US20060002968A1 - Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders - Google Patents

Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders Download PDF

Info

Publication number
US20060002968A1
US20060002968A1 US10/882,506 US88250604A US2006002968A1 US 20060002968 A1 US20060002968 A1 US 20060002968A1 US 88250604 A US88250604 A US 88250604A US 2006002968 A1 US2006002968 A1 US 2006002968A1
Authority
US
United States
Prior art keywords
drug
everolimus
stent
inflammatory
delivery system
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/882,506
Inventor
Gordon Stewart
Gina Zhang
Nancy Kristen
Paul Consigny
Stephen Dugan
Gene Park
Christopher Feezor
Wouter Roorda
Syed Hossainy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Abbott Cardiovascular Systems Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/882,506 priority Critical patent/US20060002968A1/en
Assigned to ADVANCED CARDIOVASCULAR SYSTEMS, INC. reassignment ADVANCED CARDIOVASCULAR SYSTEMS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CONSIGNY, PAUL, DUGAN, STEPHEN, FEEZOR, CHRISTOPHER, HOSSAINY, SYED F.A., KIRSTEN, NANCY, PARK, GENE, ROORDA, WOUTER, STEWART, GORDON, ZHANG, GINA
Priority to US11/090,507 priority patent/US7758881B2/en
Priority to EP05803287.1A priority patent/EP1768718B1/en
Priority to ES05803287.1T priority patent/ES2529043T3/en
Priority to PCT/US2005/022968 priority patent/WO2006014270A2/en
Priority to JP2007519366A priority patent/JP5791221B2/en
Priority to US11/322,282 priority patent/US8586069B2/en
Publication of US20060002968A1 publication Critical patent/US20060002968A1/en
Priority to US12/191,209 priority patent/US8435550B2/en
Priority to US12/838,329 priority patent/US8709469B2/en
Priority to US14/192,705 priority patent/US9138337B2/en
Priority to US14/829,558 priority patent/US9566373B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic 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
    • A61K31/33Heterocyclic 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
    • A61K31/56Compounds 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
    • A61K31/573Compounds 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 substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • 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
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2250/00Special features of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2250/0058Additional features; Implant or prostheses properties not otherwise provided for
    • A61F2250/0067Means for introducing or releasing pharmaceutical products into the body
    • 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
    • A61L2300/41Anti-inflammatory agents, e.g. NSAIDs
    • 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
    • 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
    • A61L2300/45Mixtures of two or more drugs, e.g. synergistic mixtures

Definitions

  • This invention generally relates to a drug combination including an anti-proliferative drug such as everolimus and an anti-inflammatory agent such as clobetasol for the treatment of a disorder such as restenosis and vulnerable plaque.
  • an anti-proliferative drug such as everolimus
  • an anti-inflammatory agent such as clobetasol
  • plaque has been associated with stenosis and restenosis. While treatments of plaque-induced stenosis and restenosis have advanced significantly over the last few decades, the morbidity and mortality associated with vascular plaques have remained significant. Recent work suggests that plaque may generally fall into one of two different general types: standard stenotic plaques and vulnerable plaques. Stenotic plaque, which is sometimes referred to as thrombosis-resistant plaque, can generally be treated effectively by the known intravascular lumen opening techniques. Although plaques induce stenoses, these atherosclerotic plaques themselves are often a benign and are an effectively treatable disease.
  • plaque As plaque matures, narrowing of a blood vessel by a proliferation of smooth muscle cells, matrix synthesis, and lipid accumulation may result in formation of a plaque which is quite different than a standard stenotic plaque. Such atherosclerotic plaque becomes thrombosis-prone, and can be highly dangerous. This thrombosis-prone or vulnerable plaque may be a frequent cause of acute coronary syndrome.
  • the drug-delivery system has two or more drugs for treating a vascular disorder or a related disorder.
  • the drugs can be a combination of at least one anti-proliferative agent, at least one anti-inflammatory agent, and optionally a third bioactive agent.
  • the anti-proliferative agent can be a drug such as everolimus, and the anti-inflammatory agent can be a drug such as clobetasol.
  • Methods of treating of preventing vascular disorders such as restenosis and vulnerable plaque are also disclosed by administering to the patient a combination of at least one anti-proliferative agent, at least one anti-inflammatory agent, and optionally a third bioactive agent.
  • the mode of delivery can be local or systemic.
  • FIG. 1 shows the results of 28 day quantitative coronary angioplasty (QCA) of a porcine implant study on drug-delivery systems described herein.
  • QCA quantitative coronary angioplasty
  • FIG. 2 shows 28 day histology data of a porcine implant study on drug-delivery systems described herein.
  • FIG. 3 shows the 28 day morphometry data of a porcine implant study on drug-delivery systems described herein.
  • a drug-delivery system and the method of using the drug-delivery system.
  • treatment includes prevention, reduction, delay or elimination of the vascular disorder. In some embodiments, treatment also includes repairing damage caused by the dirorder and/or the mechanical intervention.
  • the drug-delivery system has two or more drugs for treating a vascular disorder or a related disorder.
  • the drugs can be a combination of at least one anti-proliferative agent, at least one anti-inflammatory agent, and optionally a third bioactive agent.
  • the composition described herein includes an effective amount of at least one anti-inflammatory agent and an effective amount of an anti-proliferative agent. In another embodiment, the composition described herein includes an effective amount of an agent which is effective both as an anti-inflammatory agent and as an anti-proliferative agent.
  • the anti-proliferative agent can be everolimus (available under the trade name CerticanTM, Novartis Pharma A G, Germany), and the anti-inflammatory agent can be clobetasol (available under the trade name TemovateTM, Glaxosmithkline, UK).
  • the anti-proliferative agent and the anti-inflammatory agent can be in the form of a coating with and/or without a polymer matrix on a medical device or at elast one of the agents can be administered in a separate dose form such as bolus dose of a free drug, optionally with fluoroscopic dye, or bolus dose of a gel encapsulating a drug.
  • the drug-delivery system or composition may further include a third agent such as a high-density lipoproptein mimetic (HDL-mimetic).
  • HDL-mimetic high-density lipoproptein mimetic
  • an anti-inflammatory agent such as clobetasol can be delivered along with the catheter based delivery of a HDL-mimetic while everolimus is administered by a stent.
  • the drug-delivery system or composition disclosed herein can be used to treat or prevent a disorder such as thrombosis, high cholesterol, hemorrhage, vascular dissection or perforation, vascular aneurysm, vulnerable plaque, chronic total occlusion, claudication, anastomotic proliferation for vein and artificial grafts, bile duct obstruction, ureter obstruction, tumor obstruction, restenosis and progression of atherosclerosis in patient subsets including type I diabetics, type II diabetics, metabolic syndrome and syndrome X, vulnerable lesions including those with thin-capped fibroatheromatous lesions, systemic infections including gingivitis, hellobacteria, and cytomegalovirus, and combinations thereof.
  • a disorder such as thrombosis, high cholesterol, hemorrhage, vascular dissection or perforation, vascular aneurysm, vulnerable plaque, chronic total occlusion, claudication, anastomotic proliferation for vein and artificial grafts, bile
  • a common disorder in association with mechanical modification of a vessel, such as by a balloon or stenting is restenosis.
  • a number of cellular mechanisms have been proposed that lead to restenosis of a vessel. Two of these mechanisms are (1) the migration and proliferation of smooth muscle cells to and at the site of injury, and (2) the acute and chronic inflammatory response to injury and foreign body presence.
  • Inflammation is a defensive, biological response to injury, infection or an abrupt change in tissue homeostasis. Inflammation can occur anywhere in the body, and most of the time is confined to that part of the body. Well-known indicators of inflammation are pain, redness, warmth, swelling, and loss of function. In nature, inflammatory responses are designed to destroy, dilute and isolate injurious agents and then lead to recovery and repair of the affected tissue. The intensity of an inflammatory response can vary from one that is self-limiting, which requires minor therapeutic intervention, to one that is life threatening, which requires intense intervention. One drawback of the inflammatory process is its ability to become progressive, meaning tissue damage continues after the stimulus is neutralized or removed.
  • vascular inflammation is the first stage of the inflammatory response, developing after the initial contact with the stimulus and continuing sometimes for several days.
  • the presence of a stimulatory agent in the blood or in the tissue triggers the body's response through endothelial cells.
  • the endothelial cell layer is the innermost layer of larger vessels and the only cell layer of the smallest vessels, the capillaries.
  • Endothelial cells produce substances called chemokines that attract neutrophils and other white blood cells to the site of injury. Within the site, neutrophils and endothelium relay information back and forth across cell membranes through presentation of adhesion molecules and cytokines. Cellular cross-talk promotes physical interaction between the “inflamed” neutrophil and the “inflamed” endothelium.
  • endothelial cell swelling Another important pathological feature of vascular inflammation is endothelial cell swelling. This action reduces the functional vessel diameter such that the speed of blood flow falls significantly and the vessel becomes congested. When these conditions predominate, inflamed neutrophils are induced to plug the vessel. As a result, endothelial cells lose their tight connections allowing neutrophils to transmigrate into the surrounding tissue.
  • neutrophils begin to enter the tissue and may continue transmigration for many days.
  • the appearance of inflammatory cells in the surrounding tissue marks the beginning of tissue damage.
  • tissue damage is caused by direct injury of the vessels and amplified by the subsequent recruitment of neutrophils into the tissue.
  • Tissue repair is the third and final stage of inflammation. It may take several days for tissue destruction to reach full intensity before tapering off. Until then, the tissue repair process that consists of growth of new blood vessels and entry of monocytes to clean up the debris is delayed. Fibroblasts also enter the local tissue to replace the extracellular matrix and collagen. The process of tissue repair is stringently controlled within the tissue site. If the process becomes dysregulated, inappropriate tissue repair will lead to excessive scarring. Depending on the tissue and the intensity/duration of the inflammatory condition, the amount of scarring can be significant.
  • VP vulnerable plaque
  • Previous studies have demonstrated that inflammation promotes proliferation at sites of balloon angioplasty and stent placement in pigs ( Komowski, et al., Coron Artery Dis. 12(6):513-5 (2001)). Since sites of vulnerable plaque have a higher density of macrophages and lymphocytes than other types of atherosclerotic lesions, it is expected that these sites, when stented, will produce elevated amounts of the cytokines (IL-1, TNF-alpha) that promote smooth muscle cell proliferation.
  • IL-1 cytokines
  • diabetes Another example of disorders that vessel inflammation is involved is diabetes. Studies have shown that patients with type-2 diabetes have higher rates of restenosis than the general population. The diabetic patient is in pro-inflammatory state that can amplify restenosis because diabetic lesions contain a large number of inflammatory cells (e.g., macrophages, lymphocytes, etc.).
  • inflammatory cells e.g., macrophages, lymphocytes, etc.
  • the anti-proliferative agent can be a natural proteineous agent such as a cytotoxin or a synthetic molecule.
  • the active agents include antiproliferative substances such as actinomycin D, or derivatives and analogs thereof (manufactured by Sigma-Aldrich 1001 West Saint Paul Avenue, Milwaukee, Wis.
  • actinomycin D include dactinomycin, actinomycin IV, actinomycin I 1 , actinomycin X 1 , and actinomycin C 1 ), all taxoids such as taxols, docetaxel, and paclitaxel, paclitaxel derivatives, all olimus drugs such as macrolide antibiotics, rapamycin, everolimus, structural derivatives and functional analogues of rapamycin, structural derivatives and functional analogues of everolimus, FKBP-12 mediated mTOR inhibitors, biolimus, perfenidone, prodrugs thereof, co-drugs thereof, and combinations thereof.
  • rapamycin derivatives include 40-O-(3-hydroxy)propyl-rapamycin, 40-O-[2-(2-hydroxy)ethoxy]ethyl-rapamycin, or 40-O-tetrazole-rapamycin, prodrugs thereof, co-drugs thereof, and combinations thereof.
  • the anti-proliferative agent is everolimus.
  • Everolimus acts by first binding to FKBP12 to form a complex (Neuhhaus, P., et al., Liver Transpl. 2001 7(6):473-84 (2001) (Review)).
  • the everolimus /FKBP 12 complex then binds to mTOR and blocks its activity (Id.).
  • mTOR inhibition has also been shown to inhibit vascular smooth muscle migration.
  • anti-inflammatory drugs can be a steroidal anti-inflammatory agent, a nonsteroidal anti-inflammatory agent, or a combination thereof.
  • anti-inflammatory drugs include, but are not limited to, alclofenac, alclometasone dipropionate, algestone acetonide, alpha amylase, amcinafal, amcinafide, amfenac sodium, amiprilose hydrochloride, anakinra, anirolac, anitrazafen, apazone, balsalazide disodium, bendazac, benoxaprofen, benzydamine hydrochloride, bromelains, broperamole, budesonide, carprofen, cicloprofen, cintazone, cliprofen, clobetasol propionate, clobetasone butyrate, clopirac, cloticasone propionate, cormethasone acetate
  • the anti-inflammatory agent is clobetasol.
  • Clobetasol is a corticosteroid that binds to corticosteroid receptors, a class of nuclear receptor. The binding of clobetasol to the corticosteroid receptor subsequently alters gene expression in such a way that inflammation is inhibited.
  • corticosteroids inhibit the activation of NFkB, the nuclear factor that is responsible for changes in gene expression that promote inflammation. The reduction in inflammation may also inhibit the mechanisms that promote small muscle cell (SMC) hyper proliferation.
  • SMC small muscle cell
  • dexamethasone a less potent glucocorticoid as compared to clobetasol, reduces the production of PGDF and thus has anti-proliferative properties.
  • Clobetasol acts through similar pathways and is more potent than dexamethasone.
  • the dosage or concentration of the anti-proliferative and anti-inflammatory agents required to produce a favorable therapeutic effect should be less than the level at which the bioactive agent produces toxic effects and greater than the level at which non-therapeutic results are obtained.
  • the dosage or concentration of the agents required can depend upon factors such as the particular circumstances of the patient, the nature of the trauma, the nature of the therapy desired, the time over which the ingredient administered resides at the vascular site, and if other active agents are employed, the nature and type of the substance or combination of substances.
  • Therapeutic effective dosages can be determined empirically, for example by infusing vessels from suitable animal model systems and using immunohistochemical, fluorescent or electron microscopy methods to detect the agent and its effects, or by conducting suitable in vitro studies.
  • the bioactive agents can be incorporated into polymeric coating in a percent loading of between about 0.01% and less than about 100% by weight, more preferably between about 5% and about 50% by weight of the total drug-load that includes greater than about 0% to about 100% of the anti-proliferative agent and less than about 100% to greater than about 0% of the anti-inflammatory agent.
  • the relative amount of the anti-proliferative agent and anti-inflammatory agent can be determined by the type of lesions to be treated.
  • the relative amount of everolimus and clobetasol can be varied for different types of lesions, that is, the relative amount of everolimus can be higher for more proliferative lesions, and on the other hand, the relative amount of clobetasol can be higher for more inflammatory lesions.
  • bioactive agents can be any agent which is a therapeutic, prophylactic, or diagnostic agent. These agents can also have anti-proliferative and/or anti-inflammmatory properties or can have other properties such as antineoplastic, antiplatelet, anti-coagulant, anti-fibrin, antithrombonic, antimitotic, antibiotic, antiallergic, antioxidant as well as cystostatic agents.
  • suitable therapeutic and prophylactic agents include synthetic inorganic and organic compounds, proteins and peptides, polysaccharides and other sugars, lipids, and DNA and RNA nucleic acid sequences having therapeutic, prophylactic or diagnostic activities.
  • Nucleic acid sequences include genes, antisense molecules which bind to complementary DNA to inhibit transcription, and ribozymes.
  • Some other examples of other bioactive agents include antibodies, receptor ligands, enzymes, adhesion peptides, blood clotting factors, inhibitors or clot dissolving agents such as streptokinase and tissue plasminogen activator, antigens for immunization, hormones and growth factors, oligonucleotides such as antisense oligonucleotides and ribozymes and retroviral vectors for use in gene therapy.
  • antineoplastics and/or antimitotics examples include methotrexate, azathioprine, vincristine, vinblastine, fluorouracil, doxorubicin hydrochloride (e.g. Adriamycin® from Pharmacia & Upjohn, Peapack N.J.), and mitomycin (e.g. Mutamycin® from Bristol-Myers Squibb Co., Stamford, Conn.).
