WO1996025176A1 - Therapeutic inhibitor of vascular smooth muscle cells - Google Patents

Therapeutic inhibitor of vascular smooth muscle cells Download PDF

Info

Publication number
WO1996025176A1
WO1996025176A1 PCT/US1996/002125 US9602125W WO9625176A1 WO 1996025176 A1 WO1996025176 A1 WO 1996025176A1 US 9602125 W US9602125 W US 9602125W WO 9625176 A1 WO9625176 A1 WO 9625176A1
Authority
WO
WIPO (PCT)
Prior art keywords
smooth muscle
cells
stent
cytoskeletal inhibitor
analog
Prior art date
Application number
PCT/US1996/002125
Other languages
English (en)
French (fr)
Inventor
Lawrence L. Kunz
Original Assignee
Neorx Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23539404&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1996025176(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to AT96906490T priority Critical patent/ATE265233T1/de
Priority to AU49851/96A priority patent/AU4985196A/en
Priority to CA002212537A priority patent/CA2212537C/en
Priority to JP8525163A priority patent/JPH11500635A/ja
Priority to EP96906490A priority patent/EP0809515B1/en
Priority to DE69632310T priority patent/DE69632310T2/de
Application filed by Neorx Corporation filed Critical Neorx Corporation
Publication of WO1996025176A1 publication Critical patent/WO1996025176A1/en
Priority to US08/829,991 priority patent/US6306421B1/en
Priority to US08/829,685 priority patent/US5981568A/en
Priority to US09/361,194 priority patent/US6358989B1/en
Priority to US09/896,208 priority patent/US6491938B2/en
Priority to US09/995,490 priority patent/US6569441B2/en
Priority to US10/024,885 priority patent/US6663881B2/en
Priority to US11/650,059 priority patent/US8158670B2/en
Priority to US13/446,760 priority patent/US20120195951A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/04Nitro compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/131Amines acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • A61K47/6809Antibiotics, e.g. antitumor antibiotics anthracyclins, adriamycin, doxorubicin or daunomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6831Fungal toxins, e.g. alpha sarcine, mitogillin, zinniol or restrictocin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6843Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6957Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a device or a kit, e.g. stents or microdevices
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/148Materials at least partially resorbable by 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
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • 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
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/602Type of release, e.g. controlled, sustained, slow
    • 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
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/602Type of release, e.g. controlled, sustained, slow
    • A61L2300/604Biodegradation
    • 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
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/606Coatings
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/323Arteriosclerosis, Stenosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/773Nanoparticle, i.e. structure having three dimensions of 100 nm or less
    • Y10S977/775Nanosized powder or flake, e.g. nanosized catalyst
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/788Of specified organic or carbon-based composition
    • Y10S977/802Virus-based particle
    • Y10S977/806Virus-based particle with exterior chemical attachment
    • Y10S977/808Exterior attachment for targeting, e.g. drug targeting
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/905Specially adapted for travel through blood circulatory system
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/906Drug delivery
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/915Therapeutic or pharmaceutical composition
    • Y10S977/916Gene therapy

