision is made in the neck to expose the left external and common carotid arteries.
  • the balloon is inserted into the common carotid artery via the left external branch, inflated with saline, and pulled back three times through the lumen with a rotating motion to ensure maximal endothelial denudation.
  • the catheter is then removed, the external carotid artery is ligated and the wound is closed.
  • Each animal is given an injection of the antibiotic Bacillin (200.000 units/kg) and the analgesic Buprenophine (0.06 mg/kg) immediately following surgery.
  • Animals are killed at 14 days post-balloon injury. One half hour before termination blood is collected, centrifuged, and stored at -20 C C for analysis of circulating levels of compound. 5% Evans Blue is then injected intravenously to allow discrimination of re-endothelialized areas at the time of histologic processing. Animals are killed by administration of an overdose of ketamine and rompun, the osmotic pumps are recovered and the volume of remaining content is recorded to ensure that pump failure has not occurred. Carotid arteries are excised and immersion fixed, then transferred to Ringer's solution. Two samples from control blue region of each left carotid artery are imbedded in paraffin.
  • a minimum of six carotid sections, 20 ⁇ M apart are cut per animal and stained with Verhoff Elastic stain to produce a modified Verhoff stain.
  • Intimal and medial area measurements are performed with a computerized imaging system. The intimal lesion area and the medial area are determined by measurement of the internal elastic lamina, the external elastic lamina and the vessel/lumen interface.
  • 40-O-(2-hydroxyethyl)-rapamycin reduces neointimal lesion formation at 14 days post ballooning when administered locally as disclosed above at a dose of 10 to 200 ⁇ g/day. Similar good results are obtained when 40-O-(2-hydroxyethyl)-rapamycin is administered in conjunction with dexamethasone (10-250 ⁇ g/day) or a tyrosine kinase inhibitor or an anti-inflammatory agent, e.g. pimecrolimus.
  • a stent according to the invention e.g. delivering a rapamycin derivative having mTOR inhibiting properties.
  • the stents (15 mm) are delivered to the patients (3.0-3.5 mm vessel calibre) and the patients are discharged without clinical complications. At 4 months and 1 year angiographic and IVUS follow-up, no significant neo-intimal hyperplasia is detected.
  • One hundred fifty prospective dialysis patients, who undergo successful insertion of an indwelling, large bore catheter, into a vein are selected for the study. These patients are divided into two groups, and both groups do not differ significantly with sex, distribution of vascular condition or condition of lesions after insertion.
  • One group (about 50 patients) receives rapamycin or a rapamycin derivative having mTOR inhibiting properties in a daily dose of 0.75 to 20 mg (hereinafter identified as group 1), and another group (about 100 patients) does not receive the compound to be tested (hereinafter identified as group H).
  • groups may also be given a calcium antagonist, nitrates and/or anti-platelet agents. These drugs are administered for 3 consecutive months following catheter insertion.
  • the comparative clinical data collected over the observation period of 6 months demonstrate the efficacy of 3 month treatment with rapamycin or a rapamycin derivative, e.g. 40-O-(2- hydroxyethyl)-rapamycin, for the prevention or reduction of vascular access dysfunction in patients after catheter insertion.
  • rapamycin or a rapamycin derivative e.g. 40-O-(2- hydroxyethyl)-rapamycin
  • the stent is manufactered from medical 316LS stainless steel and is composed of a series of cylindrically oriented rings aligned along a common longitudinal axis. Each ring consists of 3 connecting bars and 6 expanding elements.
  • the stent is premounted on a delivery system.
  • the active agent e.g. 40-(2-hydroxyethyl)-rapamycin (0.50 mg/ml) optionally together with 2,6-di-tert.-butyl-4-methylphenol (0.001 mg/ml)
  • the active agent e.g. 40-(2-hydroxyethyl)-rapamycin (0.50 mg/ml) optionally together with 2,6-di-tert.-butyl-4-methylphenol (0.001 mg/ml)
  • the stent is coated with this matrix.
  • a stent is weighed and then mounted for coating. While the stent is rotating, a solution of polylactide glycolide, 0.75 mg/ml of 40-O-(2-hydroxyethyl)-rapamycin, 0.0015 mg/ml 2,6-di- tert.-butyl-4-methylphenol and 1 mg/ml tyrosine kinase C inhibitor dissolved in a mixture of methanol and tetrahydrofuran, is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • the tyrosine kinase inhibitor C may be replaced by taxol, paclitaxel, a VEGF receptor tyrosine kinase inhibitor, a VEGF receptor inhibitor, a compound binding to VEGF, an aldosterone synthetase inhibitor or an aldosterone receptor blocker, or a compound inhibiting the renin-angiotensin system.
  • stable release of 40-O-(2-hydroxyethyl)-rapamycin is meant less than 10% of variation of the drug release.
  • Controlled release techniques used by a person skilled in the art allow an unexpected easy adaptation of the required drug release rate.
  • by selecting appropriate amounts of reactants in the coating mixture it is possible to easily control the bioeffectiveness of the rapamycin or rapamycin derivastive coated stents.

Abstract

Provided are drug delivery systems for the prevention and treatment of proliferative diseases, particularly vascular diseases, comprising rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more active co-agents.