  • antiplatelets examples include sodium heparin, low molecular weight heparins, heparinoids, hirudin, argatroban, forskolin, vapiprost, prostacyclin and prostacyclin analogues, dextran, D-phe-pro-arg-chloromethylketone (synthetic antithrombin), dipyridamole, glycoprotein IIb/IIIa platelet membrane receptor antagonist antibody, recombinant hirudin, thrombin inhibitors such as Angiomax a (Biogen, Inc., Cambridge, Mass.), calcium channel blockers (such as nifedipine), colchicine, fibroblast growth factor (FGF) antagonists, fish oil (omega 3-fatty acid), histamine antagonists, lovastatin (an inhibitor of HMG-CoA reductase, a cholesterol lowering drug, brand name Mevacor® from Merck & Co.,
  • cytostatic substance examples include angiopeptin, angiotensin converting enzyme inhibitors such as captopril (e.g. Capoten® and Capozide® from Bristol-Myers Squibb Co., Stamford, Conn.), cilazapril or lisinopril (e.g. Prinivil® and Prinzide® from Merck & Co., Inc., Whitehouse Station, N.J.).
  • captopril e.g. Capoten® and Capozide® from Bristol-Myers Squibb Co., Stamford, Conn.
  • cilazapril or lisinopril e.g. Prinivil® and Prinzide® from Merck & Co., Inc., Whitehouse Station, N.J.
  • An example of an antiallergic agent is permirolast potassium.
  • Other therapeutic substances or agents which may be appropriate include alpha-interferon, and genetically engineered epithelial cells. The foregoing substances are listed by way
  • composition comprising both anti-proliferative agent and the anti-inflammatory agent can be formulated into any formulation suitable for delivery by any mode of delivery.
  • the composition can be formed into a coating on an implantable medical device to provide controlled release of the anti-proliferative agent and the anti-inflammatory agent.
  • the composition can also be formulated into other suitable formulations for example, bolus dose of free drug, optionally with a fluoroscopic dye, bolus dose of gel-encapsulated drug.
  • the gel can be formed of a gel-forming material or polymer such as hyaluronic acid, carboxymethyl cellulose, pectin, hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, sodium carboxymethylcellulose, hydroxyethylcellulose, polyethylene oxide, acacia, tragacanth, guar gum, xanthan gum, locust bean gum, CarbopolTM acidic carboxy polymer, polycarbophil, polyethylene oxide, poly(hydroxyalkyl methacrylate), poly(electrolyte complexes), poly(vinyl acetate) cross-linked with hydrolyzable bonds, water-swellable N-vinyl lactams polysaccharides, natural gum, agar, agarose, sodium alginate, carrageenan, fucoidan, furcellaran, laminaran, hypnea, eucheuma, gum arabic, gum ghatti, gum karaya, arbinogla
  • the gel-forming material or polymer can be hydropropyl methylcellulose having 19-24% methoxyl substitution and 7-12% hydroxypropyl substitution and a number average molecular weight of at least 20,000.
  • Such polymers include those sold by Dow Chemical Co. under the tradenames Methocel K4M, Methocel K15M and Methocel K100M.
  • the anti-inflammatory drug such as clobetasol is formulated into a bolus dose of free drug with, optionally, a fluoroscopic dye.
  • the anti-proliferative drug such as everolimus can be formulated into a coating composition with a polymeric material and then coated onto an implantable device (e.g., stent).
  • the bolus dose of anti-inflammatory drug is administered first and then the anti-proliferative drug is delivered by release from the implantable device such as a drug-delivery stent.
  • the composition may further include a third agent such as a HDL (high density lipoprotein)-mimic as described in U.S. Pat. No. 6,367,479.
  • HDL-mimic can be delivered by the stent.
  • the anti-inflammatory drug such as clobetasol is formulated into a bolus dose of gel.
  • the anti-proliferative drug such as everolimus can be formulated into a coating composition with a polymeric material and then coated onto an implantable device.
  • the bolus dose of the anti-inflammatory drug is administered first and then the anti-proliferative drug is delivered by release from the implantable device such as a drug-delivery stent.
  • the anti-inflammatory drug and the anti-proliferative drug can be included in a polymeric matrix and then coated onto a medical device such as a stent.
  • the medical device coating can be designed to have a variety of different release parameters for each of the drugs included in the coating.
  • the anti-inflammatory can have one or a combination of release profiles that include a pulse release, fast or burst release, and a sustained release.
  • the anti-proliferative drug can have one or a combination of release profiles that include a pulse release, fast or burst release, and a sustained release from the stent.
  • the combination can be delivered simultaneously or at least during the drug treatment period there is at lease some overlap between the release of the drugs.
  • the anti-inflammatory can be completely released prior to the release to the anti-proliferative or can be partially released with some or significant overlap between the release of both drugs.
  • “Pulse release” generally refers to a release profile of a drug that features a sudden surge of the release rate of the drug. The release rate surge of the drug would then disappear within a period. A more detailed definition of the term can be found in Encyclopedia of Controlled Drug Delivery, Edith Mathiowitz, Ed., Culinary and Hospitality Industry Publications Services.
  • the term “fast release” in one embodiment refers to a release profile of a drug that features a release rate in the range between about 15 to about 40 ⁇ g per day for a 18 mm stent, about 10 ⁇ g to about 27 ⁇ g per day for a 13 mm stent, and about 6.7 ⁇ g to about 17.2 ⁇ g per day for a 8 mm stent. Equivalent profiles can be derived by one having ordinary skill in the art for stents having other sizes.
  • the term “fast release” refers to an approximately 20% release in 24 hours of a drug.
  • the term “fast release” is used interchangeably with the term “burst release.”
  • sustained release generally refers to a release profile of a drug that can include zero-order release, exponential decay, step-function release or other release profiles that carry over a period of time, for example, ranging from several days to several years.
  • zero-order release “exponential decay” and “step-function release” as well as other sustained release profiles are well known in the art (see, for example, Encyclopedia of Controlled D rug Delivery, Edith Mathiowitz, Ed., Culinary and Hospitality Industry Publications Services).
  • At least one of the anti-inflammatory agent (e.g., clobetasol) and anti-proliferative agent (e.g., everolimus) is administered via a stent while the other is administered by other local means of administration or alternatively, the other is administered systemically. In other embodiments, both are administered locally, by means other than a stent, or alternatively systemically.
  • Systemic administration can be accomplished orally or parenterally including intravascularly, rectally, intranasally, intrabronchially, or transdermally.
  • Liquid carriers which are sterile solutions or suspensions can be injected intramuscularly, intraperitoneally, subcutaneously, and intravenously. Rectal administration can be in the form of conventional suppository.
  • the drug can be formulated into an aqueous or partially aqueous solution, which can then be utilized in the form of an aerosol.
  • the drug can be administered transdermally through the used of a transdermal patch and a carrier that is inert to and mutually compatible with the active component, is non-toxic to the skin, and allows for the delivery of the drug for systemic absorption into the blood stream via the skin.
  • the carrier may take any number of forms such as creams, ointments, pastes, and gels.
  • the creams and ointments may be viscous liquids or semisolid emulsions of either the oil-in-water or water-in-oil type.
  • Pastes made of absorptive powders dispersed in petroleum or hydrophilic petroleum containing the active component may also be suitable.
  • Other devices capable of releasing the drug into the blood stream include semi-permeable membranes covering a reservoir containing the drug, with or without a carrier.
  • Local administration can be accomplished by a variety of techniques which administer the active component at or near the target site.
  • the following examples of local delivery techniques are provided for illustrative purposes and are not intended to be limiting. Examples include local delivery catheters, site specific carriers, implants, direct application, or direct injection. Local delivery by a catheter allows for the administration of the drug directly to the target site.
  • Local delivery by site specific carriers is conducted by attaching the drug to a carrier which will direct or link the drug to the target cells.
  • a carrier such as a protein ligand, a monoclonal antibody or a membrane anchored linker.
  • Local delivery by an implant is the placement of a matrix carrying the drug at the site.
  • the matrix can release the active component by, for example, diffusion, degradation, chemical reaction, solvent activators, etc.
  • One example of local delivery by an implant can include direct injection of vesicles or micro-particles. These micro-particles may be composed of substances such as proteins, lipids, carbohydrates or synthetic polymers. The micro-particles can have the drug impregnated therein and/or coated thereon.
  • Application via implants is not limited to the above described routes and other techniques such as grafts, micropumps or application of a fibrin glue or hydrogel containing the active component around the exterior of a designated region of the vessel can also be implemented by one of ordinary skill in the art.
  • Local delivery by direct injection describes injecting a liquid carrier containing the drug directly into the site.
  • the liquid carrier should be inert to and mutually compatible with the drug.
  • the component can be in true solution or suspended in fine particles in the carrier.
  • a suitable example of an inert carrier includes a sterile saline solution.
  • biocompatible polymers can be used to form a coating on a stent or to provide a drug delivery particle with the anti-proliferative drug and/or anti-inflammatory drug.
  • biocompatible, bioabsorbable polymers include, but not limited to, poly(ester amide), poly(ester amide) that may contain alkyl groups, amino acid groups, or poly(ethylene glycol) (PEG) groups, polyethylene glycol (PEG), polylakanoates (PHA), poly(2-hydroxyalkanoates), poly(3-hydroxyalkanoates) such as poly(3-hydroxypropanoate), poly(3-hydroxybutyrate), poly(3-hydroxyvalerate), poly(3-hydroxyhexanoate), poly(3-hydroxyheptanoate) and poly(3-hydroxyoctanoate), poly(4-hydroxyalknaote) such as poly(4-hydroxybutyrate), poly(4-hydroxyvalerate), poly(4-hydroxyhexanote), poly(4-hydroxyheptano
  • the coating described herein can be formed by spray coating or any other coating process available in the art.
  • the coating involves dissolving or suspending the composition, or one or more components thereof, in a solvent or solvent mixture to form a solution, suspension, or dispersion of the composition or one or more components thereof, applying the solution or suspension to an implantable device, and removing the solvent or solvent mixture to form a coating or a layer of coating.
  • Suspensions or dispersions of the composition described herein can be in the form of latex or emulsion of microparticles having a size between 1 nanometer and 100 microns, preferably between 1 nanometer and 10 microns. Heat and/or pressure treatment can be applied to any of the steps involved herein.
  • the coating described here can be subjected to further heat and/or pressure treatment.
  • Some additional exemplary processes of coating an implantable device that may be used are described in, for example, Lambert T L, et al. Circulation, 1994, 90: 1003-1011; Hwang C W, et al. Circulation, 2001; 104: 600-605; Van der Giessen W J, et al. Circulation, 1996; 94: 1690-1697; Lincoff A M, et al. J Am Coll Cardiol 1997; 29: 808-816; Grube E. et al, J American College Cardiology Meeting, Mar.
  • solvent refers to a liquid substance or composition that is compatible with the polymer and is capable of dissolving or suspending the polymeric composition or one or more components thereof at a desired concentration.
  • solvents include chloroform, acetone, water (buffered saline), dimethylsulfoxide (DMSO), propylene glycol monomethyl ether (PM,) iso-propylalcohol (IPA), n-propyl alcohol, methanol, ethanol, tetrahydrofuran (THF), dimethylformamide (DMF), dimethyl acetamide (DMAC), benzene, toluene, xylene, hexane, cyclohexane, heptane, octane, nonane, decane, decalin, ethyl acetate, butyl acetate, isobutyl acetate, isopropyl acetate, butanol, diacetone alcohol
  • implantable devices include self-expandable stents, balloon-expandable stents, stent-grafts, grafts (e.g., aortic grafts), artificial heart valves, cerebrospinal fluid shunts, pacemaker electrodes, and endocardial leads (e.g., FINELINE and ENDOTAK, available from Guidant Corporation, Santa Clara, Calif.).
  • the underlying structure of the device can be of virtually any design.
  • the device can be made of a metallic material or an alloy such as, but not limited to, cobalt chromium alloy (ELGILOY), stainless steel (316L), high nitrogen stainless steel, e.g., BIODUR 108, cobalt chrome alloy L-605, “MP35N,” “MP20N,” ELASTINITE (Nitinol), tantalum, nickel-titanium alloy, platinum-iridium alloy, gold, magnesium, or combinations thereof.
  • ELGILOY cobalt chromium alloy
  • stainless steel 316L
  • high nitrogen stainless steel e.g., BIODUR 108, cobalt chrome alloy L-605, “MP35N,” “MP20N,” ELASTINITE (Nitinol), tantalum, nickel-titanium alloy, platinum-iridium alloy, gold, magnesium, or combinations thereof.
  • BIODUR 108 cobalt chrome alloy L-605, “MP35N,” “MP20N,” ELASTINITE (Nitinol)
  • tantalum nickel-t
  • the implantable device is a stent, which can be degradable stents, biodurable stents, depot stents, and metallic stens such as stents made of stainless steel or nitinol.
  • the stents can be balloon expandable or self expanding.
  • a coating of the various described embodiments can be formed on an implantable device or prosthesis, e.g., a stent.
  • the agent will be retained on the medical device such as a stent during delivery and expansion of the device, and released at a desired rate and for a predetermined duration of time at the site of implantation.
  • the medical device is a stent.
  • a stent having the above-described coating is useful for a variety of medical procedures, including, by way of example, treatment of obstructions caused by tumors in bile ducts, esophagus, trachea/bronchi and other biological passageways.
  • a stent having the above-described coating is particularly useful for treating occluded regions of blood vessels caused by abnormal or inappropriate migration and proliferation of smooth muscle cells, thrombosis, and restenosis.
  • Stents may be placed in a wide array of blood vessels, both arteries and veins. Representative examples of sites include the iliac, renal, and coronary arteries.
  • an angiogram is first performed to determine the appropriate positioning for stent therapy.
  • An angiogram is typically accomplished by injecting a radiopaque contrasting agent through a catheter inserted into an artery or vein as an x-ray is taken.
  • a guidewire is then advanced through the lesion or proposed site of treatment.
  • Over the guidewire is passed a delivery catheter which allows a stent in its collapsed configuration to be inserted into the passageway.
  • the delivery catheter is inserted either percutaneously or by surgery into the femoral artery, brachial artery, femoral vein, or brachial vein, and advanced into the appropriate blood vessel by steering the catheter through the vascular system under fluoroscopic guidance.
  • a stent having the above-described coating may then be expanded at the desired area of treatment.
  • a post-insertion angiogram may also be utilized to confirm appropriate positioning.
  • the implantable device comprising a coating described herein can be used to treat an animal having a condition or disorder that requires a treatment.
  • Such an animal can be treated by, for example, implanting a device described herein in the animal.
  • the animal is a human being.
  • Exemplary disorders or conditions that can be treated by the method disclosed herein include, but not limited to, thrombosis, high cholesterol, hemorrhage, vascular dissection or perforation, vascular aneurysm, vulnerable plaque, chronic total occlusion, claudication, anastomotic proliferation for vein and artificial grafts, bile duct obstruction, ureter obstruction, tumor obstruction, restenosis and progression of atherosclerosis in patient subsets including type I diabetics, type II diabetics, metabolic syndrome and syndrome X, vulnerable lesions including those with thin-capped fibroatheromatous lesions, systemic infections including gingivitis, hellobacteria, and cytomegalovirus, and combinations thereof.
  • Described in this example is a 28 day porcine implant study that compared the 200 ⁇ g/cm 2 dose Lemans with a clobetasol-only delivery stent, an everolimus-only stent, and an everolimus-clobetasol combination drug delivery stent.
  • the study was performed using three different drug delivery stents, Arm 1, Arm 2, and Arm 3.
  • Arm 1 is Lemans stent (a stent available from Guidant based on PVDF-co-HFP) that included 105 ⁇ g everolimus and used as a control.
  • Arm 2 was loaded with 185 ⁇ g clobetasol only, with no everolimus.
  • Arm 3 is loaded with 105 ⁇ g everolimus and 80 ⁇ g clobetasol.
  • the Arm 1, Arm 2, and Arm 3 stents were implanted in a 30% overstretch model.
  • Nine samples of each Arm stent were implanted, one for each coronary artery.
  • 24 hr Release in porcine serum data were gathered.
  • 3, 7 and 28 day in vivo release data were gathered (from the mammary arteries), as was 28 day quantitative coronary angioplasty (QCA), histology and morphometry.
  • QCA quantitative coronary angioplasty
  • the p values from a t-test of the data from FIG. 3 are summarized in Table 4.
  • a “t-test” returns the probability associated with a Student's t-Test that determines whether two samples are likely to have come from the same two underlying populations that have the same mean.
  • the value returned from the test, “p”, is the probability that the two groups of data come from the same population.
  • p Values less than or equal to 0.10 or 0.05 are generally considered significant (Zar, J H. Biostatistical Analysis . Englewood Cliffs, N.J.: Prentice-Hall Inc, 1974. pp 101-108). TABLE 4 p Values from a t-test of the data from FIG.
  • Neointi- Neointi- Media mal % mal Injury Arm Comparison Area Area Stenosis Thickness Score EVER COMBO 0.05 0.01 0.02 0.01 0.18 EVER CLOB 0.29 0.90 0.77 0.93 0.78 COMBO CLOB 0.24 0.18 0.22 0.14 0.25