Definitions

  • This invention relates generally to therapeutic methods involving surgical or intravenous introduction of binding partners directed to certain target cell populations, such as smooth muscle cells, cancer cells, somatic cells requiring modulation to ameliorate a disease state and effector cells of the immune system, particularly for treating conditions such as stenosis following vascular trauma or disease, cancer, diseases resulting from hyperactivity or hyperplasia of somatic cells and diseases that are mediated by immune system effector cells.
  • Surgical or intravenous introduction of active agents capable of altering the proliferation or migration or contraction of smooth muscle proteins is also described.
  • the invention also relates to the direct or targeted delivery of therapeutic agents to vascular smooth muscle cells that results in dilation and fixation of the vascular lumen (biological stenting effect).
  • Combined administration of a cytocidal conjugate and a sustained release dosage form of a vascular smooth muscle cell inhibitor is also disclosed.
  • PTCA Percutaneous transluminal coronary angioplasty
  • PTCA Percutaneous transluminal coronary angioplasty
  • the use of this surgical procedure has grown rapidly, with 39,000 procedures performed in 1983, nearly 150,000 in 1987, 200,000 in 1988, 250,000 in 1989, and over 500,000 PTCAs per year are estimated by 1994 (1 , 2, 3).
  • Stenosis following PTCA remains a significant problem, with from 25% to 35% of the patients developing restenosis within 1 to 3 months. Restenosis results in significant morbidity and mortality and frequently necessitates further interventions such as repeat angioplasty or coronary bypass surgery. No surgical intervention or post-surgical treatment (to date) has proven effective in preventing restenosis.
  • Heparin is an example of one such compound, which reportedly inhibits smooth muscle cell proliferation in vitro but when used in vivo has the potential adverse side effect of inhibiting coagulation.
  • Heparin peptides while having reduced anti-coagulant activity, have the undesirable pharmacological property of having a short pharmacological half-life. Attempts have been made to solve such problems by using a double balloon catheter, i.e.. for regional delivery of the therapeutic agent at the angioplasty site (e.g., 8;
  • Verrucarins and Roridins are trichothecene drugs produced as secondary metabolites by the soil fungi Myrothecium verruc ⁇ ri ⁇ and
  • Verrucarin is a macrocyclic triester.
  • Roridin is a macrocyclic diester of verrucarol (10).
  • the trichothecenes are structurally related to sesquiterpenoid mycotoxins produced by several species of fungi and characterized by the 12,13-epoxytrichothec-9-ene basic structure. Their cytotoxic activity to eukaryotic cells is closely correlated with their ability to bind to the cell, to be internalized, and to inhibit protein and macromolecular synthesis in the cell.
  • inhibitory agents may have systemic toxicity that could create an unacceptable level of risk for patients with cardiovascular disease.
  • inhibitory agents might interfere with vascular wound healing following surgery and that could either delay healing or weaken the structure or elasticity of the newly healed vessel wall.
  • inhibitory agents killing smooth muscle cells could damage surrounding endothelium and/or other medial smooth muscle cells. Dead and dying cells also release mitogenic agents that might stimulate additional smooth muscle cell proliferation and exacerbate stenosis.
  • an inhibitory agent may be problematic from several standpoints: namely, a) delivery of a large number of molecules into the intercellular spaces between smooth muscle cells may be necessary, i.e., to establish favorable conditions for allowing a therapeutical ly effective dose of molecules to cross the cell membrane; b) directing an inhibitory drug into the proper intracellular compartment, i.e., where its action is exerted, may be difficult to control; and, c) optimizing the association of the inhibitory drug with its intracellular target, e.g, a ribosome, while minimizing intercellular redistribution of the drug, e.g. to neighboring cells, may be difficult.
  • therapeutic conjugates are provided for inhibiting vascular smooth muscle cells in a mammalian host.
  • the therapeutic conjugates contain a vascular smooth muscle binding protein or peptide that binds in a specific manner to the cell membranes of a vascular smooth muscle cell or an interstitial matrix binding protein/peptide that binds in a specific manner to interstitial matrix (e.g., collagen) of the artery wall, coupled to a therapeutic agent that inhibits the activity of the cell.
  • inhibition of cellular activity results in reducing, delaying, or eliminating stenosis after angioplasty or other vascular surgical procedures.
  • the therapeutic conjugates of the invention achieve these advantageous effects by associating with vascular smooth muscle cells and pericytes, which may transform into smooth muscle cells.
  • the therapeutic conjugate may contain: (1) therapeutic agents that alter cellular metabolism or are inhibitors of protein synthesis, cellular proliferation, or cell migration; (2) microtubule and microfilament inhibitors that affect
  • the conjugates include a cytotoxic therapeutic agent that is a sesquiterpenoid mycotoxin such as a verrucarin or a roridin.
  • cytostatic therapeutic agents that inhibit DNA synthesis and proliferation at doses that have a minimal effect on protein synthesis such as protein kinase inhibitors (e.g., staurosporin), suramin, and nitric oxide releasing compounds (e.g., nitroglycerin) or analogs or functional equivalents thereof.
  • therapeutic agents that inhibit the contraction or migration of smooth muscle cells and maintain an enlarged luminal area following, for example, angioplasty trauma e.g.. the cytochalasins, such as cytochalasin B,
  • cytochalasin C cytochalasin D or the like
  • vascular smooth muscle binding proteins that specifically associate with a chondroitin sulfate proteoglycan (CSPG) expressed on the membranes of a vascular smooth muscle cell, and in a preferred embodiment this CSPG has a molecular weight of about 250 kDaltons.
  • CSPG chondroitin sulfate proteoglycan
  • the vascular smooth muscle binding protein binds to a CSPG target on the cell surface with an association constant of at least 10 -4 M.
  • the vascular smooth muscle binding protein contains a sequence of amino acids found in the Fab, Fv or CDR (complementarity determining regions) of monoclonal antibody NR-AN-01 or functional equivalents thereof.
  • aspects of the invention include methods for inhibiting stenosis, e.g., following angioplasty in a mammalian host, by administering to a human or animal subject in need of such treatment a therapeutically effective dosage of a therapeutic conjugate of the invention.
  • stenosis e.g., following angioplasty in a mammalian host
  • the dosage of therapeutic conjugate may be administered with an infusion catheter, to achieve a 10 -3 M to 10 I2 M concentration of said therapeutic conjugate at the site of administration in a blood vessel.
  • the present invention also contemplates therapeutic methods and therapeutic dosage forms involving sustained release of therapeutic agent to target cells.
  • the target cells are vascular smooth muscle cells, cancer cells, somatic cells requiring modulation to ameliorate a disease state and cells involved in immune system-mediated diseases that are accessible by local administration of the dosage form. Consequently, the methods and dosage forms of this aspect of the present invention are useful for inhibiting vascular smooth muscle cells in a mammalian host, employing a therapeutic agent that inhibits the activity of the cell (e.g., proliferation, contraction, migration or the like) but does not kill the cell and a vascular smooth muscle cell binding protein.
  • a therapeutic agent that inhibits the activity of the cell (e.g., proliferation, contraction, migration or the like) but does not kill the cell and a vascular smooth muscle cell binding protein.
  • the methods and dosage forms of this aspect of the present invention are useful for inhibiting target cell proliferation or killing such target cells, employing a therapeutic agent that inhibits proliferation or is cytotoxic to the target cells and a target cell binding protein.
  • the methods and dosage forms of this aspect of the present invention are useful for delivering cytostatic, cytocidal or metabolism modulating therapeutic agents to target cells, such as effector cells of the immune system, that are accessible by local administration of the dosage form, employing a target cell binding protein.
  • dosage forms of the present invention are useful to reduce or eliminate pathological proliferation or hyperactivity of normal tissue (i.e., somatic cells).
  • one therapeutic method of the invention involves the in vivo placement of a metallic, plastic or biodegradable intravascular stent which comprises a therapeutic agent.
  • stents comprising a therapeutic agent.
  • a preferred embodiment of the invention is a stent which comprises a therapeutic agent such as a cytoskeletal inhibitor or an inhibitor of smooth muscle cell proliferation.
  • a preferred cytoskeletal inhibitor of the invention is a cytochalasin, such as cytochalasin B or a structural analog thereof that is functionally equivalent.
  • An alternative preferred cytoskeletal inhibitor of the invention is taxol or a structural analog thereof that is functionally equivalent.
  • the stents can comprise a biodegradable coating or a porous or permeable non-biodegradable coating.
  • a preferred embodiment of the invention is a stent comprising a biodegradable coating or a porous/permeable non-biodegradable coating comprising the therapeutic agent.
  • a more preferred embodiment of the invention is a stent comprising a biodegradable coating or a porous or non-biodegradable non-biodegradable coating comprising a sustained-release dosage form of the therapeutic agent.
  • a stent e.g., a biodegradable stent
  • a biodegradable stent with the therapeutic agent impregnated therein which is further coated with a biodegradable coating or with a porous or permeable non-biodegradable coating comprising a sustained release-dosage form of a therapeutic agent is also contemplated.
  • This embodiment of the invention can provide a differential release rate of the therapeutic agent, i.e., there would be a faster release of the therapeutic agent from the coating followed by delayed release of the therapeutic agent that is impregnated in the stent matrix upon degradation of the stent matrix.
  • the intravascular stent provides a mechanical means of providing an increase in luminal area of a vessel, in addition to that provided via the biological stenting action of the cytoskeletal inhibitor, such as cytochalasin B or taxol, releasably embedded therein. Furthermore, the placement of intravascular stents comprising a therapeutic agent which is an inhibitor of smooth muscle cell proliferation provides an increased efficacy by reducing or preventing intimal proliferation. This inhibition of intimal smooth muscle cells and stroma produced by the smooth muscle allows for more rapid and complete re-endothelization following the intraventional placement of the vascular stent. The increased rate of re-endothelization and stabilization of the vessel wall following stent placement can reduce the loss of luminal area and decreased blood flow which is the primary cause of vascular stent failures.
  • the cytoskeletal inhibitor such as cytochalasin B or taxol
  • the cytochalasins such as cytochalasin B, cytochalasin C, cytochalasin D or the like
  • suramin such as nitroglycerin
  • nitric oxide- releasing compounds such as nitroglycerin, or analogs or functional equivalents thereof.
  • useful therapeutic agents inhibit proliferation or are cytotoxic to the target cells.
  • Preferred therapeutic moieties for this purpose are Roridin A and Pseudomonas exotoxin. or analogs or functional equivalents thereof.
  • useful therapeutic agents deliver cytostatic, cytocidal or metabolism-modulating therapeutic agents to target cells that are accessible by local administration of the dosage form.
  • Preferred therapeutic moieties for this purpose are Roridin A, Pseudomonas exotoxin, suramin and protein kinase inhibitors (e.g., staurosporin), sphingosine, or analogs or functional equivalents thereof.
  • anti-proliferative agents or antimigration agents are preferred (e.g., cytochalasins, taxol, somatostatin, somatostatin analogs, N-ethylmaleimide, antisense
  • the dosage forms of the present invention are targeted to a relevant target cell population by a binding protein or peptide.
  • Preferred binding proteins/peptides of the present invention are vascular smooth muscle cell binding protein, tumor cell binding protein and immune system effector cell binding protein.
  • Preferred vascular smooth muscle cell binding proteins specifically associate with a chondroitin sulfate proteoglycan (CSPG) expressed on the membranes of a vascular smooth muscle cell, and in a preferred embodiment this CSPG has a molecular weight of about 250 kDaltons.
  • CSPG chondroitin sulfate proteoglycan
  • the vascular smooth muscle binding protein binds to a CSPG target on the cell surface with an association constant of at least 10 -4 M.
  • the present invention also provides therapeutic methods and
  • therapeutic dosage forms involving administration of free (i.e., non-targeted or non-binding partner associated) therapeutic agent to target cells.
  • the target cells are vascular smooth muscle cells and the therapeutic agent is an inhibitor of vascular smooth muscle cell contraction, allowing the normal hydrostatic pressure to dilate the vascular lumen.
  • Such contraction inhibition may be achieved by actin inhibition, which is preferably achievable and sustainable at a lower dose level than that necessary to inhibit protein synthesis. Consequently, the vascular smooth muscle cells synthesize protein required to repair minor cell trauma and secrete interstitial matrix, thereby facilitating the fixation of the vascular lumen in a dilated state near its maximal systolic diameter.
  • This phenomenon constitutes a biological stenting effect that diminishes or prevents the undesirable recoil mechanism that occurs in up to 25% of the angioplasty procedures classified as successful based on an initial post-procedural angiogram.
  • Cytochalasins which inhibit the polymerization of G- to F-actin which, in turn, inhibits the migration and contraction of vascular smooth muscle cells
  • Free therapeutic agent protocols of this type effect a reduction, a delay, or an elimination of stenosis after angioplasty or other vascular surgical procedures.
  • free therapeutic agent is administered directly or substantially directly to vascular smooth muscle tissue. Such administration is preferably effected by an infusion catheter, to achieve a 10 -3 M to 10 -12 M concentration of said therapeutic agent at the site of administration in a blood vessel.
  • a cytocidal targeted conjugate to destroy proliferating vascular smooth muscle cells involved in vascular stenosis.
  • the mitogenic agents released after this biological arteromyectomy are prevented from stimulating the remaining viable vascular smooth muscle cells to proliferate and restenose the vessel by administration of the anti-contraction (anti-migration) or anti-proliferative sustained release agents of the present invention.
  • FIGURE 1 is a photomicrograph of vascular smooth muscle cells in an artery of a 24-year-old male patient with vascular smooth muscle binding protein bound to the cell surface and membrane.
  • the patient received the vascular smooth muscle binding protein by i.v. administration 4 days before the arterial tissue was prepared for histology.
  • FIGURE 1B is a photomicrograph of vascular smooth muscle cells in an artery of a 24-year-old male patient with vascular smooth muscle binding protein bound to the cell surface and membrane. The section was reacted ex vivo with HRP-conjugated goat anti-mouse IgG. This reaction was visualized by adding 4-chloro-1-naphthol. The reaction product of the substrate forms an insoluble purple or dark brown precipitate at the reaction site (shown at #2). A counter stain was used to visualize cell nuclei (shown at #1).
  • FIGURE 2 depicts a first scheme for chemical coupling of a therapeutic agent to a vascular smooth muscle binding protein.
  • FIGURE 3 depicts a second scheme for chemical coupling of a therapeutic agent to a vascular smooth muscle binding protein.
  • FIGURE 4A graphically depicts experimental data showing rapid binding of vascular smooth muscle binding protein to marker-positive test cells in vitro.
  • FIGURE 4B graphically depicts experimental data showing rapid binding of vascular smooth muscle binding protein to vascular smooth muscle cells in vitro.
  • FIGURE 5 A presents graphically experimental data showing
  • RA-NR-AN-01 therapeutic conjugate
  • free RA therapeutic agent undesirable cytotoxicity of even low levels of therapeutic conjugate (i.e., RA-NR-AN-01), and the free RA therapeutic agent, when vascular smooth muscle cells were treated for 24 hours in vitro.
  • FIGURE 5B graphically presents experimental data showing the effects of RA-NR-AN-01 therapeutic conjugate on metabolic activity of marker-positive and -negative cells.
  • the data show undesirable nonspecific cytotoxicity of the conjugate for all these cells in a 24 hour treatment in vitro.
  • the non-specificity results from extracellular hydrolysis of the coupling ligand which exposes the tested cells to free drug.
  • FIGURE 6A graphically depicts experimental data showing undesirable nonspecific cytotoxicity of PE-NR-AN-01 therapeutic conjugate for marker-positive and marker-negative test cells after 24 hours of treatment in vitro, even though the 24 hour treatment was followed by an overnight recovery period prior to testing the metabolic activity.
  • FIGURE 6B depicts experimental data showing nonspecific
  • FIGURE 7A graphically presents experimental data showing that a short 5 minute "pulse” treatment, i.e., instead of 24 hours, followed by exposure to [3H]leucine, with free RA therapeutic agent being nonspecifically cytotoxic, i.e., for control HT29 marker-negative cells, but, in contrast, the RA-NR-AN-01 therapeutic conjugate is not cytotoxic in this "pulse" treatment.
  • FIGURE 7B presents graphically experimental data showing that free
  • RA-NRAN-01 therapeutic conjugate is not cytotoxic to cells.
  • FIGURE 7C presents graphically results of experiments showing that "pulse" treatment of cells in vitro with the RA-NR-AN-01 therapeutic conjugate inhibits cellular activity in marker-positive A375 cells, as measured by protein synthesis.
  • FIGURE 7D presents graphically experimental data showing that "pulse" treatment of cells in vitro with the RA-NR-AN-01 therapeutic conjugate did not exert long-lasting inhibitory effects on cellular activity in marker-positive cells, since protein synthesis in A375 cells was not inhibited when the cells were allowed an overnight recovery period prior to testing in vitro.
  • FIGURE 8A presents graphically experimental data showing that while a "pulse" treatment of cells in vitro with free RA therapeutic agent was non-specifically cytotoxic, the RA-NR-AN-01 therapeutic conjugate did not exert long-lasting inhibitory effects on cellular activity in vascular smooth muscle cells, as evidenced by metabolic activity in BO54 cells that were allowed a 48 hour recovery period prior to testing.
  • FIGURE 8B graphically depicts experimental data similar to those presented in FIGURE 8A, above, but using a second marker-positive cell type, namely A375.
  • the data show that "pulse" treatment with the RA-NR-AN-01 therapeutic conjugate did not exert long-lasting inhibitory effects on cellular activity, as measured by metabolic activity in A375 cells that were allowed a 48 hour recovery period prior to testing.
  • FIGURE 8C graphically depicts results similar to those presented in
  • the results show that the "pulse" treatment with the RA-NR-AN-01 therapeutic conjugate did not exert long-lasting inhibitory effects on the cellular activity of marker-negative control cells, as measured by metabolic activity in HT29 cells that were allowed a 48 hour recovery period prior to testing.
  • FIGURE 9A shows stenosis due to intimal smooth muscle cell proliferation in a histological section of an untreated artery 5 weeks after angioplasty in an animal model.
  • FIGURE 9B shows inhibition of stenosis in a histological section of an artery treated with therapeutic conjugate at 5 weeks after angioplasty in an animal model.
  • FIGURE 10A graphically depicts experimental data comparing protein synthesis and DNA synthesis inhibition capability of suramin with respect to vascular smooth muscle cells.
  • FIGURE 10B graphically depicts experimental data comparing protein synthesis and DNA synthesis inhibition capability of staurosporin with respect to vascular smooth muscle cells.
  • FIGURE 10C graphically depicts experimental data comparing protein synthesis and DNA synthesis inhibition capability of nitroglycerin with respect to vascular smooth muscle cells.
  • FIGURE 1 1 shows a tangential section parallel to the inner surface of a smooth muscle cell which is magnified 62,500 times and is characterized by numerous endocytic vesicles, several of which contain antibody coated gold beads in the process of being internalized by the cell in vitro.
  • FIGURE 12 shows a smooth muscle cell which is magnified 62,500 times and is characterized by a marked accumulation of gold beads in lysosomes at 24 hours following exposure of the cell to the beads in vitro.
  • FIGURE 14 depicts an in vivo dose response study of the effect of cytochalasin B on the luminal area of pig femoral arteries. Detailed Description of the Invention
  • “Therapeutic conjugate” means a vascular smooth muscle or an interstitial matrix binding protein coupled (e.g., optionally through a linker) to a therapeutic agent.
  • Target and marker are used interchangeably in describing the conjugate aspects of the present invention to mean a molecule recognized in a specific manner by the matrix or vascular smooth muscle binding protein, e.g., an antigen, polypeptide antigen or cell surface carbohydrate (e.g., a glycolipid. glycoprotein, or proteoglycan) that is expressed on the cell surface membranes of a vascular smooth muscle cell or a matrix structure.
  • an antigen, polypeptide antigen or cell surface carbohydrate e.g., a glycolipid. glycoprotein, or proteoglycan
  • Epitope is used to refer to a specific site within the “target” molecule that is bound by the matrix or smooth muscle binding protein, e.g., a sequence of three or more amino acids or saccharides.
  • Coupled is used to mean covalent or non-covalent chemical association (i.e., hydrophobic as through van der Waals forces or
  • the binding proteins will normally be associated with the
  • Linker means an agent that couples the matrix or smooth muscle binding protein to a therapeutic agent, e.g., an organic chemical coupler.
  • Microcyclic trichothecene is intended to mean any one of the group of structurally related sesquiterpenoid macrocyclic mycotoxins produced by several species of fungi and characterized by the 12,13-epoxytrichothec-9-ene basic structure, e.g., verrucarins and roridins that are the products of secondary metabolism in the soil fungi Myrothecium verrucaria and
  • sustained release means a dosage form designed to release a therapeutic agent therefrom for a time period ranging from about 3 to about 21 days. Release over a longer time period is also contemplated as a
  • Biodegradation rate directly impacts therapeutic agent release kinetics.