Description

DRUG DELIVERY SYSTEMS FOR THE PREVENTION AND TREATMENT OF VASCULAR DISEASES COMPRISING RAPAMYCIN AND DERIVATIVES THEREOF
The present invention relates to drug delivery systems for the prevention and treatment of proliferative diseases, particularly vascular diseases.
Many humans suffer from circulatory diseases caused by a progressive blockage of the blood vessels that perfuse the heart and other major organs. Severe blockage of blood vessels in such humans often leads to ischemic injury, hypertension, stroke or myocardial infarction. Atherosclerotic lesions which limit or obstruct coronary or periphery blood flow are the major cause of ischemic disease related morbidity and mortality including coronary heart disease and stroke. To stop the disease process and prevent the more advanced disease states in which the cardiac muscle or other organs are compromised, medical revascularization procedures such as percutaneous transluminal coronary angioplasty (PCTA), percutaneous transluminal angioplasty (PTA), atherectomy, bypass grafting or other types of vascular grafting procedures are used.
Re-narrowing (restenosis) of an artherosclerotic coronary artery after various revascularization procedures occurs in 10-80% of patients undergoing this treatment, depending on the procedure used and the aterial site. Besides opening an artery obstructed by atherosclerosis, revascularization also injures endothelial cells and smooth muscle cells within the vessel wall, thus initiating a thrombotic and inflammatory response. Cell derived growth factors such as platelet derived growth factor, infiltrating macrophages, leukocytes or the smooth muscle cells themselves provoke proliferative and migratory responses in the smooth muscle cells. Simultaneous with local proliferation and migration, inflammatory cells also invade the site of vascular injury and may migrate to the deeper layers of the vessel wall. Proliferation/migration usually begins within one to two days post-injury and, depending on the revascularization procedure used, continues for days and weeks.
Both cells within the atherosclerotic lesion and those within the media migrate, proliferate and/or secrete significant amounts of extracellular matrix proteins. Proliferation, migration and extracellular matrix synthesis continue until the damaged endothelial layer is repaired at which time proliferation slows within the intima. The newly formed tissue is called neointima, intimal thickening or restenotic lesion and usually results in narrowing of the vessel lumen. Further lumen narrowing may take place due to constructive remodeling, e.g. vascular remodeling, leading to further intimal thickening or hyperplasia. Furthermore, there are also atherosclerotic lesions which do not limit or obstruct vessel blood flow but which form the so-called "vulnerable plaques". Such atherosclerotic lesions or vulnerable plaques are prone to rupture or ulcerate, which results in thrombosis and thus produces unstable angina pectoris, myocardial infarction or sudden death. Inflamed atherosclerotic plaques can be detected by thermography.
Alternatively, complications associated with vascular access treatment is a major cause of morbidity in many disease states. For example, vascular access dysfunction in hemodialysis patients is generally caused by outflow stenoses in the venous circulation (Schwam S. J., et al., Kidney Int. 36: 707-711 , 1989). Vascular access related morbidity accounts for about 23 percent of all hospital stays for advanced renal disease patients and contributes to as much as half of all hospitalization costs for such patients (Feldman H. I., J. Am. Soc. Nephrol. 7: 523 -535,1996).
Additionally, vascular access dysfunction in chemotherapy patients is generally caused by outflow stenoses in the venous circulation and results in a decreased ability to administer medications to cancer patients. Often the outflow stenoses is so severe as to require intervention.
Additionally, vascular access dysfunction in total parenteral nutrition (TPN) patients is generally caused by outflow stenoses in the venous circulation and results in reduced ability to care for these patients.
Up to the present time, there has not been any effective drug for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter, preferably a large bore catheter, into a vein in a mammal, particularly a human patient.
Survival of patients with chronic renal failure depends on optimal regular performance of dialysis. If this is not possible (for example as a result of vascular access dysfunction or failure), it leads to rapid clinical deterioration and unless the situation is remedied, these patients will die. Hemodialysis requires access to the circulation. The ideal form of hemodialysis vascular access should allow repeated access to the circulation, provide high blood flow rates, and be associated with minimal complications. At present, the three forms of vascular access are native arteriovenous fistulas (AVF), synthetic grafts, and central venous catheters. Grafts are most commonly composed of polytetrafluoroethylene (PTFE) or Gore-Tex. Each type of access has its own advantages and disadvantages. Vascular access dysfunction is the most important cause of morbidity and hospitalization in the hemodialysis population. Venous neointimal hyperplasia characterized by stenosis and subsequent thrombosis accounts for the overwhelming majority of pathology resulting in dialysis graft failure. The most common form of vascular access procedure performed in chronic hemodialysis patients in the United States is the arteriovenous PTFE graft, which accounts for approximately 70% of all hemodialysis access.
Dr. Burnett S. Kelly and Col., (Kidney International, Volume 62; Issue 6; Page 2272 - December 2002) and others have previously shown that venous neointimal hyperplasia (VNH) in the setting of arteriovenous hemodialysis grafts is characterized by smooth muscle cells, neointimal and adventitial microvessels and extracellular matrix components. However, despite a reasonable knowledge of the pathology of VNH, there are still no effective interventions for either the prevention or treatment of hemodialysis vascular access dysfunction. This is particularly unfortunate, as VNH in the setting of hemodialysis grafts appears to be a far more aggressive lesion as compared to the more common arterial neointimal hyperplasia that occurs in peripheral bypass grafts. Compare the 50% one year primary patency in PTFE dialysis access grafts with an 88% five year patency for aortoiliac grafts and a 70 to 80% one year patency for femoro-popliteal grafts. Venous stenoses in the setting of dialysis access grafts also have a poorer response to angioplasty (40% three month survival if thrombosed and a 50% six month survival if not thrombosed) as compared to arterial stenoses. They believe that the lack of effective therapies for VNH and venous stenosis in dialysis grafts such as PTFE dialysis grafts is due to (a) a lack of appreciation of the fact that venous stenosis may be very different from the more common arterial stenosis at the graft-artery anastomosis and (b) the absence of a validated large animal model of VNH to test out novel interventions.
Despite the magnitude of the problem and the enormity of the cost, there are currently no effective therapies for the prevention or treatment of venous neointimal hyperplasia in dialysis grafts.
Accordingly, there is a need for effective treatment and drug delivery systems for revascularization procedure, e.g. preventing and treating intimal thickening or restenosis that occurs after injury, e.g. vascular injury, including e.g. surgical injury, e.g. revascularization- induced injury, e.g. also in heart or other grafts, for a stabilization procedure of vulnerable plaques, or for the prevention or treatment of vascular access dysfunctions. It has now been found that rapamycin and rapamycin derivatives having mTOR inhibiting properties, optionally in conjunction with other active compounds, e.g. antiproliferative compounds, have beneficial effects on above mentioned disorders, diseases or dysfunctions.
Rapamycin is a known macrolide antibiotic produced by Streptomyces hygroscopicus, which inhibits mTOR. By rapamycin derivative having mTOR inhibiting properties is meant a substituted rapamycin, e.g. a 40-substituted-rapamycin or a 16-substituted rapamycin, or a 32-hydrogenated rapamycin, for example a compound of formula I
Figure imgf000005_0001
wherein
Figure imgf000005_0002
R2 is H, -CH2-CH2-OH, 3-hydroxy-2-(hydroxymethyl)-2-methyl-propanoyl or tetrazolyl, and X is =O, (H,H) or (H,OH) provided that R2 is other than H when X is =O and R1 is CH3, or a prodrug thereof when R2 is -CH2-CH2-OH, e.g. a physiologically hydrolysable ether thereof.
Representative rapamycin derivatives of formula I are e.g. 32-deoxorapamycin, 16-pent-2- ynyloxy-32-deoxorapamycin, 16-pent-2-ynyloxy-32(S or R)-dihydro-rapamycin, 16-pent-2- ynyloxy-32(S or R)-dihydro-40-O-(2-hydroxyethyl)-rapamycin, 40-[3-hydroxy-2- (hydroxymethyl)-2-methylpropanoate]-rapamycin (also called CCI779) or 40-epi-(tetrazolyl)- rapamycin (also called ABT578). A preferred compound is e.g. 40-0-(2-hydroxyethyl)-rapamycin disclosed in Example 8 in WO 94/09010, or 32- deoxorapamycin or 16-pent-2-ynyloxy-32(S)-dihydro-rapamycin as disclosed in WO 96/41807. Rapamycin derivatives may also include the so-called rapalogs, e.g. as disclosed in WO 98/02441 and WO01/14387, e.g. AP23573.
According to the invention, rapamycin or a rapamycin derivative having mTOR inhibiting properties may be applied as the sole active ingredient or in conjunction with one or more active co-agents selected from a) an immunosuppressive agent, e.g. a calcineurin inhibitor, e.g. a cyclosporin, for example cyclosporin A, ISA tx 247 or FK506, b) an EDG-receptor agonist having lymphocyte depleting properties, e.g. FTY720 (2-amino- 2-[2-(4-octylphenyI) ethyl]propane-1 ,3-diol in free form or in a pharmaceutically acceptable salt form, e.g. the hydrochloride) or an analogue such as described in WO96/06068 or WO 98/45249, e.g. 2-amino-2-{2-[4-(1-oxo-5- phenylpentyl)phenyl]ethyl}propane-1 ,3-diol or 2-amino-4-(4-heptyloxyphenyl)-2-methyl- butanol in free form or in a pharmaceutically acceptable salt form, c) an anti-inflammatory agent, e.g. a steroid, e.g. a corticosteroid, e.g. dexamethasone or prednisone, a NSAID, e.g. a cyclooxygenase inhibitor, e.g. a cox-2 inhibitor, e.g. celecoxib, rofecoxib, etoricoxib or valdecoxib, an ascomycin, e.g. ASM981 (or pimecrolimus), a cytokine inhibitor, e.g. a lymphokine inhibitor, e.g. an IL-1 , -2 or -6 inhibitor, for example pralnacasan or anakinra, or a TNF inhibitor, for instance Etanercept, or a chemokine inhibitor; d) an anti- thrombotic or anti-coagulant agent, e.g. heparin or a glycoprotein llb/llla inhibitor, e.g. abciximab, eptifibatide or tirofibran; e) an antiproliferative agent, e.g. a microtubule stabilizing or destabilizing agent including but not limited to taxanes, e.g. taxol, paclitaxel or docetaxel, vinca alkaloids, e.g. vinblastine, especially vinblastine sulfate, vincristine especially vincristine sulfate, and vinorelbine, discodermolides or epothilones or a derivative thereof.e.g. epothilone B or a derivative thereof; a protein tyrosine kinase inhibitor, e.g. protein kinase C or Pl(3) kinase inhibitor, for example staurosporin and related small molecules, e.g. UCN-01 , BAY 43-9006, Bryostatin 1 , Perifosine, Limofosine, midostaurin, CGP52421 , RO318220, RO320432, GO 6976, Isis 3521 , LY333531 , LY379196, SU5416, SU6668, AG1296, imatinib, etc.; a compound or antibody which inhibits the PDGF receptor tyrosine kinase or a compound which binds to PDGF or reduces expression of the PDGF receptor e.g. a N-phenyl-2- pyrimidine-amine derivative, e.g. imatinib, CT52923, RP-1776, GFB-111 , a pyrrolo[3,4-c]- beta-carboline-dione, etc.; a compound or antibody which inhibits the EGF receptor tyrosine kinase or a compound which binds to EGF or reduces expression of the EGF receptor e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the compound of ex. 39, or in EP 0 564 409, WO 99/03854, EP 0520722, EP 0 566 226, EP 0 787 722, EP 0 837 063, US 5,747,498, WO 98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and, especially, WO 96/30347 (e.g. compound known as CP 358774), WO 96/33980 (e.g. compound ZD 1839, Iressa) and WO 95/03283 (e.g. compound ZM105180); e.g. trastuzumab (HerpetinR), cetuximab, OSI-774, CI-1033, EKB-569, GW-2016, E1.1 , E2.4, E2.5, E6.2, E6.4, E2.11 , E6.3 or E7.6.3, retinoic acid, alpha-, gamma- or delta-tocopherol or alpha-, gamma- or delta-tocotrienol, or compounds affecting GRB2, IMC-C225; or a compound or antibody which inhibits the VEGF receptor tyrosine kinase or a VEGF receptor or a compound which binds to VEGF, e.g. proteins, small molecules or monoclonal antibodies generically and specifically disclosed in WO 98/35958, e.g. 1-(4- chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g. the succinate, or in WO 00/09495, WO 00/27820, WO 00/59509, WO 98/11223, WO 00/27819, WO 00/37502, WO 94/10202 and EP 0 769 947, those as described by M. Prewett et al in Cancer Research 59 (1999) 5209-5218, by F. Yuan et al in Proc. Natl. Acad. Sci. USA, vol. 93, pp. 14765-14770, Dec. 1996, by Z. Zhu et al in Cancer Res. 58, 1998, 3209-3214, by J. Mordenti et al in Toxicologic Pathology, Vol. 27, no. 1 , pp 14-21 , 1999, Angiostatin™, described by M. S. O'Reilly et al, Cell 79, 1994, 315- 328, Endostatin™, described by M. S. O'Reilly et al, Cell 88, 1997, 277-285, anthranilic acid amides, ZD4190; ZD6474, SU5416, SU6668 or anti-VEGF antibodies or anti-VEGF receptor antibodies, e.g. RhuMab; f) a statin, e.g. having HMG-CoA reductase inhibition activity, e.g. fluvastatin, lovastatin, simvastatin, pravastatin, atorvastatin, cerivastatin, pitavastatin, rosuvastatin or nivastatin; g) a compound, protein, growth factor or compound stimulating growth factor production that will enhance endothelial regrowth of the luminal endothelium, e.g. FGF, IGF; h) a matrix metalloproteinase inhibitor, e.g. batimistat, marimistat, trocade, CGS 27023, RS 130830 or AG3340; k) a modulator (i.e. antagonists or agonists) of kinases, e.g. JNK, ERK1/2, MAPK or STAT;