Abstract

Drug-delivery systems such as drug-delivery stents having an anti-proliferative agent such as everolimus and an anti-flammatory agent such as clobetasol are provided. Also disclosed are methods of treating a vascular impairment such as restenosis or vulnerable plaque.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • This invention generally relates to a drug combination including an anti-proliferative drug such as everolimus and an anti-inflammatory agent such as clobetasol for the treatment of a disorder such as restenosis and vulnerable plaque.
  • 2. Description of the Background
  • Plaques have been associated with stenosis and restenosis. While treatments of plaque-induced stenosis and restenosis have advanced significantly over the last few decades, the morbidity and mortality associated with vascular plaques have remained significant. Recent work suggests that plaque may generally fall into one of two different general types: standard stenotic plaques and vulnerable plaques. Stenotic plaque, which is sometimes referred to as thrombosis-resistant plaque, can generally be treated effectively by the known intravascular lumen opening techniques. Although plaques induce stenoses, these atherosclerotic plaques themselves are often a benign and are an effectively treatable disease.
  • Unfortunately, as plaque matures, narrowing of a blood vessel by a proliferation of smooth muscle cells, matrix synthesis, and lipid accumulation may result in formation of a plaque which is quite different than a standard stenotic plaque. Such atherosclerotic plaque becomes thrombosis-prone, and can be highly dangerous. This thrombosis-prone or vulnerable plaque may be a frequent cause of acute coronary syndrome.
  • While the known procedures for treating plaque have gained wide acceptance and shown good efficacy for treatment of standard stenotic plaques, they may be ineffective (and possibly dangerous) when thrombotic conditions are superimposed on atherosclerotic plaques. Specifically, mechanical stresses caused by primary treatments like percutaneous transluminal intervention (PTI), such as stenting, may actually trigger release of fluids and/or solids from a vulnerable plaque into the blood stream, thereby potentially causing a coronary thrombotic occlusion. For example, rupture of the fibrous cap that overlies the thrombogenic necrotic core is presently believed to play an important role in acute ischemic events, such as stroke, transient ischemic attack, myocardial infarction, and unstable angina (Virmani R, et al. Arterioscler Thromb Vasc Biol. 20: 1262-1275 (2000)). There is evidence that fibrous cap can be ruptured during stent deployment. Human data from various sources have indicated that lipid rich and/or positively remodeled and/or echolucent lesions in sysmptomatic coronary atherosclerosis have higher likelihood for restenosis (See, for example, J. Am. Coll. Cardiol. 21(2):298-307 (1993); Am. J. Cardiol. 89(5):505 (2002); Circ. 94(12):3098-102 (1996)). Therefore, there is a need for the treatment of vulnerable plaques and restenosis.
  • The embodiments of the present invention address these and other needs.
  • SUMMARY OF THE INVENTION
  • Described herein are a drug-delivery system and the method of using the drug-delivery system. The drug-delivery system has two or more drugs for treating a vascular disorder or a related disorder. The drugs can be a combination of at least one anti-proliferative agent, at least one anti-inflammatory agent, and optionally a third bioactive agent. In one embodiment, the anti-proliferative agent can be a drug such as everolimus, and the anti-inflammatory agent can be a drug such as clobetasol.
  • Methods of treating of preventing vascular disorders such as restenosis and vulnerable plaque are also disclosed by administering to the patient a combination of at least one anti-proliferative agent, at least one anti-inflammatory agent, and optionally a third bioactive agent. The mode of delivery can be local or systemic.
  • BRIEF DESCRIPTION OF THE FIGURE
  • FIG. 1 shows the results of 28 day quantitative coronary angioplasty (QCA) of a porcine implant study on drug-delivery systems described herein.
  • FIG. 2 shows 28 day histology data of a porcine implant study on drug-delivery systems described herein.
  • FIG. 3 shows the 28 day morphometry data of a porcine implant study on drug-delivery systems described herein.
  • DETAILED DESCRIPTION Anti-Proliferative Agents and Anti-Inflammatory Agents
  • In accordance with one embodiment, described herein are a drug-delivery system and the method of using the drug-delivery system. The term “treatment” includes prevention, reduction, delay or elimination of the vascular disorder. In some embodiments, treatment also includes repairing damage caused by the dirorder and/or the mechanical intervention. The drug-delivery system has two or more drugs for treating a vascular disorder or a related disorder. The drugs can be a combination of at least one anti-proliferative agent, at least one anti-inflammatory agent, and optionally a third bioactive agent.
  • In one embodiment, the composition described herein includes an effective amount of at least one anti-inflammatory agent and an effective amount of an anti-proliferative agent. In another embodiment, the composition described herein includes an effective amount of an agent which is effective both as an anti-inflammatory agent and as an anti-proliferative agent.
  • In some embodiments, the anti-proliferative agent can be everolimus (available under the trade name Certican™, Novartis Pharma A G, Germany), and the anti-inflammatory agent can be clobetasol (available under the trade name Temovate™, Glaxosmithkline, UK).
  • The anti-proliferative agent and the anti-inflammatory agent can be in the form of a coating with and/or without a polymer matrix on a medical device or at elast one of the agents can be administered in a separate dose form such as bolus dose of a free drug, optionally with fluoroscopic dye, or bolus dose of a gel encapsulating a drug. The drug-delivery system or composition may further include a third agent such as a high-density lipoproptein mimetic (HDL-mimetic). For example, an anti-inflammatory agent such as clobetasol can be delivered along with the catheter based delivery of a HDL-mimetic while everolimus is administered by a stent.
  • The drug-delivery system or composition disclosed herein can be used to treat or prevent a disorder such as thrombosis, high cholesterol, hemorrhage, vascular dissection or perforation, vascular aneurysm, vulnerable plaque, chronic total occlusion, claudication, anastomotic proliferation for vein and artificial grafts, bile duct obstruction, ureter obstruction, tumor obstruction, restenosis and progression of atherosclerosis in patient subsets including type I diabetics, type II diabetics, metabolic syndrome and syndrome X, vulnerable lesions including those with thin-capped fibroatheromatous lesions, systemic infections including gingivitis, hellobacteria, and cytomegalovirus, and combinations thereof.
  • Inflammation in Stenting a Vessel
  • A common disorder in association with mechanical modification of a vessel, such as by a balloon or stenting is restenosis. A number of cellular mechanisms have been proposed that lead to restenosis of a vessel. Two of these mechanisms are (1) the migration and proliferation of smooth muscle cells to and at the site of injury, and (2) the acute and chronic inflammatory response to injury and foreign body presence.
  • Inflammation is a defensive, biological response to injury, infection or an abrupt change in tissue homeostasis. Inflammation can occur anywhere in the body, and most of the time is confined to that part of the body. Well-known indicators of inflammation are pain, redness, warmth, swelling, and loss of function. In nature, inflammatory responses are designed to destroy, dilute and isolate injurious agents and then lead to recovery and repair of the affected tissue. The intensity of an inflammatory response can vary from one that is self-limiting, which requires minor therapeutic intervention, to one that is life threatening, which requires intense intervention. One drawback of the inflammatory process is its ability to become progressive, meaning tissue damage continues after the stimulus is neutralized or removed.
  • Vascular inflammation is the first stage of the inflammatory response, developing after the initial contact with the stimulus and continuing sometimes for several days. The presence of a stimulatory agent in the blood or in the tissue triggers the body's response through endothelial cells. The endothelial cell layer is the innermost layer of larger vessels and the only cell layer of the smallest vessels, the capillaries. Endothelial cells produce substances called chemokines that attract neutrophils and other white blood cells to the site of injury. Within the site, neutrophils and endothelium relay information back and forth across cell membranes through presentation of adhesion molecules and cytokines. Cellular cross-talk promotes physical interaction between the “inflamed” neutrophil and the “inflamed” endothelium.
  • Another important pathological feature of vascular inflammation is endothelial cell swelling. This action reduces the functional vessel diameter such that the speed of blood flow falls significantly and the vessel becomes congested. When these conditions predominate, inflamed neutrophils are induced to plug the vessel. As a result, endothelial cells lose their tight connections allowing neutrophils to transmigrate into the surrounding tissue.
  • Within hours of the initial stimulus, neutrophils begin to enter the tissue and may continue transmigration for many days. The appearance of inflammatory cells in the surrounding tissue marks the beginning of tissue damage. In some inflammatory conditions, tissue damage is caused by direct injury of the vessels and amplified by the subsequent recruitment of neutrophils into the tissue.
  • Activated by local mediators, neutrophils and tissue macrophages are triggered to release agents that destroy toxins and clean up dead cells in the area. Unfortunately, these same agents also cause collateral damage to healthy cells, which further extends the borders of the initial tissue destruction.
  • Tissue repair is the third and final stage of inflammation. It may take several days for tissue destruction to reach full intensity before tapering off. Until then, the tissue repair process that consists of growth of new blood vessels and entry of monocytes to clean up the debris is delayed. Fibroblasts also enter the local tissue to replace the extracellular matrix and collagen. The process of tissue repair is stringently controlled within the tissue site. If the process becomes dysregulated, inappropriate tissue repair will lead to excessive scarring. Depending on the tissue and the intensity/duration of the inflammatory condition, the amount of scarring can be significant.
  • An example of disorders that vessel inflammation is involved is vulnerable plaque (VP) rupture. Previous studies have demonstrated that inflammation promotes proliferation at sites of balloon angioplasty and stent placement in pigs (Komowski, et al., Coron Artery Dis. 12(6):513-5 (2001)). Since sites of vulnerable plaque have a higher density of macrophages and lymphocytes than other types of atherosclerotic lesions, it is expected that these sites, when stented, will produce elevated amounts of the cytokines (IL-1, TNF-alpha) that promote smooth muscle cell proliferation.
  • Another example of disorders that vessel inflammation is involved is diabetes. Studies have shown that patients with type-2 diabetes have higher rates of restenosis than the general population. The diabetic patient is in pro-inflammatory state that can amplify restenosis because diabetic lesions contain a large number of inflammatory cells (e.g., macrophages, lymphocytes, etc.).
  • Anti-Proliferative Agents
  • Any drugs having anti-proliferative effects can be used in the present invention. The anti-proliferative agent can be a natural proteineous agent such as a cytotoxin or a synthetic molecule. Preferably, the active agents include antiproliferative substances such as actinomycin D, or derivatives and analogs thereof (manufactured by Sigma-Aldrich 1001 West Saint Paul Avenue, Milwaukee, Wis. 53233; or COSMEGEN available from Merck) (synonyms of actinomycin D include dactinomycin, actinomycin IV, actinomycin I1, actinomycin X1, and actinomycin C1), all taxoids such as taxols, docetaxel, and paclitaxel, paclitaxel derivatives, all olimus drugs such as macrolide antibiotics, rapamycin, everolimus, structural derivatives and functional analogues of rapamycin, structural derivatives and functional analogues of everolimus, FKBP-12 mediated mTOR inhibitors, biolimus, perfenidone, prodrugs thereof, co-drugs thereof, and combinations thereof. Representative rapamycin derivatives include 40-O-(3-hydroxy)propyl-rapamycin, 40-O-[2-(2-hydroxy)ethoxy]ethyl-rapamycin, or 40-O-tetrazole-rapamycin, prodrugs thereof, co-drugs thereof, and combinations thereof.
  • In one embodiment, the anti-proliferative agent is everolimus. Everolimus acts by first binding to FKBP12 to form a complex (Neuhhaus, P., et al., Liver Transpl. 2001 7(6):473-84 (2001) (Review)). The everolimus /FKBP 12 complex then binds to mTOR and blocks its activity (Id.). By blocking mTOR activity, cells are unable to pass through GI of the cell cycle and as a result, proliferation is inhibited. mTOR inhibition has also been shown to inhibit vascular smooth muscle migration.
  • Anti-Inflammatory Agents
  • Any drugs having anti-inflammatory effects can be used in the present invention. The anti-inflammatory drug can be a steroidal anti-inflammatory agent, a nonsteroidal anti-inflammatory agent, or a combination thereof. In some embodiments, anti-inflammatory drugs include, but are not limited to, alclofenac, alclometasone dipropionate, algestone acetonide, alpha amylase, amcinafal, amcinafide, amfenac sodium, amiprilose hydrochloride, anakinra, anirolac, anitrazafen, apazone, balsalazide disodium, bendazac, benoxaprofen, benzydamine hydrochloride, bromelains, broperamole, budesonide, carprofen, cicloprofen, cintazone, cliprofen, clobetasol propionate, clobetasone butyrate, clopirac, cloticasone propionate, cormethasone acetate, cortodoxone, deflazacort, desonide, desoximetasone, dexamethasone dipropionate, diclofenac potassium, diclofenac sodium, diflorasone diacetate, diflumidone sodium, diflunisal, difluprednate, diftalone, dimethyl sulfoxide, drocinonide, endrysone, enlimomab, enolicam sodium, epirizole, etodolac, etofenamate, felbinac, fenamole, fenbufen, fenclofenac, fenclorac, fendosal, fenpipalone, fentiazac, flazalone, fluazacort, flufenamic acid, flumizole, flunisolide acetate, flunixin, flunixin meglumine, fluocortin butyl, fluorometholone acetate, fluquazone, flurbiprofen, fluretofen, fluticasone propionate, furaprofen, furobufen, halcinonide, halobetasol propionate, halopredone acetate, ibufenac, ibuprofen, ibuprofen aluminum, ibuprofen piconol, ilonidap, indomethacin, indomethacin sodium, indoprofen, indoxole, intrazole, isoflupredone acetate, isoxepac, isoxicam, ketoprofen, lofemizole hydrochloride, lomoxicam, loteprednol etabonate, meclofenamate sodium, meclofenamic acid, meclorisone dibutyrate, mefenamic acid, mesalamine, meseclazone, methylprednisolone suleptanate, momiflumate, nabumetone, naproxen, naproxen sodium, naproxol, nimazone, olsalazine sodium, orgotein, orpanoxin, oxaprozin, oxyphenbutazone, paranyline hydrochloride, pentosan polysulfate sodium, phenbutazone sodium glycerate, pirfenidone, piroxicam, piroxicam cinnamate, piroxicam olamine, pirprofen, prednazate, prifelone, prodolic acid, proquazone, proxazole, proxazole citrate, rimexolone, romazarit, salcolex, salnacedin, salsalate, sanguinarium chloride, seclazone, sermetacin, sudoxicam, sulindac, suprofen, talmetacin, talniflumate, talosalate, tebufelone, tenidap, tenidap sodium, tenoxicam, tesicam, tesimide, tetrydamine, tiopinac, tixocortol pivalate, tolmetin, tolmetin sodium, triclonide, triflumidate, zidometacin, zomepirac sodium, aspirin (acetylsalicylic acid), salicylic acid, corticosteroids, glucocorticoids, tacrolimus, pimecorlimus, prodrugs thereof, co-drugs thereof, and combinations thereof.
  • In one embodiment, the anti-inflammatory agent is clobetasol. Clobetasol is a corticosteroid that binds to corticosteroid receptors, a class of nuclear receptor. The binding of clobetasol to the corticosteroid receptor subsequently alters gene expression in such a way that inflammation is inhibited. For example, corticosteroids inhibit the activation of NFkB, the nuclear factor that is responsible for changes in gene expression that promote inflammation. The reduction in inflammation may also inhibit the mechanisms that promote small muscle cell (SMC) hyper proliferation. This is shown in that dexamethasone, a less potent glucocorticoid as compared to clobetasol, reduces the production of PGDF and thus has anti-proliferative properties. Clobetasol acts through similar pathways and is more potent than dexamethasone.
  • Dosage
  • The dosage or concentration of the anti-proliferative and anti-inflammatory agents required to produce a favorable therapeutic effect should be less than the level at which the bioactive agent produces toxic effects and greater than the level at which non-therapeutic results are obtained. The dosage or concentration of the agents required can depend upon factors such as the particular circumstances of the patient, the nature of the trauma, the nature of the therapy desired, the time over which the ingredient administered resides at the vascular site, and if other active agents are employed, the nature and type of the substance or combination of substances. Therapeutic effective dosages can be determined empirically, for example by infusing vessels from suitable animal model systems and using immunohistochemical, fluorescent or electron microscopy methods to detect the agent and its effects, or by conducting suitable in vitro studies.
  • In one embodiment, the bioactive agents can be incorporated into polymeric coating in a percent loading of between about 0.01% and less than about 100% by weight, more preferably between about 5% and about 50% by weight of the total drug-load that includes greater than about 0% to about 100% of the anti-proliferative agent and less than about 100% to greater than about 0% of the anti-inflammatory agent. The relative amount of the anti-proliferative agent and anti-inflammatory agent can be determined by the type of lesions to be treated. For example, where everolimus is used as the anti-proliferative agent and clobetasol is used as the anti-inflammatory agent, the relative amount of everolimus and clobetasol can be varied for different types of lesions, that is, the relative amount of everolimus can be higher for more proliferative lesions, and on the other hand, the relative amount of clobetasol can be higher for more inflammatory lesions.
  • Other Bioactive Agents
  • In some embodiments, other agents can be used in combination with the anti-proliferative agent and the anti-inflammatory agents: These bioactive agents can be any agent which is a therapeutic, prophylactic, or diagnostic agent. These agents can also have anti-proliferative and/or anti-inflammmatory properties or can have other properties such as antineoplastic, antiplatelet, anti-coagulant, anti-fibrin, antithrombonic, antimitotic, antibiotic, antiallergic, antioxidant as well as cystostatic agents. Examples of suitable therapeutic and prophylactic agents include synthetic inorganic and organic compounds, proteins and peptides, polysaccharides and other sugars, lipids, and DNA and RNA nucleic acid sequences having therapeutic, prophylactic or diagnostic activities. Nucleic acid sequences include genes, antisense molecules which bind to complementary DNA to inhibit transcription, and ribozymes. Some other examples of other bioactive agents include antibodies, receptor ligands, enzymes, adhesion peptides, blood clotting factors, inhibitors or clot dissolving agents such as streptokinase and tissue plasminogen activator, antigens for immunization, hormones and growth factors, oligonucleotides such as antisense oligonucleotides and ribozymes and retroviral vectors for use in gene therapy. Examples of antineoplastics and/or antimitotics include methotrexate, azathioprine, vincristine, vinblastine, fluorouracil, doxorubicin hydrochloride (e.g. Adriamycin® from Pharmacia & Upjohn, Peapack N.J.), and mitomycin (e.g. Mutamycin® from Bristol-Myers Squibb Co., Stamford, Conn.). Examples of such antiplatelets, anticoagulants, antifibrin, and antithrombins include sodium heparin, low molecular weight heparins, heparinoids, hirudin, argatroban, forskolin, vapiprost, prostacyclin and prostacyclin analogues, dextran, D-phe-pro-arg-chloromethylketone (synthetic antithrombin), dipyridamole, glycoprotein IIb/IIIa platelet membrane receptor antagonist antibody, recombinant hirudin, thrombin inhibitors such as Angiomax a (Biogen, Inc., Cambridge, Mass.), calcium channel blockers (such as nifedipine), colchicine, fibroblast growth factor (FGF) antagonists, fish oil (omega 3-fatty acid), histamine antagonists, lovastatin (an inhibitor of HMG-CoA reductase, a cholesterol lowering drug, brand name Mevacor® from Merck & Co., Inc., Whitehouse Station, N.J.), monoclonal antibodies (such as those specific for Platelet-Derived Growth Factor (PDGF) receptors), nitroprusside, phosphodiesterase inhibitors, prostaglandin inhibitors, suramin, serotonin blockers, steroids, thioprotease inhibitors, triazolopyrimidine (a PDGF antagonist), nitric oxide or nitric oxide donors, super oxide dismutases, super oxide dismutase mimetic, 4-amino-2,2,6,6-tetramethylpiperidine-1-oxyl (4-amino-TEMPO), estradiol, anticancer agents, dietary supplements such as various vitamins, and a combination thereof. Examples of such cytostatic substance include angiopeptin, angiotensin converting enzyme inhibitors such as captopril (e.g. Capoten® and Capozide® from Bristol-Myers Squibb Co., Stamford, Conn.), cilazapril or lisinopril (e.g. Prinivil® and Prinzide® from Merck & Co., Inc., Whitehouse Station, N.J.). An example of an antiallergic agent is permirolast potassium. Other therapeutic substances or agents which may be appropriate include alpha-interferon, and genetically engineered epithelial cells. The foregoing substances are listed by way of example and are not meant to be limiting. Other active agents which are currently available or that may be developed in the future are equally applicable.
  • Delivery Formulations
  • The composition comprising both anti-proliferative agent and the anti-inflammatory agent can be formulated into any formulation suitable for delivery by any mode of delivery. For example, the composition can be formed into a coating on an implantable medical device to provide controlled release of the anti-proliferative agent and the anti-inflammatory agent. The composition can also be formulated into other suitable formulations for example, bolus dose of free drug, optionally with a fluoroscopic dye, bolus dose of gel-encapsulated drug.