  • the biodegradation rate is regulable by alteration of the composition or structure of the sustained release dosage form.
  • alteration of the lactide/glycolide ratio in preferred dosage forms of the present invention can be conducted, as described by Tice et al., "Biodegradable Controlled-Release Parenteral Systems," Pharmaceutical Technology, pp. 26-35, 1984; by inclusion of polymer hydrolysis modifying agents, such as citric acid and sodium carbonate, as described by Kent et al., "Microencapsulation of Water Soluble Active Polypeptides," U.S. Patent No.
  • vascular smooth muscle binding proteins include antibodies (e.g., monoclonal and polyclonal affinity-purified antibodies, F(ab'), Fab', Fab. and Fv fragments and/or complementarity determining regions (CDR) of antibodies or functional equivalents thereof, (e.g., binding peptides and the like)); growth factors, cytokines, and polypeptide hormones and the like; and macromolecules recognizing extracellular matrix receptors (e.g., integrin and fibronectin receptors and the like).
  • antibodies e.g., monoclonal and polyclonal affinity-purified antibodies, F(ab'), Fab', Fab. and Fv fragments and/or complementarity determining regions (CDR) of antibodies or functional equivalents thereof, (e.g., binding peptides and the like)); growth factors, cytokines, and polypeptide hormones and the like; and macromolecules recognizing extracellular matrix receptors (e.g., integrin and fibronect
  • trichothecenes There are two broad classes of trichothecenes: those that have only a central sesquiterpenoid structure and those that have an additional macrocyclic ring (simple and macrocyclic trichothecenes, respectively).
  • the simple trichothecenes may be subdivided into three groups (i.e.. Group A, B, and C) as described in U.S. Patent Nos. 4,744,981 and 4.906,452 (incorporated herein by reference).
  • Representative examples of Group A simple trichothecenes include: Scirpene, Roridin C, dihydrotrichothecene. Scirpen-4. 8-diol,
  • Group C simple trichothecenes include: Crotocol and Crotocin.
  • Representative macrocyclic trichothecenes include Verrucarin A, Verrucarin B, Verrucarin J (Satratoxin C), Roridin A, Roridin D, Roridin E (Satratoxin D), Roridin H, Satratoxin F, Satratoxin G, Satratoxin H, Vertisporin, Mytoxin A, Mytoxin C, Mytoxin B, Myrotoxin A, Myrotoxin B, Myrotoxin C,
  • Preferred agents for these embodiments include Roridin A, Pseudomonas exotoxin, suramin, protein kinase inhibitors (e.g., staurosporin) and the like, or analogs or functional equivalents thereof.
  • compositions useful in the practice of the present invention include moieties capable of reducing or eliminating pathological proliferation, migration or hyperactivity of normal tissues.
  • exemplary of such therapeutic agents are those capable of reducing or eliminating hyperactivity of corneal epithelium and stroma, pathological proliferation or prolonged contraction of smooth muscle cells or pericytes of the intraocular vasculature implicated in degenerative eye disease resulting from hyperplasia or decreased vascular lumen area.
  • Preferred agents for this purpose are staurosporin and cytochalasin B.
  • Vascular smooth muscle binding proteins of the invention bind to targets on the surface of vascular smooth muscle cells.
  • targets e.g., polypeptides or carbohydrates, proteoglycans and the like, that are associated with the cell membranes of vascular smooth muscle cells are useful for selecting (e.g., by cloning) or constructing (e.g., by genetic engineering or chemical synthesis) appropriately specific vascular smooth muscle binding proteins.
  • Particularly useful "targets” are internalized by smooth muscle cells, e.g., as membrane constituent antigen turnover occurs in renewal. Internalization by cells may also be by mechanisms involving phagolysosomes, clathrin-coated pits, receptor-mediated redistribution or endocytosis and the like.
  • such a “target” is exemplified by chondroitin sulfate proteoglycans (CSPGs) synthesized by vascular smooth muscle cells and pericytes, and a discrete portion (termed an epitope herein) of the CSPG molecule having an apparent molecular weight of about 250 kD is especially preferred.
  • the 250 kD target is an N-linked glycoprotein that is a component of a larger 400 kD proteoglycan complex (14).
  • NR-ML-05 binding to smooth muscle cells has been disclosed (Fritzberg et al., U.S. Pat. No. 4,879.225).
  • Monoclonal antibody NR-ML-05 and subculture NR-ML-05 No. 85-41-4I-A2, freeze # A2106, have both been deposited with the American Type Culture Collection, Rockville, MD and granted Accession Nos. HB-5350 and HB-9350, respectively.
  • NR-ML-05 is the parent of, and structurally and functionally equivalent to, subclone
  • the binding-site amino acids and three dimensional model of the NR-AN-01 antigen binding site serve as a molecular model for constructing functional equivalents, e.g., short polypeptides ("minimal polypeptides"), that have binding affinity for a CSPG synthesized by vascular smooth muscle cells and pericytes.
  • functional equivalents e.g., short polypeptides ("minimal polypeptides")
  • binding proteins e.g., antibodies or fragments
  • selected binding proteins for use in the practice of the invention have a binding affinity of >10 4 liter/mole for the vascular smooth muscle 250 kD CSPG, and also the ability to be bound to and internalized by smooth muscle cells or pericytes.
  • murine monoclonal antibody may be "chimerized” by genetically recombining the nucleotide sequence encoding the murine Fv region (i.e., containing the antigen binding sites) with the nucleotide sequence encoding a human constant domain region and an Fc region, e.g., in a manner similar to that disclosed in European Patent Application No. 0,41 1,893 A2.
  • Humanized vascular smooth muscle binding partners will be recognized to have the advantage of decreasing the immunoreactivity of the antibody or polypeptide in the host recipient, which may thereby be useful for increasing the in vivo half-life and reducing the possibility of adverse immune reactions.
  • binding peptides localize to the surface membrane of intact cells and internal epitopes of disrupted cells, respectively, and deliver the therapeutic agent for assimilation into the target cells.
  • Minimal peptides, mimetic organic compounds and human or humanized antibodies that localize to the requisite tumor cell types are also useful as binding peptides of the present invention.
  • binding peptides may be identified and constructed or isolated in accordance with known techniques.
  • Preferred binding peptides of these embodiments of the present invention bind to a target epitope with an association constant of at least about 10 -6 M.
  • binding peptides are also useful as binding peptides of the present invention. Such binding peptides may be identified and constructed or isolated in accordance with known techniques. Preferred binding peptides of these embodiments of the present invention bind to a target epitope with an association constant of at least about 10 -6 M.
  • binding proteins or peptides useful in the practice of the present invention include moieties capable of localizing to pathologically proliferating normal tissues, such as pericytes of the intraocular vasculature implicated in degenerative eye disease.
  • the therapeutic agent is delivered to target cells for internalization therein by such sustained release dosage forms.
  • Minimal peptides, mimetic organic compounds and human or humanized antibodies that localize to the requisite pathologically proliferating normal cell types are also useful as binding peptides of the present invention.
  • binding peptides may be identified and constructed or isolated in accordance with known techniques.
  • Preferred binding peptides of these embodiments of the present invention bind to a target epitope with an association constant of at least about 10 -6 M.
  • Coupled methods for linking the therapeutic agent through covalent or non-covalent bonds to the vascular smooth muscle binding protein include chemical cross-linkers and heterobifunctional cross-linking compounds (i.e., "linkers") that react to form a bond between reactive groups (such as hydroxyl, amino, amido, or sulfhydryl groups) in a therapeutic agent and other reactive groups (of a similar nature) in the vascular smooth muscle binding protein.
  • This bond may be, for example, a peptide bond, disulfide bond, thioester bond, amide bond, thioether bond, and the like.
  • the sustained release embodiment of the present invention includes a therapeutic agent dispersed within a non-biodegradable or biodegradable polymeric structure. Such dispersion is conducted in accordance with the procedure described by Cowsar et al., "Poly(Lactide-Co-Glycolide)
  • nucleophilic groups include lysine epsilon amino groups (amide linkage), serine hydroxyl groups (ester linkage) or cysteine mercaptan groups (thioester linkage). Reactions with particular groups depend upon pH and the reduction state of the reaction conditions.
  • the benztriazole-derivatized particulates i.e., activated imidate-bearing moieties
  • a protein/peptide nucleophilic moiety such as an available epsilon amino moiety.
  • p-nitrophenol p-nitrophenol
  • binding protein/peptide nucleophilic moieties include hydroxyl groups of serine, endogenous free thiols of cysteine, thiol groups resulting from reduction of binding
  • lactic/glycolic acid particulates are activatable by reaction with thionyl chloride to form an acyl chloride derivatized moiety.
  • the derivatized particulates are then reacted with binding peptide/protein nucleophilic groups to form targeted dosage forms of the present invention.
  • the particulate carboxy groups are biotinylated (e.g., through carbodiimide activation of the carboxy group and subsequent reaction with amino alkyl biotinamide).
  • the biotinylated particulates are then incubated with a saturating concentration (i.e., a molar excess) of avidin or streptavidin to form protein coated particulates having free biotin binding sites.
  • a saturating concentration i.e., a molar excess
  • Such coated particulates are then capable of reaction with a molar excess of biotinylated binding protein formed as described above.
  • Another option involves avidin or streptavidin bound binding peptide or protein attachment to biotinylated particulates.
  • binding protein/peptide may be partially entrapped in the particulate polymeric matrix upon formation thereof.
  • entrapped binding protein/peptide provides residual selective binding character to the particulate. Mild particulate formation conditions, such as those employed by Cohen et al., Pharmaceutical Research, 8: 713-720 (1991), are preferred for this embodiment of the present invention. Such entrapped binding protein is also useful in target cell reattachment of a partially degraded particulate that has undergone exocytosis.
  • Other polymeric particulate dosage forms e.g., non-biodegradable dosage forms having different exposed functional groups can be bound to binding proteins or peptides in accordance with the principles discussed above.
  • targeting is specific for potentially proliferating cells that result in increased smooth muscle in the intimal region of a traumatized vascular site, e.g.. following angioplasty, e.g., pericytes and vascular smooth muscle cells.
  • angioplasty e.g., pericytes and vascular smooth muscle cells.
  • the therapeutic conjugates or dosage forms of the invention may be administered to the host using an infusion catheter, such as produced by C.R. Bard Inc., Billerica, MA, or that disclosed by Wolinsky (7; U.S. Patent No. 4,824,436) or Spears (U.S. Patent No. 4,512,762).
  • an infusion catheter such as produced by C.R. Bard Inc., Billerica, MA, or that disclosed by Wolinsky (7; U.S. Patent No. 4,824,436) or Spears (U.S. Patent No. 4,512,762).
  • a therapeutically effective dosage of the therapeutic conjugate will be typically reached when the concentration of conjugate in the fluid space between the balloons of the catheter is in the range of about 10 -3 to 10 -12 M.
  • therapeutic conjugates of the invention may only need to be delivered in an anti-proliferative therapeutic dosage sufficient to expose the proximal (6 to 9) cell layers of the intimal or tunica media cells lining the lumen to the therapeutic anti-proliferative conjugate, whereas the anti-contractile therapeutic dosage needs to expose the entire tunica media, and further that this dosage can be determined empirically, e.g., by a) infusing vessels from suitable animal model systems and using immunohistochemical methods to detect the conjugate and its effects (e.g., such as exemplified in the EXAMPLES below); and b) conducting suitable in vitro studies such as exemplified in EXAMPLES 3, 4, and 5, below).
  • this therapeutically effective dosage is achieved by preparing 10 ml of a 200 ⁇ g/ml therapeutic conjugate solution, wherein the vascular smooth muscle protein binding protein is NR-AN-01 and the therapeutic agent is Roridin A, a trichothecene drug.
  • 10 ml will commonly be sufficient volume to fill the catheter and infuse 1 to 1.5 ml into one to three traumatic lesion sites in the vessel wall.
  • Sustained release dosage forms of an embodiment of the invention may only need to be delivered in an anti-proliferative therapeutic dosage sufficient to expose the proximal (6 to 9) cell layers of the tunica media smooth muscle cells lining the lumen to the dosage form.
  • This dosage is determinable empirically, e.g., by a) infusing vessels from suitable animal model systems and using immunohistochemical, fluorescent or electron microscopy methods to detect the dosage form and its effects; and b) conducting suitable in vitro studies.
  • this therapeutically effective dosage is achieved by determining in smooth muscle cell tissue culture the pericellular agent dosage, which at a continuous exposure results in a therapeutic effect between the toxic and minimal effective doses.
  • This therapeutic level is obtained in vivo by determining the size, number and therapeutic agent concentration and release rate required for particulates infused between the smooth muscle cells of the artery wall to maintain this pericellular therapeutic dosage.
  • the dosage form should release the therapeutic agent at a rate that approximates the pericellular dose of the following exemplary therapeutic agents: from about 0.01 to about 100 micrograms/ml nitroglycerin, from about 1.0 to about 1000 micrograms/ml of suramin, from about 0.001 to about 100 micrograms/ml for cytochalasin. and from about 0.01 to about 10 5 nanograms/ml of staurosporin.
  • therapeutically effective dosages of the sustained release dosage form of the invention will be dependent on several factors, including, e.g.: a) the binding affinity of the binding protein associated with the dosage form; b) the atmospheric pressure and duration of the infusion; c) the time over which the dosage form administered resides at the target site; d) the rate of therapeutic agent release from the particulate dosage form; e) the nature of the therapeutic agent employed; f) the nature of the trauma and/or therapy desired; and/or g) the intercellular and/or intracellular localization of the particulate dosage form.
  • Those skilled practitioners trained to deliver drugs at therapeutically effective dosages are capable of determining the optimal dosage for an individual patient based on experience and professional judgment.
  • about 0.3 atm (i.e., 300 mm of Hg) to about 3 atm of pressure applied for 15 seconds to 3 minutes to the arterial wall is adequate to achieve infiltration of a sustained release dosage form bound to the NR-AN-01 binding protein into the smooth muscle layers of a mammalian artery wall.
  • Wolinsky et al. "Direct Intraarterial Wall Injection of Microparticles Via a Catheter: A Potential Drug Delivery Strategy Following Angioplasty," Am. Heart Jour., 122(4):1 136-1 140, 1991.
  • Those skilled in the art will recognize that infiltration of a sustained release dosage form into a target cell population will probably be variable and will need to be determined on an individual basis.
  • therapeutic conjugates radiolabeled with alpha-, beta- or gamma-emitters of known specific activities are useful for determining the therapeutically effective dosage by using them in animal studies and human trials with quantitative imaging or autoradiography of histological tissue sections to determine the concentration of therapeutic conjugate that is required by the therapeutic protocol.
  • a therapeutically effective dosage of the therapeutic conjugate or dosage form will be reached when at least three conditions are met: namely.
  • the therapeutic dosage required to achieve the desired inhibitory activity for a therapeutic conjugate or dosage form can also be anticipated through the use of in vitro studies.
  • the infusion catheter may be conveniently a double balloon or quadruple balloon catheter with a permeable membrane.
  • a therapeutically effective dosage of a therapeutic conjugate or dosage form is useful in treating vascular trauma resulting from disease (e.g., atherosclerosis, aneurysm, or the like) or vascular surgical procedures such as angioplasty, atheroectomy, placement of a stent (e.g., in a vessel), thrombectomy, and grafting.
  • Atheroectomy may be performed, for example, by surgical excision, ultrasound or laser treatment, or by high pressure fluid flow.
  • Grafting may be, for example, vascular grafting using natural or synthetic materials or surgical anastomosis of vessels such as, e.g., during organ grafting.
  • a therapeutic conjugate containing Roridin A and NR-AN-01 achieved a therapeutically effective dosage in vivo at a concentration which inhibited cellular protein synthesis in test cells in vitro by at least 5 to 50%, as judged by incorporation of radiolabeled amino acids.
  • cell migration and cell adherence in in vitro assays may be used for determining the concentration at which a therapeutically effective dosage will be reached in the fluid space created by the infusion catheter in the vessel wall.
  • While one representative embodiment of the invention relates to therapeutic methods employing an infusion catheter, it will be recognized that other methods for drug delivery or routes of administration may also be useful, e.g., injection by the intravenous, intralymphatic, intrathecal.
  • intraarterial local delivery by implanted osmotic pumps or other intracavity routes.
  • nanoparticulate dosage forms of the present invention are preferred.
  • Intravenous administration of nanoparticulates is useful, for example, where vascular permeability is increased in tumors for leakage, especially in necrotic areas of tumors having damaged vessels which allow the leakage of particles into the interstitial fluid, and where artery walls have been denuded and traumatized allowing the particles to enter the interstitial area of the tunica media.
  • non-coupled vascular smooth muscle cell binding protein e.g., free NR-AN-01 antibody
  • non-coupled vascular smooth muscle cell binding protein is administered prior to administration of the therapeutic agent conjugate or dosage form to provide a blocker of non-specific binding to cross-reactive sites. Blocking of such sites is important because vascular smooth muscle cell binding proteins will generally have some low level of cross-reactivity with cells in tissues other than the desired smooth muscle cells. Such blocking can improve localization of the therapeutic conjugate or dosage form at the specific vascular site, e.g., by making more of the therapeutic conjugate available to the cells.
  • non-coupled vascular smooth muscle binding protein is administered from about 5 minutes to about 48 hours, most preferably from about 5 minutes to about 30 minutes, prior to administration of the therapeutic conjugate or dosage form.
  • the unlabeled specific "blocker" is a monovalent or bivalent form of an antibody (e.g., a whole antibody or an F(ab)' 2 , Fab. Fab', or Fv fragment of an antibody).
  • the monovalent form of the antibody has the advantage of minimizing displacement of the therapeutic conjugate or dosage form while maximizing blocking of the non-specific cross-reactive sites.
  • the non-coupled vascular smooth muscle cell binding protein is administered in an amount effective to blocking binding of a least a portion of the non-specific cross-reactive sites in a patient. The amount may vary according to such factors as the weight of the patient and the nature of the binding protein. In general, about 0.06 mg to 0.20 mg per kg body weight or more of the unlabeled specific blocker is administered to a human.
  • a second irrelevant vascular smooth muscle cell binding protein may optionally be administered to a patient prior to administration of the therapeutic conjugate or dosage form to reduce non-specific binding of the therapeutic conjugate or dosage form to tissues.
  • the irrelevant binding protein may be an antibody which does not bind to sites in the patient through antigen-specific binding, but instead binds in a non-specific manner, e.g., through Fc receptor binding reticuloendothelial cells, asialo-receptor binding, and by binding to ubiquitin-expressing cells.
  • the irrelevant "blocker” decreases non-specific binding of the therapeutic conjugate or dosage form and thus reduces side-effects, e.g., tissue toxicity, associated with the use of the therapeutic conjugate or dosage form.
  • the irrelevant "blocker” is advantageously administered from 5 minutes to
  • irrelevant "blockers” include antibodies that are nonreactive with human tissues and receptors or cellular and serum proteins prepared from animal sources that when tested are found not to bind in a specific manner (e.g., with a Ka ⁇ 10 3 M -1 ) to human cell membrane targets.
  • conjugates and dosage forms of the invention are not restricted in use for therapy following angioplasty; rather, the usefulness of the therapeutic conjugates and dosage forms will be proscribed by their ability to inhibit cellular activities of smooth muscle cells and pericytes in the vascular wall.
  • other aspects of the invention include therapeutic conjugates and dosage forms and protocols useful in early therapeutic intervention for reducing, delaying, or eliminating (and even reversing) atherosclerotic plaques and areas of vascular wall hypertrophy and/or hyperplasia.
  • Therapeutic conjugates and dosage forms of the invention also find utility for early intervention in pre-atherosclerotic conditions, e.g., they are useful in patients at a high risk of developing atherosclerosis or with signs of hypertension resulting from atherosclerotic changes in vessels or vessel stenosis due to hypertrophy of the vessel wall.
  • the stent preferably comprises a biodegradable coating or a porous or permeable non-biodegradable coating comprising the therapeutic agent.
  • a more preferred embodiment of the invention is a coated stent wherein the coating comprises a sustained-release dosage form of the therapeutic agent.
  • a biodegradable stent may also have the therapeutic agent impregnated therein, i.e., in the stent matrix.
  • the intravascular stent thus provides a mechanical means of providing an increase in luminal area of a vessel, in addition to that provided via the biological stenting action of the cytoskeletal inhibitor, such as cytochalasin B or taxol, releasably embedded therein.
  • the cytoskeletal inhibitor such as cytochalasin B or taxol
  • intravascular stents comprising a therapeutic agent which is an inhibitor of smooth muscle cell proliferation
  • a stent which further comprises a cytochalasin to inhibit the proliferation and migration of pericytes, which can transform into smooth muscle cells and contribute to intimal thickening, is also an embodiment of the invention.