I) a compound stimulating the release of (NO) or a NO donor, e.g. diazeniumdiolates, S- nitrosothiols, mesoionic oxatriazoles, isosorbide or a combination thereof, e.g. mononitrate and/or dinitrate; m)a somatostatin analogue, e.g. octreotide, lanreotide, vapreotide or a cyclohexapeptide having somatostatin agonist properties, e.g. cyclo[4-(NH2-C2H4-NH-CO-O)Pro-Phg-DTrp- Lys-Tyr(Bzl)-Phe]; or a modified GH analogue chemically linked to PEG, e.g. Pegvisomant; n) an altosterone synthetase inhibitor or aldosterone receptor blocker, e.g. eplerenone, or a compound inhibiting the renin-angiotensin system, e.g. a renin inhibitor, e.g. SPP100, an ACE inhibitor, e.g. captopril, enalapril, lisinopril, fosinopril, benazepril, quinapril, ramipril, imidapril, perindopril erbumine, trandolapril or moexipril, or an ACE receptor blocker, e.g. losartan, irbesartan, candesartan cilexetil, valsartan or olmesartan medoxomil; o) mycophenolic acid or a salt thereof, e.g. sodium mycophenolate, or a prodrug thereof, e.g. mycophenolate mofetil.
Are comprised also in the above list the pharmaceutically acceptable salts, the corresponding racemates, diastereoisomers, enantiomers, tautomers as well as the corresponding crystal modifications of above disclosed compounds where present, e.g. solvates, hydrates and polymorphs.
By antibody is meant monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
A pharmaceutical combination comprising i) rapamycin or a rapamycin derivative having mTOR properties and ii) pimecrolimus, also form part of the present invention.
According to the invention, rapamycin is preferably locally administered or delivered in conjunction with one or more co-agents selected from b), e), f), g), h), k), m), n), o), a cox-2 inhibitor, a cytokine inhibitor or a chemokine inhibitor, as defined above.
In accordance with the particular findings of the present invention, there is provided 1.1 A method for preventing or treating smooth muscle cell proliferation and migration in hollow tubes, or increased cell proliferation or decreased apoptosis or increased matrix deposition in a subject in need thereof, comprising local administration of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.2 A method for the prevention or treatment of intimal thickening in vessel walls comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
Preferably the intimal thickening in vessel walls is stenosis, restenosis, e.g. following revascularization or neovascularization, and/or inflammation and/or thrombosis.
1.3 A method for the prevention or treatment of inflammatory disorders, e.g. T-cell induced inflammation, in hollow tubes comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.4 A method for stabilizing vulnerable plaques in blood vessels of a subject in need of such a stabilization comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.5 A method as defined in 1.1 to 1.4 associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of rapamycin or a derivative thereof having mTOR inhibiting properties, e.g. a compound of formula I. Preferably rapamycin or the derivative thereof, e.g. of formula I, is administered orally.
Alternatively, a method as defined in 1.1 to 1.4 may be associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of the co- agent. 1.6 A method for preventing or treating restenosis in diabetic patients comprising administering to said patients a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.7 A method for preventing or treating restenosis in diabetic patients comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.8 A method comprising a combination of method steps as disclosed above under 1.6 and 1.7.
1.9 A method for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter, preferably a large bore catheter, into a vein or artery, or actual treatment, in a subject in need thereof, which comprises administering to the subject rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above, or a controlled delivery from a drug delivery medical device or system of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
Preferably the invention relates to the prevention or reduction of vascular access dysfunction in hemodialysis.
1.10 A method for the stabilization or repair of arterial or venous aneurisms in a subject comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.11 A method for the prevention or treatment of anastomic hyperplasia in a subject comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above. 1.12 A method for the prevention or treatment of arterial, e.g. aortic, by-pass anastomosis in a subject comprising the controlled delivery from any catheter-based device, intraluminal medical device or adventitial medical device of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents, e.g. as disclosed above.
1.13 A method as defined in 1.9 to 1.12 associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of rapamycin or a derivative thereof, e.g. a compound of formula I. Preferably rapamycin or the derivative thereof, e.g. of formula I, is administered orally.
Alternatively, a method as defined in 1.9 to 1.12 may be associated, simultaneously or sequentially, with the administration of a therapeutically effective amount of the co- agent.
2.1 A drug delivery device or system comprising i) a medical device adapted for local application or administration in hollow tubes, e.g. a catheter-based delivery device or a medical device intraluminal or outside of hollow tubes such as an implant or a sheath placed within the adventitia, and ii) a therapeutic dosage of a rapamycin derivative having mTOR inhibiting properties or rapamycin, optionally in conjunction with a therapeutic dosage of one or more other active co-agents, e.g. as disclosed above, each being releasably affixed to the delivery device or system.
2.2 A device as defined herein for use in any method as defined under 1.1 to 1.12.
3.1 Use of rapamycin or a rapamycin derivative having mTOR inhibiting properties in any of the method as defined under 1.4, 1.6 or 1.9 optionally in conjunction with one or more other active co-agent, or in the manufacture of a medicament for use in any of the method as defined under 1.4, 1.6 or 1.9 optionally in conjunction with one or more other active co-agent.
3.2 Use of a rapamycin derivative having mTOR inhibiting properties, optionally in combination with an active co-agent as defined herein, in the manufacture of a device as defined herein for use in any method as defined under 1.1 to 1.12.
3.3 Use of indwelling shunt, fistula or catheter coated by, impregnated with or incorporating rapamycin or a rapamycin derivative having mTOR inhibiting properties (i.e. being releasably affixed to the medical device) as described herein, for the manufacture of a medicament for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter into a vein or artery, in a subject in need thereof.
4. A pharmaceutical composition for use in any method as defined under 1.4, 1.6 or 1.9 comprising rapamycin or a derivative thereof having mTOR properties, e.g. CCI779, ABT578, a rapalog or a compound of formula I, together with one or more pharmaceutically acceptable diluents or carriers therefor.
A local delivery device or system according to the invention can be used to reduce stenosis or restenosis as an adjunct to revascularization, bypass or grafting procedures performed in any vascular location including coronary arteries, carotid arteries, renal arteries, peripheral arteries, cerebral arteries or any other arterial or venous location, to reduce anastomic stenosis or hyperplasia including in the case of arterial-venous dialysis access with or without PTFE or e.g. Gore-Tex grafting and with or without stenting, or in conjunction with any other heart or transplantation procedures, or congenital vascular interventions.
In a preferred embodiment, the present invention also provides a drug delivery system or device as disclosed above additionally comprising a source delivering a therapeutic dosage of a compound or antibody which inhibits the PDGF receptor tyrosine kinase or a compound which binds to PDGF or reduces expression of the PDGF receptor e.g. as disclosed above, a compound or antibody which inhibits the EGF receptor tyrosine kinase or a compound which binds to EGF or reduces expression of the EGF receptor e.g. as disclosed above, a compound or antibody which inhibits the VEGF receptor tyrosine kinase or a VEGF receptor or a compound which binds to VEGF, e.g. as disclosed above, each being releasably affixed to the catheter-based delivery device or medical device.
Rapamycin or rapamycin derivative having mTOR inhibiting properties will be referred to hereinafter as "active agent". "Drug(s)" means active agent or the active agent and the active co-agent.
The local administration preferably takes place at or near the lesion sites, e.g. vascular lesion sites.
The local administration may be by one or more of the following routes: via catheter or other intravascular delivery system, intranasally, intrabronchially, interperitoneally or eosophagal, or via delivery balloons used in the musculature. Hollow tubes include natural body vessels or ducts, e.g. circulatory system vessels such as blood vessels (arteries or veins), tissue lumen, lymphatic pathways, digestive tract including alimentary duct, e.g. esophagus or biliary ducts, respiratory tract, e.g. trachea, excretory system tubes, e.g. intestines, ureters or urethra-prostate, reproductive system tubes and ducts, body cavity tubes, etc. Local administration or application of the drug(s) may afford concentrated delivery of said drug(s), achieving tissue levels in target tissues not otherwise obtainable through other administration route. Additionally local administration or application may reduce the risk of remote or systemic toxicity. Preferably the smooth muscle cell proliferation or migration is inhibited or reduced according to the invention immediately proximal or distal to the locally treated or stented area.
Means for local delivery of the drug(s) to hollow tubes can be by physical delivery of the drug(s) either internally or externally to the hollow tube. Local drug(s) delivery includes catheter delivery systems, local injection devices or systems or indwelling devices. Such devices or systems would include, but not be limited to, stents, coated stents, endolumenal sleeves, stent-grafts, sheathes, balloons, liposomes, controlled release matrices, polymeric endoluminal paving, or other endovascular devices, embolic delivery particles, cell targeting such as affinity based delivery, internal patches around the hollow tube, external patches around the hollow tube, hollow tube cuff, external paving, external stent sleeves, and the like. See, Eccleston et al. (1995) Interventional Cardiology Monitor 1 :33-40-41 , Slepian, N.J. (1996) Interventional Cardiol. 1 :103-116, or Regar E, Sianos G, Serruys PW, Stent development and local drug delivery, Br Med Bull 2001 ,59:227-48, which disclosures are herein incorporated by reference.