  • The gel can be formed of a gel-forming material or polymer such as hyaluronic acid, carboxymethyl cellulose, pectin, hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, sodium carboxymethylcellulose, hydroxyethylcellulose, polyethylene oxide, acacia, tragacanth, guar gum, xanthan gum, locust bean gum, Carbopol™ acidic carboxy polymer, polycarbophil, polyethylene oxide, poly(hydroxyalkyl methacrylate), poly(electrolyte complexes), poly(vinyl acetate) cross-linked with hydrolyzable bonds, water-swellable N-vinyl lactams polysaccharides, natural gum, agar, agarose, sodium alginate, carrageenan, fucoidan, furcellaran, laminaran, hypnea, eucheuma, gum arabic, gum ghatti, gum karaya, arbinoglactan, amylopectin, gelatin, hydrophilic colloids such as carboxylmethyl cellulose gum or alginate gum, including both non-crosslinked and crosslinked alginate gums, where the crosslinked alginate gums may be crosslinked with di- or trivalent ions, polyols such as propylene glycol, or other crosslinking agents, Cyanamer™ polyacrylamides, Good-rite™ polyacrylic acid, starch graft copolymers, Aqua-Keeps™ acrylate polymer, ester crosslinked polyglucan, and the like, and combinations thereof. Some of the gel-forming materials are discussed in U.S. patents, U.S. Pat. Nos. 3,640,741, 3,865,108, 3,992,562, 4,002,173, 4,014,335, and 4,207,893. Hydrogels also are discussed in the Handbook of Common Polymers, by Scott and Roff, published by the Chemical Rubber Company, Cleveland, Ohio. For any given gel-forming material or polymer, use of a material with higher average molecular weight provides higher viscosity in aqueous solution of any given concentration. Therefore, using a higher molecular weight generally enables use of a lesser quantity of polymer to accomplish the required retardation of dissolution. In some embodiments, the gel-forming material or polymer can be hydropropyl methylcellulose having 19-24% methoxyl substitution and 7-12% hydroxypropyl substitution and a number average molecular weight of at least 20,000. Such polymers include those sold by Dow Chemical Co. under the tradenames Methocel K4M, Methocel K15M and Methocel K100M.
  • Modes of Delivery
  • In one embodiment, the anti-inflammatory drug such as clobetasol is formulated into a bolus dose of free drug with, optionally, a fluoroscopic dye. The anti-proliferative drug such as everolimus can be formulated into a coating composition with a polymeric material and then coated onto an implantable device (e.g., stent). The bolus dose of anti-inflammatory drug is administered first and then the anti-proliferative drug is delivered by release from the implantable device such as a drug-delivery stent. The composition may further include a third agent such as a HDL (high density lipoprotein)-mimic as described in U.S. Pat. No. 6,367,479. Alternatively, HDL-mimic can be delivered by the stent.
  • In another embodiment, the anti-inflammatory drug such as clobetasol is formulated into a bolus dose of gel. The anti-proliferative drug such as everolimus can be formulated into a coating composition with a polymeric material and then coated onto an implantable device. The bolus dose of the anti-inflammatory drug is administered first and then the anti-proliferative drug is delivered by release from the implantable device such as a drug-delivery stent.
  • In a further embodiment, the anti-inflammatory drug and the anti-proliferative drug can be included in a polymeric matrix and then coated onto a medical device such as a stent. The medical device coating can be designed to have a variety of different release parameters for each of the drugs included in the coating. For example, the anti-inflammatory can have one or a combination of release profiles that include a pulse release, fast or burst release, and a sustained release. Similarly, the anti-proliferative drug can have one or a combination of release profiles that include a pulse release, fast or burst release, and a sustained release from the stent. In some embodiments, the combination can be delivered simultaneously or at least during the drug treatment period there is at lease some overlap between the release of the drugs. In some embodiments, the anti-inflammatory can be completely released prior to the release to the anti-proliferative or can be partially released with some or significant overlap between the release of both drugs. “Pulse release” generally refers to a release profile of a drug that features a sudden surge of the release rate of the drug. The release rate surge of the drug would then disappear within a period. A more detailed definition of the term can be found in Encyclopedia of Controlled Drug Delivery, Edith Mathiowitz, Ed., Culinary and Hospitality Industry Publications Services.
  • As used herein, the term “fast release” in one embodiment refers to a release profile of a drug that features a release rate in the range between about 15 to about 40 μg per day for a 18 mm stent, about 10 μg to about 27 μg per day for a 13 mm stent, and about 6.7 μg to about 17.2 μg per day for a 8 mm stent. Equivalent profiles can be derived by one having ordinary skill in the art for stents having other sizes. In another embodiment, the term “fast release” refers to an approximately 20% release in 24 hours of a drug. The term “fast release” is used interchangeably with the term “burst release.”
  • As used herein, the term “sustained release” generally refers to a release profile of a drug that can include zero-order release, exponential decay, step-function release or other release profiles that carry over a period of time, for example, ranging from several days to several years. The terms “zero-order release”, “exponential decay” and “step-function release” as well as other sustained release profiles are well known in the art (see, for example, Encyclopedia of Controlled D rug Delivery, Edith Mathiowitz, Ed., Culinary and Hospitality Industry Publications Services).
  • In one embodiment, at least one of the anti-inflammatory agent (e.g., clobetasol) and anti-proliferative agent (e.g., everolimus) is administered via a stent while the other is administered by other local means of administration or alternatively, the other is administered systemically. In other embodiments, both are administered locally, by means other than a stent, or alternatively systemically. Systemic administration can be accomplished orally or parenterally including intravascularly, rectally, intranasally, intrabronchially, or transdermally. Liquid carriers which are sterile solutions or suspensions can be injected intramuscularly, intraperitoneally, subcutaneously, and intravenously. Rectal administration can be in the form of conventional suppository. For adminsitration by intranasal or intrabronchial inhalation or insufflation, the drug can be formulated into an aqueous or partially aqueous solution, which can then be utilized in the form of an aerosol. The drug can be administered transdermally through the used of a transdermal patch and a carrier that is inert to and mutually compatible with the active component, is non-toxic to the skin, and allows for the delivery of the drug for systemic absorption into the blood stream via the skin. The carrier may take any number of forms such as creams, ointments, pastes, and gels. The creams and ointments may be viscous liquids or semisolid emulsions of either the oil-in-water or water-in-oil type. Pastes made of absorptive powders dispersed in petroleum or hydrophilic petroleum containing the active component may also be suitable. Other devices capable of releasing the drug into the blood stream include semi-permeable membranes covering a reservoir containing the drug, with or without a carrier.
  • Local administration can be accomplished by a variety of techniques which administer the active component at or near the target site. The following examples of local delivery techniques are provided for illustrative purposes and are not intended to be limiting. Examples include local delivery catheters, site specific carriers, implants, direct application, or direct injection. Local delivery by a catheter allows for the administration of the drug directly to the target site.
  • Local delivery by site specific carriers is conducted by attaching the drug to a carrier which will direct or link the drug to the target cells. Examples of this delivery technique include the use of carrier such as a protein ligand, a monoclonal antibody or a membrane anchored linker.
  • Local delivery by an implant (other than a stent) is the placement of a matrix carrying the drug at the site. The matrix can release the active component by, for example, diffusion, degradation, chemical reaction, solvent activators, etc. One example of local delivery by an implant can include direct injection of vesicles or micro-particles. These micro-particles may be composed of substances such as proteins, lipids, carbohydrates or synthetic polymers. The micro-particles can have the drug impregnated therein and/or coated thereon. Application via implants is not limited to the above described routes and other techniques such as grafts, micropumps or application of a fibrin glue or hydrogel containing the active component around the exterior of a designated region of the vessel can also be implemented by one of ordinary skill in the art.
  • Local delivery by direct injection describes injecting a liquid carrier containing the drug directly into the site. The liquid carrier should be inert to and mutually compatible with the drug. The component can be in true solution or suspended in fine particles in the carrier. A suitable example of an inert carrier includes a sterile saline solution.
  • Biocompatible Polymers
  • Any biocompatible polymers can be used to form a coating on a stent or to provide a drug delivery particle with the anti-proliferative drug and/or anti-inflammatory drug. Such biocompatible, bioabsorbable polymers include, but not limited to, poly(ester amide), poly(ester amide) that may contain alkyl groups, amino acid groups, or poly(ethylene glycol) (PEG) groups, polyethylene glycol (PEG), polylakanoates (PHA), poly(2-hydroxyalkanoates), poly(3-hydroxyalkanoates) such as poly(3-hydroxypropanoate), poly(3-hydroxybutyrate), poly(3-hydroxyvalerate), poly(3-hydroxyhexanoate), poly(3-hydroxyheptanoate) and poly(3-hydroxyoctanoate), poly(4-hydroxyalknaote) such as poly(4-hydroxybutyrate), poly(4-hydroxyvalerate), poly(4-hydroxyhexanote), poly(4-hydroxyheptanoate), poly(4-hydroxyoctanoate) and copolymers comprising any of the 3-hydroxyalkanoate or 4-hydroxyalkanoate monomers described herein or blends thereof, polyesters, poly(D,L-lactide), poly(L-lactide), polyglycolide, poly(D,L-lactide-co-glycolide), polycaprolactone, poly(D,L-lactide-co-caprolactone), poly(glycolide-co-caprolactone), poly(dioxanone), poly(ortho esters), poly(anhydrides), poly(tyrosine carbonates) and derivatives thereof, poly(tyrosine ester) and derivatives thereof, poly(imino carbonates), poly(phosphoesters), polyphosphazenes, poly(amino acids), polysaccharides, collagen, chitosan, alginate, polyethers, polyamides, polyurethanes, polyalkylenes, polyalkylene oxides, polyethylene oxide, polypropylene oxide, polyethylene glycol (PEG), PHA-PEG, polyvinylpyrrolidone (PVP), alkylene vinyl acetate copolymers such as ethylene vinyl acetate (EVA), alkylene vinyl alcohol copolymers such as ethylene vinyl alcohol (EVOH or EVAL), poly(n-butyl methacrylate) (PBMA), SOLEF™ polymers such as poly(vinylidene fluoride-co-hexafluoropropene) (PVDF-co-HFP) and poly(vinylidene fluoride) (PVDF) and combinations thereof.
  • Method of Coating A Device
  • The coating described herein can be formed by spray coating or any other coating process available in the art. Generally, the coating involves dissolving or suspending the composition, or one or more components thereof, in a solvent or solvent mixture to form a solution, suspension, or dispersion of the composition or one or more components thereof, applying the solution or suspension to an implantable device, and removing the solvent or solvent mixture to form a coating or a layer of coating. Suspensions or dispersions of the composition described herein can be in the form of latex or emulsion of microparticles having a size between 1 nanometer and 100 microns, preferably between 1 nanometer and 10 microns. Heat and/or pressure treatment can be applied to any of the steps involved herein. In addition, if desirable, the coating described here can be subjected to further heat and/or pressure treatment. Some additional exemplary processes of coating an implantable device that may be used are described in, for example, Lambert T L, et al. Circulation, 1994, 90: 1003-1011; Hwang C W, et al. Circulation, 2001; 104: 600-605; Van der Giessen W J, et al. Circulation, 1996; 94: 1690-1697; Lincoff A M, et al. J Am Coll Cardiol 1997; 29: 808-816; Grube E. et al, J American College Cardiology Meeting, Mar. 6, 2002, ACCIS2002, poster 1174-15; Grube E, et al, Circulation, 2003, 107: 1, 38-42; Bullesfeld L, et al. Z Kardiol, 2003, 92: 10, 825-832; and Tanabe K, et al. Circulation 2003, 107: 4, 559-64.
  • As used herein, the term “solvent” refers to a liquid substance or composition that is compatible with the polymer and is capable of dissolving or suspending the polymeric composition or one or more components thereof at a desired concentration. Representative examples of solvents include chloroform, acetone, water (buffered saline), dimethylsulfoxide (DMSO), propylene glycol monomethyl ether (PM,) iso-propylalcohol (IPA), n-propyl alcohol, methanol, ethanol, tetrahydrofuran (THF), dimethylformamide (DMF), dimethyl acetamide (DMAC), benzene, toluene, xylene, hexane, cyclohexane, heptane, octane, nonane, decane, decalin, ethyl acetate, butyl acetate, isobutyl acetate, isopropyl acetate, butanol, diacetone alcohol, benzyl alcohol, 2-butanone, cyclohexanone, dioxane, methylene chloride, carbon tetrachloride, tetrachloroethylene, tetrachloro ethane, chlorobenzene, 1,1,1-trichloroethane, 1,1,2-trichloroethane, formamide, hexafluoroisopropanol, 1,1,1-trifluoroethanol, and hexamethyl phosphoramide and a combination thereof.
  • Examples of such implantable devices include self-expandable stents, balloon-expandable stents, stent-grafts, grafts (e.g., aortic grafts), artificial heart valves, cerebrospinal fluid shunts, pacemaker electrodes, and endocardial leads (e.g., FINELINE and ENDOTAK, available from Guidant Corporation, Santa Clara, Calif.). The underlying structure of the device can be of virtually any design. The device can be made of a metallic material or an alloy such as, but not limited to, cobalt chromium alloy (ELGILOY), stainless steel (316L), high nitrogen stainless steel, e.g., BIODUR 108, cobalt chrome alloy L-605, “MP35N,” “MP20N,” ELASTINITE (Nitinol), tantalum, nickel-titanium alloy, platinum-iridium alloy, gold, magnesium, or combinations thereof. “MP35N” and “MP20N” are trade names for alloys of cobalt, nickel, chromium and molybdenum available from Standard Press Steel Co., Jenkintown, Pa. “MP35N” consists of 35% cobalt, 35% nickel, 20% chromium, and 10% molybdenum. “MP20N” consists of 50% cobalt, 20% nickel, 20% chromium, and 10% molybdenum. Devices made from bioabsorbable or biostable polymers could also be used with the embodiments of the present invention. In one embodiment, the implantable device is a stent, which can be degradable stents, biodurable stents, depot stents, and metallic stens such as stents made of stainless steel or nitinol. The stents can be balloon expandable or self expanding.
  • Method of Use
  • In accordance with embodiments of the invention, a coating of the various described embodiments can be formed on an implantable device or prosthesis, e.g., a stent. For coatings including one or more active agents, the agent will be retained on the medical device such as a stent during delivery and expansion of the device, and released at a desired rate and for a predetermined duration of time at the site of implantation. Preferably, the medical device is a stent. A stent having the above-described coating is useful for a variety of medical procedures, including, by way of example, treatment of obstructions caused by tumors in bile ducts, esophagus, trachea/bronchi and other biological passageways. A stent having the above-described coating is particularly useful for treating occluded regions of blood vessels caused by abnormal or inappropriate migration and proliferation of smooth muscle cells, thrombosis, and restenosis. Stents may be placed in a wide array of blood vessels, both arteries and veins. Representative examples of sites include the iliac, renal, and coronary arteries.
  • For implantation of a stent, an angiogram is first performed to determine the appropriate positioning for stent therapy. An angiogram is typically accomplished by injecting a radiopaque contrasting agent through a catheter inserted into an artery or vein as an x-ray is taken. A guidewire is then advanced through the lesion or proposed site of treatment. Over the guidewire is passed a delivery catheter which allows a stent in its collapsed configuration to be inserted into the passageway. The delivery catheter is inserted either percutaneously or by surgery into the femoral artery, brachial artery, femoral vein, or brachial vein, and advanced into the appropriate blood vessel by steering the catheter through the vascular system under fluoroscopic guidance. A stent having the above-described coating may then be expanded at the desired area of treatment. A post-insertion angiogram may also be utilized to confirm appropriate positioning.
  • The implantable device comprising a coating described herein can be used to treat an animal having a condition or disorder that requires a treatment. Such an animal can be treated by, for example, implanting a device described herein in the animal. Preferably, the animal is a human being. Exemplary disorders or conditions that can be treated by the method disclosed herein include, but not limited to, thrombosis, high cholesterol, hemorrhage, vascular dissection or perforation, vascular aneurysm, vulnerable plaque, chronic total occlusion, claudication, anastomotic proliferation for vein and artificial grafts, bile duct obstruction, ureter obstruction, tumor obstruction, restenosis and progression of atherosclerosis in patient subsets including type I diabetics, type II diabetics, metabolic syndrome and syndrome X, vulnerable lesions including those with thin-capped fibroatheromatous lesions, systemic infections including gingivitis, hellobacteria, and cytomegalovirus, and combinations thereof.
  • EXAMPLES
  • The embodiments of the present invention will be illustrated by the following set forth examples. All parameters and data are not to be construed to unduly limit the scope of the embodiments of the invention.
  • Example 1 Porcine Implant Study
  • Described in this example is a 28 day porcine implant study that compared the 200 μg/cm2 dose Lemans with a clobetasol-only delivery stent, an everolimus-only stent, and an everolimus-clobetasol combination drug delivery stent. The study was performed using three different drug delivery stents, Arm 1, Arm 2, and Arm 3. Arm 1 is Lemans stent (a stent available from Guidant based on PVDF-co-HFP) that included 105 μg everolimus and used as a control. Arm 2 was loaded with 185 μg clobetasol only, with no everolimus. Arm 3 is loaded with 105 μg everolimus and 80 μg clobetasol.
  • The Arm 1, Arm 2, and Arm 3 stents were implanted in a 30% overstretch model. Nine samples of each Arm stent were implanted, one for each coronary artery. 24 hr Release in porcine serum data were gathered. 3, 7 and 28 day in vivo release data were gathered (from the mammary arteries), as was 28 day quantitative coronary angioplasty (QCA), histology and morphometry.
  • In this study, 12 mm Vision Small stents (available from Guidant) were used. All drug solutions were sprayed in a 2% Solef™ in acetone/cyclohexanone formulation. All stents had a 100 μg PBMA primer. Table 1 shows the coating design of the stents used in this study.
    TABLE 1
    Coating design
    Polymer Evererolimus Clobetasol Solid Target
    Drug (D) (P) D:P Drug % Target (μg) Target (μg) (μg)
    Arm 1 Everolimus Solef ™ 1:3   25.0 105 420
    Arm 2 Clobetasol Solef ™ 1:4.2  19.2 185 962
    Arm 3 Ever & Clob Solef ™ 1:3.49 22.2 105  80 833
  • The release rate data are shown in Table 2. As can be seen from Table 2, a coating based on Solef™ is capable of simultaneous release of both everolimus and clobetasol.
    TABLE 2
    Release rate data
    In vivo In vivo In vivo In vitro In vivo In vivo In vivo In vitro
    Day 3 Day 7 Day 28 24 hr Day 3 Day 7 Day 28 24 hr
    % % % % % % % %
    Clobetasol Clobetasol Clobetasol Clobetasol Everolimus Everolimus Everolimus Everolimus
    Release Release Release Release in Release Release Release Release in
    Arm (n = 2) (n = 3) (n = 4) PS (n = 3) (n = 2) (n = 3) (n = 4) PS (n = 3)
    1 - Everolimus 37.6% 49.3% 66.7% 30.0%
    2 - Clobetasol 32.5% 43.1% 60.6% 26.7%
    3 - Everolimus +
      Clobetasol 40.9% 50.2% 71.9% 30.1% 35.1% 43.6% 60.4% 24.8%
  • The results of 28 day QCA are shown in FIG. 1, the 28 day histology data are in FIG. 2, and the 28 day morphometry data are shown in FIG. 3 and summarized in Table 3 below.
    TABLE 3
    28 Day morphometry data from FIG. 3
    AVERAGE STANDARD DEVIATION
    Media Neointimal Media Neointimal Neointimal
    Area Neointimal % Thickness Injury Area Area % Thickness Injury
    (mm{circumflex over ( )}2) Area (mm{circumflex over ( )}2) Stenosis (mm) Score (mm{circumflex over ( )}2) (mm{circumflex over ( )}2) Stenosis (mm) Score
    Everolimus 1.21 1.81 27.83 0.28 1.83 Everolimus 0.23 0.72 13.18 0.11 0.23
    Clobetasol 1.09 1.73 24.86 0.29 1.79 Clobetasol 0.18 1.57 23.27 0.23 0.22
    Clobetasol/ 0.97 0.82 12.39 0.14 1.62 Clobetasol/ 0.18 0.39  7.54 0.04 0.29
    Everolimus Everolimus
  • The p values from a t-test of the data from FIG. 3 are summarized in Table 4. A “t-test” returns the probability associated with a Student's t-Test that determines whether two samples are likely to have come from the same two underlying populations that have the same mean. The value returned from the test, “p”, is the probability that the two groups of data come from the same population. p Values less than or equal to 0.10 or 0.05 are generally considered significant (Zar, J H. Biostatistical Analysis. Englewood Cliffs, N.J.: Prentice-Hall Inc, 1974. pp 101-108).
    TABLE 4
    p Values from a t-test of the data from FIG. 3
    Neointi- Neointi-
    Media mal % mal Injury
    Arm Comparison Area Area Stenosis Thickness Score
    EVER COMBO 0.05 0.01 0.02 0.01 0.18
    EVER CLOB 0.29 0.90 0.77 0.93 0.78
    COMBO CLOB 0.24 0.18 0.22 0.14 0.25
  • While particular embodiments of the present invention have been shown and described, it will be obvious to those skilled in the art that changes and modifications can be made without departing from this invention in its broader aspects. Therefore, the appended claims are to encompass within their scope all such changes and modifications as fall within the true spirit and scope of this invention.