  • This inhibition of intimal smooth muscle cells and stroma produced by the smooth muscle and pericytes can allow more rapid and complete re-endothelization following the intraventional placement of the vascular stent.
  • the increased rate of re-endothelization and stabilization of the vessel wall following stent placement can reduce the loss of luminal area and decreased blood flow which is the primary cause of vascular stent failures.
  • the biodegradable microparticles containing the therapeutic agent are from about 1 to 50 microns.
  • the microparticles would biodegrade over a period of 30 to 120 days, releasing into the tunica media and intima a sustained cellular concentration of approximately from about 0.05 ⁇ g/ml to about 0.25 ⁇ g/ml of cytochalasin B into the cytosol, thus providing the diffusion of therapeutic levels of cytochalasin B without toxicity to cells adjacent to the stent/vessel wall interface.
  • the therapeutic conjugates and dosage forms of the invention may also be used in therapeutic modalities for enhancing the regrowth of endothelial cells in injured vascular tissues and in many kinds of wound sites including epithelial wounds.
  • the therapeutic conjugates and dosage forms of the invention find utility in inhibiting the migration and/or proliferation of smooth muscle cells or pericytes. Smooth muscle cells and pericytes have been implicated in the production of factors in vitro that inhibit endothelial cell proliferation, and their proliferation can also result in a physical barrier to establishing a continuous endothelium.
  • the therapeutic conjugates and dosage forms of the invention find utility in promoting neo-angiogenesis and increased re-endothelialization, e.g.. during wound healing, vessel grafts and following vascular surgery.
  • the dosage forms may also release therapeutic modalities that stimulate or speed up re-endothelialization of the damaged vessel wall.
  • An exemplary therapeutic agent for this purpose is vascular permeability factor.
  • Still other aspects of the invention relate to therapeutic modalities for enhancing wound healing in a vascular site and improving the structural and elastic properties of healed vascular tissues.
  • these therapeutic modalities using the therapeutic conjugate or dosage form of the invention i.e.. to inhibit the migration and proliferation of smooth muscle cells or pericytes in a vessel wall
  • the strength and quality of healing of the vessel wall are improved.
  • Smooth muscle cells in the vascular wound site contribute to the normal process of contraction of the wound site which promotes wound healing. It is presently believed that migration and proliferation of smooth muscle cells and matrix secretion by transformed smooth muscle cells may detract from this normal process and impair the long-term structural and elastic qualities of the healed vessel.
  • other aspects of the invention provide for therapeutic conjugates and dosage forms that inhibit smooth muscle and pericyte proliferation and migration as well as morphological transformation, and improve the quality of the healed vasculature.
  • one embodiment of the present invention comprises the in vivo or ex vivo infusion of a solution of a therapeutic agent such as cytochalasin B into the walls of isolated vessels (arteries or veins) to be used for vascular grafts.
  • a therapeutic agent such as cytochalasin B
  • the vessel that is to serve as the graft is excised or isolated and subsequently distended by an infusion of a solution of a therapeutic agent.
  • the infusion is accomplished by a pressure infusion at a pressure of about 0.2 to 1 atmosphere for a time period of from about 2 to about 4 minutes. This infusion regime will result in the penetration of an efficacious dose of the therapeutic agent to the smooth muscle cells of the vessel wall.
  • the therapeutic agent will be at a concentration of from about 0.1 ⁇ g/ml to about 0.8 ⁇ g/ml of infusate.
  • the therapeutic agent will be a cytochalasin, and most preferably, the therapeutic agent employed will be cytochalasin B, or a functionally equivalent analogue thereof.
  • cytochalasin B in this manner will prevent the constriction or spasm that frequently occurs after vascular grafts are anastomosed to both their proximal and distal locations, that can lead to impaired function, if not total failure, of vascular grafts.
  • the vessel stenting produced by cytochalasin b should decrease the incidence of spasms, which can occur from a few days to several months following the graft procedure.
  • the present invention also provides a combination therapeutic method involving a cytocidal therapeutic conjugate and a cytostatic therapeutic agent.
  • the cytocidal conjugate includes a binding partner (such as a protein or peptide) capable of specifically localizing to vascular smooth muscle cells and an active agent capable of killing such cells.
  • the cytocidal conjugate is administered, preferably intravenously or through any other convenient route therefor, localizes to the target smooth muscle cells, and destroys proliferating cells involved in stenotic or restenotic events. This cellular destruction causes the release of mitogens and other metabolic events, which events generally lead, in turn, to vascular smooth muscle cell proliferation.
  • the present invention also provides methods for the treatment of cancer and immune system-mediated diseases through local administration of a targeted particulate dosage form.
  • the particulate dosage form is, for example, administered locally into primary and/or metastatic foci of cancerous target cells.
  • Local administration is preferably conducted using an infusion needle or intraluminal administration route, infusing the particulate dosage form in the intercellular region of the tumor tissue or in luminal fluid surrounding the tumor cells.
  • Primary foci introduction is preferably conducted with respect to target cells that are generally situated in confined areas within a mammal, e.g., ovarian carcinomas located in the abdominal cavity.
  • the dosage form of the present invention binds to the target cell population and, optionally, is internalized therein for release of the therapeutic agent over time.
  • Local administration of dosage forms of the present invention to such primary foci results in a localized effect on such target cells, with limited exposure of other sensitive organs, e.g., the bone marrow and kidneys, to the therapeutic agent.
  • the administered microparticles and larger nanoparticles are primarily bound to the target cells situated near the infusion site and are, optionally, internalized for release of the therapeutic agent, thereby generating a marked and localized effect on the target cells immediately surrounding the infusion site.
  • smaller nanoparticles follow interstitial fluid flow or lymphatic drainage channels and bind to target cells that are distal to the infusion site and undergoing lymphatic metastasis.
  • the targeted dosage forms of this embodiment of the present invention can be used in combination with more commonly employed immunoconjugate therapy.
  • the immunoconjugate achieves a systemic effect within the limits of systemic toxicity, while the dosage form of the present invention delivers a concentrated and sustained dose of therapeutic agent to the primary and metastatic foci, which often receive an inadequate therapeutic dose from such "systemic" immunoconjugate administration alone, and avoids or minimizes systemic toxic effects.
  • target cell population can be accessed by local
  • the dosage forms of the present invention are utilized to control immune system-mediated diseases.
  • diseases are arthritis, sprue, uveitis, endophthalmitis, keratitis and the like.
  • the target cell populations implicated in these embodiments of the present invention are confined to a body cavity or space, such as joint capsules, pleural and abdominal cavity, eye and sub-conjunctival space, respectively.
  • Local administration is preferably conducted using an infusion needle for a intrapleural, intraperitoneal, intraocular or sub-conjunctival administration route.
  • This embodiment of the present invention provides a more intense, localized modulation of immune system cells with minimal effect on the systemic immune system cells.
  • the systemic cells of the immune system are simultaneously treatable with a chemotherapeutic agent conjugated to a binding protein or peptide.
  • a conjugate preferably penetrates from the vascular lumen into target immune system cells.
  • the local particulate dosage form administration may also localize to normal tissues that have been stimulated to proliferate, thereby reducing or eliminating such pathological (i.e., hyperactive) conditions.
  • An example of this embodiment of the present invention involves intraocular administration of a particulate dosage form coated with a binding protein or peptide that localizes to pericytes and smooth muscle cells of neovascularizing tissue. Proliferation of these pericytes causes degenerative eye disease.
  • Preferred dosage forms of the present invention release compounds capable of suppressing the pathological proliferation of the target cell population.
  • the preferred dosage forms can also release compounds that increase vessel lumen area and blood flow, reducing the pathological alterations produced by this reduced blood supply.
  • Still another aspect of the present invention relates to therapeutic modalities for maintaining an expanded luminal volume following angioplasty or other vessel trauma.
  • a therapeutic agent capable of inhibiting the ability of vascular smooth muscle cells to contract.
  • agents useful in the practice of this aspect of the present invention are those capable of causing a traumatized artery to lose vascular tone, such that normal vascular hydrostatic pressure (i.e., blood pressure) expands the flaccid vessel to or near to its maximal physiological diameter.
  • Loss of vascular tone may be caused by agents that interfere with the formation or function of contractile proteins (e.g., actin, myosin, tropomyosin, caldesmon, calponin or the like). This interference can occur directly or indirectly through, for example, inhibition of calcium modulation, phosphorylation or other metabolic pathways implicated in contraction of vascular smooth muscle cells.
  • Inhibition of cellular contraction may operate through two mechanisms to reduce the degree of vascular stenosis.
  • Second, inhibition of cellular contraction causes the smooth muscle wall to relax and dilate under normal vascular hydrostatic pressure (i.e., blood pressure).
  • the clinician can eliminate over-inflation and its resultant trauma shock as a means to diminish or delay the vessel spasm or elastic recoil. Reduction or elimination of over-inflation decreases trauma to the muscular wall of the vessel, thereby reducing the determinants of smooth muscle cell proliferation in the intima and, therefore, reducing the incidence or severity of restenosis.
  • Cytochalasins are exemplary therapeutic agents capable of generating a biological stenting effect on vascular smooth muscle cells. Cytochalasins are thought to inhibit both migration and contraction of vascular smooth muscle cells by interacting with actin. The cytochalasins interact with the ends of filamentous actin to inhibit the elongation of the actin filaments. Low doses of cytochalasins (e.g., cytochalasin B) also disrupt microfilament networks of actin. In vitro data indicate that after vascular smooth muscle cells clear cytochalasin B, the cells regenerate enough polymerized actin to resume migration within about 24 hours. In vivo assessments reveal that vascular smooth muscle cells regain vascular tone within 2 to 4 days. It is during this recuperative period that the lumen diameter fixation and biological stenting effect occurs.
  • cytochalasins are thought to inhibit both migration and contraction of vascular smooth muscle cells by interacting with actin. The cytochalasins interact with the ends of filamentous
  • the therapeutic agent may be targeted, but is preferably administered directly to the traumatized vessel following the angioplasty or other traumatic event.
  • the biological stenting effect of cytochalasin B is achievable using a single infusion of the therapeutic agent into the traumatized region of the vessel wall at a dose concentration ranging from about 0.1 microgram/ml to about 1.0 micrograms/ml.
  • vascular smooth muscle cell migration has been demonstrated in the same dose range (Example 1 1 ); however, a sustained exposure of the vessel to the therapeutic agent is preferable in order to maximize these anti-migratory effects. If the vascular smooth muscle cells cannot migrate into the intima, they cannot proliferate there. Should vascular smooth muscle cells migrate to the intima, a subsequently administered anti-proliferative sustained release dosage form inhibits the intimal proliferation.
  • the sustained release dosage form of the present invention incorporating a cytochalasin or other anti-proliferative therapeutic agent, can be administered in combination with a free cytochalasin therapeutic agent. In this manner, the biological stenting effect, as well as an anti-proliferative or anti-migratory effect, can be achieved in a single administration protocol.
  • Agents useful in the protocols of the present invention are identifiable. for example, in accordance with the following procedures.
  • a potential agent for free agent (i.e., non-targeted) administration exhibits one or more of the following characteristics:
  • angioplasty e.g., PTCA, percutaneous transluminal angioplasty (PTA) or the like
  • PTA percutaneous transluminal angioplasty
  • atheroectomy e.g., rotoblater, laser and the like
  • coronary artery bypass procedures e.g., coronary artery bypass procedures or the like
  • vascular disease e.g., atherosclerosis, eye diseases secondary to vascular stenosis or atrophy, cerebral vascular stenotic diseases or the like
  • a therapeutic agent employed herein will have all four properties; however, the first and third are more important than the second and fourth for practice of the present invention.
  • Cytochalasin B for example, was evaluated to determine suitability for use in free therapeutic agent protocols. The biological stenting effect of cytochalasin B is achievable using a single infusion of the therapeutic agent into the traumatized region of the vessel wall at a dose concentration ranging from about 0.1 microgram/ml to about 1.0 micrograms/ml.
  • An agent useful in the sustained release embodiments of the present invention exhibits one or more of the following characteristics:
  • angioplasty e.g., PTCA, percutaneous transluminal angioplasty (PTA) or the like
  • PTA percutaneous transluminal angioplasty
  • atheroectomy e.g., rotoblater, laser and the like
  • coronary artery bypass procedures e.g., coronary artery bypass procedures or the like
  • vascular disease e.g., atherosclerosis, eye diseases secondary to vascular stenosis or atrophy, cerebral vascular stenotic diseases or the like
  • target cell proliferation e.g., following 5 minute and 24 hour exposure to the agent, in vitro vascular smooth muscle tissue cultures demonstrate a level of inhibition of 3 H-thymidine uptake and. preferably, display relatively less inhibition of 3 H-leucine uptake;
  • (v) is cytostatic.
  • the agent Upon identification of a therapeutic agent exhibiting one or more of the preceding attributes, the agent is subjected to a second testing protocol that involves longer exposure of vascular smooth muscle cells to the therapeutic agent.
  • FIGURE 3 shows diagrammatically the second reaction scheme for synthesis of Roridin A hemisuccinyl succinimidate (RA-HS-NHS) through a five step process with reagents: t-butyl dimethyl silyl chloride (TBMS-C1) and imidazole in dimethylformamide (DMF) at room temperature (RT); acetic anhydride, triethylamine (TEA), and diethylaminopyridine in dichloromethane (CH 2 Cl 2 ) at RT; succinic anhydride, triethylamine (TEA) and
  • the binding of the NR-AN-01 MAb to the A375 cells was quantified by washing to remove unbound antibody, adding HRP-conjugated goat anti-mouse IgG to react with the cell-bound MAb, washing to remove unbound goat second antibody, and adding 2,2'-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS) substrate for peroxidase. Color development was monitored after 30 minutes at both 415 nm and 490 nm (ABS415.490).
  • the HT29 and Ml 4 cells were seeded in 96-well microtiter plates at a concentration of 5.0 ⁇ 10 3 cells/well, and the BO54 cells were seeded at 2.5 ⁇ 10 3 cells/well.
  • Serial two-fold dilutions of free Roridin A and 2'RA-HS-NR-AN-01 i.e., Roridin A coupled through a hemisuccinate (HS) coupling agent at the 2' position to NR-AN-01
  • HS hemisuccinate
  • RA-conjugate concentration of >10 ng/ml.
  • PE Pseudomonas exotoxin
  • FIGURE 6B graphically depicts the results of in vitro studies conducted in manner similar to those discussed above in regard to FIGURE 6A. but designed to compare the effects obtained with free PE (PE) to those obtained above, i.e., in FIGURE 6A. with PE-NR-AN-01.
  • PE free PE
  • FIGURE 6B graphically depicts the results of in vitro studies conducted in manner similar to those discussed above in regard to FIGURE 6A. but designed to compare the effects obtained with free PE (PE) to those obtained above, i.e., in FIGURE 6A. with PE-NR-AN-01.
  • the cells, culture conditions, calculations, and presentation of the results are the same as in FIGURE 6A, above.
  • FIGURE 7A graphically depicts the results of in vitro studies conducted to investigate the effects on control HT29 marker-negative cells of a 5 minute exposure to different concentrations of Roridin A (Free RA; open squares, FIGURE 7A), or 2'RA-NR-AN-01 (2'RA-NRAN01 ; closed squares, FIGURE 7A), or 13'RA-NR-AN-01 (13'RA-NRAN01 ; closed triangles, FIGURE 7A) conjugates.
  • Roridin A Free RA; open squares, FIGURE 7A
  • 2'RA-NR-AN-01 (2'RA-NRAN01 ; closed squares, FIGURE 7A
  • 13'RA-NR-AN-01 13'RA-NRAN01 ; closed triangles, FIGURE 7A
  • the concentrations of Free RA, 2'RA-NR-AN-01 or 13'NR-AN-01 are expressed as the calculated concentration of Roridin A in the assay (in ⁇ g/ml plotted on a log scale), i.e., rather than the total ⁇ g/ml of NR-AN-01 protein, so that direct comparisons of the results can be made.
  • the cells were treated for 5 minutes, washed, and then returned to culture for 4 hours, during which time cellular protein synthesis was evaluated by adding 0.5 mCi/ml of 3 H-leucine to the culture medium. At the end of the 4 hour period, cellular proteins were collected and radioactivity was determined. The results are expressed as the percentage of the radioactivity recorded in a control (non-treated) HT29 cell culture (i.e., %control).
  • FIGURE 7B graphically depicts the results of in vitro studies investigating the effects on control HT29 marker-negative cells of a 5 minute expose to different concentrations of Free RA (open squares,FIGURE 7B), 2'RA-NRAN01 ( closed squares, FIGURE 7B), or 13'RA-NRAN01 ( closed triangles, FIGURE 7B), as described above in regard to FIGURE 7A, but in the present experiments the cells were incubated for a 16-18 hour recovery period (i.e., overnight; o/n) prior to testing protein synthesis in a four hour 3 H-leucine protein synthesis assay. The results are presented in a manner similar to those above in FIGURE 7A.
  • FIGURE 7D graphically depicts the results of in vitro studies investigating the effects on A375 m/ml marker-positive cells of a 5 minute exposure to different concentrations of Free RA (open squares ,FIGURE 7D), 2'RA-NRAN01 (closed squares, FIGURE 7D), 13'RA-NRAN01 (closed triangles, FIGURE 7D), as described above in regard to FIGURE 7B.
  • the A375 cells were incubated for 5 minutes in the test agent, washed, and then returned to culture for a 16-18 hour recovery period (i.e., overnight; o/n Recovery), after which time protein synthesis was evaluated during a 4 hour 3 H-leucine protein synthesis assay.
  • the results of the experiments are plotted in a manner similar to those described above in regard to FIGURE 7A.
  • FIGURE 7A and FIGURE 7B, above that its effect on the cells is manifest even after a 16-18 hour recovery period.
  • the specific effects of the RA-NR-AN-01 conjugates on target cells during a "pulse" treatment appear to be a property of the NR-AN-01 binding protein.
  • MTT assays were conducted at 48 hours following a 5-minute exposure of target and non-target cells to RA or RA-NR-AN-01 conjugates.
  • Target cells in these studies included BO54 and A375, and non-target cells included HT29 cells.
  • Sterile 96 well microtiter plates were seeded with 2500 cells/well, wrapped in aluminum foil and incubated in a humidified chamber containing 5% CO 2 /95% air for 16-18 hours.
  • the plates were covered and incubated at 37°C for 4 hours and then the reaction was developed as described above (see EXAMPLE 4, above).
  • the dark blue solubilized formazan reaction product was developed at room temperature after a 16-18 hour incubation.
  • the samples were quantified using an ELISA microtiter plate reader at an absorbance of 570 nm.
  • FIGURE 8A graphically depicts the results of in vitro studies investigating the effects on BO54 marker-positive smooth muscle cells of a 5 minute exposure to different concentrations of Roridin A (open squares, FIGURE 8A), 2'RA-NR-AN-01 (NRAN01-2'RA; closed diamonds, FIGURE 8A), or 13' RA-NR-AN-01 (NRAN01-13'RA; closed squares, FIGURE 8A).
  • the experiments were conducted in a manner similar to those described above in regard to FIGURE 7B, but metabolic activity was assayed by MTT assay, i.e., rather than protein synthesis as in FIGURE 7B, and cells were also given 48 hours to recover (rather than 24 hours, as in FIGURE 7B).
  • the results of the experiments are plotted in a manner similar to those described (above) in regard to FIGURE 7A .
  • FIGURE 8B graphically depicts the results of in vitro studies investigating the effects on A375 m/m marker-positive cells of a 5 minute exposure to different concentrations of Roridin A (open squares, FIGURE 8B), 2'RA-NR-AN-01 (NRAN01-2'RA; closed diamonds, FIGURE 8B), 13'RA-NR-AN-01 (NRAN01-13'RA; closed squares, FIGURE 8B).
  • the experiments were conducted (and the results plotted) in a manner similar to those described above in regard to FIGURE 8A.
  • FIGURE 8C graphically depicts the results of in vitro studies investigating the effects on HT29 marker-negative cells of a 5 minute exposure to different concentrations of Roridin A (open squares, FIGURE 8C), 2'RA-NR-AN-01 (NRAN01 -2'RA; closed diamonds, FIGURE 8C), 13'RA-NR-AN-01 (NRAN01-13'RA; closed squares, FIGURE 8C).
  • the experiments were conducted (and the results plotted) in a manner similar to those described above in regard to FIGURE 8A.
  • FIGURES 8A-8C show slight differences between the different RA-NR-AN-01 conjugates at the highest doses, but at the lower doses the 2' and 13'RA-NR-AN-01 did not significantly inhibit target cell (i.e., BO54 and A375) or non-target cell (i.e., HT29) metabolic activity over the long-term (i.e., 48 hours).
  • target cell i.e., BO54 and A375
  • non-target cell i.e., HT29
  • the therapeutic conjugates of the invention are useful for inhibiting stenosis following vascular trauma or disease.
  • vascular trauma that is induced during angioplasty is treated during the surgical procedure by removing the catheter used to perform the angioplasty, and inserting a balloon infusion catheter into the vessel.
  • the infusion catheter is positioned with the instillation port (or, alternatively, a permeable membrane region) in the traumatized area of the vessel, and then pressure is applied to introduce the therapeutic conjugate.
  • an infusion catheter with two balloons may be used, and when one balloon is inflated on either side of the trauma site a fluid space is created that can be filled with a suitable infusion fluid containing the therapeutic conjugate.