Preferably the delivery device or system fulfils pharmacological, pharmacokinetic and mechanical requirements. Preferably it also is suitable for sterilisation.
The stent according to the invention can be any stent, including self-expanding stent, or a stent that is radially expandable by inflating a balloon or expanded by an expansion member, or a stent that is expanded by the use of radio frequency which provides heat to cause the stent to change its size. A stent composed of or coated with a polymer or other biocompatible materials, e.g. porous ceramic, e.g. nanoporous ceramic, into which the drug(s) has been impregnated or incorporated can be used. Stents can be biodegradable or can be made of metal or alloy, e.g. Ni and Ti, or another stable substance when intented for permanent use. The drug(s) may also be entrapped into the metal of the stent or graft body which has been modified to contain micropores or channels. Also lumenal and/or ablumenal coating or external sleeve made of polymer or other biocompatible materials, e.g. as disclosed below, that contain the drug(s) can also be used for local delivery.
By "biocompatible" is meant a material which elicits no or minimal negative tissue reaction including e.g. thrombus formation and/or inflammation.
Stents may commonly be used as a tubular structure left inside the lumen of a duct to relieve an obstruction. They may be inserted into the duct lumen in a non-expanded form and are then expanded autonomously (self-expanding stents) or with the aid of a second device in situ, e.g. a catheter-mounted angioplasty balloon which is inflated within the stenosed vessel or body passageway in order to shear and disrupt the obstructions associated with the wall components of the vessel and to obtain an enlarged lumen. Alternatively, stents being easily deformed at lower temperature to be inserted in the hollow tubes may be used: after deployment at site, such stents recover their original shape and exert a retentive and gentle force on the internal wall of the hollow tubes, e.g. of the esophagus or trachea.
The drug(s) may be incorporated into or affixed to the stent in a number of ways and utilizing any biocompatible materials; it may be incorporated into e.g. a polymer or a polymeric matrix and sprayed onto the outer surface of the stent. A mixture of the drug(s) and the polymeric material may be prepared in a solvent or a mixture of solvents and applied to the surfaces of the stents also by dip-coating, brush coating and/or dip/spin coating, the solvent (s) being allowed to evaporate to leave a film with entrapped drug(s). In the case of stents where the drug(s) is delivered from micropores, struts or channels, a solution of a polymer may additionally be applied as an outlayer to control the drug(s) release; alternatively, the active agent may be comprised in the micropores, struts or channels and the active co-agent may be incorporated in the outlayer, or vice versa. The active agent may also be affixed in an inner layer of the stent and the active co-agent in an outer layer, or vice versa. The drug(s) may also be attached by a covalent bond, e.g. esters, amides or anhydrides, to the stent surface, involving chemical derivatization. The drug(s) may also be incorporated into a biocompatible porous ceramic coating, e.g. a nanoporous ceramic coating. The medical device of the invention is configured to release the active co-agent concurrent with or subsequent to the release of the active agent.
Examples of polymeric materials include hydrophilic, hydrophobic or biocompatible biodegradable materials, e.g. polycarboxylic acids; cellulosic polymers; starch; collagen; hyaluronic acid; gelatin; lactone-based polyesters or copolyesters, e.g. polylactide; polyglycolide; polylactide-glycolide; polycaprolactone; polycaprolactone-glycolide; poly(hydroxybutyrate); poly(hydroxyvalerate); polyhydroxy(butyrate-co-valerate); polyglycolide-co-trimethylene carbonate; poly(diaxanone); polyorthoesters; polyanhydrides; polyaminoacids; polysaccharides; polyphospoeters; polyphosphoester-urethane; polycyanoacrylates; polyphosphazenes; poly(ether-ester) copolymers, e.g. PEO-PLLA, fibrin; fibrinogen; or mixtures thereof; and biocompatible non-degrading materials, e.g. polyurethane; polyolefins; polyesters; polyamides; polycaprolactame; polyimide; polyvinyl chloride; polyvinyl methyl ether; polyvinyl alcohol or vinyl alcohol/olefin copolymers, e.g. vinyl alcohol/ethylene copolymers; polyacrylonitrile; polystyrene copolymers of vinyl monomers with olefins, e.g. styrene acrylonitrile copolymers, ethylene methyl methacrylate copolymers; polydimethylsiloxane; poly(ethylene-vinylacetate); acrylate based polymers or coplymers, e.g. polybutylmethacrylate, poly(hydroxyethyl methylmethacrylate); polyvinyl pyrrolidinone; fluorinated polymers such as polytetrafluoethylene; cellulose esters e.g. cellulose acetate, cellulose nitrate or cellulose propionate; or mixtures thereof.
When a polymeric matrix is used, it may comprise 2 layers, e.g. a base layer in which the drug(s) is/are incorporated, e.g. ethylene-co-vinylacetate and polybutylmethacrylate, and a top coat, e.g. polybutylmethacrylate, which is drug(s)-free and acts as a diffusion-control of the drug(s). Alternatively, the active agent may be comprised in the base layer and the active co-agent may be incorporated in the outlayer, or vice versa. Total thickness of the polymeric matrix may be from about 1 to 20μ or greater.
According to the method of the invention or in the device or system of the invention, the drug(s) may elute passively, actively or under activation, e.g. light-activation.
The drug(s) elutes from the polymeric material or the stent over time and enters the surrounding tissue, e.g. up to ca. 1 month to 1 year. The local delivery according to the present invention allows for high concentration of the drug(s) at the disease site with low concentration of circulating compound. The amount of drug(s) used for local delivery applications will vary depending on the compounds used, the condition to be treated and the desired effect. For purposes of the invention, a therapeutically effective amount will be administered; for example, the drug delivery device or system is configured to release the active agent and/or the active co-agent at a rate of 0.001 to 200 μg/day. By therapeutically effective amount is intended an amount sufficient to inhibit cellular proliferation and resulting in the prevention and treatment of the disease state. Specifically, for the prevention or treatment of restenosis e.g. after revascularization, or antitumor treatment, local delivery may require less compound than systemic administration. A contemplated treatment period for use in the prevention or reduction of vascular access dysfunction of the present invention is about 85, e.g. 70, preferably 50, e.g. 28, more preferably 28 days in association with the insertion or repair of an indwelling shunt, fistula or catheter, or actual treatment.
A preferred method of use in the prevention or reduction of vascular access dysfunction is a method for preventing or reducing vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling access clotting shunt, fistula or catheter associated with insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, in dialysis patients.
A preferred method of use in the prevention or reduction of vascular access dysfunction is a method for preventing or reducing vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling vascular access shunt, fistula or catheter associated with insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, in cancer patients.
A preferred method of use in the prevention or reduction of vascular access dysfunction is a method for preventing or reducing vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling vascular access shunt, fistula or catheter associated with insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, in total parenteral nutrition (TPN) patients.
By "prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter" as used herein, is meant that the incidence of vascular thrombosis and/or fistula failure and/or shunt failure and/or vascular access clotting and/or stenosis and/or restenosis and/or the need for declotting an indwelling vascular access shunt, fistula or catheter in patients treated according to the invention collected over the observation period are prevented or reduced in comparison to untreated patients.
By "in association with the insertion or repair of an indwelling shunt, fistula or catheter" as used herein, is meant that the treatment according to the invention can commence immediately, for example within 4 to 8 hours, after insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, such as dialysis treatment; within a few days, for example about 7 days, preferably about 1 or 2 days, after insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, such as dialysis treatment; or for a period of days, for example about 30 days, preferably about 14 days, preferably about 7 days, prior to insertion or repair of the indwelling shunt, fistula or catheter, or actual treatment, such as dialysis treatment. Also contemplated within the phrase "in association with the insertion or repair of an indwelling shunt, fistula or catheter" is a dosing protocol in which a dose or several doses, are skipped, for example in the morning of or on the day of insertion, repair or treatment. Also contemplated within the phrase "in association with the insertion or repair of an indwelling shunt, fistula or catheter" is a dosing protocol in which a day of drug treatment or several days of drug treatment, are skipped.
Included in term "treatment", when used herein to refer surgical procedures, are procedures selected from access surgery, placement of fistula or shunt, catheter insertion, actual disease treatment, such as dialysis treatment, and declotting of an access shunt, fistula or catheter. Further, treatment for insertion access also includes repair/revision of the access. For example, a patient experiencing a failure in a dialysis access shunt will have the access repaired, for instance, by angioplasty.
By the term "collected over the observation period" as used herein, means a period of up to or about 12 months, preferably 12 months.
When rapamycin or a rapamycin derivative having mTOR inhibiting properties is administered systemically or is additionally administered by systemic application, e.g. in the prevention or reduction of vascular access dysfunction, according to the invention.daily dosages required in practicing the method of the present invention will vary depending upon, for example, the compound used, the host, the mode of administration and the severity of the condition to be treated. A preferred daily dosage range is about from 0.1 to 25 mg as a single dose or in divided doses. Suitable daily dosages for patients are on the order of from e.g. 0.1 to 25 mg p.o. The compound may be administered by any conventional route, in particular enterally, e.g. orally, e.g. in the form of tablets, capsules, drink solutions, nasally, pulmonary (by inhalation) or parenterally, e.g. in the form of injectable solutions or suspensions. Suitable unit dosage forms for oral administration comprise from ca. 0.05 to 12.5 mg, usually 0.25 to 10 mg compound, together with one or more pharmaceutically acceptable diluents or carriers therefor.
Preferred combinations according to the invention are those comprising a compound of formula I, e.g. 40-0-(2-hydroxyethyl)-rapamycin or 32-deoxorapamycin, or CCI-779, ABT578 or a rapalog in conjunction or association with a compound having antiproliferative properties, e.g. taxol, paclitaxel, docetaxel, an epothilone, a tyrosine kinase inhibitor, e.g. a protein kinase C or Pl(3) kinase inhibitor, for example staurosporin or a related small molecule, a PDGF receptor tyrosine kinase inhibitor, a PDGF receptor inhibitor, a compound binding to PDGF, e.g. imatinib, a VEGF receptor tyrosine kinase inhibitor, a VEGF receptor inhibitor, a compound binding to VEGF, e.g. 1-(4-chloroanilino)-4-(4- pyridylmethyl)phtalazine, a cox-2 inhibitor, an ascomycin, e.g. pimecrolimus, or a calcineurin inhibitor, e.g. CysA, ISA tx 247 or FK506. A combination of rapamycin or a rapamycin derivative as mentioned above with a compound having anti-inflammatory properties, pimecrolimus, or an EDG-receptor agonist having lymphocyte depleting properties, has particularly beneficial effects when used in the treatment or prevention of restenosis in diabetic patients. A combination of rapamycin or a rapamycin derivative as mentioned above with a statin or an aldosterone synthetase inhibitor or an aldosterone receptor blocker, or with a compound inhibiting the renin-angiotensin system has also beneficial properties; such a combination also forms part of the invention.