Claims (21)

1. A drug-delivery system, comprising an effective amount of everolimus, or derivatives thereof, and an effective amount of clobetasol for the treatment or prevention of a vascular disorder or a related disorder.
2. The drug-delivery system of claim 1, wherein the system is a stent.
3. The drug-delivery system of claim 1, wherein the disorder is restenosis.
4. The drug-delivery system of claim 1, wherein the disorder is restenosis and/or progression of atherosclerosis in patient subsets including type I diabetics and type II diabetics.
5. The drug-delivery system of claim 1, wherein the disorder is vulnerable plaque.
6. The drug-delivery system of claim 1, wherein the system is a polymer or a polymeric coating.
7. A drug-delivery system, comprising
an effective amount of everolimus, rapamycin, or derivatives of everolimus or rapamycin, and
an effective amount of a steroidal anti-inflammatory agent or a non steroidal anti-inflammatory agent, wherein the combination is for treatment or prevention of a vascular disorder.
8. The drug-delivery system of claim 7, wherein the anti-inflammatory agent is clobetasol.
9. The drug-delivery system of claim 7, wherein the system is a stent.
10. The drug-delivery system of claim 7, wherein the disorder is restenosis.
11. The drug-delivery system of claim 7, wherein the disorder is restenosis and/or progression of atherosclerosis in patient subsets including type I diabetics and type II diabetics.
12. The drug-delivery system of claim 7, wherein the disorder is vulnerable plaque.
13. The drug-delivery system of claim 7, wherein the system is a polymer or a polymeric coating.
14. A method of treating restenosis of a blood vessel comprising administering to a patient an effective amount of everolimus, rapamycin, or derivatives of everolimus or rapamycin and an effective amount of a steroidal anti-inflammatory agent or a non steroidal anti-inflammatory agent, wherein the combination is for treatment or prevention of the vascular disorder.
15. The method of claim 14, wherein the anti-inflammatory is clobetasol.
16. The method of claim 14, wherein everolimus, rapamycin, or derivatives of everolimus or rapamycin is delivered via a stent and the anti-inflammatory is delivered by other local means or by systemic means.
17. The method of claim 14, wherein the combination of the drugs are administered by a drug-delivery stent.
18. A method of treating vulnerable plaque of a blood vessel comprising
administering to a patient an effective amount of everolimus, rapamycin, or derivatives of everolimus or rapamycin and an effective amount of a steroidal anti-inflammatory agent or a non steroidal anti-inflammatory agent, wherein the combination is for treatment or prevention of the vascular disorder.
19. The method of claim 18, wherein the anti-inflammatory is clobetasol.
20. The method of claim 18, wherein everolimus, rapamycin, or derivatives of everolimus or rapamycin is delivered via a stent and the anti-inflammatory is delivered by local means other than a stent or by systemic means.
21. The method of claim 18, wherein the combination of the drugs are administered by a drug-delivery stent.
US10/882,506 2002-12-16 2004-06-30 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders Abandoned US20060002968A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US10/882,506 US20060002968A1 (en) 2004-06-30 2004-06-30 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US11/090,507 US7758881B2 (en) 2004-06-30 2005-03-24 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
JP2007519366A JP5791221B2 (en) 2004-06-30 2005-06-28 Combination of antiproliferative and anti-inflammatory drugs for the treatment of vascular diseases
PCT/US2005/022968 WO2006014270A2 (en) 2004-06-30 2005-06-28 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
ES05803287.1T ES2529043T3 (en) 2004-06-30 2005-06-28 Combination of an antiproliferative and anti-inflammatory agent for the treatment of vascular disorders
EP05803287.1A EP1768718B1 (en) 2004-06-30 2005-06-28 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US11/322,282 US8586069B2 (en) 2002-12-16 2005-12-29 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US12/191,209 US8435550B2 (en) 2002-12-16 2008-08-13 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US12/838,329 US8709469B2 (en) 2004-06-30 2010-07-16 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US14/192,705 US9138337B2 (en) 2004-06-30 2014-02-27 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US14/829,558 US9566373B2 (en) 2004-06-30 2015-08-18 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/882,506 US20060002968A1 (en) 2004-06-30 2004-06-30 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/089,763 Continuation-In-Part US20050238686A1 (en) 1999-12-23 2005-03-25 Coating for implantable devices and a method of forming the same

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US11/090,507 Continuation-In-Part US7758881B2 (en) 2002-12-16 2005-03-24 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US11/090,507 Continuation US7758881B2 (en) 2002-12-16 2005-03-24 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US11/322,282 Continuation US8586069B2 (en) 2002-12-16 2005-12-29 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders

Publications (1)

Publication Number Publication Date
US20060002968A1 true US20060002968A1 (en) 2006-01-05

Family

ID=35514191

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/882,506 Abandoned US20060002968A1 (en) 2002-12-16 2004-06-30 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US11/322,282 Expired - Fee Related US8586069B2 (en) 2002-12-16 2005-12-29 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/322,282 Expired - Fee Related US8586069B2 (en) 2002-12-16 2005-12-29 Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders

Country Status (1)

Country Link
US (2) US20060002968A1 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050208093A1 (en) * 2004-03-22 2005-09-22 Thierry Glauser Phosphoryl choline coating compositions
US20060069427A1 (en) * 2004-09-24 2006-03-30 Savage Douglas R Drug-delivery endovascular stent and method for treating restenosis
US20070299511A1 (en) * 2006-06-27 2007-12-27 Gale David C Thin stent coating
US20080299164A1 (en) * 2007-05-30 2008-12-04 Trollsas Mikael O Substituted polycaprolactone for coating
US20090110711A1 (en) * 2007-10-31 2009-04-30 Trollsas Mikael O Implantable device having a slow dissolving polymer
US20090259302A1 (en) * 2008-04-11 2009-10-15 Mikael Trollsas Coating comprising poly (ethylene glycol)-poly (lactide-glycolide-caprolactone) interpenetrating network
US20090263457A1 (en) * 2008-04-18 2009-10-22 Trollsas Mikael O Block copolymer comprising at least one polyester block and a poly(ethylene glycol) block
US20090285873A1 (en) * 2008-04-18 2009-11-19 Abbott Cardiovascular Systems Inc. Implantable medical devices and coatings therefor comprising block copolymers of poly(ethylene glycol) and a poly(lactide-glycolide)
US20090297578A1 (en) * 2008-06-03 2009-12-03 Trollsas Mikael O Biosoluble coating comprising anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US20090297584A1 (en) * 2008-04-18 2009-12-03 Florencia Lim Biosoluble coating with linear over time mass loss
US20090306120A1 (en) * 2007-10-23 2009-12-10 Florencia Lim Terpolymers containing lactide and glycolide
US20100063585A1 (en) * 2006-07-03 2010-03-11 Hemoteq Ag Manufacture, method and use of active substance-releasing medical products for permanently keeping blood vessels open
US20100168842A1 (en) * 2007-02-14 2010-07-01 Junbo Ge Coronary stent with asymmetric drug releasing controlled coating
US20100179475A1 (en) * 2007-01-21 2010-07-15 Erika Hoffmann Medical product for treating stenosis of body passages and for preventing threatening restenosis
US20100209476A1 (en) * 2008-05-21 2010-08-19 Abbott Cardiovascular Systems Inc. Coating comprising a terpolymer comprising caprolactone and glycolide
US20100272773A1 (en) * 2009-04-24 2010-10-28 Boston Scientific Scimed, Inc. Use of Drug Polymorphs to Achieve Controlled Drug Delivery From a Coated Medical Device
US20100322992A1 (en) * 2004-06-30 2010-12-23 Stephen Dugan Anti-Proliferative And Anti-Inflammatory Agent Combination For Treatment Of Vascular Disorders With An Implantable Medical Device
US20110008260A1 (en) * 2009-07-10 2011-01-13 Boston Scientific Scimed, Inc. Use of Nanocrystals for Drug Delivery from a Balloon
US20110015664A1 (en) * 2009-07-17 2011-01-20 Boston Scientific Scimed, Inc. Nucleation of Drug Delivery Balloons to Provide Improved Crystal Size and Density
US20110160698A1 (en) * 2007-07-03 2011-06-30 Hemoteq Ag Balloon Catheter for Treating Stenosis of Body Passages and for Preventing Threatening Restenosis
US20110301697A1 (en) * 2009-04-10 2011-12-08 Hemoteq Ag Manufacture, method and use of drug-eluting medical devices for permanently keeping blood vessels open
US8669360B2 (en) 2011-08-05 2014-03-11 Boston Scientific Scimed, Inc. Methods of converting amorphous drug substance into crystalline form
US8685430B1 (en) 2006-07-14 2014-04-01 Abbott Cardiovascular Systems Inc. Tailored aliphatic polyesters for stent coatings
US8697110B2 (en) 2009-05-14 2014-04-15 Abbott Cardiovascular Systems Inc. Polymers comprising amorphous terpolymers and semicrystalline blocks
US8889211B2 (en) 2010-09-02 2014-11-18 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
US9056152B2 (en) 2011-08-25 2015-06-16 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
US9090745B2 (en) 2007-06-29 2015-07-28 Abbott Cardiovascular Systems Inc. Biodegradable triblock copolymers for implantable devices
US9220759B2 (en) 2012-02-23 2015-12-29 Abbott Cardiovascular Systems Inc. Treatment of diabetic patients with a drug eluting stent and adjunctive therapy
US9539332B2 (en) 2004-08-05 2017-01-10 Abbott Cardiovascular Systems Inc. Plasticizers for coating compositions
US9737638B2 (en) 2007-06-20 2017-08-22 Abbott Cardiovascular Systems, Inc. Polyester amide copolymers having free carboxylic acid pendant groups
US9750627B2 (en) 2012-03-30 2017-09-05 Abbott Cardiovascular Systems Inc. Treatment of diabetic patients with a stent and locally administered adjunctive therapy
US9814553B1 (en) 2007-10-10 2017-11-14 Abbott Cardiovascular Systems Inc. Bioabsorbable semi-crystalline polymer for controlling release of drug from a coating

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6790228B2 (en) * 1999-12-23 2004-09-14 Advanced Cardiovascular Systems, Inc. Coating for implantable devices and a method of forming the same
US7807211B2 (en) * 1999-09-03 2010-10-05 Advanced Cardiovascular Systems, Inc. Thermal treatment of an implantable medical device
US20060002968A1 (en) 2004-06-30 2006-01-05 Gordon Stewart Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US8435550B2 (en) 2002-12-16 2013-05-07 Abbot Cardiovascular Systems Inc. Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US7758881B2 (en) 2004-06-30 2010-07-20 Advanced Cardiovascular Systems, Inc. Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US8016879B2 (en) 2006-08-01 2011-09-13 Abbott Cardiovascular Systems Inc. Drug delivery after biodegradation of the stent scaffolding
US20090104240A1 (en) * 2007-10-19 2009-04-23 Abbott Cardiovascular Systems Inc. Dual Drug Formulations For Implantable Medical Devices For Treatment of Vascular Diseases
WO2009057174A1 (en) * 2007-11-02 2009-05-07 Nobil Bio Ricerche S.R.L. Intracorporeal medical device
US9839644B2 (en) 2014-09-09 2017-12-12 ARKAY Therapeutics, LLC Formulations and methods for treatment of metabolic syndrome

Citations (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2072303A (en) * 1932-10-18 1937-03-02 Chemische Forschungs Gmbh Artificial threads, bands, tubes, and the like for surgical and other purposes
US4656242A (en) * 1985-06-07 1987-04-07 Henkel Corporation Poly(ester-amide) compositions
US4733665A (en) * 1985-11-07 1988-03-29 Expandable Grafts Partnership Expandable intraluminal graft, and method and apparatus for implanting an expandable intraluminal graft
US4800882A (en) * 1987-03-13 1989-01-31 Cook Incorporated Endovascular stent and delivery system
US5100992A (en) * 1989-05-04 1992-03-31 Biomedical Polymers International, Ltd. Polyurethane-based polymeric materials and biomedical articles and pharmaceutical compositions utilizing the same
US5292516A (en) * 1990-05-01 1994-03-08 Mediventures, Inc. Body cavity drug delivery with thermoreversible gels containing polyoxyalkylene copolymers
US5298260A (en) * 1990-05-01 1994-03-29 Mediventures, Inc. Topical drug delivery with polyoxyalkylene polymer thermoreversible gels adjustable for pH and osmolality
US5300295A (en) * 1990-05-01 1994-04-05 Mediventures, Inc. Ophthalmic drug delivery with thermoreversible polyoxyalkylene gels adjustable for pH
US5306786A (en) * 1990-12-21 1994-04-26 U C B S.A. Carboxyl group-terminated polyesteramides
US5306501A (en) * 1990-05-01 1994-04-26 Mediventures, Inc. Drug delivery by injection with thermoreversible gels containing polyoxyalkylene copolymers
US5380299A (en) * 1993-08-30 1995-01-10 Med Institute, Inc. Thrombolytic treated intravascular medical device
US5485496A (en) * 1994-09-22 1996-01-16 Cornell Research Foundation, Inc. Gamma irradiation sterilizing of biomaterial medical devices or products, with improved degradation and mechanical properties
US5605696A (en) * 1995-03-30 1997-02-25 Advanced Cardiovascular Systems, Inc. Drug loaded polymeric material and method of manufacture
US5607467A (en) * 1990-09-14 1997-03-04 Froix; Michael Expandable polymeric stent with memory and delivery apparatus and method
US5609629A (en) * 1995-06-07 1997-03-11 Med Institute, Inc. Coated implantable medical device
US5610241A (en) * 1996-05-07 1997-03-11 Cornell Research Foundation, Inc. Reactive graft polymer with biodegradable polymer backbone and method for preparing reactive biodegradable polymers
US5616338A (en) * 1988-02-11 1997-04-01 Trustees Of Columbia University In The City Of New York Infection-resistant compositions, medical devices and surfaces and methods for preparing and using same
US5624411A (en) * 1993-04-26 1997-04-29 Medtronic, Inc. Intravascular stent and method
US5711958A (en) * 1996-07-11 1998-01-27 Life Medical Sciences, Inc. Methods for reducing or eliminating post-surgical adhesion formation
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US5721131A (en) * 1987-03-06 1998-02-24 United States Of America As Represented By The Secretary Of The Navy Surface modification of polymers with self-assembled monolayers that promote adhesion, outgrowth and differentiation of biological cells
US5723219A (en) * 1995-12-19 1998-03-03 Talison Research Plasma deposited film networks
US5735897A (en) * 1993-10-19 1998-04-07 Scimed Life Systems, Inc. Intravascular stent pump
US5858746A (en) * 1992-04-20 1999-01-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5865814A (en) * 1995-06-07 1999-02-02 Medtronic, Inc. Blood contacting medical device and method
US5869127A (en) * 1995-02-22 1999-02-09 Boston Scientific Corporation Method of providing a substrate with a bio-active/biocompatible coating
US5873904A (en) * 1995-06-07 1999-02-23 Cook Incorporated Silver implantable medical device
US5876433A (en) * 1996-05-29 1999-03-02 Ethicon, Inc. Stent and method of varying amounts of heparin coated thereon to control treatment
US5877224A (en) * 1995-07-28 1999-03-02 Rutgers, The State University Of New Jersey Polymeric drug formulations
US5879713A (en) * 1994-10-12 1999-03-09 Focal, Inc. Targeted delivery via biodegradable polymers
US6011125A (en) * 1998-09-25 2000-01-04 General Electric Company Amide modified polyesters
US6010530A (en) * 1995-06-07 2000-01-04 Boston Scientific Technology, Inc. Self-expanding endoluminal prosthesis
US6015541A (en) * 1997-11-03 2000-01-18 Micro Therapeutics, Inc. Radioactive embolizing compositions
US6034204A (en) * 1997-08-08 2000-03-07 Basf Aktiengesellschaft Condensation products of basic amino acids with copolymerizable compounds and a process for their production
US6033582A (en) * 1996-01-22 2000-03-07 Etex Corporation Surface modification of medical implants
US6042875A (en) * 1997-04-30 2000-03-28 Schneider (Usa) Inc. Drug-releasing coatings for medical devices
US6051576A (en) * 1994-01-28 2000-04-18 University Of Kentucky Research Foundation Means to achieve sustained release of synergistic drugs by conjugation
US6051648A (en) * 1995-12-18 2000-04-18 Cohesion Technologies, Inc. Crosslinked polymer compositions and methods for their use
US6054553A (en) * 1996-01-29 2000-04-25 Bayer Ag Process for the preparation of polymers having recurring agents
US6172167B1 (en) * 1996-06-28 2001-01-09 Universiteit Twente Copoly(ester-amides) and copoly(ester-urethanes)
US6177523B1 (en) * 1999-07-14 2001-01-23 Cardiotech International, Inc. Functionalized polyurethanes
US6180632B1 (en) * 1997-05-28 2001-01-30 Aventis Pharmaceuticals Products Inc. Quinoline and quinoxaline compounds which inhibit platelet-derived growth factor and/or p56lck tyrosine kinases
US6203551B1 (en) * 1999-10-04 2001-03-20 Advanced Cardiovascular Systems, Inc. Chamber for applying therapeutic substances to an implant device
US6211249B1 (en) * 1997-07-11 2001-04-03 Life Medical Sciences, Inc. Polyester polyether block copolymers
US6214901B1 (en) * 1998-04-27 2001-04-10 Surmodics, Inc. Bioactive agent release coating
US6335029B1 (en) * 1998-08-28 2002-01-01 Scimed Life Systems, Inc. Polymeric coatings for controlled delivery of active agents
US20020007215A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020005206A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Antiproliferative drug and delivery device
US20020007214A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020007213A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020009604A1 (en) * 1999-12-22 2002-01-24 Zamora Paul O. Plasma-deposited coatings, devices and methods
US20020016625A1 (en) * 2000-05-12 2002-02-07 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020032414A1 (en) * 1998-08-20 2002-03-14 Ragheb Anthony O. Coated implantable medical device
US6358556B1 (en) * 1995-04-19 2002-03-19 Boston Scientific Corporation Drug release stent coating
US6379381B1 (en) * 1999-09-03 2002-04-30 Advanced Cardiovascular Systems, Inc. Porous prosthesis and a method of depositing substances into the pores
US20030004141A1 (en) * 2001-03-08 2003-01-02 Brown David L. Medical devices, compositions and methods for treating vulnerable plaque
US6503954B1 (en) * 2000-03-31 2003-01-07 Advanced Cardiovascular Systems, Inc. Biocompatible carrier containing actinomycin D and a method of forming the same
US6503556B2 (en) * 2000-12-28 2003-01-07 Advanced Cardiovascular Systems, Inc. Methods of forming a coating for a prosthesis
US6503538B1 (en) * 2000-08-30 2003-01-07 Cornell Research Foundation, Inc. Elastomeric functional biodegradable copolyester amides and copolyester urethanes
US20030028244A1 (en) * 1995-06-07 2003-02-06 Cook Incorporated Coated implantable medical device
US20030028243A1 (en) * 1995-06-07 2003-02-06 Cook Incorporated Coated implantable medical device
US20030032767A1 (en) * 2001-02-05 2003-02-13 Yasuhiro Tada High-strength polyester-amide fiber and process for producing the same
US20030036794A1 (en) * 1995-06-07 2003-02-20 Cook Incorporated Coated implantable medical device
US6524347B1 (en) * 1997-05-28 2003-02-25 Avantis Pharmaceuticals Inc. Quinoline and quinoxaline compounds which inhibit platelet-derived growth factor and/or p56lck tyrosine kinases
US20030040712A1 (en) * 1999-07-13 2003-02-27 Pinaki Ray Substance delivery apparatus and a method of delivering a therapeutic substance to an anatomical passageway
US20030040790A1 (en) * 1998-04-15 2003-02-27 Furst Joseph G. Stent coating
US20030039689A1 (en) * 2001-04-26 2003-02-27 Jianbing Chen Polymer-based, sustained release drug delivery system
US6527863B1 (en) * 2001-06-29 2003-03-04 Advanced Cardiovascular Systems, Inc. Support device for a stent and a method of using the same to coat a stent
US6530950B1 (en) * 1999-01-12 2003-03-11 Quanam Medical Corporation Intraluminal stent having coaxial polymer member
US6530951B1 (en) * 1996-10-24 2003-03-11 Cook Incorporated Silver implantable medical device
US20030059520A1 (en) * 2001-09-27 2003-03-27 Yung-Ming Chen Apparatus for regulating temperature of a composition and a method of coating implantable devices
US20030060877A1 (en) * 2001-09-25 2003-03-27 Robert Falotico Coated medical devices for the treatment of vascular disease
US6540776B2 (en) * 2000-12-28 2003-04-01 Advanced Cardiovascular Systems, Inc. Sheath for a prosthesis and methods of forming the same
US20030065377A1 (en) * 2001-09-28 2003-04-03 Davila Luis A. Coated medical devices
US6544223B1 (en) * 2001-01-05 2003-04-08 Advanced Cardiovascular Systems, Inc. Balloon catheter for delivering therapeutic agents
US6544543B1 (en) * 2000-12-27 2003-04-08 Advanced Cardiovascular Systems, Inc. Periodic constriction of vessels to treat ischemic tissue
US6544582B1 (en) * 2001-01-05 2003-04-08 Advanced Cardiovascular Systems, Inc. Method and apparatus for coating an implantable device
US20030073961A1 (en) * 2001-09-28 2003-04-17 Happ Dorrie M. Medical device containing light-protected therapeutic agent and a method for fabricating thereof
US6555157B1 (en) * 2000-07-25 2003-04-29 Advanced Cardiovascular Systems, Inc. Method for coating an implantable device and system for performing the method
US6673154B1 (en) * 2001-06-28 2004-01-06 Advanced Cardiovascular Systems, Inc. Stent mounting device to coat a stent
US6673385B1 (en) * 2000-05-31 2004-01-06 Advanced Cardiovascular Systems, Inc. Methods for polymeric coatings stents
US6689099B2 (en) * 1999-07-13 2004-02-10 Advanced Cardiovascular Systems, Inc. Local drug delivery injection catheter
US20040029952A1 (en) * 1999-09-03 2004-02-12 Yung-Ming Chen Ethylene vinyl alcohol composition and coating
US6695920B1 (en) * 2001-06-27 2004-02-24 Advanced Cardiovascular Systems, Inc. Mandrel for supporting a stent and a method of using the mandrel to coat a stent
US20040047980A1 (en) * 2000-12-28 2004-03-11 Pacetti Stephen D. Method of forming a diffusion barrier layer for implantable devices
US20040047978A1 (en) * 2000-08-04 2004-03-11 Hossainy Syed F.A. Composition for coating an implantable prosthesis
US6706013B1 (en) * 2001-06-29 2004-03-16 Advanced Cardiovascular Systems, Inc. Variable length drug delivery catheter
US20040054104A1 (en) * 2002-09-05 2004-03-18 Pacetti Stephen D. Coatings for drug delivery devices comprising modified poly(ethylene-co-vinyl alcohol)
US20040052859A1 (en) * 2001-05-09 2004-03-18 Wu Steven Z. Microparticle coated medical device
US6709514B1 (en) * 2001-12-28 2004-03-23 Advanced Cardiovascular Systems, Inc. Rotary coating apparatus for coating implantable medical devices
US6712845B2 (en) * 2001-04-24 2004-03-30 Advanced Cardiovascular Systems, Inc. Coating for a stent and a method of forming the same
US6713119B2 (en) * 1999-09-03 2004-03-30 Advanced Cardiovascular Systems, Inc. Biocompatible coating for a prosthesis and a method of forming the same