  • HRP horseradish peroxidase
  • the coronary and femoral arteries of domestic pigs or non-human primates were infused with NR-AN-01 for 45 seconds to 3 minutes at multiple pressures in the range of about
  • NR-AN-01 target antigen with man.
  • NR-AN-01 could bind in a specific manner to its target antigen in vivo
  • the coronary and femoral arteries of domestic pigs were infused with therapeutic conjugates using an infusion catheter, the infusion sites were flushed with sterile saline, the surgical sites were then closed, and the animals were maintained for an additional 3-5 days.
  • the vascular infusion sites were excised and prepared for immunohistology, once again using goat anti-mouse IgG to identify
  • NR-AN-01 was identified in the vessel wall of swine coronary and femoral arteries 3-5 days after surgery, and the NR-AN-01 appeared to be associated only with vascular smooth muscle cells. These findings suggest that NR-AN-01 is capable of specifically binding to its target antigen in vivo.
  • Intimal smooth muscle proliferation that follows balloon catheter-induced trauma is a good model to evaluate the therapeutic efficacy of conjugates for inhibiting smooth muscle cell activity in vivo in response to vascular trauma, including restenosis following angioplasty.
  • Domestic pigs were used to study the effects of NR-AN-01 (i.e., termed vascular smooth muscle binding protein or simply VSMBP in these studies; and therapeutic conjugates with Roridin A are termed VSMBP - RA).
  • VSMBP - RA vascular smooth muscle binding protein
  • test protocol was conducted in domestic pigs with NR-AN-01-Roridin A conjugates.
  • Localized administration of the test conjugates i.e., through a catheter into a region of traumatized vessel confined by temporary slip ligatures, was designed to reduce systemic toxicity while providing a high level of exposure for the target smooth muscle cells.
  • This intra-artery route of administration in animal model studies simulates the proposed route in human coronary arteries.
  • the test protocol was designed as an initial in vivo screening of intra-arteriolar, site specific, catheter
  • vascular smooth muscle binding protein VSMBP conjugates.
  • Toxicity of free drug was also evaluated, i.e., for pathobiological effects on arteriolar smooth muscle cells.
  • the therapeutically effective dosage of the Roridin A-NR-AN-01 conjugate was determined by in vitro studies, and the proper intra-arteriolar administration pressure was determined by administering free MAb and MAb conjugates to animals, as described above in Example 7.
  • Test conjugates and control compounds were administered as a single intra-artery infusion at the site of endothelial denuding and trauma induced by a balloon catheter. Both the carotid and femoral arteries were abraded over 1 cm to 2 cm of endothelium by intraluminal passage of a 23 cm, size 3 (femoral) and size 4 (carotid) Uresil Vascu-Flo® silicone occlusion balloon catheter (Uresil Technology Center, Skokie, IL), sufficiently distended with saline to generate slight resistance. This technique produced slight distension of the artery.
  • proximal and distal slip ligatures were placed near the ends of the abraded region, and a size 8 French, Infant Feeding Catheter (Cutter-Resiflex. Berkeley, CA) attached to an Inflation Pro® (USCI, C.R. Bard, Inc., Billerica, MA) pressure syringe was used to administer the test conjugates and control compounds directly to the denuded segment at a pressure of three atmospheres for three minutes.
  • the slip ligatures were removed after the three minute exposure period and arterial blood flow was re-established.
  • branches of the femoral or carotid arteries were ligated with 00 silk suture as required to attain pressurized infusion in the treated region.
  • the largest distal branch of the femoral artery (the saphenous artery) was incised and used as an entry site for the catheters which were then passed into the main femoral artery. Following this catheterization procedure in the main femoral artery, the secondary branch was ligated. In these cases, ligation or incision was used to allow entry of the catheters and the opening was then closed with 3 to 4 sutures of
  • the pigs were kept in 3 ⁇ 5 foot indoor runs with cement floors during the quarantine and surgical recovery periods. They were then transferred to indoor/outdoor pens for the remainder of the five week healing period prior to collection of tissues for histology.
  • the animals recovered normally from surgery with no evidence of hemorrhage or inflammation at the surgical sites. All six animals were examined 5 to 6 days after treatment with a doppler stethoscope, and all arteries in each of the animals were patent. Post treatment all animals had normal appetite, activity and weight gain.
  • heparinized i.v. 2 ml sodium heparin, 1000 units/ml
  • euthanized by i.v. pentobarbital Both the right and left carotid and femoral arteries were removed with normal vessel included both proximal and distal to the treated segment. The arteries were measured and the location of ligatures and gross abnormalities noted. The arteries were transected at 2 mm intervals and arranged in order in cryomolds with O.C.T. (optimum cutting temperature) compound (Tissue Tek®, Miles Laboratories Inc., Elkhart, IN) and frozen in liquid nitrogen. The blocks were sectioned at 5 microns and stained with
  • Sections were also used for immunohistological staining of vascular smooth muscle.
  • FIGURES 9A and 9B are identical to FIGURES 9A and 9B.
  • FIGURE 9A show (at 160x magnification) a cross-sectional of an untreated artery 5 weeks after angioplasty.
  • Dominant histological features of the artery include displacement of the endothelium (see #1 in FIGURE 9A) away from the internal elastic lamina (see #2, FIGURE 9A), apparently due to intimal smooth muscle proliferation (see #3, FIGURE 9A).
  • FIGURE 9B show (at 160x magnification) a cross-section of a treated artery 5 weeks after angioplasty and infusion of the RA-NR-AN-01 therapeutic conjugate.
  • the vessel in this section was subjected to greater mechanical stresses than the vessel shown in FIGURE 9A, with multiple sites where the external elastic membrane was ruptured and associated proliferation of smooth muscle cells in the outer layers of the media was observed (i.e., see #4 in FIGURE 9B).
  • Treatment with therapeutic conjugate inhibited intimal hypertrophy, as evidenced by the lack of displacement of the endothelium (see #1 , FIGURE 9B) from the internal elastic lamina (see #2, FIGURE 9B).
  • this inhibitory effect on intimal smooth muscle cells was accomplished without inhibiting hypertrophy of medial smooth muscle cells in the areas where the external elastic membrane was ruptured (see #4, FIGURE 9B).
  • FIGURE 9B therapeutic conjugate administered at the site following angioplasty resulted in approximately 95% inhibition of the smooth muscle hypertrophy that restricted the lumen of the untreated vessel
  • FIG. 9A Significantly, the therapeutic conjugate exerted its effects on the smooth muscle cells migrating from the medial smooth muscle layers into the intima, without affecting either endothelium, or producing any signs of necrosis (i.e., cell death) in the smooth muscle cells in the medial layers of the arterial wall. Studies also failed to show any histological signs of
  • H-leucine uptake Vascular smooth muscle cells at 40,000 cells/ml were seeded in sterile 24 well plates at 1 ml/well. The plates were incubated overnight at 37°C, 5% CO 2 , 95% air in a humidified atmosphere (saturation). Log dilutions of the therapeutic agent of interest were incubated with the vascular smooth muscle cells for 5 minutes or 24 hours. Samples of the therapeutic agents were diluted in DMEM :F- 12 medium (Whittaker Bioproducts, Walkersville, Maryland) with 5% fetal bovine serum (FBS, Gibco BRL, Gaithersburg, MD) and 5% Serum Plus® (JRH Biosciences, Lenexa, KS).
  • micrograms/ml transferrin (5 micrograms/ml) and sodium selenite (5 nanograms/ml), available from Sigma Chemical, St. Louis, Missouri) was added. Cells were incubated in this medium for 24 hours. For a 5 minute therapeutic agent exposure, log dilutions of the therapeutic agent were incubated with the cells in complete medium. After 5 minutes and medium aspiration, 1 ml/well of 1.0 microcurie/ml 3 H-thymidine dispersed in complete medium was added. The 24 hour exposure involved incubation of the cells with 1 ml/well of 1.0 microcurie/ml of 3 H-thymidine dispersed in complete medium and log dilutions of the therapeutic agent being tested.
  • Vascular smooth muscle cells were seeded at 4.0 ⁇ 10 4 cells/ml medium/well on a commercially prepared four well slide (Nunc. Inc., Naperville. Illinois).
  • the therapeutic agent medium was aspirated from each well, including the untreated well.
  • One milliliter of fresh medium was then added to replace the aspirated medium.
  • Re-incubation followed until each of the incremented evaluation points were achieved.
  • the medium was aspirated and subsequently replaced with 1 ml of 10% neutral buffered formalin for one hour to allow for proper fixation.
  • These fixed slides were stained by hematoxylin (nuclear) and cosin (cytoplasmic) for morphologic evaluation and grading.
  • results of the 24 hour 3 H-leucine protein inhibition assay and the 24 hour 3 H-thymidine DNA synthesis inhibition assay are shown in Figs. 10A-10D for suramin, staurosporin. nitroglycerin and cytochalasin B. respectively. All of the tested compounds showed an available therapeutic range (area under the curve of 3 H-leucine assay is greater than that resulting from the 3 H-thymidine assay), indicating usefulness in the practice of sustained release dosage form embodiments of the present invention. More specifically, the compounds inhibited the ability of vascular smooth muscle cells to undergo DNA synthesis in the presence of 5% FBS to a greater extent than they inhibited protein synthesis of vascular smooth muscle cells.
  • the protein and DNA synthesis inhibitory effects of suramin, staurosporin, nitroglycerin and cytochalasin B during a 5 minute and 24 hour pulsed exposure are shown in Figure 10 A-D, respectively.
  • vascular smooth muscle cells The ability of vascular smooth muscle cells to bind and internalize particles coated with binding protein or peptide was demonstrated with monoclonal antibody (NR-AN-01 ) coated gold beads both in vitro and in vivo.
  • the cells were exposed to the beads as monolayer and cell suspension cultures, and were examined at six time points (i.e.. 1 minute, 5 minutes, 15 minutes, 30 minutes. 60 minutes and 24 hours) for binding and internalization by electron microscopy.
  • Table 3 shows the results of the experimentation, indicating that the binding to the cell surface is specific.
  • NR-AN-01 internalized by macro and micro phagocytosis. When the cells were maintained in a cell suspension, non-specific internalization was minimal or absent. Non-specific adherence of gold beads devoid of NR-AN-01 to surface mucin produced by HT29 cells was observed, resulting in modest non-specific internalization thereof. Vascular smooth muscle cell uptake of NR-AN-01 targeted gold beads was highly specific in cell suspension cultures.
  • FIGURE 1 1 shows a tangential section parallel to the inner surface of a smooth muscle cell characterized by numerous endocytic vesicles, several of which contain antibody coated gold beads in the process of being internalized by the cell. These endocytic vesicles with particles attached to cell surface antigens were stimulated to fuse with lysosomes at a higher than expected rate for normal cell surface membrane recycling. The resultant marked
  • NR-AN-01 coated gold beads were infused via intravascular catheter, open ended with treated area occluded proximally and distally with slip ligatures, at 3 atm pressure applied for 3 minutes into the wall of a pig femoral artery
  • FIGURE 13 shows particulate concentration in the lysosomes in vivo at one week following bead
  • Vascular smooth muscle cells at 40.000-50,000 cells/ml were seeded and processed as described in Example 8. "5 minute exposure; 3 H-leucine uptake.”
  • Log dilutions of staurosporin 200 ng/ml, 20 ng/ml, 2 ng/ml, 0.2 ng/ml and 0.02 ng/ml
  • cytochalasin B log dilutions at 20 ⁇ g/ml, 2.0 ⁇ g/ml, 0.2 ⁇ g/ml, 0.02 ⁇ g/ml and 0.002 ⁇ g/ml were dispersed in complete medium. Complete medium was then added to the control wells.
  • Protein Synthesis Assay vascular smooth muscle (BO54) cells at 20,000 cells/ml were seeded in sterile 24 well plates and incubated in complete medium (1 ml/well) overnight at 37°C, 5% CO 2 , 95% air in a humidified atmosphere (saturation). Log dilutions of staurosporin (100 ng/ml, 10 ng/ml, 1 ng/ml, 0.1 ng/ml and 0.01 ng/ml) were dispersed sequentially in the two media, as described below.
  • cytochalasin B log dilutions at 10 ⁇ g/ml, 1.0 ⁇ g/ml, 0.1 ⁇ g/ml, 0.01 ⁇ g/ml and 0.001 ⁇ g/ml were dispersed sequentially in the two media, as described below:
  • each therapeutic agent was diluted in Medium (2), as noted above.
  • Medium (1) was aspirated from the wells, and aliquots of therapeutic agent dilutions in Medium (2) were added in
  • each therapeutic agent was diluted in Medium (1), as noted above.
  • Medium (1) was aspirated from the wells, and aliquots of therapeutic agent dilutions in Medium (1 ) were added in quadruplicate to the appropriate wells.
  • Medium (1 ) was then added to the control wells. The medium was changed every 24 hours, and fresh therapeutic agent was added to the test wells.
  • each therapeutic agent was diluted in Medium (2), as noted above.
  • Medium (1 ) was aspirated from the wells, and aliquots of therapeutic agent dilutions in Medium (2) were added in quadruplicate to the appropriate wells.
  • Medium (2) was then added to the control wells.
  • test agents in 3 H-leucine were incubated overnight at 37°C, 5% CO 2 in a humidified atmosphere.
  • the toxic effect of the therapeutic agents was then determined, as described in the 5 Minute
  • the minimum effective dose (MED) of each agent was determined as a percentage of the control that was treated with medium only; 50% of control values was chosen as the cytotoxicity benchmark.
  • MED minimum effective dose
  • staurosporin demonstrated an MED of 100 ng/ml in the protein synthesis assay and 1 ng/ml in the DNA assay.
  • the 24 hour MED for staurosporin was 10 ng/ml in the protein synthesis assay and 1 ng/ml in the DNA synthesis assay. Both assays gave an MED of 1 ng/ml for a 120 hour exposure of staurosporin.
  • cytochalasin B demonstrated an MED of 10 ⁇ g/ml in the protein synthesis assay as well as in the DNA assay.
  • the 24 hour MED for cytochalasin B was 1.0 ⁇ g/ml in the protein synthesis assay and 0.1 ⁇ g/ml in the DNA synthesis assay. Both assays gave an MED of approximately 0.1 ⁇ g/ml for a 120 hour exposure of staurosporin.
  • Cytochalasin C and cytochalasin D therapeutic agents were tested at 24 and 48 hour exposures using the same dilutions as described for cytochalasin B, above.
  • cytochalasin C demonstrated an MED of 1.0 ⁇ g/ml in the protein synthesis assay and an MED of 0.01 ⁇ g/ml in the DNA synthesis assay.
  • cytochalasin C demonstrated an MED of 0.1 ⁇ g/ml in the protein synthesis assay and 0.01 ⁇ g/ml in the DNA synthesis assay.
  • Cytochalasin D demonstrated an MED of 1.0 ⁇ g/ml in the 24 hour protein synthesis assay and an MED of 0.1 ⁇ g/ml in the 24 hr DNA synthesis assay.
  • a 48 hour exposure to cytochalasin D gave an MED ranging between 0.1 and 0.01 ⁇ g/ml in both the protein synthesis and DNA synthesis assays.
  • Vascular smooth muscle cells were derived from explants of baboon aortic smooth muscle, as described in Example 10. The cells were grown in flat bottom, six well tissue culture plates, which hold about 5 ml of medium. The vascular smooth muscle cells were plated at 200,000 cells/well and placed at 37°C in a humidified 5% CO 2 incubator for 18 hours. The wells were then scratched with a sterile portion of a single edge razor blade that was held by clamp or pliers and was brought aseptically into contact with the bottom of the well at a 90° angle. The cells from a small area along the scratch were removed by a sterile cotton tipped applicator while the blade was in contact with the bottom of the well. After incubation, the presence of cells in the "scratched" area is indicative of cell migration across the scratch line. A control incubation showed significant cellular migration, and serves as the standard against which the migration of cells exposed to the therapeutic agent is compared.
  • cytochalasin B Sigma Chemical Co.
  • DMSO dimethyl sulfoxide
  • a set Test agent exposure for 1 , 3, 6, 8 and 10 days only; and B set: Test agent exposure for 1, 3, 6, 8 and 10 days, followed by a seven day recovery time with control medium.
  • Both sets of plates were fixed (10% formalin in PBS) and stained (0.02% crystal violet) at the end of the timed exposures.
  • Test concentrations for cytochalasin B were 1, 0.1 and 0.01 ⁇ g/ml, and a negative medium control was included. Fresh medium and drug were supplied 3 times per week.
  • Table 4 shows the results of these experiments.
  • the migration results are expressed as "Grade in the Cleared Area of the Well / Grade in an Undisturbed Region of the Well.”
  • the toxicity values represent a grade for all cells in each well.
  • BRDU assay In vivo vascular smooth muscle proliferation was quantitated by measuring incorporation of the base analog 5-bromo-2'-deoxyuridine (BRDU. available from Sigma Chemical Co.) into DNA during cellular DNA synthesis and proliferation. BRDU incorporation was demonstrated histochemically using commercially available anti-BRDU monoclonal antibodies. The 1 hour pulse labeling permits assessment of the number of cells undergoing division during the pulse period.
  • BRDU base analog 5-bromo-2'-deoxyuridine
  • BRDU was then administered intravenously via the lateral ear vein.
  • Two ml of BRDU at a concentration of 50 mg/ml was administered to each 30-40 lb pig.
  • Test artery segments were then removed (a segment included normal vessel located proximally and, if possible, distally with respect to the treated artery segment).
  • the artery segments were transected at 2 mm intervals; arranged in order in cryomolds with O.C.T.
  • BRDU-labeled cell detection After BRDU (1 g BRDU diluted in 17 ml sterile water and 3 ml 1 N NaOH) pulse labeling and test artery segment removal and sectioning (as above), immunohistochemical staining with anti-BRDU monoclonal antibody provides a visual means of determining a mitotic index over a specified time period.
  • the immunohistochemical staining method was performed as follows:
  • test artery 5 ⁇ m sections of test artery were dehydrated in cold acetone (- 20°C) for 10 minutes;
  • Sections were mounted on glass microscope slides, and the slides were dried in a 37°C oven for 10 minutes;
  • Balloon traumatized pig arteries that had been treated with cytochalasin B displayed a larger luminal area at the 4 day and 3 week post-treatment time points, as compared to arteries treated with other test agents or controls.
  • Ten femoral arteries two arteries obtained from each of the 5 pigs that were treated according to the single dose protocol described in Example 7) were evaluated histologically. The maximal luminal area of each artery was measured and calculated from digitized microscopic images by a BQ System IV computerized morphometric analysis system (R & M Biometrics, Inc., Nashville, TN).
  • the luminal area of the traumatized and cytochalasin B-treated segments of the arteries were also compared to the luminal area of the normal, untreated region of the femoral artery proximal to the test area.
  • the results showed that the lumen area in the test region was approximately two times as large as the area of the normal control segment of the same artery.
  • the negative control agents, PBS and monoclonal antibody NR-AN-01 showed no increase or a slight decrease in lumen area as compared to the normal control segment of the same artery.
  • cytochalasin B dose response study was then conducted on 10 pigs, following the experimental protocol described in Example 7. Briefly, both arteries in each of 2 pigs were treated with one of the following doses of cytochalasin B: 0.0 ⁇ g/ml (i.e., PBS negative control); 0.01 ⁇ g/ml; 0.10 ⁇ g/ml; 1.0 ⁇ g/ml; and 10.0 ⁇ g/ml.
  • the agent was delivered by intraluminal catheter at 1 atm pressure for 3 min. and the arteries were evaluated 3 weeks later by the morphometric analysis system described above. The ratio of treated artery luminal area to proximal normal artery luminal area was determined as a percent change in treated vs. normal area.
  • Antibody-coated latex particles (a model of an antibody- coated, sustained release dosage form) may be obtained using the following aseptic technique:
  • Sizing Particle size homogeneity is assessed by laser anisotropy or, for particles larger than 1 ⁇ m, by microscopic examination.
  • Specific Binding Assessment Specific binding to smooth muscle cells is determined by histological examination of tissue or cell pellet microtome slices after incubation of protein/peptide conjugates with conjugated particles, with or without blocker protein/peptide included in the incubation mixture.
  • Preferred detection techniques include second antibody assays (i.e., anti-mouse Ig) or competition assays (i.e., radioscintigraphic detection in conjunction with radioisotopically labeled protein/peptide conjugates).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Organic Chemistry (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Vascular Medicine (AREA)
  • Surgery (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Biophysics (AREA)
  • Inorganic Chemistry (AREA)
  • Medical Informatics (AREA)
  • Cardiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Ceramic Engineering (AREA)
  • Dermatology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Neurosurgery (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
PCT/US1996/002125 1992-09-25 1996-02-15 Therapeutic inhibitor of vascular smooth muscle cells WO1996025176A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
AT96906490T ATE265233T1 (de) 1995-02-15 1996-02-15 Therapeutische inhibitoren der zellen der glatten gefässmuskulatur
AU49851/96A AU4985196A (en) 1995-02-15 1996-02-15 Therapeutic inhibitor of vascular smooth muscle cells
CA002212537A CA2212537C (en) 1995-02-15 1996-02-15 Therapeutic inhibitor of vascular smooth muscle cells
JP8525163A JPH11500635A (ja) 1995-02-15 1996-02-15 血管平滑筋細胞の治療インヒビター
EP96906490A EP0809515B1 (en) 1995-02-15 1996-02-15 Therapeutic inhibitor of vascular smooth muscle cells
DE69632310T DE69632310T2 (de) 1995-02-15 1996-02-15 Therapeutische inhibitoren der zellen der glatten gefässmuskulatur
US08/829,685 US5981568A (en) 1993-01-28 1997-03-31 Therapeutic inhibitor of vascular smooth muscle cells
US08/829,991 US6306421B1 (en) 1992-09-25 1997-03-31 Therapeutic inhibitor of vascular smooth muscle cells
US09/361,194 US6358989B1 (en) 1993-05-13 1999-07-26 Therapeutic inhibitor of vascular smooth muscle cells
US09/896,208 US6491938B2 (en) 1993-05-13 2001-06-29 Therapeutic inhibitor of vascular smooth muscle cells
US09/995,490 US6569441B2 (en) 1993-01-28 2001-11-27 Therapeutic inhibitor of vascular smooth muscle cells
US10/024,885 US6663881B2 (en) 1993-01-28 2001-12-18 Therapeutic inhibitor of vascular smooth muscle cells
US11/650,059 US8158670B2 (en) 1995-02-15 2007-01-04 Therapeutic inhibitor of vascular smooth muscle cells
US13/446,760 US20120195951A1 (en) 1995-02-15 2012-04-13 Therapeutic Inhibitor of Vascular Smooth Muscle Cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/389,712 1995-02-15
US08/389,712 US6515009B1 (en) 1991-09-27 1995-02-15 Therapeutic inhibitor of vascular smooth muscle cells