Rapamycin or the rapamycin derivative having mTOR inhibiting properties may also be applied to the drug delivery device or system in admixture with an antioxidant, e.g. 2,6-di- tert.-butyl-4-methylphenol, e.g. at an amount up to 0.5% by weight, preferably 0.2% by weight.
Utility of the drug(s) may be demonstrated in animal test methods as well as in clinic, for example in accordance with the methods hereinafter described.
A1. Inhibition of late neointimal lesion formation in the 28 day rat carotid artery balloon injury model
Numerous compounds have been shown to inhibit intimal lesion formation at 2 weeks in the rat ballooned carotid model, while only few compounds prove effective at 4 weeks. Compounds of formula I are tested in the following rat model.
Rats are dosed orally with placebo or a compound of formula I. Daily dosing starts 3 days prior to surgery and continues for 31 days. Rat carotid arteries are balloon injured using a method described by Clowes et al. Lab. Invest. 1983;49;208-215. Following sacrifice at 28 days post-balloon injury, carotid arteries are removed and processed for histologic and morphometric evaluation. In this assay the compounds of formula I, e.g. 40-O-(2- hydroxyethyl)-rapamycin, significantly reduce neointimal lesion formation at 28 days following balloon injury when administered at a dose of from 0.5 to 2.0 mg/kg. For example for 40-O-(2-hydroxyethyl)-rapamycin administered at 0.5, 1.0, and 2.0 mg/kg , the percent inhibition is similar at all three doses: inhibition is 31% at the lowest dose (0.5 mg/kg) and 39% at the highest dose (2.0 mg/kg). Compounds of formula I, e.g. 40-O-(2-hydroxyethyl)- rapamycin, have the beneficial effect to inhibit lesions at 4 weeks post-ballooning.
A.2 Inhibition of restenosis at 28 days in the rabbit iliac stent model
A combined angioplasty and stenting procedure is performed in New Zealand White rabbit iliac arteries. Iliac artery balloon injury is performed by inflating a 3.0 x 9.0 mm angioplasty balloon in the mid-portion of the artery followed by "pull-back" of the catheter for 1 balloon length. Balloon injury is repeated 2 times, and a 3.0 x 12 mm stent is deployed at 6 atm for 30 seconds in the iliac artery. Balloon injury and stent placement is then performed on the contralateral iliac artery in the same manner. A post-stent deployment angiogram is performed. All animals receive oral aspirin 40 mg/day daily as anti-platelet therapy and are fed standard low-cholesterol rabbit chow. Twenty-eight days after stenting, animals are anesthetized and euthanized and the arterial tree is perfused at 100 mmHg with lactated Ringer's for several minutes, then perfused with 10% formalin at 100 mmHg for 15 minutes. The vascular section between the distal aorta and the proximal femoral arteries is excised and cleaned of periadventitial tissue. The stented section of artery is embedded in plastic and sections are taken from the proximal, middle, and distal portions of each stent. All sections are stained with hematoxylin-eosin and Movat pentachrome stains. Computerized planimetry is performed to determine the area of the internal elastic lamina (IEL), external elastic lamina (EEL) and lumen. The neointima and neointimal thickness is measured both at and between the stent struts. The vessel area is measured as the area within the EEL. Data are expressed as mean ± SEM. Statistical analysis of the histologic data is accomplished using analysis of variance (ANOVA) due to the fact that two stented arteries are measured per animal with a mean generated per animal. A P < 0.05 is considered statistically significant.
A compound of formula I, e.g. 40-O-(2-hydroxyethyl)-rapamycin, is administered orally by gavage at a loading dose of 1.5 mg/kg one day prior to stenting, then dosed at 0.75 mg/kg/day from the day of stenting until day 27 post-stenting. In this model, the treatment with the compounds of formula I results in a marked reduction in the extent of restenotic lesion formation: for example, the treatment with 40-O-(2-hydroxyethyl)-rapamycin produces a significant (P< 0.03) reduction in neointimal thickness (40% reduction), neointimal area (24% reduction), and percent arterial stenosis (26% reduction) with a significant 32% increase in lumen area. There is extensive neointimal formation in placebo-treated animals at 28 days, with the lesions consisting of abundant smooth muscle cells in proteoglycan/collagen matrix and apparent full endothelial healing. In the majority of arterial segments from the animals treated with 40-O-(2-hydroxyethyl)-rapamycin, the intima is well healed, characterized by a compact neointimal consisting of smooth muscle cells and endothelium both over stent struts and between struts. Scanning electron microscopic analysis shows that stented arteries from the animals treated with 40-O-(2-hydroxyethyl)- rapamycin (n = 4 arteries) was 84% endothelialized.
A.3 Inhibition of restenosis at 14 days in the rat carotid stent model
Male Sprague Dawley rats weighing 250 to 500 mg are housed individually and allowed to acclimate prior to surgery. All animals receive standard rat chow and water ad libitum. Group size is 12 animals per group.
The drug(s) administration is perivascular. A segment of ballooned carotid is encircled with a 1 cm length of silastic tubing (0.25 inch inside diameter, 0.47 inch outside diameter) to which is attached a catheter which feeds into an osmotic pump containing either compound or vehicle. This delivery system provides continuous, local delivery to the adventitia of the wrapped portion of vessel. Local drug(s) administration ranges between 5 μg and 10 mg, locally per day, depending on the solubility characteristics of the individual compounds.
The left common carotid arteries are denuded of endothelium using a 2F Fogarty catheter as previously described (Prescott Am. J. Pathol. (1991) 139:1291-1296, Clowes et al., (1983) Lab Invest. 49:327-333). Briefly, rats are anesthetized with ketamine (50 mg/ml) and rompun (10mg/ml) administered intraperitoneally at a dose of 1.5 ml/kg. A midline incision is made in the neck to expose the left external and common carotid arteries. The balloon is inserted into the common carotid artery via the left external branch, inflated with saline, and pulled back three times through the lumen with a rotating motion to ensure maximal endothelial denudation. The catheter is then removed, the external carotid artery is ligated and the wound is closed. Each animal is given an injection of the antibiotic Bacillin (200.000 units/kg) and the analgesic Buprenophine (0.06 mg/kg) immediately following surgery.
Animals are killed at 14 days post-balloon injury. One half hour before termination blood is collected, centrifuged, and stored at -20CC for analysis of circulating levels of compound. 5% Evans Blue is then injected intravenously to allow discrimination of re-endothelialized areas at the time of histologic processing. Animals are killed by administration of an overdose of ketamine and rompun, the osmotic pumps are recovered and the volume of remaining content is recorded to ensure that pump failure has not occurred. Carotid arteries are excised and immersion fixed, then transferred to Ringer's solution. Two samples from control blue region of each left carotid artery are imbedded in paraffin. A minimum of six carotid sections, 20 μM apart are cut per animal and stained with Verhoff Elastic stain to produce a modified Verhoff stain. Intimal and medial area measurements are performed with a computerized imaging system. The intimal lesion area and the medial area are determined by measurement of the internal elastic lamina, the external elastic lamina and the vessel/lumen interface.
In this assay, 40-O-(2-hydroxyethyl)-rapamycin reduces neointimal lesion formation at 14 days post ballooning when administered locally as disclosed above at a dose of 10 to 200μg/day. Similar good results are obtained when 40-O-(2-hydroxyethyl)-rapamycin is administered in conjunction with dexamethasone (10-250μg/day) or a tyrosine kinase inhibitor or an anti-inflammatory agent, e.g. pimecrolimus.
A4. Treatment of Angina Pectoris Patients
25 patients with angina pectoris are treated with a stent according to the invention, e.g. delivering a rapamycin derivative having mTOR inhibiting properties. The stents (15 mm) are delivered to the patients (3.0-3.5 mm vessel calibre) and the patients are discharged without clinical complications. At 4 months and 1 year angiographic and IVUS follow-up, no significant neo-intimal hyperplasia is detected.
In this trial, when a stent delivering rapamycin or a derivative thereof having mTOR inhibiting properties in conjunction with pimecrolimus or midostaurin is used, beneficials effects are obtained.
A5. Prevention or reduction of vascular access dysfunction in association with the insertion of an indwelling catheter into the vein of a patient
One hundred fifty prospective dialysis patients, who undergo successful insertion of an indwelling, large bore catheter, into a vein are selected for the study. These patients are divided into two groups, and both groups do not differ significantly with sex, distribution of vascular condition or condition of lesions after insertion. One group (about 50 patients) receives rapamycin or a rapamycin derivative having mTOR inhibiting properties in a daily dose of 0.75 to 20 mg (hereinafter identified as group 1), and another group (about 100 patients) does not receive the compound to be tested (hereinafter identified as group H). In addition, patients may also be given a calcium antagonist, nitrates and/or anti-platelet agents. These drugs are administered for 3 consecutive months following catheter insertion. The comparative clinical data collected over the observation period of 6 months demonstrate the efficacy of 3 month treatment with rapamycin or a rapamycin derivative, e.g. 40-O-(2- hydroxyethyl)-rapamycin, for the prevention or reduction of vascular access dysfunction in patients after catheter insertion.
The following examples are illustrative of the invention without limitating it.
Example 1
The stent is manufactered from medical 316LS stainless steel and is composed of a series of cylindrically oriented rings aligned along a common longitudinal axis. Each ring consists of 3 connecting bars and 6 expanding elements. The stent is premounted on a delivery system. The active agent, e.g. 40-(2-hydroxyethyl)-rapamycin (0.50 mg/ml) optionally together with 2,6-di-tert.-butyl-4-methylphenol (0.001 mg/ml), is incorporated into a polymer matrix based on a semicrystalline ethylene-vinyl alcohol copolymer. The stent is coated with this matrix.
Example 2
A stent is weighed and then mounted for coating. While the stent is rotating, a solution of polylactide glycolide, 0.75 mg/ml of 40-O-(2-hydroxyethyl)-rapamycin, 0.0015 mg/ml 2,6-di- tert.-butyl-4-methylphenol and 1 mg/ml tyrosine kinase C inhibitor dissolved in a mixture of methanol and tetrahydrofuran, is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
The tyrosine kinase inhibitor C may be replaced by taxol, paclitaxel, a VEGF receptor tyrosine kinase inhibitor, a VEGF receptor inhibitor, a compound binding to VEGF, an aldosterone synthetase inhibitor or an aldosterone receptor blocker, or a compound inhibiting the renin-angiotensin system.
Example 3
Four 2 cm pieces of coated stents as described above are placed into 100 ml of phosphate buffer solution (PBS) having a pH of 7.4. Another 4 pieces from each series are placed into 100 ml polyethylene glycol (PEG)/water solution (40/60 v/v, MW of PEG=400). The stent pieces are incubated at 37°C in a shaker. The buffer and PEG solutions are changed daily and different assays are performed on the solution to determine the released 40-O-(2- hydroxyethyl)-rapamycin concentrations. Such assays can show a stable release of 40-O-(2- hydroxyethyl)-rapamycin from coated stents for more than 45 days. By the term "stable release of 40-O-(2-hydroxyethyl)-rapamycin" is meant less than 10% of variation of the drug release. Controlled release techniques used by a person skilled in the art allow an unexpected easy adaptation of the required drug release rate. Thus, by selecting appropriate amounts of reactants in the coating mixture it is possible to easily control the bioeffectiveness of the rapamycin or rapamycin derivastive coated stents.