Family Cites Families (204)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2386454A (en) 1940-11-22 1945-10-09 Bell Telephone Labor Inc High molecular weight linear polyester-amides
US3849514A (en) 1967-11-17 1974-11-19 Eastman Kodak Co Block polyester-polyamide copolymers
US3773737A (en) 1971-06-09 1973-11-20 Sutures Inc Hydrolyzable polymers of amino acid and hydroxy acids
US4329383A (en) * 1979-07-24 1982-05-11 Nippon Zeon Co., Ltd. Non-thrombogenic material comprising substrate which has been reacted with heparin
SU790725A1 (en) 1979-07-27 1983-01-23 Ордена Ленина Институт Элементоорганических Соединений Ан Ссср Process for preparing alkylaromatic polyimides
US4226243A (en) 1979-07-27 1980-10-07 Ethicon, Inc. Surgical devices of polyesteramides derived from bis-oxamidodiols and dicarboxylic acids
SU872531A1 (en) 1979-08-07 1981-10-15 Институт Физиологии Им.И.С.Бериташвили Ан Гсср Method of producing polyurethans
SU811750A1 (en) 1979-08-07 1983-09-23 Институт Физиологии Им.С.И.Бериташвили Bis-bicarbonates of aliphatic diols as monomers for preparing polyurethanes and process for producing the same
SU876663A1 (en) 1979-11-11 1981-10-30 Институт Физиологии Им. Академика И.С.Бериташвили Ан Гсср Method of producing polyarylates
US4343931A (en) 1979-12-17 1982-08-10 Minnesota Mining And Manufacturing Company Synthetic absorbable surgical devices of poly(esteramides)
US4529792A (en) 1979-12-17 1985-07-16 Minnesota Mining And Manufacturing Company Process for preparing synthetic absorbable poly(esteramides)
SU1016314A1 (en) 1979-12-17 1983-05-07 Институт Физиологии Им.И.С.Бериташвили Process for producing polyester urethanes
SU905228A1 (en) 1980-03-06 1982-02-15 Институт Физиологии Им. Акад.И.С. Бериташвили Ан Гсср Method for preparing thiourea
SU1293518A1 (en) 1985-04-11 1987-02-28 Тбилисский зональный научно-исследовательский и проектный институт типового и экспериментального проектирования жилых и общественных зданий Installation for testing specimen of cross-shaped structure
US4611051A (en) 1985-12-31 1986-09-09 Union Camp Corporation Novel poly(ester-amide) hot-melt adhesives
US4882168A (en) 1986-09-05 1989-11-21 American Cyanamid Company Polyesters containing alkylene oxide blocks as drug delivery systems
JPH0696023B2 (en) 1986-11-10 1994-11-30 宇部日東化成株式会社 Artificial blood vessel and method for producing the same
US6387379B1 (en) * 1987-04-10 2002-05-14 University Of Florida Biofunctional surface modified ocular implants, surgical instruments, medical devices, prostheses, contact lenses and the like
US4894231A (en) 1987-07-28 1990-01-16 Biomeasure, Inc. Therapeutic agent delivery system
US4886062A (en) 1987-10-19 1989-12-12 Medtronic, Inc. Intravascular radially expandable stent and method of implant
JP2561309B2 (en) 1988-03-28 1996-12-04 テルモ株式会社 Medical material and manufacturing method thereof
US4931287A (en) 1988-06-14 1990-06-05 University Of Utah Heterogeneous interpenetrating polymer networks for the controlled release of drugs
US5328471A (en) 1990-02-26 1994-07-12 Endoluminal Therapeutics, Inc. Method and apparatus for treatment of focal disease in hollow tubular organs and other tissue lumens
US4977901A (en) 1988-11-23 1990-12-18 Minnesota Mining And Manufacturing Company Article having non-crosslinked crystallized polymer coatings
US5540931A (en) * 1989-03-03 1996-07-30 Charles W. Hewitt Methods for inducing site-specific immunosuppression and compositions of site specific immunosuppressants
US5272012A (en) 1989-06-23 1993-12-21 C. R. Bard, Inc. Medical apparatus having protective, lubricious coating
US5971954A (en) 1990-01-10 1999-10-26 Rochester Medical Corporation Method of making catheter
US5496557A (en) 1990-01-30 1996-03-05 Akzo N.V. Article for the controlled delivery of an active substance, comprising a hollow space fully enclosed by a wall and filled in full or in part with one or more active substances
WO1991017724A1 (en) 1990-05-17 1991-11-28 Harbor Medical Devices, Inc. Medical device polymer
CA2038605C (en) 1990-06-15 2000-06-27 Leonard Pinchuk Crack-resistant polycarbonate urethane polymer prostheses and the like
US6060451A (en) * 1990-06-15 2000-05-09 The National Research Council Of Canada Thrombin inhibitors based on the amino acid sequence of hirudin
WO1991019529A1 (en) * 1990-06-15 1991-12-26 Cortrak Medical, Inc. Drug delivery apparatus and method
US5112457A (en) * 1990-07-23 1992-05-12 Case Western Reserve University Process for producing hydroxylated plasma-polymerized films and the use of the films for enhancing the compatiblity of biomedical implants
US5455040A (en) 1990-07-26 1995-10-03 Case Western Reserve University Anticoagulant plasma polymer-modified substrate
US6248129B1 (en) 1990-09-14 2001-06-19 Quanam Medical Corporation Expandable polymeric stent with memory and delivery apparatus and method
US5163952A (en) 1990-09-14 1992-11-17 Michael Froix Expandable polymeric stent with memory and delivery apparatus and method
US5462990A (en) 1990-10-15 1995-10-31 Board Of Regents, The University Of Texas System Multifunctional organic polymers
US5330768A (en) 1991-07-05 1994-07-19 Massachusetts Institute Of Technology Controlled drug delivery using polymer/pluronic blends
US5500013A (en) * 1991-10-04 1996-03-19 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
US5599352A (en) 1992-03-19 1997-02-04 Medtronic, Inc. Method of making a drug eluting stent
GB9206736D0 (en) 1992-03-27 1992-05-13 Sandoz Ltd Improvements of organic compounds and their use in pharmaceutical compositions
US5219980A (en) 1992-04-16 1993-06-15 Sri International Polymers biodegradable or bioerodiable into amino acids
DE69325845T2 (en) * 1992-04-28 2000-01-05 Terumo Corp Thermoplastic polymer composition and medical devices made therefrom
DE4224401A1 (en) 1992-07-21 1994-01-27 Pharmatech Gmbh New biodegradable homo- and co-polymer(s) for pharmaceutical use - produced by polycondensation of prod. from heterolytic cleavage of aliphatic polyester with functionalised (cyclo)aliphatic cpd.
FR2699168B1 (en) * 1992-12-11 1995-01-13 Rhone Poulenc Chimie Method of treating a material comprising a polymer by hydrolysis.
EP0604022A1 (en) 1992-12-22 1994-06-29 Advanced Cardiovascular Systems, Inc. Multilayered biodegradable stent and method for its manufacture
US20020055710A1 (en) 1998-04-30 2002-05-09 Ronald J. Tuch Medical device for delivering a therapeutic agent and method of preparation
US5824048A (en) 1993-04-26 1998-10-20 Medtronic, Inc. Method for delivering a therapeutic substance to a body lumen
JPH0767895A (en) 1993-06-25 1995-03-14 Sumitomo Electric Ind Ltd Antimicrobial artificial blood vessel and suture yarn for antimicrobial operation
EG20321A (en) 1993-07-21 1998-10-31 Otsuka Pharma Co Ltd Medical material and process for producing the same
DE4327024A1 (en) 1993-08-12 1995-02-16 Bayer Ag Thermoplastically processable and biodegradable aliphatic polyesteramides
US5723004A (en) 1993-10-21 1998-03-03 Corvita Corporation Expandable supportive endoluminal grafts
WO1995019796A1 (en) 1994-01-21 1995-07-27 Brown University Research Foundation Biocompatible implants
CA2190121A1 (en) 1994-03-15 1995-09-21 Edith Mathiowitz Polymeric gene delivery system
US5567410A (en) * 1994-06-24 1996-10-22 The General Hospital Corporation Composotions and methods for radiographic imaging
US5857998A (en) * 1994-06-30 1999-01-12 Boston Scientific Corporation Stent and therapeutic delivery system
US5670558A (en) 1994-07-07 1997-09-23 Terumo Kabushiki Kaisha Medical instruments that exhibit surface lubricity when wetted
US5788979A (en) * 1994-07-22 1998-08-04 Inflow Dynamics Inc. Biodegradable coating with inhibitory properties for application to biocompatible materials
US5516881A (en) * 1994-08-10 1996-05-14 Cornell Research Foundation, Inc. Aminoxyl-containing radical spin labeling in polymers and copolymers
US5578073A (en) 1994-09-16 1996-11-26 Ramot Of Tel Aviv University Thromboresistant surface treatment for biomaterials
US5649977A (en) 1994-09-22 1997-07-22 Advanced Cardiovascular Systems, Inc. Metal reinforced polymer stent
FR2724938A1 (en) 1994-09-28 1996-03-29 Lvmh Rech POLYMERS FUNCTIONALIZED BY AMINO ACIDS OR AMINO ACID DERIVATIVES, THEIR USE AS SURFACTANTS, IN PARTICULAR, IN COSMETIC COMPOSITIONS AND IN PARTICULAR NAIL POLISH.
US5637113A (en) 1994-12-13 1997-06-10 Advanced Cardiovascular Systems, Inc. Polymer film for wrapping a stent structure
US5569198A (en) 1995-01-23 1996-10-29 Cortrak Medical Inc. Microporous catheter
US6017577A (en) 1995-02-01 2000-01-25 Schneider (Usa) Inc. Slippery, tenaciously adhering hydrophilic polyurethane hydrogel coatings, coated polymer substrate materials, and coated medical devices
US5919570A (en) 1995-02-01 1999-07-06 Schneider Inc. Slippery, tenaciously adhering hydrogel coatings containing a polyurethane-urea polymer hydrogel commingled with a poly(N-vinylpyrrolidone) polymer hydrogel, coated polymer and metal substrate materials, and coated medical devices
US6231600B1 (en) * 1995-02-22 2001-05-15 Scimed Life Systems, Inc. Stents with hybrid coating for medical devices
US5702754A (en) 1995-02-22 1997-12-30 Meadox Medicals, Inc. Method of providing a substrate with a hydrophilic coating and substrates, particularly medical devices, provided with such coatings
US5854376A (en) 1995-03-09 1998-12-29 Sekisui Kaseihin Kogyo Kabushiki Kaisha Aliphatic ester-amide copolymer resins
RU2169742C2 (en) 1995-04-19 2001-06-27 Катаока Казунори Heterotelochelate block copolymer and method of preparation thereof
US20020091433A1 (en) 1995-04-19 2002-07-11 Ni Ding Drug release coated stent
US6099562A (en) 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US6120536A (en) 1995-04-19 2000-09-19 Schneider (Usa) Inc. Medical devices with long term non-thrombogenic coatings
US5674242A (en) 1995-06-06 1997-10-07 Quanam Medical Corporation Endoprosthetic device with therapeutic compound
US6129761A (en) 1995-06-07 2000-10-10 Reprogenesis, Inc. Injectable hydrogel compositions
US5667767A (en) 1995-07-27 1997-09-16 Micro Therapeutics, Inc. Compositions for use in embolizing blood vessels
US5658995A (en) 1995-11-27 1997-08-19 Rutgers, The State University Copolymers of tyrosine-based polycarbonate and poly(alkylene oxide)
DE19545678A1 (en) 1995-12-07 1997-06-12 Goldschmidt Ag Th Copolymers of polyamino acid esters
US5932299A (en) 1996-04-23 1999-08-03 Katoot; Mohammad W. Method for modifying the surface of an object
US5955509A (en) 1996-05-01 1999-09-21 Board Of Regents, The University Of Texas System pH dependent polymer micelles
US5874165A (en) 1996-06-03 1999-02-23 Gore Enterprise Holdings, Inc. Materials and method for the immobilization of bioactive species onto polymeric subtrates
US5830178A (en) 1996-10-11 1998-11-03 Micro Therapeutics, Inc. Methods for embolizing vascular sites with an emboilizing composition comprising dimethylsulfoxide
US6060518A (en) * 1996-08-16 2000-05-09 Supratek Pharma Inc. Polymer compositions for chemotherapy and methods of treatment using the same
US5783657A (en) 1996-10-18 1998-07-21 Union Camp Corporation Ester-terminated polyamides of polymerized fatty acids useful in formulating transparent gels in low polarity liquids
US6120491A (en) 1997-11-07 2000-09-19 The State University Rutgers Biodegradable, anionic polymers derived from the amino acid L-tyrosine
US5871437A (en) * 1996-12-10 1999-02-16 Inflow Dynamics, Inc. Radioactive stent for treating blood vessels to prevent restenosis
US5980972A (en) 1996-12-20 1999-11-09 Schneider (Usa) Inc Method of applying drug-release coatings
US5997517A (en) 1997-01-27 1999-12-07 Sts Biopolymers, Inc. Bonding layers for medical device surface coatings
DE69826639T2 (en) 1997-01-28 2005-10-06 United States Surgical Corp., Norwalk SURGICAL ARTICLES MADE FROM POLYESTERAMIDES WITH GROUPS DERIVED FROM AMINO ACIDS AND ALTERNATIVELY WITH GROUPS DERIVED FROM ALPHA HYDROXYLIC ACIDS
DE69828387T2 (en) 1997-01-28 2005-12-08 United States Surgical Corp., Norwalk POLYESTERAMIDE, ITS PRESENTATION AND SURGICAL FABRICATED SURGICAL ARTICLES
WO1998032779A1 (en) * 1997-01-28 1998-07-30 United States Surgical Corporation Polyesteramide, its preparation and surgical devices fabricated therefrom
US6240616B1 (en) * 1997-04-15 2001-06-05 Advanced Cardiovascular Systems, Inc. Method of manufacturing a medicated porous metal prosthesis
US6273913B1 (en) 1997-04-18 2001-08-14 Cordis Corporation Modified stent useful for delivery of drugs along stent strut
US6245760B1 (en) 1997-05-28 2001-06-12 Aventis Pharmaceuticals Products, Inc Quinoline and quinoxaline compounds which inhibit platelet-derived growth factor and/or p56lck tyrosine kinases
US6056993A (en) * 1997-05-30 2000-05-02 Schneider (Usa) Inc. Porous protheses and methods for making the same wherein the protheses are formed by spraying water soluble and water insoluble fibers onto a rotating mandrel
US6110483A (en) 1997-06-23 2000-08-29 Sts Biopolymers, Inc. Adherent, flexible hydrogel and medicated coatings
US5980928A (en) 1997-07-29 1999-11-09 Terry; Paul B. Implant for preventing conjunctivitis in cattle
US6121027A (en) 1997-08-15 2000-09-19 Surmodics, Inc. Polybifunctional reagent having a polymeric backbone and photoreactive moieties and bioactive groups
US6316522B1 (en) 1997-08-18 2001-11-13 Scimed Life Systems, Inc. Bioresorbable hydrogel compositions for implantable prostheses
US6890546B2 (en) 1998-09-24 2005-05-10 Abbott Laboratories Medical devices containing rapamycin analogs
US6120788A (en) 1997-10-16 2000-09-19 Bioamide, Inc. Bioabsorbable triglycolic acid poly(ester-amide)s
US6110188A (en) 1998-03-09 2000-08-29 Corvascular, Inc. Anastomosis method
US6258371B1 (en) 1998-04-03 2001-07-10 Medtronic Inc Method for making biocompatible medical article
US20010029351A1 (en) 1998-04-16 2001-10-11 Robert Falotico Drug combinations and delivery devices for the prevention and treatment of vascular disease
US7658727B1 (en) 1998-04-20 2010-02-09 Medtronic, Inc Implantable medical device with enhanced biocompatibility and biostability
US20020188037A1 (en) 1999-04-15 2002-12-12 Chudzik Stephen J. Method and system for providing bioactive agent release coating
US6113629A (en) 1998-05-01 2000-09-05 Micrus Corporation Hydrogel for the therapeutic treatment of aneurysms
KR100314496B1 (en) 1998-05-28 2001-11-22 윤동진 Non-thrombogenic heparin derivatives, process for preparation and use thereof
US6153252A (en) 1998-06-30 2000-11-28 Ethicon, Inc. Process for coating stents
US6248127B1 (en) 1998-08-21 2001-06-19 Medtronic Ave, Inc. Thromboresistant coated medical device
US6419692B1 (en) 1999-02-03 2002-07-16 Scimed Life Systems, Inc. Surface protection method for stents and balloon catheters for drug delivery
US6143354A (en) 1999-02-08 2000-11-07 Medtronic Inc. One-step method for attachment of biomolecules to substrate surfaces
US6258121B1 (en) 1999-07-02 2001-07-10 Scimed Life Systems, Inc. Stent coating
US6790228B2 (en) 1999-12-23 2004-09-14 Advanced Cardiovascular Systems, Inc. Coating for implantable devices and a method of forming the same
US6287628B1 (en) 1999-09-03 2001-09-11 Advanced Cardiovascular Systems, Inc. Porous prosthesis and a method of depositing substances into the pores
US6759054B2 (en) 1999-09-03 2004-07-06 Advanced Cardiovascular Systems, Inc. Ethylene vinyl alcohol composition and coating
US6749626B1 (en) 2000-03-31 2004-06-15 Advanced Cardiovascular Systems, Inc. Actinomycin D for the treatment of vascular disease
US6204245B1 (en) * 1999-09-17 2001-03-20 The Regents Of The University Of California Treatment of narcolepsy with immunosuppressants
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
US6251136B1 (en) 1999-12-08 2001-06-26 Advanced Cardiovascular Systems, Inc. Method of layering a three-coated stent using pharmacological and polymeric agents
US6908624B2 (en) * 1999-12-23 2005-06-21 Advanced Cardiovascular Systems, Inc. Coating for implantable devices and a method of forming the same
US6283949B1 (en) 1999-12-27 2001-09-04 Advanced Cardiovascular Systems, Inc. Refillable implantable drug delivery pump
AU2599501A (en) 1999-12-29 2001-07-09 Advanced Cardiovascular Systems Inc. Device and active component for inhibiting formation of thrombus-inflammatory cell matrix
AU2623201A (en) 1999-12-30 2001-07-16 Kam W Leong Controlled delivery of therapeutic agents by insertable medical devices
US6527801B1 (en) 2000-04-13 2003-03-04 Advanced Cardiovascular Systems, Inc. Biodegradable drug delivery material for stent
US6270779B1 (en) 2000-05-10 2001-08-07 United States Of America Nitric oxide-releasing metallic medical devices
US6395326B1 (en) 2000-05-31 2002-05-28 Advanced Cardiovascular Systems, Inc. Apparatus and method for depositing a coating onto a surface of a prosthesis
US6585765B1 (en) 2000-06-29 2003-07-01 Advanced Cardiovascular Systems, Inc. Implantable device having substances impregnated therein and a method of impregnating the same
US20020077693A1 (en) 2000-12-19 2002-06-20 Barclay Bruce J. Covered, coiled drug delivery stent and method
WO2002009768A2 (en) 2000-07-27 2002-02-07 Rutgers, The State University Therapeutic polyesters and polyamides
US6585926B1 (en) 2000-08-31 2003-07-01 Advanced Cardiovascular Systems, Inc. Method of manufacturing a porous balloon
US6716444B1 (en) * 2000-09-28 2004-04-06 Advanced Cardiovascular Systems, Inc. Barriers for polymer-coated implantable medical devices and methods for making the same
US6254632B1 (en) 2000-09-28 2001-07-03 Advanced Cardiovascular Systems, Inc. Implantable medical device having protruding surface structures for drug delivery and cover attachment
US7261735B2 (en) 2001-05-07 2007-08-28 Cordis Corporation Local drug delivery devices and methods for maintaining the drug coatings thereon
US6746773B2 (en) 2000-09-29 2004-06-08 Ethicon, Inc. Coatings for medical devices
US20020111590A1 (en) 2000-09-29 2002-08-15 Davila Luis A. Medical devices, drug coatings and methods for maintaining the drug coatings thereon
US20020051730A1 (en) 2000-09-29 2002-05-02 Stanko Bodnar Coated medical devices and sterilization thereof
US6506437B1 (en) * 2000-10-17 2003-01-14 Advanced Cardiovascular Systems, Inc. Methods of coating an implantable device having depots formed in a surface thereof
US6558733B1 (en) 2000-10-26 2003-05-06 Advanced Cardiovascular Systems, Inc. Method for etching a micropatterned microdepot prosthesis
US6758859B1 (en) 2000-10-30 2004-07-06 Kenny L. Dang Increased drug-loading and reduced stress drug delivery device
US7077859B2 (en) 2000-12-22 2006-07-18 Avantec Vascular Corporation Apparatus and methods for variably controlled substance delivery from implanted prostheses
US6824559B2 (en) 2000-12-22 2004-11-30 Advanced Cardiovascular Systems, Inc. Ethylene-carboxyl copolymers as drug delivery matrices
US20020082679A1 (en) 2000-12-22 2002-06-27 Avantec Vascular Corporation Delivery or therapeutic capable agents
US20020087123A1 (en) 2001-01-02 2002-07-04 Hossainy Syed F.A. Adhesion of heparin-containing coatings to blood-contacting surfaces of medical devices
US6645195B1 (en) 2001-01-05 2003-11-11 Advanced Cardiovascular Systems, Inc. Intraventricularly guided agent delivery system and method of use
US6740040B1 (en) 2001-01-30 2004-05-25 Advanced Cardiovascular Systems, Inc. Ultrasound energy driven intraventricular catheter to treat ischemia
WO2002064014A2 (en) 2001-02-09 2002-08-22 Endoluminal Therapeutics, Inc. Endomural therapy
US6613077B2 (en) * 2001-03-27 2003-09-02 Scimed Life Systems, Inc. Stent with controlled expansion
US6623448B2 (en) 2001-03-30 2003-09-23 Advanced Cardiovascular Systems, Inc. Steerable drug delivery device
US6645135B1 (en) 2001-03-30 2003-11-11 Advanced Cardiovascular Systems, Inc. Intravascular catheter device and method for simultaneous local delivery of radiation and a therapeutic substance
US6780424B2 (en) 2001-03-30 2004-08-24 Charles David Claude Controlled morphologies in polymer drug for release of drugs from polymer films
US6625486B2 (en) 2001-04-11 2003-09-23 Advanced Cardiovascular Systems, Inc. Method and apparatus for intracellular delivery of an agent
US6764505B1 (en) 2001-04-12 2004-07-20 Advanced Cardiovascular Systems, Inc. Variable surface area stent
US6660034B1 (en) 2001-04-30 2003-12-09 Advanced Cardiovascular Systems, Inc. Stent for increasing blood flow to ischemic tissues and a method of using the same
US7651695B2 (en) * 2001-05-18 2010-01-26 Advanced Cardiovascular Systems, Inc. Medicated stents for the treatment of vascular disease
US6605154B1 (en) 2001-05-31 2003-08-12 Advanced Cardiovascular Systems, Inc. Stent mounting device
US6743462B1 (en) 2001-05-31 2004-06-01 Advanced Cardiovascular Systems, Inc. Apparatus and method for coating implantable devices
US7862495B2 (en) 2001-05-31 2011-01-04 Advanced Cardiovascular Systems, Inc. Radiation or drug delivery source with activity gradient to minimize edge effects
US6666880B1 (en) 2001-06-19 2003-12-23 Advised Cardiovascular Systems, Inc. Method and system for securing a coated stent to a balloon catheter
US6572644B1 (en) 2001-06-27 2003-06-03 Advanced Cardiovascular Systems, Inc. Stent mounting device and a method of using the same to coat a stent
US6565659B1 (en) 2001-06-28 2003-05-20 Advanced Cardiovascular Systems, Inc. Stent mounting assembly and a method of using the same to coat a stent
US6585755B2 (en) 2001-06-29 2003-07-01 Advanced Cardiovascular Polymeric stent suitable for imaging by MRI and fluoroscopy
US6656216B1 (en) 2001-06-29 2003-12-02 Advanced Cardiovascular Systems, Inc. Composite stent with regioselective material
EP1273314A1 (en) 2001-07-06 2003-01-08 Terumo Kabushiki Kaisha Stent
US6641611B2 (en) 2001-11-26 2003-11-04 Swaminathan Jayaraman Therapeutic coating for an intravascular implant
IN2014DN10834A (en) * 2001-09-17 2015-09-04 Psivida Inc
US20030083739A1 (en) 2001-09-24 2003-05-01 Robert Cafferata Rational drug therapy device and methods
US6753071B1 (en) 2001-09-27 2004-06-22 Advanced Cardiovascular Systems, Inc. Rate-reducing membrane for release of an agent
US6939376B2 (en) * 2001-11-05 2005-09-06 Sun Biomedical, Ltd. Drug-delivery endovascular stent and method for treating restenosis
US7585516B2 (en) 2001-11-12 2009-09-08 Advanced Cardiovascular Systems, Inc. Coatings for drug delivery devices
US6663880B1 (en) 2001-11-30 2003-12-16 Advanced Cardiovascular Systems, Inc. Permeabilizing reagents to increase drug delivery and a method of local delivery
US7445629B2 (en) 2002-01-31 2008-11-04 Boston Scientific Scimed, Inc. Medical device for delivering biologically active material
US6887270B2 (en) 2002-02-08 2005-05-03 Boston Scientific Scimed, Inc. Implantable or insertable medical device resistant to microbial growth and biofilm formation
US6743463B2 (en) * 2002-03-28 2004-06-01 Scimed Life Systems, Inc. Method for spray-coating a medical device having a tubular wall such as a stent
US20030203915A1 (en) * 2002-04-05 2003-10-30 Xinqin Fang Nitric oxide donors, compositions and methods of use related applications
US7008979B2 (en) * 2002-04-30 2006-03-07 Hydromer, Inc. Coating composition for multiple hydrophilic applications
US6865810B2 (en) * 2002-06-27 2005-03-15 Scimed Life Systems, Inc. Methods of making medical devices
US20040063805A1 (en) 2002-09-19 2004-04-01 Pacetti Stephen D. Coatings for implantable medical devices and methods for fabrication thereof
US7087263B2 (en) 2002-10-09 2006-08-08 Advanced Cardiovascular Systems, Inc. Rare limiting barriers for implantable medical devices
US20060002968A1 (en) 2004-06-30 2006-01-05 Gordon Stewart Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US8088404B2 (en) 2003-03-20 2012-01-03 Medtronic Vasular, Inc. Biocompatible controlled release coatings for medical devices and related methods
US7318944B2 (en) 2003-08-07 2008-01-15 Medtronic Vascular, Inc. Extrusion process for coating stents
US20050038497A1 (en) 2003-08-11 2005-02-17 Scimed Life Systems, Inc. Deformation medical device without material deformation
US20050037052A1 (en) 2003-08-13 2005-02-17 Medtronic Vascular, Inc. Stent coating with gradient porosity
US20050043786A1 (en) 2003-08-18 2005-02-24 Medtronic Ave, Inc. Methods and apparatus for treatment of aneurysmal tissue
US20050049693A1 (en) 2003-08-25 2005-03-03 Medtronic Vascular Inc. Medical devices and compositions for delivering biophosphonates to anatomical sites at risk for vascular disease
US20050055078A1 (en) 2003-09-04 2005-03-10 Medtronic Vascular, Inc. Stent with outer slough coating
US20050054774A1 (en) 2003-09-09 2005-03-10 Scimed Life Systems, Inc. Lubricious coating
US7544381B2 (en) 2003-09-09 2009-06-09 Boston Scientific Scimed, Inc. Lubricious coatings for medical device
US20050060020A1 (en) 2003-09-17 2005-03-17 Scimed Life Systems, Inc. Covered stent with biologically active material
US7371228B2 (en) 2003-09-19 2008-05-13 Medtronic Vascular, Inc. Delivery of therapeutics to treat aneurysms
US20050065501A1 (en) 2003-09-23 2005-03-24 Scimed Life Systems, Inc. Energy activated vaso-occlusive devices
US7789891B2 (en) 2003-09-23 2010-09-07 Boston Scientific Scimed, Inc. External activation of vaso-occlusive implants
US8801692B2 (en) 2003-09-24 2014-08-12 Medtronic Vascular, Inc. Gradient coated stent and method of fabrication
US7060319B2 (en) 2003-09-24 2006-06-13 Boston Scientific Scimed, Inc. method for using an ultrasonic nozzle to coat a medical appliance
US7055237B2 (en) 2003-09-29 2006-06-06 Medtronic Vascular, Inc. Method of forming a drug eluting stent
US20050074406A1 (en) 2003-10-03 2005-04-07 Scimed Life Systems, Inc. Ultrasound coating for enhancing visualization of medical device in ultrasound images
US6984411B2 (en) 2003-10-14 2006-01-10 Boston Scientific Scimed, Inc. Method for roll coating multiple stents
EP1604697A1 (en) * 2004-06-09 2005-12-14 J.A.C.C. GmbH Implantable device
US7563780B1 (en) * 2004-06-18 2009-07-21 Advanced Cardiovascular Systems, Inc. Heparin prodrugs and drug delivery stents formed therefrom
US7244443B2 (en) 2004-08-31 2007-07-17 Advanced Cardiovascular Systems, Inc. Polymers of fluorinated monomers and hydrophilic monomers
US7166680B2 (en) * 2004-10-06 2007-01-23 Advanced Cardiovascular Systems, Inc. Blends of poly(ester amide) polymers
US7214759B2 (en) 2004-11-24 2007-05-08 Advanced Cardiovascular Systems, Inc. Biologically absorbable coatings for implantable devices based on polyesters and methods for fabricating the same
US7202325B2 (en) 2005-01-14 2007-04-10 Advanced Cardiovascular Systems, Inc. Poly(hydroxyalkanoate-co-ester amides) and agents for use with medical articles