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US08/389,712 Continuation-In-Part US6515009B1 (en) 1991-09-27 1995-02-15 Therapeutic inhibitor of vascular smooth muscle cells
US08/450,793 Continuation-In-Part US5811447A (en) 1991-09-27 1995-05-25 Therapeutic inhibitor of vascular smooth muscle cells

Related Child Applications (8)

Application Number Title Priority Date Filing Date
PCT/US1992/008220 Continuation-In-Part WO1994007529A1 (en) 1991-09-27 1992-09-25 Therapeutic inhibitor of vascular smooth muscle cells
US6245193A Continuation-In-Part 1991-09-27 1993-05-13
US08/450,793 Continuation US5811447A (en) 1991-09-27 1995-05-25 Therapeutic inhibitor of vascular smooth muscle cells
US08/829,991 Continuation-In-Part US6306421B1 (en) 1992-09-25 1997-03-31 Therapeutic inhibitor of vascular smooth muscle cells
US08/829,685 Continuation-In-Part US5981568A (en) 1993-01-28 1997-03-31 Therapeutic inhibitor of vascular smooth muscle cells
US08894350 A-371-Of-International 1997-10-10
US09/995,490 Continuation-In-Part US6569441B2 (en) 1993-01-28 2001-11-27 Therapeutic inhibitor of vascular smooth muscle cells
US10/190,211 Continuation US20030083733A1 (en) 1995-02-15 2002-07-03 Therapeutic inhibitor of vascular smooth muscle cells

Publications (1)

Publication Number Publication Date
WO1996025176A1 true WO1996025176A1 (en) 1996-08-22

Family

ID=23539404

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/002125 WO1996025176A1 (en) 1992-09-25 1996-02-15 Therapeutic inhibitor of vascular smooth muscle cells

Country Status (9)

Country Link
US (7) US6515009B1 (US06515009-20030204-C00004.png)
EP (3) EP1407786B1 (US06515009-20030204-C00004.png)
JP (2) JPH11500635A (US06515009-20030204-C00004.png)
AT (2) ATE321573T1 (US06515009-20030204-C00004.png)
AU (1) AU4985196A (US06515009-20030204-C00004.png)
CA (2) CA2212537C (US06515009-20030204-C00004.png)
DE (2) DE69632310T2 (US06515009-20030204-C00004.png)
ES (1) ES2217306T3 (US06515009-20030204-C00004.png)
WO (1) WO1996025176A1 (US06515009-20030204-C00004.png)