Claims

1. A pharmaceutical composition for stabilizing vulnerable plaques in blood vessels of a subject in need of such a stabilization, for preventing or treating restenosis in diabetic patients, or for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter in a subject in need of a dialysis, comprising rapamycin or a derivative thereof having mTOR properties, together with one or more pharmaceutically acceptable diluents or carriers therefor.
2. Use of rapamycin or a derivative thereof having mTOR inhibiting properties for the manufacture of a pharmaceutical for stabilizing vulnerable plaques in blood vessels of a subject in need of such a stabilization, for preventing or treating restenosis in diabetic patients, or for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter in a subject in need of dialysis.
3. Use or composition according to claim 1 or 2, for use in conjunction with one or more active co-agents.
4. A drug delivery device or system comprising i) a medical device adapted for local application or administration in hollow tubes and ii) a therapeutic dosage of a rapamycin derivative having mTOR inhibiting properties or rapamycin, in conjunction with a therapeutic dosage of one or more active co-agents selected from an EDG-receptor agonist having lymphocyte depleting properties, a cox-2 inhibitor, pimecrolimus, a cytokine inhibitor, a chemokine inhibitor, an antiproliferative agent, a statin, a protein, growth factor or compound stimulating growth factor production that will enhance endothelial regrowth of the luminal endothelium, a matrix metalloproteinase inhibitor, a somatostatin analogue, an aldosterone synthetase inhibitor or aldosterone receptor blocker and a compound inhibiting the renin- angiotensin system, each being releasably affixed to the drug delivery device or system.
5. A drug delivery device or system comprising i) a medical device adapted for local application or administration in hollow tubes and ii) a therapeutic dosage of a rapamycin derivative having mTOR inhibiting properties, in conjunction with a therapeutic dosage of one or more active co-agents selected from a calcineurin inhibitor and mycophenolic acid or a salt thereof or prodrug thereof, each being releasably affixed to the drug delivery device or system.
6. A drug delivery device or system according to claim 4 or 5, for preventing or treating smooth muscle cell proliferation and migration in hollow tubes, or increased cell proliferation or decreased apoptosis or increased matrix deposition in a subject in need thereof.
7. A drug delivery device or system according to claim 4 or 5, for stabilizing vulnerable plaques in blood vessels, for preventing or treating restenosis in diabetic patients or for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or cathether in a dialysis patient.
8. A drug delivery device or system comprising i) a medical device adapted for local application or administration in hollow tubes and ii) a therapeutic dosage of a rapamycin derivative having mTOR inhibiting properties or rapamycin, each being releasably affixed to the catheter-based delivery device or system, for use in stabilizing vulnerable plaques in blood vessels, for preventing or treating restenosis in diabetic patients or for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or cathether in a dialysis patient.
9. A combination of rapamycin or a rapamycin derivative having mTOR inhibiting properties with pimecrolimus, an aldosterone synthetase inhibitor or an aldosterone receptor blocker, or with a compound inhibiting the renin-angiotensin system.
10. A method for preventing or treating smooth muscle cell proliferation and migration in hollow tubes, or increased cell proliferation or decreased apoptosis or increased matrix deposition in a subject in need thereof, comprising local administration of a therapeutically effective amount of a rapamycin derivative having mTOR inhibiting properties or rapamycin in conjunction with one or more active co-agents selected from an EDG-receptor agonist having lymphocyte depleting properties, a cox-2 inhibitor, pimecrolimus, a cytokine inhibitor, a chemokine inhibitor, an antiproliferative agent, a statin, a protein, growth factor or compound stimulating growth factor production that will enhance endothelial regrowth of the luminal endothelium, a matrix metalloproteinase inhibitor, a somatostatin analogue, an aldosterone synthetase inhibitor or aldosterone receptor blocker and a compound inhibiting the renin-angiotensin system.
11. A method for stabilizing vulnerable plaques in blood vessels of a subject in need of such a stabilization comprising the controlled delivery from a drug delivery device or system of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more active co-agents.
12. A method for preventing or treating restenosis in diabetic patients comprising administering to said patients a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more active co-agents, or the controlled delivery from a drug delivery device or system of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more active co-agents.
13. A method for the prevention or reduction of vascular access dysfunction in association with the insertion or repair of an indwelling shunt, fistula or catheter, or actual treatment, in a subject in need thereof, which comprises administering to the subject rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more active co-agents, or a controlled delivery from a drug delivery medical device or system of a therapeutically effective amount of rapamycin or a rapamycin derivative having mTOR inhibiting properties, optionally in conjunction with one or more other active co-agents.
PCT/EP2003/000153 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof WO2003057218A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
EP03702413A EP1465624A1 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
JP2003557576A JP2005514411A (en) 2002-01-10 2003-01-09 Drug delivery system for the prevention and treatment of vascular diseases comprising rapamycin and its derivatives
AU2003205586A AU2003205586A1 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
CA002472198A CA2472198A1 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
US10/501,210 US20050020614A1 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
MXPA04006731A MXPA04006731A (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof.
IL16271903A IL162719A0 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin andderivatives thereof
BR0306858-7A BR0306858A (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
KR10-2004-7010710A KR20040076278A (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
HU0402594A HUP0402594A3 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
NO20043309A NO20043309L (en) 2002-01-10 2004-08-09 Drug delivery systems for the prevention and treatment of vascular diseases including rapamycin and its derivatives
US12/242,980 US20090036352A1 (en) 2002-01-10 2008-10-01 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US34726402P 2002-01-10 2002-01-10
US60/347,264 2002-01-10

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/242,980 Continuation US20090036352A1 (en) 2002-01-10 2008-10-01 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof

Publications (1)

Publication Number Publication Date
WO2003057218A1 true WO2003057218A1 (en) 2003-07-17

Family

ID=23363001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/000153 WO2003057218A1 (en) 2002-01-10 2003-01-09 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof

Country Status (19)

Country Link
US (5) US20050020614A1 (en)
EP (1) EP1465624A1 (en)
JP (1) JP2005514411A (en)
KR (1) KR20040076278A (en)
CN (1) CN1615137A (en)
AU (1) AU2003205586A1 (en)
BR (1) BR0306858A (en)
CA (1) CA2472198A1 (en)
CO (1) CO5601015A2 (en)
EC (1) ECSP085181A (en)
HU (1) HUP0402594A3 (en)
IL (1) IL162719A0 (en)
MX (1) MXPA04006731A (en)
NO (1) NO20043309L (en)
PL (1) PL369671A1 (en)
RU (3) RU2004124387A (en)
TW (2) TW200730152A (en)
WO (1) WO2003057218A1 (en)
ZA (1) ZA200405118B (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1362602A1 (en) * 2002-04-15 2003-11-19 Cordis Corporation Coated medical devices for the treatment of vascular disease
WO2004082681A1 (en) * 2003-03-17 2004-09-30 Novartis Ag Pharmaceutical compositions comprising a combination of rapamycin or its derivative and pimecrolimus for the treatment of inflammation- and ummunologically-mediated diseases
US6808536B2 (en) 1997-04-18 2004-10-26 Carol Wright Stent containing rapamycin or its analogs using a modified stent
WO2004073768A3 (en) * 2003-02-18 2005-04-21 Medtronic Inc Occlusion resistant hydrocephalic shunt
WO2005042567A1 (en) * 2003-11-03 2005-05-12 Altachem Pharma Ltd. Rapamycin peptides conjugates: synthesis and uses thereof
WO2005049021A1 (en) * 2003-11-03 2005-06-02 Oy Helsinki Transplantation R & D Ltd Materials and methods for inhibiting neointimal hyperplasia
WO2005053661A2 (en) * 2003-12-01 2005-06-16 Novartis Ag Pharmaceutical combinations
EP1557183A1 (en) * 2004-01-20 2005-07-27 Cordis Corporation Local vascular delivery of mycophenolic acid in combination with rapamycin to prevent restenosis
JP2005288170A (en) * 2004-03-31 2005-10-20 Cordis Corp Medicament distribution device
WO2005110480A2 (en) * 2004-05-17 2005-11-24 Novartis Ag Combination of organic compounds
WO2006053754A1 (en) * 2004-11-19 2006-05-26 Novartis Ag COMBINATIONS OF ANTI-ATHEROSCLEROTIC PEPTIDES AND AN mTOR INHIBITING AGENT AND THEIR METHODS OF USE
WO2006102359A3 (en) * 2005-03-23 2007-05-10 Abbott Lab Delivery of highly lipophilic agents via medical devices
WO2007111885A3 (en) * 2006-03-22 2007-12-06 Abbott Lab Delivery of highly lipophilic agents via medical devices
EP1868656A1 (en) * 2005-04-04 2007-12-26 Medivas, LLC Bioactive stents for type ii diabetics and methods for use thereof
JP2008504890A (en) * 2004-06-29 2008-02-21 アドヴァンスド カーディオヴァスキュラー システムズ, インコーポレイテッド Drug delivery stent formulation for restenosis and vulnerable plaque
WO2008027322A1 (en) * 2006-08-28 2008-03-06 Wyeth Implantable shunt or catheter enabling gradual delivery of therapeutic agents
US7399480B2 (en) 1997-09-26 2008-07-15 Abbott Laboratories Methods of administering tetrazole-containing rapamycin analogs with other therapeutic substances using medical devices
US7455853B2 (en) 1998-09-24 2008-11-25 Abbott Cardiovascular Systems Inc. Medical devices containing rapamycin analogs
US7544689B2 (en) 2002-08-15 2009-06-09 Cyclacel Limited Purine derivatives
US20100086579A1 (en) * 2008-10-03 2010-04-08 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
US7819912B2 (en) 1998-03-30 2010-10-26 Innovational Holdings Llc Expandable medical device with beneficial agent delivery mechanism
US7842083B2 (en) 2001-08-20 2010-11-30 Innovational Holdings, Llc. Expandable medical device with improved spatial distribution
US7850728B2 (en) 2000-10-16 2010-12-14 Innovational Holdings Llc Expandable medical device for delivery of beneficial agent
US7850727B2 (en) 2001-08-20 2010-12-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US8182527B2 (en) 2001-05-07 2012-05-22 Cordis Corporation Heparin barrier coating for controlled drug release
US20120172389A1 (en) * 2006-08-22 2012-07-05 Ruey-Shiuan Tsai Treatment of fibrosing disorders
US10058641B2 (en) 2001-09-10 2018-08-28 Abbott Laboratories Medical devices containing rapamycin analogs
US11278657B2 (en) 2019-04-11 2022-03-22 Enclear Therapies, Inc. Methods of amelioration of cerebrospinal fluid and devices and systems therefor
US11419921B2 (en) 2018-07-23 2022-08-23 Enclear Therapies, Inc. Methods of treating neurological disorders
US11752200B2 (en) 2018-07-23 2023-09-12 Enclear Therapies Inc. Methods of treating neurological disorders