Patent Citations (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2072303A (en) * 1932-10-18 1937-03-02 Chemische Forschungs Gmbh Artificial threads, bands, tubes, and the like for surgical and other purposes
US4656242A (en) * 1985-06-07 1987-04-07 Henkel Corporation Poly(ester-amide) compositions
US4733665A (en) * 1985-11-07 1988-03-29 Expandable Grafts Partnership Expandable intraluminal graft, and method and apparatus for implanting an expandable intraluminal graft
US4733665B1 (en) * 1985-11-07 1994-01-11 Expandable Grafts Partnership Expandable intraluminal graft,and method and apparatus for implanting an expandable intraluminal graft
US4733665C2 (en) * 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US5721131A (en) * 1987-03-06 1998-02-24 United States Of America As Represented By The Secretary Of The Navy Surface modification of polymers with self-assembled monolayers that promote adhesion, outgrowth and differentiation of biological cells
US4800882A (en) * 1987-03-13 1989-01-31 Cook Incorporated Endovascular stent and delivery system
US5616338A (en) * 1988-02-11 1997-04-01 Trustees Of Columbia University In The City Of New York Infection-resistant compositions, medical devices and surfaces and methods for preparing and using same
US5100992A (en) * 1989-05-04 1992-03-31 Biomedical Polymers International, Ltd. Polyurethane-based polymeric materials and biomedical articles and pharmaceutical compositions utilizing the same
US5292516A (en) * 1990-05-01 1994-03-08 Mediventures, Inc. Body cavity drug delivery with thermoreversible gels containing polyoxyalkylene copolymers
US5306501A (en) * 1990-05-01 1994-04-26 Mediventures, Inc. Drug delivery by injection with thermoreversible gels containing polyoxyalkylene copolymers
US5300295A (en) * 1990-05-01 1994-04-05 Mediventures, Inc. Ophthalmic drug delivery with thermoreversible polyoxyalkylene gels adjustable for pH
US5298260A (en) * 1990-05-01 1994-03-29 Mediventures, Inc. Topical drug delivery with polyoxyalkylene polymer thermoreversible gels adjustable for pH and osmolality
US5607467A (en) * 1990-09-14 1997-03-04 Froix; Michael Expandable polymeric stent with memory and delivery apparatus and method
US5306786A (en) * 1990-12-21 1994-04-26 U C B S.A. Carboxyl group-terminated polyesteramides
US5858746A (en) * 1992-04-20 1999-01-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5624411A (en) * 1993-04-26 1997-04-29 Medtronic, Inc. Intravascular stent and method
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US5380299A (en) * 1993-08-30 1995-01-10 Med Institute, Inc. Thrombolytic treated intravascular medical device
US5735897A (en) * 1993-10-19 1998-04-07 Scimed Life Systems, Inc. Intravascular stent pump
US6051576A (en) * 1994-01-28 2000-04-18 University Of Kentucky Research Foundation Means to achieve sustained release of synergistic drugs by conjugation
US5485496A (en) * 1994-09-22 1996-01-16 Cornell Research Foundation, Inc. Gamma irradiation sterilizing of biomaterial medical devices or products, with improved degradation and mechanical properties
US5879713A (en) * 1994-10-12 1999-03-09 Focal, Inc. Targeted delivery via biodegradable polymers
US5869127A (en) * 1995-02-22 1999-02-09 Boston Scientific Corporation Method of providing a substrate with a bio-active/biocompatible coating
US5605696A (en) * 1995-03-30 1997-02-25 Advanced Cardiovascular Systems, Inc. Drug loaded polymeric material and method of manufacture
US6358556B1 (en) * 1995-04-19 2002-03-19 Boston Scientific Corporation Drug release stent coating
US20030028243A1 (en) * 1995-06-07 2003-02-06 Cook Incorporated Coated implantable medical device
US20030028244A1 (en) * 1995-06-07 2003-02-06 Cook Incorporated Coated implantable medical device
US5865814A (en) * 1995-06-07 1999-02-02 Medtronic, Inc. Blood contacting medical device and method
US5873904A (en) * 1995-06-07 1999-02-23 Cook Incorporated Silver implantable medical device
US20030036794A1 (en) * 1995-06-07 2003-02-20 Cook Incorporated Coated implantable medical device
US5609629A (en) * 1995-06-07 1997-03-11 Med Institute, Inc. Coated implantable medical device
US6010530A (en) * 1995-06-07 2000-01-04 Boston Scientific Technology, Inc. Self-expanding endoluminal prosthesis
US5877224A (en) * 1995-07-28 1999-03-02 Rutgers, The State University Of New Jersey Polymeric drug formulations
US6051648A (en) * 1995-12-18 2000-04-18 Cohesion Technologies, Inc. Crosslinked polymer compositions and methods for their use
US5723219A (en) * 1995-12-19 1998-03-03 Talison Research Plasma deposited film networks
US6033582A (en) * 1996-01-22 2000-03-07 Etex Corporation Surface modification of medical implants
US6054553A (en) * 1996-01-29 2000-04-25 Bayer Ag Process for the preparation of polymers having recurring agents
US5610241A (en) * 1996-05-07 1997-03-11 Cornell Research Foundation, Inc. Reactive graft polymer with biodegradable polymer backbone and method for preparing reactive biodegradable polymers
US5876433A (en) * 1996-05-29 1999-03-02 Ethicon, Inc. Stent and method of varying amounts of heparin coated thereon to control treatment
US6172167B1 (en) * 1996-06-28 2001-01-09 Universiteit Twente Copoly(ester-amides) and copoly(ester-urethanes)
US5711958A (en) * 1996-07-11 1998-01-27 Life Medical Sciences, Inc. Methods for reducing or eliminating post-surgical adhesion formation
US6530951B1 (en) * 1996-10-24 2003-03-11 Cook Incorporated Silver implantable medical device
US6042875A (en) * 1997-04-30 2000-03-28 Schneider (Usa) Inc. Drug-releasing coatings for medical devices
US6180632B1 (en) * 1997-05-28 2001-01-30 Aventis Pharmaceuticals Products Inc. Quinoline and quinoxaline compounds which inhibit platelet-derived growth factor and/or p56lck tyrosine kinases
US6528526B1 (en) * 1997-05-28 2003-03-04 Aventis Pharmaceuticals Inc. Quinoline and quinoxaline compounds which inhibit platelet-derived growth factor and/or p56lck tyrosine kinases
US6524347B1 (en) * 1997-05-28 2003-02-25 Avantis Pharmaceuticals Inc. Quinoline and quinoxaline compounds which inhibit platelet-derived growth factor and/or p56lck tyrosine kinases
US6211249B1 (en) * 1997-07-11 2001-04-03 Life Medical Sciences, Inc. Polyester polyether block copolymers
US6034204A (en) * 1997-08-08 2000-03-07 Basf Aktiengesellschaft Condensation products of basic amino acids with copolymerizable compounds and a process for their production
US6015541A (en) * 1997-11-03 2000-01-18 Micro Therapeutics, Inc. Radioactive embolizing compositions
US20030040790A1 (en) * 1998-04-15 2003-02-27 Furst Joseph G. Stent coating
US6214901B1 (en) * 1998-04-27 2001-04-10 Surmodics, Inc. Bioactive agent release coating
US20030031780A1 (en) * 1998-04-27 2003-02-13 Chudzik Stephen J. Bioactive agent release coating
US6344035B1 (en) * 1998-04-27 2002-02-05 Surmodics, Inc. Bioactive agent release coating
US20020032434A1 (en) * 1998-04-27 2002-03-14 Chudzik Stephen J. Bioactive agent release coating
US20020032414A1 (en) * 1998-08-20 2002-03-14 Ragheb Anthony O. Coated implantable medical device
US6335029B1 (en) * 1998-08-28 2002-01-01 Scimed Life Systems, Inc. Polymeric coatings for controlled delivery of active agents
US6011125A (en) * 1998-09-25 2000-01-04 General Electric Company Amide modified polyesters
US6530950B1 (en) * 1999-01-12 2003-03-11 Quanam Medical Corporation Intraluminal stent having coaxial polymer member
US20030040712A1 (en) * 1999-07-13 2003-02-27 Pinaki Ray Substance delivery apparatus and a method of delivering a therapeutic substance to an anatomical passageway
US6689099B2 (en) * 1999-07-13 2004-02-10 Advanced Cardiovascular Systems, Inc. Local drug delivery injection catheter
US6177523B1 (en) * 1999-07-14 2001-01-23 Cardiotech International, Inc. Functionalized polyurethanes
US6713119B2 (en) * 1999-09-03 2004-03-30 Advanced Cardiovascular Systems, Inc. Biocompatible coating for a prosthesis and a method of forming the same
US20040029952A1 (en) * 1999-09-03 2004-02-12 Yung-Ming Chen Ethylene vinyl alcohol composition and coating
US6379381B1 (en) * 1999-09-03 2002-04-30 Advanced Cardiovascular Systems, Inc. Porous prosthesis and a method of depositing substances into the pores
US6346110B2 (en) * 1999-10-04 2002-02-12 Advanced Cardiovascular Systems, Inc. Chamber for applying therapeutic substances to an implantable device
US6203551B1 (en) * 1999-10-04 2001-03-20 Advanced Cardiovascular Systems, Inc. Chamber for applying therapeutic substances to an implant device
US20020009604A1 (en) * 1999-12-22 2002-01-24 Zamora Paul O. Plasma-deposited coatings, devices and methods
US6503954B1 (en) * 2000-03-31 2003-01-07 Advanced Cardiovascular Systems, Inc. Biocompatible carrier containing actinomycin D and a method of forming the same
US20020016625A1 (en) * 2000-05-12 2002-02-07 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020007214A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020007215A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020005206A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Antiproliferative drug and delivery device
US20020007213A1 (en) * 2000-05-19 2002-01-17 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US6673385B1 (en) * 2000-05-31 2004-01-06 Advanced Cardiovascular Systems, Inc. Methods for polymeric coatings stents
US6555157B1 (en) * 2000-07-25 2003-04-29 Advanced Cardiovascular Systems, Inc. Method for coating an implantable device and system for performing the method
US20040047978A1 (en) * 2000-08-04 2004-03-11 Hossainy Syed F.A. Composition for coating an implantable prosthesis
US6503538B1 (en) * 2000-08-30 2003-01-07 Cornell Research Foundation, Inc. Elastomeric functional biodegradable copolyester amides and copolyester urethanes
US6544543B1 (en) * 2000-12-27 2003-04-08 Advanced Cardiovascular Systems, Inc. Periodic constriction of vessels to treat ischemic tissue
US20040047980A1 (en) * 2000-12-28 2004-03-11 Pacetti Stephen D. Method of forming a diffusion barrier layer for implantable devices
US6503556B2 (en) * 2000-12-28 2003-01-07 Advanced Cardiovascular Systems, Inc. Methods of forming a coating for a prosthesis
US20030072868A1 (en) * 2000-12-28 2003-04-17 Sameer Harish Methods of forming a coating for a prosthesis
US6540776B2 (en) * 2000-12-28 2003-04-01 Advanced Cardiovascular Systems, Inc. Sheath for a prosthesis and methods of forming the same
US6544223B1 (en) * 2001-01-05 2003-04-08 Advanced Cardiovascular Systems, Inc. Balloon catheter for delivering therapeutic agents
US6544582B1 (en) * 2001-01-05 2003-04-08 Advanced Cardiovascular Systems, Inc. Method and apparatus for coating an implantable device
US20030032767A1 (en) * 2001-02-05 2003-02-13 Yasuhiro Tada High-strength polyester-amide fiber and process for producing the same
US20030004141A1 (en) * 2001-03-08 2003-01-02 Brown David L. Medical devices, compositions and methods for treating vulnerable plaque
US6712845B2 (en) * 2001-04-24 2004-03-30 Advanced Cardiovascular Systems, Inc. Coating for a stent and a method of forming the same
US20030039689A1 (en) * 2001-04-26 2003-02-27 Jianbing Chen Polymer-based, sustained release drug delivery system
US20040052858A1 (en) * 2001-05-09 2004-03-18 Wu Steven Z. Microparticle coated medical device
US20040052859A1 (en) * 2001-05-09 2004-03-18 Wu Steven Z. Microparticle coated medical device
US6695920B1 (en) * 2001-06-27 2004-02-24 Advanced Cardiovascular Systems, Inc. Mandrel for supporting a stent and a method of using the mandrel to coat a stent
US6673154B1 (en) * 2001-06-28 2004-01-06 Advanced Cardiovascular Systems, Inc. Stent mounting device to coat a stent
US6527863B1 (en) * 2001-06-29 2003-03-04 Advanced Cardiovascular Systems, Inc. Support device for a stent and a method of using the same to coat a stent
US6706013B1 (en) * 2001-06-29 2004-03-16 Advanced Cardiovascular Systems, Inc. Variable length drug delivery catheter
US20030060877A1 (en) * 2001-09-25 2003-03-27 Robert Falotico Coated medical devices for the treatment of vascular disease
US20030059520A1 (en) * 2001-09-27 2003-03-27 Yung-Ming Chen Apparatus for regulating temperature of a composition and a method of coating implantable devices
US20030065377A1 (en) * 2001-09-28 2003-04-03 Davila Luis A. Coated medical devices
US20030073961A1 (en) * 2001-09-28 2003-04-17 Happ Dorrie M. Medical device containing light-protected therapeutic agent and a method for fabricating thereof
US6709514B1 (en) * 2001-12-28 2004-03-23 Advanced Cardiovascular Systems, Inc. Rotary coating apparatus for coating implantable medical devices
US20040054104A1 (en) * 2002-09-05 2004-03-18 Pacetti Stephen D. Coatings for drug delivery devices comprising modified poly(ethylene-co-vinyl alcohol)