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998043618A2 (en) * 1997-03-31 1998-10-08 Neorx Corporation Use of cytoskeletal inhibitors for the prevention of restenosis
WO1998056312A1 (en) * 1997-06-13 1998-12-17 Scimed Life Systems, Inc. Stents having multiple layers of biodegradable polymeric composition
WO1999008729A1 (en) * 1997-08-13 1999-02-25 Boston Scientific Limited Loading and release of water-insoluble drugs
US5977163A (en) * 1996-03-12 1999-11-02 Pg-Txl Company, L. P. Water soluble paclitaxel prodrugs
WO2000012147A1 (en) * 1998-09-02 2000-03-09 Scimed Life Systems, Inc. Drug delivery device for stent
WO2000032238A1 (en) * 1998-12-03 2000-06-08 Scimed Life Systems, Inc. Stent having drug crystals thereon
WO2000006244A3 (en) * 1998-07-30 2000-06-15 James F Hainfeld Loading metal particles into cell membrane vesicles and metal particle use for imaging and therapy
WO2000044443A2 (en) * 1999-01-29 2000-08-03 Angiotech Pharmaceuticals Inc. Intra-pericardial delivery of anti-microtubule agents
US6120847A (en) * 1999-01-08 2000-09-19 Scimed Life Systems, Inc. Surface treatment method for stent coating
US6156373A (en) * 1999-05-03 2000-12-05 Scimed Life Systems, Inc. Medical device coating methods and devices
WO2001036007A2 (en) * 1999-11-12 2001-05-25 Angiotech Pharmaceuticals, Inc. Compositions of a combination of radioactive therapy and cell-cycle inhibitors
WO2001070295A1 (en) * 2000-03-22 2001-09-27 Zenon Kyriakides Coronary artery stent covered with endothelin receptor antagonist
US6306166B1 (en) 1997-08-13 2001-10-23 Scimed Life Systems, Inc. Loading and release of water-insoluble drugs
US6419692B1 (en) 1999-02-03 2002-07-16 Scimed Life Systems, Inc. Surface protection method for stents and balloon catheters for drug delivery
EP1221997A1 (en) * 1999-10-06 2002-07-17 The Penn State Research Foundation System and device for preventing restenosis in body vessels
US6441025B2 (en) 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US6485514B1 (en) 1997-12-12 2002-11-26 Supergen, Inc. Local delivery of therapeutic agents
WO2003041756A1 (en) * 2001-11-08 2003-05-22 Dsb Invest Holding Sa Endoluminal devices coated with latrunculin to prevent ingrowth of cells
US6730064B2 (en) 1998-08-20 2004-05-04 Cook Incorporated Coated implantable medical device
US6774278B1 (en) 1995-06-07 2004-08-10 Cook Incorporated Coated implantable medical device
US7153864B2 (en) 2000-03-17 2006-12-26 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US7265199B2 (en) 2000-04-11 2007-09-04 Celonova Biosciences Germany Gmbh Poly-tri-fluoro-ethoxypolyphosphazene coverings and films
EP2098230A1 (en) * 1997-03-31 2009-09-09 Boston Scientific Scimed Limited Use of cytoskeletal inhibitors in crystalline form for the inhibition or prevention of restenosis
SG165156A1 (en) * 1997-06-27 2010-10-28 Abraxis Bioscience Llc Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US7842083B2 (en) 2001-08-20 2010-11-30 Innovational Holdings, Llc. Expandable medical device with improved spatial distribution
US7850727B2 (en) 2001-08-20 2010-12-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US7862605B2 (en) 1995-06-07 2011-01-04 Med Institute, Inc. Coated implantable medical device
US7922764B2 (en) 2006-10-10 2011-04-12 Celonova Bioscience, Inc. Bioprosthetic heart valve with polyphosphazene
US8052734B2 (en) 1998-03-30 2011-11-08 Innovational Holdings, Llc Expandable medical device with beneficial agent delivery mechanism
US8097236B2 (en) 2001-02-22 2012-01-17 Psimedica Limited Devices and methods for the treatment of cancer
US8101275B2 (en) 2001-08-17 2012-01-24 Celonova Biosciences, Inc. Device based on nitinol, a process for its production, and its use
US8197881B2 (en) 2003-09-22 2012-06-12 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US8349390B2 (en) 2002-09-20 2013-01-08 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US8361537B2 (en) 1998-03-30 2013-01-29 Innovational Holdings, Llc Expandable medical device with beneficial agent concentration gradient
US8389043B2 (en) 2001-03-26 2013-03-05 Bayer Pharma Aktiengesellschaft Preparation for restenosis prevention
US8439968B2 (en) 1998-03-30 2013-05-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US8889211B2 (en) 2010-09-02 2014-11-18 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
US8945206B2 (en) 1995-06-07 2015-02-03 Cook Medical Technologies Llc Methods for making implantable medical devices
US8974522B2 (en) 2000-10-31 2015-03-10 Cook Medical Technologies Llc Coated medical device
US9034245B2 (en) 2010-03-04 2015-05-19 Icon Medical Corp. Method for forming a tubular medical device
US9056152B2 (en) 2011-08-25 2015-06-16 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
US9080146B2 (en) 2001-01-11 2015-07-14 Celonova Biosciences, Inc. Substrates containing polyphosphazene as matrices and substrates containing polyphosphazene with a micro-structured surface
US9107850B2 (en) 2004-10-25 2015-08-18 Celonova Biosciences, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US9107899B2 (en) 2005-03-03 2015-08-18 Icon Medical Corporation Metal alloys for medical devices
US9114162B2 (en) 2004-10-25 2015-08-25 Celonova Biosciences, Inc. Loadable polymeric particles for enhanced imaging in clinical applications and methods of preparing and using the same
US9126025B2 (en) 2008-05-01 2015-09-08 Bayer Intellectual Property Gmbh Method of coating a folded catheter balloon
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
US9216272B2 (en) 2002-09-20 2015-12-22 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US10080821B2 (en) 2009-07-17 2018-09-25 Boston Scientific Scimed, Inc. Nucleation of drug delivery balloons to provide improved crystal size and density
US10369256B2 (en) 2009-07-10 2019-08-06 Boston Scientific Scimed, Inc. Use of nanocrystals for drug delivery from a balloon
US10973770B2 (en) 2004-10-25 2021-04-13 Varian Medical Systems, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US11766506B2 (en) 2016-03-04 2023-09-26 Mirus Llc Stent device for spinal fusion
US11779685B2 (en) 2014-06-24 2023-10-10 Mirus Llc Metal alloys for medical devices