Families Citing this family (144)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030129215A1 (en) * 1998-09-24 2003-07-10 T-Ram, Inc. Medical devices containing rapamycin analogs
US7713297B2 (en) * 1998-04-11 2010-05-11 Boston Scientific Scimed, Inc. Drug-releasing stent with ceramic-containing layer
US20050084514A1 (en) * 2000-11-06 2005-04-21 Afmedica, Inc. Combination drug therapy for reducing scar tissue formation
US7077859B2 (en) * 2000-12-22 2006-07-18 Avantec Vascular Corporation Apparatus and methods for variably controlled substance delivery from implanted prostheses
US20020082679A1 (en) * 2000-12-22 2002-06-27 Avantec Vascular Corporation Delivery or therapeutic capable agents
US20050203612A1 (en) * 2000-12-22 2005-09-15 Avantec Vascular Corporation Devices delivering therapeutic agents and methods regarding the same
US20030033007A1 (en) * 2000-12-22 2003-02-13 Avantec Vascular Corporation Methods and devices for delivery of therapeutic capable agents with variable release profile
US20030050692A1 (en) * 2000-12-22 2003-03-13 Avantec Vascular Corporation Delivery of therapeutic capable agents
GB0100761D0 (en) 2001-01-11 2001-02-21 Biocompatibles Ltd Drug delivery from stents
NZ540000A (en) * 2001-02-16 2007-05-31 Abbott Lab Vascular Entpr Ltd Implants with tacrolimus
US20080145402A1 (en) * 2001-09-10 2008-06-19 Abbott Cardiovascular Systems Inc. Medical Devices Containing Rapamycin Analogs
US10441747B2 (en) * 2002-01-22 2019-10-15 Mercator Medsystems, Inc. Methods and systems for inhibiting vascular inflammation
IL162734A0 (en) 2002-02-01 2005-11-20 Ariad Gene Therapeutics Inc Phosphorus-containing compounds & uses thereof
AU2003220390A1 (en) * 2002-03-18 2003-10-08 Medtronic Ave Inc. Medical devices for delivering anti-proliferative compositions to anatomical sites at risk for restenosis
US20040133270A1 (en) * 2002-07-08 2004-07-08 Axel Grandt Drug eluting stent and methods of manufacture
US20050214343A1 (en) * 2002-07-18 2005-09-29 Patrice Tremble Medical devices comprising a protein-tyrosine kinase inhibitor to inhibit restonosis
EP2263709A1 (en) * 2002-09-06 2010-12-22 Abbott Laboratories Medical device having hydration inhibitor
US9770349B2 (en) * 2002-11-13 2017-09-26 University Of Virginia Patent Foundation Nanoporous stents with enhanced cellular adhesion and reduced neointimal formation
US8088404B2 (en) * 2003-03-20 2012-01-03 Medtronic Vasular, Inc. Biocompatible controlled release coatings for medical devices and related methods
US20040254629A1 (en) * 2003-04-25 2004-12-16 Brian Fernandes Methods and apparatus for treatment of aneurysmal tissue
US20050033417A1 (en) * 2003-07-31 2005-02-10 John Borges Coating for controlled release of a therapeutic agent
EP1656163B1 (en) * 2003-08-13 2014-08-06 Poly-Med, Inc. Biocompatible controlled release coatings for medical devices and related methods
AR045957A1 (en) * 2003-10-03 2005-11-16 Novartis Ag PHARMACEUTICAL COMPOSITION AND COMBINATION
WO2005051229A2 (en) * 2003-11-24 2005-06-09 Avantec Vascular Corporation Devices delivering therapeutic agents and methods regarding the same
WO2005075003A1 (en) * 2004-01-21 2005-08-18 Medtronic Vascular Inc. Implantable medical devices for treating or preventing restenosis
CN1917865A (en) * 2004-01-22 2007-02-21 诺瓦提斯公司 Combination of organic compounds
US8431145B2 (en) * 2004-03-19 2013-04-30 Abbott Laboratories Multiple drug delivery from a balloon and a prosthesis
US20070027523A1 (en) * 2004-03-19 2007-02-01 Toner John L Method of treating vascular disease at a bifurcated vessel using coated balloon
US8551512B2 (en) * 2004-03-22 2013-10-08 Advanced Cardiovascular Systems, Inc. Polyethylene glycol/poly(butylene terephthalate) copolymer coated devices including EVEROLIMUS
US8778014B1 (en) 2004-03-31 2014-07-15 Advanced Cardiovascular Systems, Inc. Coatings for preventing balloon damage to polymer coated stents
US8003122B2 (en) * 2004-03-31 2011-08-23 Cordis Corporation Device for local and/or regional delivery employing liquid formulations of therapeutic agents
US7846940B2 (en) * 2004-03-31 2010-12-07 Cordis Corporation Solution formulations of sirolimus and its analogs for CAD treatment
US8293890B2 (en) 2004-04-30 2012-10-23 Advanced Cardiovascular Systems, Inc. Hyaluronic acid based copolymers
WO2006020755A2 (en) * 2004-08-10 2006-02-23 Beth Israel Deaconess Medical Center, Inc. Methods for identifying inhibitors of the mtor pathway as diabetes therapeutics
US20060051338A1 (en) * 2004-08-20 2006-03-09 New York University Inhibition of mitogen-activated protein kinases in cardiovascular disease
US20090041778A1 (en) * 2004-11-22 2009-02-12 Sukhatme Vikas P Methods And Compositions For The Treatment Of Graft Failure
US20060127443A1 (en) * 2004-12-09 2006-06-15 Helmus Michael N Medical devices having vapor deposited nanoporous coatings for controlled therapeutic agent delivery
US20060129215A1 (en) * 2004-12-09 2006-06-15 Helmus Michael N Medical devices having nanostructured regions for controlled tissue biocompatibility and drug delivery
WO2006099381A1 (en) * 2005-03-14 2006-09-21 Conor Medsystems, Inc. Drug delivery stent with extended in vivo and in vitro release of anti-inflammatory
US7252834B2 (en) * 2005-04-25 2007-08-07 Clemson University Research Foundation (Curf) Elastin stabilization of connective tissue
CN101227927A (en) * 2005-05-31 2008-07-23 诺瓦提斯公司 Combination of HMG-CoA reductase inhibitors and mTOR inhibitors
US20070009564A1 (en) * 2005-06-22 2007-01-11 Mcclain James B Drug/polymer composite materials and methods of making the same
US20070038176A1 (en) * 2005-07-05 2007-02-15 Jan Weber Medical devices with machined layers for controlled communications with underlying regions
KR101406415B1 (en) 2005-07-15 2014-06-19 미셀 테크놀로지즈, 인코포레이티드 Polymer coatings containing drug powder of controlled morphology
WO2007011708A2 (en) 2005-07-15 2007-01-25 Micell Technologies, Inc. Stent with polymer coating containing amorphous rapamycin
WO2007053578A2 (en) * 2005-10-31 2007-05-10 Amulet Pharmaceuticals, Inc. Multi-phasic nitric oxide and drug co-eluting stent coatings
US20070116736A1 (en) * 2005-11-23 2007-05-24 Argentieri Dennis C Local vascular delivery of PI3 kinase inhibitors alone or in combination with sirolimus to prevent restinosis following vascular injury
EP1968615A4 (en) * 2005-12-06 2010-08-04 Amulet Pharmaceuticals Inc Nitric oxide-releasing polymers
US20070203171A1 (en) * 2006-02-28 2007-08-30 Zhao Jonathon Z Combination of rapamycin and its tetrazole isomers and epimers, methods of making and using the same
US20070224235A1 (en) * 2006-03-24 2007-09-27 Barron Tenney Medical devices having nanoporous coatings for controlled therapeutic agent delivery
US8187620B2 (en) * 2006-03-27 2012-05-29 Boston Scientific Scimed, Inc. Medical devices comprising a porous metal oxide or metal material and a polymer coating for delivering therapeutic agents
WO2007127363A2 (en) 2006-04-26 2007-11-08 Micell Technologies, Inc. Coatings containing multiple drugs
US20080051335A1 (en) * 2006-05-02 2008-02-28 Kleiner Lothar W Methods, compositions and devices for treating lesioned sites using bioabsorbable carriers
US7868531B2 (en) * 2006-05-05 2011-01-11 Brother International Corporation Carbon nanotube arrays for field electron emission
US20070264303A1 (en) * 2006-05-12 2007-11-15 Liliana Atanasoska Coating for medical devices comprising an inorganic or ceramic oxide and a therapeutic agent
US20080004695A1 (en) * 2006-06-28 2008-01-03 Abbott Cardiovascular Systems Inc. Everolimus/pimecrolimus-eluting implantable medical devices
US8815275B2 (en) 2006-06-28 2014-08-26 Boston Scientific Scimed, Inc. Coatings for medical devices comprising a therapeutic agent and a metallic material
WO2008002778A2 (en) * 2006-06-29 2008-01-03 Boston Scientific Limited Medical devices with selective coating
US20080039362A1 (en) * 2006-08-09 2008-02-14 Afmedica, Inc. Combination drug therapy for reducing scar tissue formation
US9173733B1 (en) 2006-08-21 2015-11-03 Abbott Cardiovascular Systems Inc. Tracheobronchial implantable medical device and methods of use
JP2010503469A (en) 2006-09-14 2010-02-04 ボストン サイエンティフィック リミテッド Medical device having drug-eluting film
EP2084310A1 (en) * 2006-10-05 2009-08-05 Boston Scientific Limited Polymer-free coatings for medical devices formed by plasma electrolytic deposition
CA2667228C (en) * 2006-10-23 2015-07-14 Micell Technologies, Inc. Holder for electrically charging a substrate during coating
US7981150B2 (en) 2006-11-09 2011-07-19 Boston Scientific Scimed, Inc. Endoprosthesis with coatings
US20080161335A1 (en) * 2006-11-14 2008-07-03 Vladyka Ronald S Oral formulations
US7713541B1 (en) 2006-11-21 2010-05-11 Abbott Cardiovascular Systems Inc. Zwitterionic terpolymers, method of making and use on medical devices
US11426494B2 (en) 2007-01-08 2022-08-30 MT Acquisition Holdings LLC Stents having biodegradable layers
WO2008086369A1 (en) * 2007-01-08 2008-07-17 Micell Technologies, Inc. Stents having biodegradable layers
EP2125895B1 (en) 2007-02-02 2015-04-08 Vegenics Pty Ltd Vegf receptor antagonists for treating organ transplant alloimmunity and arteriosclerosis
US8070797B2 (en) 2007-03-01 2011-12-06 Boston Scientific Scimed, Inc. Medical device with a porous surface for delivery of a therapeutic agent
US8431149B2 (en) 2007-03-01 2013-04-30 Boston Scientific Scimed, Inc. Coated medical devices for abluminal drug delivery
US20080241215A1 (en) * 2007-03-28 2008-10-02 Robert Falotico Local vascular delivery of probucol alone or in combination with sirolimus to treat restenosis, vulnerable plaque, aaa and stroke
US8067054B2 (en) 2007-04-05 2011-11-29 Boston Scientific Scimed, Inc. Stents with ceramic drug reservoir layer and methods of making and using the same
NZ580469A (en) * 2007-04-17 2012-05-25 Micell Technologies Inc Coronary stents having biodegradable layers
US7976915B2 (en) * 2007-05-23 2011-07-12 Boston Scientific Scimed, Inc. Endoprosthesis with select ceramic morphology
WO2008148013A1 (en) * 2007-05-25 2008-12-04 Micell Technologies, Inc. Polymer films for medical device coating
US7942926B2 (en) * 2007-07-11 2011-05-17 Boston Scientific Scimed, Inc. Endoprosthesis coating
US8002823B2 (en) * 2007-07-11 2011-08-23 Boston Scientific Scimed, Inc. Endoprosthesis coating
EP2187988B1 (en) 2007-07-19 2013-08-21 Boston Scientific Limited Endoprosthesis having a non-fouling surface
US7931683B2 (en) 2007-07-27 2011-04-26 Boston Scientific Scimed, Inc. Articles having ceramic coated surfaces
US8815273B2 (en) * 2007-07-27 2014-08-26 Boston Scientific Scimed, Inc. Drug eluting medical devices having porous layers
CN101969959A (en) * 2007-07-31 2011-02-09 利默里克生物制药公司 Phosphorylated pyrone analogs and methods
US8221822B2 (en) * 2007-07-31 2012-07-17 Boston Scientific Scimed, Inc. Medical device coating by laser cladding
WO2009020520A1 (en) * 2007-08-03 2009-02-12 Boston Scientific Scimed, Inc. Coating for medical device having increased surface area
US20090104240A1 (en) * 2007-10-19 2009-04-23 Abbott Cardiovascular Systems Inc. Dual Drug Formulations For Implantable Medical Devices For Treatment of Vascular Diseases
WO2009051780A1 (en) * 2007-10-19 2009-04-23 Micell Technologies, Inc. Drug coated stents
US20090118809A1 (en) * 2007-11-02 2009-05-07 Torsten Scheuermann Endoprosthesis with porous reservoir and non-polymer diffusion layer
US7938855B2 (en) * 2007-11-02 2011-05-10 Boston Scientific Scimed, Inc. Deformable underlayer for stent
US8216632B2 (en) 2007-11-02 2012-07-10 Boston Scientific Scimed, Inc. Endoprosthesis coating
US8029554B2 (en) * 2007-11-02 2011-10-04 Boston Scientific Scimed, Inc. Stent with embedded material
US20090118818A1 (en) * 2007-11-02 2009-05-07 Boston Scientific Scimed, Inc. Endoprosthesis with coating
WO2009064806A1 (en) * 2007-11-12 2009-05-22 Endologix, Inc. Method and agent for in-situ stabilization of vascular tissue
US20090214654A1 (en) * 2008-02-21 2009-08-27 Isenburg Jason C Treatment of aneurysm with application of connective tissue stabilization agent in combination with a delivery vehicle
US8420110B2 (en) 2008-03-31 2013-04-16 Cordis Corporation Drug coated expandable devices
US8409601B2 (en) 2008-03-31 2013-04-02 Cordis Corporation Rapamycin coated expandable devices
JP5608160B2 (en) 2008-04-17 2014-10-15 ミセル テクノロジーズ、インコーポレイテッド Stent with bioabsorbable layer
WO2009131911A2 (en) 2008-04-22 2009-10-29 Boston Scientific Scimed, Inc. Medical devices having a coating of inorganic material
WO2009132176A2 (en) 2008-04-24 2009-10-29 Boston Scientific Scimed, Inc. Medical devices having inorganic particle layers
EP2303350A2 (en) 2008-06-18 2011-04-06 Boston Scientific Scimed, Inc. Endoprosthesis coating
WO2009158031A2 (en) * 2008-06-27 2009-12-30 Limerick Biopharma, Inc. Methods and compositions for therapeutic treatment
US20100016833A1 (en) * 2008-07-15 2010-01-21 Ogle Matthew F Devices for the Treatment of Vascular Aneurysm
CA2946195A1 (en) 2008-07-17 2010-01-21 Micell Technologies, Inc. Drug delivery medical device
US20100119605A1 (en) * 2008-11-12 2010-05-13 Isenburg Jason C Compositions for tissue stabilization
US8231980B2 (en) * 2008-12-03 2012-07-31 Boston Scientific Scimed, Inc. Medical implants including iridium oxide
US8834913B2 (en) * 2008-12-26 2014-09-16 Battelle Memorial Institute Medical implants and methods of making medical implants
US8071156B2 (en) * 2009-03-04 2011-12-06 Boston Scientific Scimed, Inc. Endoprostheses
US20100256746A1 (en) * 2009-03-23 2010-10-07 Micell Technologies, Inc. Biodegradable polymers
EP2410954A4 (en) * 2009-03-23 2014-03-05 Micell Technologies Inc Peripheral stents having layers
EP2413847A4 (en) 2009-04-01 2013-11-27 Micell Technologies Inc Coated stents
EP3366326A1 (en) 2009-04-17 2018-08-29 Micell Technologies, Inc. Stents having controlled elution
US20100274352A1 (en) * 2009-04-24 2010-10-28 Boston Scientific Scrimed, Inc. Endoprosthesis with Selective Drug Coatings
US8287937B2 (en) * 2009-04-24 2012-10-16 Boston Scientific Scimed, Inc. Endoprosthese
EP2453834A4 (en) 2009-07-16 2014-04-16 Micell Technologies Inc Drug delivery medical device
WO2011014563A1 (en) 2009-07-29 2011-02-03 Vatrix Medical, Inc. Tissue stabilization for heart failure
WO2011044455A1 (en) * 2009-10-09 2011-04-14 Vatrix Medical, Inc. In vivo chemical stabilization of vulnerable plaque
US8444624B2 (en) * 2009-10-19 2013-05-21 Vatrix Medical, Inc. Vascular medical devices with sealing elements and procedures for the treatment of isolated vessel sections
US8951595B2 (en) * 2009-12-11 2015-02-10 Abbott Cardiovascular Systems Inc. Coatings with tunable molecular architecture for drug-coated balloon
US20110144577A1 (en) * 2009-12-11 2011-06-16 John Stankus Hydrophilic coatings with tunable composition for drug coated balloon
US8480620B2 (en) * 2009-12-11 2013-07-09 Abbott Cardiovascular Systems Inc. Coatings with tunable solubility profile for drug-coated balloon
US11369498B2 (en) * 2010-02-02 2022-06-28 MT Acquisition Holdings LLC Stent and stent delivery system with improved deliverability
US8795762B2 (en) * 2010-03-26 2014-08-05 Battelle Memorial Institute System and method for enhanced electrostatic deposition and surface coatings
CA2794704C (en) * 2010-04-16 2019-09-17 Micell Technologies, Inc. Stents having controlled elution
CA2797110C (en) 2010-04-22 2020-07-21 Micell Technologies, Inc. Stents and other devices having extracellular matrix coating
CA2805631C (en) 2010-07-16 2018-07-31 Micell Technologies, Inc. Drug delivery medical device
CA2810842C (en) 2010-09-09 2018-06-26 Micell Technologies, Inc. Macrolide dosage forms
US20130230571A1 (en) * 2010-10-04 2013-09-05 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
US8911468B2 (en) 2011-01-31 2014-12-16 Vatrix Medical, Inc. Devices, therapeutic compositions and corresponding percutaneous treatment methods for aortic dissection
EP2709711B8 (en) 2011-05-18 2017-03-22 Vatrix Medical, Inc. Coated balloons for blood vessel stabilization
US10464100B2 (en) 2011-05-31 2019-11-05 Micell Technologies, Inc. System and process for formation of a time-released, drug-eluting transferable coating
US10117972B2 (en) 2011-07-15 2018-11-06 Micell Technologies, Inc. Drug delivery medical device
US9220716B2 (en) * 2011-07-26 2015-12-29 Children's Medical Center Corporation Methods and compositions for the treatment of proliferative vascular disorders
US10188772B2 (en) 2011-10-18 2019-01-29 Micell Technologies, Inc. Drug delivery medical device
US20140094900A1 (en) * 2012-10-01 2014-04-03 Brigham Young University Compliant biocompatible device and method of manufacture
JP6330024B2 (en) 2013-03-12 2018-05-23 マイセル・テクノロジーズ,インコーポレイテッド Bioabsorbable biomedical implant
WO2014160358A1 (en) * 2013-03-14 2014-10-02 Thomas Cooper Woods Use of mir-221 and 222 lowering agents to prevent cardiovascular disease in diabetic subjects
US9522130B2 (en) 2013-03-14 2016-12-20 Thomas Cooper Woods Use of miR-221 and 222 lowering agents to prevent cardiovascular disease in diabetic subjects
US10274503B2 (en) 2013-05-08 2019-04-30 Vegenics Pty Limited Methods of using VEGF-C biomarkers for age-related macular degeneration (AMD) diagnosis
JP2016519965A (en) 2013-05-15 2016-07-11 マイセル・テクノロジーズ,インコーポレイテッド Bioabsorbable biomedical implant
WO2016100782A1 (en) * 2014-12-18 2016-06-23 University Of South Carolina Suppression of neointimal formation following vascular surgery using cdk8 inhibitors
CN116585311A (en) 2016-09-22 2023-08-15 墨卡托医疗系统公司 Treatment of restenosis using temsirolimus
KR20200008166A (en) 2017-05-26 2020-01-23 머케이터 메드시스템즈, 인크. Combination therapy for the treatment of restenosis
CN109010931B (en) * 2017-06-09 2022-03-11 上海微创医疗器械(集团)有限公司 Interventional medical device and application of aphidicolin
WO2019104065A1 (en) * 2017-11-22 2019-05-31 Turrinii Pharmaceutical, Llc Anti-aging methods and compositions
WO2019178228A1 (en) 2018-03-14 2019-09-19 Mercator Medsystems, Inc. Medical instrument and medical method for localized drug delivery