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9468706B2 (en) 2004-03-22 2016-10-18 Abbott Cardiovascular Systems Inc. Phosphoryl choline coating compositions
US20050208093A1 (en) * 2004-03-22 2005-09-22 Thierry Glauser Phosphoryl choline coating compositions
US8709469B2 (en) 2004-06-30 2014-04-29 Abbott Cardiovascular Systems Inc. Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US9566373B2 (en) 2004-06-30 2017-02-14 Abbott Cardiovascular Systems Inc. Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US20100322992A1 (en) * 2004-06-30 2010-12-23 Stephen Dugan Anti-Proliferative And Anti-Inflammatory Agent Combination For Treatment Of Vascular Disorders With An Implantable Medical Device
US9138337B2 (en) 2004-06-30 2015-09-22 Abbott Cardiovascular Systems Inc. Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US9539332B2 (en) 2004-08-05 2017-01-10 Abbott Cardiovascular Systems Inc. Plasticizers for coating compositions
US20060069427A1 (en) * 2004-09-24 2006-03-30 Savage Douglas R Drug-delivery endovascular stent and method for treating restenosis
US7901451B2 (en) * 2004-09-24 2011-03-08 Biosensors International Group, Ltd. Drug-delivery endovascular stent and method for treating restenosis
US8871292B2 (en) * 2004-09-24 2014-10-28 Biosensors International Group, Ltd. Drug-delivery endovascular stent and method for treating restenosis
US20120290076A1 (en) * 2004-09-24 2012-11-15 Biosensors International Group, Ltd. Drug-delivery endovascular stent and method for treating restenosis
US8252047B2 (en) 2004-09-24 2012-08-28 Biosensors International Group, Ltd. Drug-delivery endovascular stent and method for treating restenosis
US20070299511A1 (en) * 2006-06-27 2007-12-27 Gale David C Thin stent coating
US20100063585A1 (en) * 2006-07-03 2010-03-11 Hemoteq Ag Manufacture, method and use of active substance-releasing medical products for permanently keeping blood vessels open
US8685430B1 (en) 2006-07-14 2014-04-01 Abbott Cardiovascular Systems Inc. Tailored aliphatic polyesters for stent coatings
US8597720B2 (en) 2007-01-21 2013-12-03 Hemoteq Ag Medical product for treating stenosis of body passages and for preventing threatening restenosis
US20100179475A1 (en) * 2007-01-21 2010-07-15 Erika Hoffmann Medical product for treating stenosis of body passages and for preventing threatening restenosis
US8591931B2 (en) * 2007-02-14 2013-11-26 Shangdong Intech Medical Technology Co., Ltd Coronary stent with asymmetric drug releasing controlled coating
US20100168842A1 (en) * 2007-02-14 2010-07-01 Junbo Ge Coronary stent with asymmetric drug releasing controlled coating
US10155881B2 (en) 2007-05-30 2018-12-18 Abbott Cardiovascular Systems Inc. Substituted polycaprolactone for coating
US20080299164A1 (en) * 2007-05-30 2008-12-04 Trollsas Mikael O Substituted polycaprolactone for coating
US9737638B2 (en) 2007-06-20 2017-08-22 Abbott Cardiovascular Systems, Inc. Polyester amide copolymers having free carboxylic acid pendant groups
US9090745B2 (en) 2007-06-29 2015-07-28 Abbott Cardiovascular Systems Inc. Biodegradable triblock copolymers for implantable devices
US9468707B2 (en) 2007-06-29 2016-10-18 Abbott Cardiovascular Systems Inc. Biodegradable triblock copolymers for implantable devices
US20110160698A1 (en) * 2007-07-03 2011-06-30 Hemoteq Ag Balloon Catheter for Treating Stenosis of Body Passages and for Preventing Threatening Restenosis
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
US9814553B1 (en) 2007-10-10 2017-11-14 Abbott Cardiovascular Systems Inc. Bioabsorbable semi-crystalline polymer for controlling release of drug from a coating
US20090306120A1 (en) * 2007-10-23 2009-12-10 Florencia Lim Terpolymers containing lactide and glycolide
US20090110711A1 (en) * 2007-10-31 2009-04-30 Trollsas Mikael O Implantable device having a slow dissolving polymer
US8889170B2 (en) 2007-10-31 2014-11-18 Abbott Cardiovascular Systems Inc. Implantable device having a coating with a triblock copolymer
US9629944B2 (en) 2007-10-31 2017-04-25 Abbott Cardiovascular Systems Inc. Implantable device with a triblock polymer coating
US9345668B2 (en) 2007-10-31 2016-05-24 Abbott Cardiovascular Systems Inc. Implantable device having a slow dissolving polymer
US8642062B2 (en) 2007-10-31 2014-02-04 Abbott Cardiovascular Systems Inc. Implantable device having a slow dissolving polymer
US20090259302A1 (en) * 2008-04-11 2009-10-15 Mikael Trollsas Coating comprising poly (ethylene glycol)-poly (lactide-glycolide-caprolactone) interpenetrating network
US8128983B2 (en) 2008-04-11 2012-03-06 Abbott Cardiovascular Systems Inc. Coating comprising poly(ethylene glycol)-poly(lactide-glycolide-caprolactone) interpenetrating network
US20090297584A1 (en) * 2008-04-18 2009-12-03 Florencia Lim Biosoluble coating with linear over time mass loss
US8916188B2 (en) 2008-04-18 2014-12-23 Abbott Cardiovascular Systems Inc. Block copolymer comprising at least one polyester block and a poly (ethylene glycol) block
US20090285873A1 (en) * 2008-04-18 2009-11-19 Abbott Cardiovascular Systems Inc. Implantable medical devices and coatings therefor comprising block copolymers of poly(ethylene glycol) and a poly(lactide-glycolide)
US20090263457A1 (en) * 2008-04-18 2009-10-22 Trollsas Mikael O Block copolymer comprising at least one polyester block and a poly(ethylene glycol) block
US20100209476A1 (en) * 2008-05-21 2010-08-19 Abbott Cardiovascular Systems Inc. Coating comprising a terpolymer comprising caprolactone and glycolide
US8697113B2 (en) 2008-05-21 2014-04-15 Abbott Cardiovascular Systems Inc. Coating comprising a terpolymer comprising caprolactone and glycolide
US20090297578A1 (en) * 2008-06-03 2009-12-03 Trollsas Mikael O Biosoluble coating comprising anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US20110301697A1 (en) * 2009-04-10 2011-12-08 Hemoteq Ag Manufacture, method and use of drug-eluting medical devices for permanently keeping blood vessels open
US20100272773A1 (en) * 2009-04-24 2010-10-28 Boston Scientific Scimed, Inc. Use of Drug Polymorphs to Achieve Controlled Drug Delivery From a Coated Medical Device
US8697110B2 (en) 2009-05-14 2014-04-15 Abbott Cardiovascular Systems Inc. Polymers comprising amorphous terpolymers and semicrystalline blocks
US20110008260A1 (en) * 2009-07-10 2011-01-13 Boston Scientific Scimed, Inc. Use of Nanocrystals for Drug Delivery from a Balloon
US10369256B2 (en) 2009-07-10 2019-08-06 Boston Scientific Scimed, Inc. Use of nanocrystals for drug delivery from a balloon
US11278648B2 (en) 2009-07-10 2022-03-22 Boston Scientific Scimed, Inc. Use of nanocrystals for drug delivery from a balloon
US20110015664A1 (en) * 2009-07-17 2011-01-20 Boston Scientific Scimed, Inc. Nucleation of Drug Delivery Balloons to Provide Improved Crystal Size and Density
US10080821B2 (en) 2009-07-17 2018-09-25 Boston Scientific Scimed, Inc. Nucleation of drug delivery balloons to provide improved crystal size and density
US8889211B2 (en) 2010-09-02 2014-11-18 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
US8669360B2 (en) 2011-08-05 2014-03-11 Boston Scientific Scimed, Inc. Methods of converting amorphous drug substance into crystalline form
US9056152B2 (en) 2011-08-25 2015-06-16 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
US9220759B2 (en) 2012-02-23 2015-12-29 Abbott Cardiovascular Systems Inc. Treatment of diabetic patients with a drug eluting stent and adjunctive therapy
US9750627B2 (en) 2012-03-30 2017-09-05 Abbott Cardiovascular Systems Inc. Treatment of diabetic patients with a stent and locally administered adjunctive therapy

Also Published As

Publication number Publication date
US8586069B2 (en) 2013-11-19
US20060105019A1 (en) 2006-05-18

Similar Documents

Publication Publication Date Title
US8586069B2 (en) Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US7758881B2 (en) Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US8435550B2 (en) Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US20090297578A1 (en) Biosoluble coating comprising anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders
US8367090B2 (en) Coating on a balloon comprising a polymer and a drug
US8961589B2 (en) Bioabsorbable coating with tunable hydrophobicity
US20110137243A1 (en) Coating On A Balloon Device
US8343529B2 (en) Implantable drug delivery devices having alternating hydrophilic and amphiphillic polymer layers
US20120330405A1 (en) Implantable Medical Devices With A Topcoat Layer Of Phosphoryl Choline For Reduced Thrombosis, And Improved Mechanical Properties
US20050287184A1 (en) Drug-delivery stent formulations for restenosis and vulnerable plaque
EP2061530B1 (en) Implantable devices containing nuclear receptor ligands for the treatment of vascular and related disorders
US20150086603A1 (en) Dual-targeted drug carriers
US20140161862A1 (en) Absorbable coating for implantable device
WO2009158068A2 (en) Methods of application of coatings composed of hydrophobic, high glass transition polymers with tunable drug release rates
US9220759B2 (en) Treatment of diabetic patients with a drug eluting stent and adjunctive therapy
US9669137B2 (en) Modified polylactide polymers
US9566373B2 (en) Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
US20090104240A1 (en) Dual Drug Formulations For Implantable Medical Devices For Treatment of Vascular Diseases

Legal Events

Date Code Title Description
AS Assignment

Owner name: ADVANCED CARDIOVASCULAR SYSTEMS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STEWART, GORDON;ZHANG, GINA;KIRSTEN, NANCY;AND OTHERS;REEL/FRAME:015544/0423

Effective date: 20040629

STCB Information on status: application discontinuation

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