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010029351A1 (en) * 1998-04-16 2001-10-11 Robert Falotico Drug combinations and delivery devices for the prevention and treatment of vascular disease
WO2001087263A2 (en) * 2000-05-12 2001-11-22 Cordis Corporation Delivery systems for treatment of vascular disease
WO2001087372A1 (en) * 2000-05-12 2001-11-22 Cordis Corporation Drug combinations useful for prevention of restenosis

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US27340A (en) * 1860-03-06 Improvement in steam-boilers
US82680A (en) * 1868-10-06 Rufus e
US123505A (en) * 1872-02-06 Improvement in wheel-plows
US7213A (en) * 1850-03-26 Improvement in seed-planters
ZA737247B (en) * 1972-09-29 1975-04-30 Ayerst Mckenna & Harrison Rapamycin and process of preparation
US5516781A (en) * 1992-01-09 1996-05-14 American Home Products Corporation Method of treating restenosis with rapamycin
US6273913B1 (en) * 1997-04-18 2001-08-14 Cordis Corporation Modified stent useful for delivery of drugs along stent strut
NL1006553C2 (en) * 1997-07-11 1999-01-12 Hoogovens Staal Bv Method for controlling a melt reduction process.
US6890546B2 (en) * 1998-09-24 2005-05-10 Abbott Laboratories Medical devices containing rapamycin analogs
US7208010B2 (en) * 2000-10-16 2007-04-24 Conor Medsystems, Inc. Expandable medical device for delivery of beneficial agent
WO2000030636A1 (en) * 1998-11-20 2000-06-02 The General Hospital Corporation Use of pyrethroid compounds to promote hair growth
US20020007213A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US6641611B2 (en) * 2001-11-26 2003-11-04 Swaminathan Jayaraman Therapeutic coating for an intravascular implant
US6805703B2 (en) * 2001-09-18 2004-10-19 Scimed Life Systems, Inc. Protective membrane for reconfiguring a workpiece
US7025734B1 (en) * 2001-09-28 2006-04-11 Advanced Cardiovascular Systmes, Inc. Guidewire with chemical sensing capabilities
WO2003072159A1 (en) * 2002-02-28 2003-09-04 Novartis Ag N-{5-[4-(4-methyl-piperazino-methyl)­-benzoylamido]-2-methylphenyl}-4-(3-pyridyl)-2-pyrimidine-amine coated stents

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010029351A1 (en) * 1998-04-16 2001-10-11 Robert Falotico Drug combinations and delivery devices for the prevention and treatment of vascular disease
WO2001087263A2 (en) * 2000-05-12 2001-11-22 Cordis Corporation Delivery systems for treatment of vascular disease
WO2001087372A1 (en) * 2000-05-12 2001-11-22 Cordis Corporation Drug combinations useful for prevention of restenosis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SOUSA J E ET AL: "Sustained suppression of neointimal proliferation by sirolimus-eluting stents: one-year angiographic and intravascular ultrasound follow-up.", CIRCULATION. UNITED STATES 23 OCT 2001, vol. 104, no. 17, 23 October 2001 (2001-10-23), pages 2007 - 2011, XP002239133, ISSN: 1524-4539 *

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7666222B2 (en) 1997-04-18 2010-02-23 Cordis Corporation Methods and devices for delivering therapeutic agents to target vessels
US6808536B2 (en) 1997-04-18 2004-10-26 Carol Wright Stent containing rapamycin or its analogs using a modified stent
US7399480B2 (en) 1997-09-26 2008-07-15 Abbott Laboratories Methods of administering tetrazole-containing rapamycin analogs with other therapeutic substances using medical devices
US8153150B2 (en) 1997-09-26 2012-04-10 Abbott Laboratories Methods of administering tetrazole-containing rapamycin analogs with other therapeutic substances for treatment of vascular disorder
US7896912B2 (en) 1998-03-30 2011-03-01 Innovational Holdings, Llc Expandable medical device with S-shaped bridging elements
US7819912B2 (en) 1998-03-30 2010-10-26 Innovational Holdings Llc Expandable medical device with beneficial agent delivery mechanism
US8052735B2 (en) 1998-03-30 2011-11-08 Innovational Holdings, Llc Expandable medical device with ductile hinges
US8439968B2 (en) 1998-03-30 2013-05-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US7455853B2 (en) 1998-09-24 2008-11-25 Abbott Cardiovascular Systems Inc. Medical devices containing rapamycin analogs
US8187321B2 (en) 2000-10-16 2012-05-29 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US7850728B2 (en) 2000-10-16 2010-12-14 Innovational Holdings Llc Expandable medical device for delivery of beneficial agent
US8182527B2 (en) 2001-05-07 2012-05-22 Cordis Corporation Heparin barrier coating for controlled drug release
US7850727B2 (en) 2001-08-20 2010-12-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US7842083B2 (en) 2001-08-20 2010-11-30 Innovational Holdings, Llc. Expandable medical device with improved spatial distribution
US10058641B2 (en) 2001-09-10 2018-08-28 Abbott Laboratories Medical devices containing rapamycin analogs
EP1362602A1 (en) * 2002-04-15 2003-11-19 Cordis Corporation Coated medical devices for the treatment of vascular disease
AU2003203722B2 (en) * 2002-04-15 2008-04-17 Cardinal Health 529, Llc Coated medical devices for the treatment of vascular disease
US7544689B2 (en) 2002-08-15 2009-06-09 Cyclacel Limited Purine derivatives
WO2004073768A3 (en) * 2003-02-18 2005-04-21 Medtronic Inc Occlusion resistant hydrocephalic shunt
US7582068B2 (en) 2003-02-18 2009-09-01 Medtronic, Inc. Occlusion resistant hydrocephalic shunt
WO2004082681A1 (en) * 2003-03-17 2004-09-30 Novartis Ag Pharmaceutical compositions comprising a combination of rapamycin or its derivative and pimecrolimus for the treatment of inflammation- and ummunologically-mediated diseases
WO2005049021A1 (en) * 2003-11-03 2005-06-02 Oy Helsinki Transplantation R & D Ltd Materials and methods for inhibiting neointimal hyperplasia
WO2005042567A1 (en) * 2003-11-03 2005-05-12 Altachem Pharma Ltd. Rapamycin peptides conjugates: synthesis and uses thereof
US7659244B2 (en) 2003-11-03 2010-02-09 Quest Pharmatech, Inc. Rapamycin peptides conjugates: synthesis and uses thereof
WO2005053661A3 (en) * 2003-12-01 2005-12-29 Novartis Ag Pharmaceutical combinations
WO2005053661A2 (en) * 2003-12-01 2005-06-16 Novartis Ag Pharmaceutical combinations
AU2004294282B2 (en) * 2003-12-01 2009-05-07 Novartis Ag Pharmaceutical combinations
JP2005237951A (en) * 2004-01-20 2005-09-08 Cordis Corp Local vascular delivery of mycophenolic acid in combination with rapamycin to prevent restenosis after trauma of vessel
EP2286848A3 (en) * 2004-01-20 2012-08-29 Cordis Corporation Local vascular delivery of mycophenolic acid in combination with rapamycin to prevent restenosis
EP1557183A1 (en) * 2004-01-20 2005-07-27 Cordis Corporation Local vascular delivery of mycophenolic acid in combination with rapamycin to prevent restenosis
JP2005288170A (en) * 2004-03-31 2005-10-20 Cordis Corp Medicament distribution device
WO2005110480A3 (en) * 2004-05-17 2006-07-27 Novartis Ag Combination of organic compounds
WO2005110480A2 (en) * 2004-05-17 2005-11-24 Novartis Ag Combination of organic compounds
JP2008504890A (en) * 2004-06-29 2008-02-21 アドヴァンスド カーディオヴァスキュラー システムズ, インコーポレイテッド Drug delivery stent formulation for restenosis and vulnerable plaque
WO2006053754A1 (en) * 2004-11-19 2006-05-26 Novartis Ag COMBINATIONS OF ANTI-ATHEROSCLEROTIC PEPTIDES AND AN mTOR INHIBITING AGENT AND THEIR METHODS OF USE
WO2006102359A3 (en) * 2005-03-23 2007-05-10 Abbott Lab Delivery of highly lipophilic agents via medical devices
EP2327429A1 (en) * 2005-03-23 2011-06-01 Abbott Laboratories Delivery of highly lipophilic agents via medical devices
EP1868656A4 (en) * 2005-04-04 2011-11-02 Medivas Llc Bioactive stents for type ii diabetics and methods for use thereof
EP1868656A1 (en) * 2005-04-04 2007-12-26 Medivas, LLC Bioactive stents for type ii diabetics and methods for use thereof
WO2007111885A3 (en) * 2006-03-22 2007-12-06 Abbott Lab Delivery of highly lipophilic agents via medical devices
US20120172389A1 (en) * 2006-08-22 2012-07-05 Ruey-Shiuan Tsai Treatment of fibrosing disorders
WO2008027322A1 (en) * 2006-08-28 2008-03-06 Wyeth Implantable shunt or catheter enabling gradual delivery of therapeutic agents
US20100086579A1 (en) * 2008-10-03 2010-04-08 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
EP2352459A1 (en) * 2008-10-03 2011-08-10 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
EP2352459A4 (en) * 2008-10-03 2013-09-25 Elixir Medical Corp Macrocyclic lactone compounds and methods for their use
CN102231969A (en) * 2008-10-03 2011-11-02 万能医药公司 Macrocyclic lactone compounds and methods for their use
US11419921B2 (en) 2018-07-23 2022-08-23 Enclear Therapies, Inc. Methods of treating neurological disorders
US11752200B2 (en) 2018-07-23 2023-09-12 Enclear Therapies Inc. Methods of treating neurological disorders
US11278657B2 (en) 2019-04-11 2022-03-22 Enclear Therapies, Inc. Methods of amelioration of cerebrospinal fluid and devices and systems therefor

Also Published As

Publication number Publication date
ECSP085181A (en) 2008-09-29
CN1615137A (en) 2005-05-11
US20060127440A1 (en) 2006-06-15
TW200730152A (en) 2007-08-16
JP2005514411A (en) 2005-05-19
RU2008107255A (en) 2009-09-10
TW200306826A (en) 2003-12-01
RU2008150750A (en) 2010-06-27
PL369671A1 (en) 2005-05-02
EP1465624A1 (en) 2004-10-13
US20050020614A1 (en) 2005-01-27
AU2003205586A1 (en) 2003-07-24
HUP0402594A2 (en) 2005-10-28
MXPA04006731A (en) 2004-10-04
RU2004124387A (en) 2005-06-10
US20090043379A1 (en) 2009-02-12
CO5601015A2 (en) 2006-01-31
HUP0402594A3 (en) 2006-01-30
US20090036352A1 (en) 2009-02-05
CA2472198A1 (en) 2003-07-17
IL162719A0 (en) 2005-11-20
BR0306858A (en) 2004-11-03
ZA200405118B (en) 2005-06-21
KR20040076278A (en) 2004-08-31
US20030170287A1 (en) 2003-09-11
NO20043309L (en) 2004-08-09

Similar Documents

Publication Publication Date Title
US20030170287A1 (en) Drug delivery systems for the prevention and treatment of vascular diseases
AU2003214079B2 (en) N-{5-[4-(4-methyl-piperazino-methyl)-benzoylamido]-2-methylphenyl}-4-(3-pyridyl)-2-pyrimidine-amine coated stents
EP1699503B1 (en) Devices coated with PEC polymers
AU2003283399B2 (en) Drug delivery system
WO2005094916A1 (en) Vegf receptor tyrosine kinase inhibitor coated stent
WO2005099695A1 (en) Drug delivery systems for the prevention and treatment of vascular diseases
AU2009201507A1 (en) Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
AU2007200756A1 (en) Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
EP1572193A1 (en) Microtubule stabilisers for treating stenosis in stents
MXPA06007319A (en) Parmaceutical compositions

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LT LU LV MA MD MK MN MX NO NZ OM PH PL PT RO RU SE SG SK TJ TM TN TR TT UA US UZ VC VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT SE SI SK TR

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

Ref document number: 1-2004-501000

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2003702413

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 162719

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2003205586

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004/05118

Country of ref document: ZA

Ref document number: 200405118

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2472198

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 533944

Country of ref document: NZ

Ref document number: 1500/CHENP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2004/006731

Country of ref document: MX

Ref document number: 20038020807

Country of ref document: CN

Ref document number: 2003557576

Country of ref document: JP

Ref document number: 10501210

Country of ref document: US

Ref document number: 1020047010710

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2004124387

Country of ref document: RU

Ref document number: 1200700370

Country of ref document: VN

Ref document number: 1200400752

Country of ref document: VN

WWP Wipo information: published in national office

Ref document number: 2003702413

Country of ref document: EP