Families Citing this family (330)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811447A (en) * 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6515009B1 (en) 1991-09-27 2003-02-04 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6663881B2 (en) 1993-01-28 2003-12-16 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US20070117863A1 (en) * 1993-02-22 2007-05-24 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20030203976A1 (en) 1993-07-19 2003-10-30 William L. Hunter Anti-angiogenic compositions and methods of use
EP1632259B1 (en) * 1993-07-19 2011-12-21 Angiotech Pharmaceuticals, Inc. Anti-angiogene compositions and methods of use
US20030083733A1 (en) * 1997-10-10 2003-05-01 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6099562A (en) * 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US5837313A (en) * 1995-04-19 1998-11-17 Schneider (Usa) Inc Drug release stent coating process
US20020091433A1 (en) * 1995-04-19 2002-07-11 Ni Ding Drug release coated stent
AU6959898A (en) * 1997-04-11 1998-11-11 David J. Grainger Compounds and therapies for the prevention of vascular and non-vascular pathol ogies
US10028851B2 (en) * 1997-04-15 2018-07-24 Advanced Cardiovascular Systems, Inc. Coatings for controlling erosion of a substrate of an implantable medical device
US6240616B1 (en) * 1997-04-15 2001-06-05 Advanced Cardiovascular Systems, Inc. Method of manufacturing a medicated porous metal prosthesis
US8172897B2 (en) * 1997-04-15 2012-05-08 Advanced Cardiovascular Systems, Inc. Polymer and metal composite implantable medical devices
US6776792B1 (en) * 1997-04-24 2004-08-17 Advanced Cardiovascular Systems Inc. Coated endovascular stent
US20030199425A1 (en) * 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
US7208011B2 (en) * 2001-08-20 2007-04-24 Conor Medsystems, Inc. Implantable medical device with drug filled holes
US7713297B2 (en) * 1998-04-11 2010-05-11 Boston Scientific Scimed, Inc. Drug-releasing stent with ceramic-containing layer
US20020099438A1 (en) * 1998-04-15 2002-07-25 Furst Joseph G. Irradiated stent coating
US20030040790A1 (en) * 1998-04-15 2003-02-27 Furst Joseph G. Stent coating
US7967855B2 (en) * 1998-07-27 2011-06-28 Icon Interventional Systems, Inc. Coated medical device
US8070796B2 (en) * 1998-07-27 2011-12-06 Icon Interventional Systems, Inc. Thrombosis inhibiting graft
US20020065546A1 (en) * 1998-12-31 2002-05-30 Machan Lindsay S. Stent grafts with bioactive coatings
US20050171594A1 (en) * 1998-12-31 2005-08-04 Angiotech International Ag Stent grafts with bioactive coatings
US6790228B2 (en) * 1999-12-23 2004-09-14 Advanced Cardiovascular Systems, Inc. Coating for implantable devices and a method of forming the same
US20050271701A1 (en) * 2000-03-15 2005-12-08 Orbus Medical Technologies, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20030229393A1 (en) * 2001-03-15 2003-12-11 Kutryk Michael J. B. Medical device with coating that promotes cell adherence and differentiation
US20070055367A1 (en) * 2000-03-15 2007-03-08 Orbus Medical Technologies, Inc. Medical device with coating that promotes endothelial cell adherence and differentiation
US8088060B2 (en) 2000-03-15 2012-01-03 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
EP1263484B1 (en) * 2000-03-15 2007-05-16 OrbusNeich Medical, Inc. Coating which promotes endothelial cell adherence
US8460367B2 (en) * 2000-03-15 2013-06-11 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20160287708A9 (en) * 2000-03-15 2016-10-06 Orbusneich Medical, Inc. Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US20070141107A1 (en) * 2000-03-15 2007-06-21 Orbusneich Medical, Inc. Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US9522217B2 (en) 2000-03-15 2016-12-20 Orbusneich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods for using same
US6527801B1 (en) * 2000-04-13 2003-03-04 Advanced Cardiovascular Systems, Inc. Biodegradable drug delivery material for stent
US8109994B2 (en) 2003-01-10 2012-02-07 Abbott Cardiovascular Systems, Inc. Biodegradable drug delivery material for stent
US7875283B2 (en) * 2000-04-13 2011-01-25 Advanced Cardiovascular Systems, Inc. Biodegradable polymers for use with implantable medical devices
AU2001253479A1 (en) * 2000-04-13 2001-10-30 Sts Biopolymers, Inc. Targeted therapeutic agent release devices and methods of making and using the same
US20040243097A1 (en) * 2000-05-12 2004-12-02 Robert Falotico Antiproliferative drug and delivery device
US20050002986A1 (en) * 2000-05-12 2005-01-06 Robert Falotico Drug/drug delivery systems for the prevention and treatment of vascular disease
US8236048B2 (en) * 2000-05-12 2012-08-07 Cordis Corporation Drug/drug delivery systems for the prevention and treatment of vascular disease
US6776796B2 (en) 2000-05-12 2004-08-17 Cordis Corportation Antiinflammatory drug and delivery device
US7261735B2 (en) * 2001-05-07 2007-08-28 Cordis Corporation Local drug delivery devices and methods for maintaining the drug coatings thereon
US20020051730A1 (en) * 2000-09-29 2002-05-02 Stanko Bodnar Coated medical devices and sterilization thereof
AU2001294869B2 (en) * 2000-09-29 2006-06-15 Cardinal Health 529, Llc Coated 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
US6783793B1 (en) * 2000-10-26 2004-08-31 Advanced Cardiovascular Systems, Inc. Selective coating of medical devices
WO2002062335A2 (en) 2001-01-16 2002-08-15 Vascular Therapies, Llc Implantable device containing resorbable matrix material and anti-proliferative drugs for preventing or treating failure of hemodialysis vascular access and other vascular grafts
US20040204756A1 (en) * 2004-02-11 2004-10-14 Diaz Stephen Hunter Absorbent article with improved liquid acquisition capacity
US7771468B2 (en) * 2001-03-16 2010-08-10 Angiotech Biocoatings Corp. Medicated stent having multi-layer polymer coating
US6613083B2 (en) * 2001-05-02 2003-09-02 Eckhard Alt Stent device and method
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
US20040249443A1 (en) * 2001-08-20 2004-12-09 Shanley John F. Expandable medical device for treating cardiac arrhythmias
US7056338B2 (en) * 2003-03-28 2006-06-06 Conor Medsystems, Inc. Therapeutic agent delivery device with controlled therapeutic agent release rates
JP2003079647A (ja) * 2001-09-10 2003-03-18 Yasuyoshi Uchida 血管治療用器具
US7989018B2 (en) * 2001-09-17 2011-08-02 Advanced Cardiovascular Systems, Inc. Fluid treatment of a polymeric coating on an implantable medical device
US7285304B1 (en) * 2003-06-25 2007-10-23 Advanced Cardiovascular Systems, Inc. Fluid treatment of a polymeric coating on an implantable medical device
US7776379B2 (en) * 2001-09-19 2010-08-17 Medlogics Device Corporation Metallic structures incorporating bioactive materials and methods for creating the same
US20030060873A1 (en) * 2001-09-19 2003-03-27 Nanomedical Technologies, Inc. Metallic structures incorporating bioactive materials and methods for creating the same
US6863683B2 (en) 2001-09-19 2005-03-08 Abbott Laboratoris Vascular Entities Limited Cold-molding process for loading a stent onto a stent delivery system
US7195640B2 (en) * 2001-09-25 2007-03-27 Cordis Corporation Coated medical devices for the treatment of vulnerable plaque
US20030065345A1 (en) * 2001-09-28 2003-04-03 Kevin Weadock Anastomosis devices and methods for treating anastomotic sites
US20030065377A1 (en) * 2001-09-28 2003-04-03 Davila Luis A. Coated medical devices
US7108701B2 (en) * 2001-09-28 2006-09-19 Ethicon, Inc. Drug releasing anastomosis devices and methods for treating anastomotic sites
US20030077310A1 (en) * 2001-10-22 2003-04-24 Chandrashekhar Pathak Stent coatings containing HMG-CoA reductase inhibitors
US20030077279A1 (en) * 2001-10-24 2003-04-24 Cedars-Sinai Medical Center Methods for treating vascular disease by inhibiting toll-like receptor-4
US8740973B2 (en) * 2001-10-26 2014-06-03 Icon Medical Corp. Polymer biodegradable medical device
US20030088307A1 (en) * 2001-11-05 2003-05-08 Shulze John E. Potent coatings for stents
US6939376B2 (en) 2001-11-05 2005-09-06 Sun Biomedical, Ltd. Drug-delivery endovascular stent and method for treating restenosis
WO2003051396A1 (en) * 2001-12-17 2003-06-26 Cedars-Sinai Medical Center Treating vascular disease by inhibiting myeloid differentiation factor 88
US20030194421A1 (en) * 2001-12-28 2003-10-16 Angiotech Pharmaceuticals, Inc. Treatment of uveitis
US8016881B2 (en) * 2002-07-31 2011-09-13 Icon Interventional Systems, Inc. Sutures and surgical staples for anastamoses, wound closures, and surgical closures
JP3887588B2 (ja) * 2002-08-30 2007-02-28 株式会社リガク X線回折による応力測定法
US7396538B2 (en) * 2002-09-26 2008-07-08 Endovascular Devices, Inc. Apparatus and method for delivery of mitomycin through an eluting biocompatible implantable medical device
JP2006515186A (ja) * 2002-09-26 2006-05-25 アンジオテック インターナショナル アクツィエン ゲゼルシャフト 血管周囲ラップ
EP1549254A4 (en) * 2002-09-27 2007-11-07 Medlogics Device Corp IMPLANTABLE STENT WITH MODIFIED ENDS
US20060271168A1 (en) * 2002-10-30 2006-11-30 Klaus Kleine Degradable medical device
US20040142014A1 (en) * 2002-11-08 2004-07-22 Conor Medsystems, Inc. Method and apparatus for reducing tissue damage after ischemic injury
EP1575638A1 (en) * 2002-11-08 2005-09-21 Conor Medsystems, Inc. Expandable medical device and method for treating chronic total occlusions with local delivery of an angiogenic factor
AU2003285195A1 (en) * 2002-11-08 2004-06-03 Innovational Holdings, Llc Method and apparatus for treating vulnerable artherosclerotic plaque
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
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
CN1732022A (zh) * 2002-12-30 2006-02-08 血管技术国际股份公司 含有丝的支架移植物
AU2004226327A1 (en) * 2003-03-28 2004-10-14 Innovational Holdings, Llc Implantable medical device with beneficial agent concentration gradient
US20040202692A1 (en) * 2003-03-28 2004-10-14 Conor Medsystems, Inc. Implantable medical device and method for in situ selective modulation of agent delivery
US20050010170A1 (en) * 2004-02-11 2005-01-13 Shanley John F Implantable medical device with beneficial agent concentration gradient
US8109987B2 (en) 2003-04-14 2012-02-07 Tryton Medical, Inc. Method of treating a lumenal bifurcation
US7306580B2 (en) * 2003-04-16 2007-12-11 Cook Incorporated Medical device with therapeutic agents
US7972616B2 (en) * 2003-04-17 2011-07-05 Nanosys, Inc. Medical device applications of nanostructured surfaces
US20050038498A1 (en) * 2003-04-17 2005-02-17 Nanosys, Inc. Medical device applications of nanostructured surfaces
US20050221072A1 (en) * 2003-04-17 2005-10-06 Nanosys, Inc. Medical device applications of nanostructured surfaces
US7803574B2 (en) * 2003-05-05 2010-09-28 Nanosys, Inc. Medical device applications of nanostructured surfaces
US7186789B2 (en) * 2003-06-11 2007-03-06 Advanced Cardiovascular Systems, Inc. Bioabsorbable, biobeneficial polyester polymers for use in drug eluting stent coatings
US20050063937A1 (en) * 2003-09-16 2005-03-24 Cheng Li Multiple-arm peptide compounds, methods of manufacture and use in therapy
US7198675B2 (en) * 2003-09-30 2007-04-03 Advanced Cardiovascular Systems Stent mandrel fixture and method for selectively coating surfaces of a stent
US7208172B2 (en) * 2003-11-03 2007-04-24 Medlogics Device Corporation Metallic composite coating for delivery of therapeutic agents from the surface of implantable devices
WO2005049105A2 (en) * 2003-11-10 2005-06-02 Angiotech International Ag Medical implants and anti-scarring agents
US20050100577A1 (en) * 2003-11-10 2005-05-12 Parker Theodore L. Expandable medical device with beneficial agent matrix formed by a multi solvent system
WO2005046747A2 (en) * 2003-11-10 2005-05-26 Angiotech International Ag Intravascular devices and fibrosis-inducing agents
US20060085062A1 (en) * 2003-11-28 2006-04-20 Medlogics Device Corporation Implantable stent with endothelialization factor
US20050119723A1 (en) * 2003-11-28 2005-06-02 Medlogics Device Corporation Medical device with porous surface containing bioerodable bioactive composites and related methods
JP2007517550A (ja) * 2004-01-02 2007-07-05 アドヴァンスド カーディオヴァスキュラー システムズ, インコーポレイテッド 高比重リポ蛋白をコーティングした医療デバイス
US7211108B2 (en) * 2004-01-23 2007-05-01 Icon Medical Corp. Vascular grafts with amphiphilic block copolymer coatings
EP1753476B1 (en) * 2004-03-10 2015-07-29 OrbusNeich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20050214339A1 (en) * 2004-03-29 2005-09-29 Yiwen Tang Biologically degradable compositions for medical applications
US20050220835A1 (en) * 2004-03-30 2005-10-06 Jayaraman Ramesh B Agent eluting bioimplantable devices and polymer systems for their preparation
US20050220836A1 (en) * 2004-03-31 2005-10-06 Robert Falotico Drug delivery device
US20050288481A1 (en) * 2004-04-30 2005-12-29 Desnoyer Jessica R Design of poly(ester amides) for the control of agent-release from polymeric compositions
TW200605910A (en) * 2004-04-30 2006-02-16 Orbus Medical Technologies Inc Medical device with coating for capturing genetically-altered cells and methods for using same
US20050245905A1 (en) * 2004-04-30 2005-11-03 Schmidt Steven P Local drug-delivery system
EP1604697A1 (en) 2004-06-09 2005-12-14 J.A.C.C. GmbH Implantable device
US20050277696A1 (en) * 2004-06-14 2005-12-15 Igor Sokolov Novel use of cytotoxic drugs for treatment and prophylasxis of aging diseases by reversing the loss of elasticity of epithelial cells due to aging
US20050287287A1 (en) * 2004-06-24 2005-12-29 Parker Theodore L Methods and systems for loading an implantable medical device with beneficial agent
US20080280973A1 (en) * 2004-06-28 2008-11-13 Wender Paul A Laulimalide Analogues as Therapeutic Agents
US8568469B1 (en) 2004-06-28 2013-10-29 Advanced Cardiovascular Systems, Inc. Stent locking element and a method of securing a stent on a delivery system
US8241554B1 (en) 2004-06-29 2012-08-14 Advanced Cardiovascular Systems, Inc. Method of forming a stent pattern on a tube
US20060009839A1 (en) * 2004-07-12 2006-01-12 Scimed Life Systems, Inc. Composite vascular graft including bioactive agent coating and biodegradable sheath
US8778256B1 (en) 2004-09-30 2014-07-15 Advanced Cardiovascular Systems, Inc. Deformation of a polymer tube in the fabrication of a medical article
US8747879B2 (en) * 2006-04-28 2014-06-10 Advanced Cardiovascular Systems, Inc. Method of fabricating an implantable medical device to reduce chance of late inflammatory response
US7971333B2 (en) * 2006-05-30 2011-07-05 Advanced Cardiovascular Systems, Inc. Manufacturing process for polymetric stents
US20060020330A1 (en) * 2004-07-26 2006-01-26 Bin Huang Method of fabricating an implantable medical device with biaxially oriented polymers
US8747878B2 (en) 2006-04-28 2014-06-10 Advanced Cardiovascular Systems, Inc. Method of fabricating an implantable medical device by controlling crystalline structure
US7731890B2 (en) * 2006-06-15 2010-06-08 Advanced Cardiovascular Systems, Inc. Methods of fabricating stents with enhanced fracture toughness
US20060041102A1 (en) * 2004-08-23 2006-02-23 Advanced Cardiovascular Systems, Inc. Implantable devices comprising biologically absorbable polymers having constant rate of degradation and methods for fabricating the same
US9283099B2 (en) * 2004-08-25 2016-03-15 Advanced Cardiovascular Systems, Inc. Stent-catheter assembly with a releasable connection for stent retention
EP1789107B1 (en) * 2004-08-30 2009-05-27 Interstitial Therapeutics Medical stent provided with inhibitors of atp synthesis
US7229471B2 (en) * 2004-09-10 2007-06-12 Advanced Cardiovascular Systems, Inc. Compositions containing fast-leaching plasticizers for improved performance of medical devices
US7901451B2 (en) 2004-09-24 2011-03-08 Biosensors International Group, Ltd. Drug-delivery endovascular stent and method for treating restenosis
US7875233B2 (en) 2004-09-30 2011-01-25 Advanced Cardiovascular Systems, Inc. Method of fabricating a biaxially oriented implantable medical device
US8173062B1 (en) 2004-09-30 2012-05-08 Advanced Cardiovascular Systems, Inc. Controlled deformation of a polymer tube in fabricating a medical article
US8043553B1 (en) 2004-09-30 2011-10-25 Advanced Cardiovascular Systems, Inc. Controlled deformation of a polymer tube with a restraining surface in fabricating a medical article
US20060088571A1 (en) * 2004-10-21 2006-04-27 Medtronic Vascular, Inc. Biocompatible and hemocompatible polymer compositions
US7176261B2 (en) * 2004-10-21 2007-02-13 Medtronic, Inc. Angiotensin-(1-7) eluting polymer-coated medical device to reduce restenosis and improve endothelial cell function
US20060093647A1 (en) * 2004-10-29 2006-05-04 Villafana Manuel A Multiple layer coating composition
US20060127443A1 (en) * 2004-12-09 2006-06-15 Helmus Michael N Medical devices having vapor deposited nanoporous coatings for controlled therapeutic agent delivery
USH2260H1 (en) 2005-02-17 2011-07-05 Angiotech International Ag Stents combined with paclitaxel derivatives
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
KR101914254B1 (ko) 2005-02-18 2018-11-02 아브락시스 바이오사이언스, 엘엘씨 치료제의 조합 및 투여 방식, 및 조합 요법
AU2006221046B2 (en) * 2005-03-03 2012-02-02 Icon Medical Corp. Improved metal alloys for medical device
US20060264914A1 (en) * 2005-03-03 2006-11-23 Icon Medical Corp. Metal alloys for medical devices
US20060201601A1 (en) * 2005-03-03 2006-09-14 Icon Interventional Systems, Inc. Flexible markers
WO2006110197A2 (en) * 2005-03-03 2006-10-19 Icon Medical Corp. Polymer biodegradable medical device
US20060200048A1 (en) * 2005-03-03 2006-09-07 Icon Medical Corp. Removable sheath for device protection
US8323333B2 (en) * 2005-03-03 2012-12-04 Icon Medical Corp. Fragile structure protective coating
US7540995B2 (en) 2005-03-03 2009-06-02 Icon Medical Corp. Process for forming an improved metal alloy stent
US20060216431A1 (en) * 2005-03-28 2006-09-28 Kerrigan Cameron K Electrostatic abluminal coating of a stent crimped on a balloon catheter
US20060224226A1 (en) * 2005-03-31 2006-10-05 Bin Huang In-vivo radial orientation of a polymeric implantable medical device
US7381048B2 (en) * 2005-04-12 2008-06-03 Advanced Cardiovascular Systems, Inc. Stents with profiles for gripping a balloon catheter and molds for fabricating stents
US7291166B2 (en) * 2005-05-18 2007-11-06 Advanced Cardiovascular Systems, Inc. Polymeric stent patterns
US7622070B2 (en) * 2005-06-20 2009-11-24 Advanced Cardiovascular Systems, Inc. Method of manufacturing an implantable polymeric medical device
US20070038176A1 (en) * 2005-07-05 2007-02-15 Jan Weber Medical devices with machined layers for controlled communications with underlying regions
US7658880B2 (en) * 2005-07-29 2010-02-09 Advanced Cardiovascular Systems, Inc. Polymeric stent polishing method and apparatus
US7297758B2 (en) * 2005-08-02 2007-11-20 Advanced Cardiovascular Systems, Inc. Method for extending shelf-life of constructs of semi-crystallizable polymers
US20070038290A1 (en) * 2005-08-15 2007-02-15 Bin Huang Fiber reinforced composite stents
US7476245B2 (en) * 2005-08-16 2009-01-13 Advanced Cardiovascular Systems, Inc. Polymeric stent patterns
US9248034B2 (en) * 2005-08-23 2016-02-02 Advanced Cardiovascular Systems, Inc. Controlled disintegrating implantable medical devices
US20070045255A1 (en) * 2005-08-23 2007-03-01 Klaus Kleine Laser induced plasma machining with an optimized process gas
US20070045252A1 (en) * 2005-08-23 2007-03-01 Klaus Kleine Laser induced plasma machining with a process gas
US7452372B2 (en) * 2005-09-22 2008-11-18 Boston Scientific Scimed, Inc. Bifurcated stent
US7867547B2 (en) 2005-12-19 2011-01-11 Advanced Cardiovascular Systems, Inc. Selectively coating luminal surfaces of stents
US20070151961A1 (en) * 2006-01-03 2007-07-05 Klaus Kleine Fabrication of an implantable medical device with a modified laser beam
US20070156230A1 (en) * 2006-01-04 2007-07-05 Dugan Stephen R Stents with radiopaque markers
US7951185B1 (en) 2006-01-06 2011-05-31 Advanced Cardiovascular Systems, Inc. Delivery of a stent at an elevated temperature
US20070179219A1 (en) * 2006-01-31 2007-08-02 Bin Huang Method of fabricating an implantable medical device using gel extrusion and charge induced orientation
US20070224235A1 (en) * 2006-03-24 2007-09-27 Barron Tenney Medical devices having nanoporous coatings for controlled therapeutic agent delivery
US8187620B2 (en) * 2006-03-27 2012-05-29 Boston Scientific Scimed, Inc. Medical devices comprising a porous metal oxide or metal material and a polymer coating for delivering therapeutic agents
US20070231361A1 (en) * 2006-03-28 2007-10-04 Medtronic Vascular, Inc. Use of Fatty Acids to Inhibit the Growth of Aneurysms
US7964210B2 (en) * 2006-03-31 2011-06-21 Abbott Cardiovascular Systems Inc. Degradable polymeric implantable medical devices with a continuous phase and discrete phase
US20070254012A1 (en) * 2006-04-28 2007-11-01 Ludwig Florian N Controlled degradation and drug release in stents
US7985441B1 (en) 2006-05-04 2011-07-26 Yiwen Tang Purification of polymers for coating applications
US8003156B2 (en) 2006-05-04 2011-08-23 Advanced Cardiovascular Systems, Inc. Rotatable support elements for stents
US8304012B2 (en) 2006-05-04 2012-11-06 Advanced Cardiovascular Systems, Inc. Method for drying a stent
US20070264303A1 (en) * 2006-05-12 2007-11-15 Liliana Atanasoska Coating for medical devices comprising an inorganic or ceramic oxide and a therapeutic agent
US7761968B2 (en) * 2006-05-25 2010-07-27 Advanced Cardiovascular Systems, Inc. Method of crimping a polymeric stent
US20130325105A1 (en) 2006-05-26 2013-12-05 Abbott Cardiovascular Systems Inc. Stents With Radiopaque Markers
US7951194B2 (en) 2006-05-26 2011-05-31 Abbott Cardiovascular Sysetms Inc. Bioabsorbable stent with radiopaque coating
US20070282434A1 (en) * 2006-05-30 2007-12-06 Yunbing Wang Copolymer-bioceramic composite implantable medical devices
US7959940B2 (en) * 2006-05-30 2011-06-14 Advanced Cardiovascular Systems, Inc. Polymer-bioceramic composite implantable medical devices
US8343530B2 (en) * 2006-05-30 2013-01-01 Abbott Cardiovascular Systems Inc. Polymer-and polymer blend-bioceramic composite implantable medical devices
US7842737B2 (en) 2006-09-29 2010-11-30 Abbott Cardiovascular Systems Inc. Polymer blend-bioceramic composite implantable medical devices
US20080058916A1 (en) * 2006-05-31 2008-03-06 Bin Huang Method of fabricating polymeric self-expandable stent
US20070281073A1 (en) * 2006-06-01 2007-12-06 Gale David C Enhanced adhesion of drug delivery coatings on stents
US20070282433A1 (en) * 2006-06-01 2007-12-06 Limon Timothy A Stent with retention protrusions formed during crimping
US8034287B2 (en) 2006-06-01 2011-10-11 Abbott Cardiovascular Systems Inc. Radiation sterilization of medical devices
US8486135B2 (en) 2006-06-01 2013-07-16 Abbott Cardiovascular Systems Inc. Implantable medical devices fabricated from branched polymers
US20080124372A1 (en) * 2006-06-06 2008-05-29 Hossainy Syed F A Morphology profiles for control of agent release rates from polymer matrices
US20070286941A1 (en) * 2006-06-13 2007-12-13 Bin Huang Surface treatment of a polymeric stent
US8603530B2 (en) * 2006-06-14 2013-12-10 Abbott Cardiovascular Systems Inc. Nanoshell therapy
US8048448B2 (en) * 2006-06-15 2011-11-01 Abbott Cardiovascular Systems Inc. Nanoshells for drug delivery
US8535372B1 (en) 2006-06-16 2013-09-17 Abbott Cardiovascular Systems Inc. Bioabsorbable stent with prohealing layer
US8333000B2 (en) 2006-06-19 2012-12-18 Advanced Cardiovascular Systems, Inc. Methods for improving stent retention on a balloon catheter
US20070290412A1 (en) * 2006-06-19 2007-12-20 John Capek Fabricating a stent with selected properties in the radial and axial directions
US8017237B2 (en) 2006-06-23 2011-09-13 Abbott Cardiovascular Systems, Inc. Nanoshells on polymers
US9072820B2 (en) * 2006-06-26 2015-07-07 Advanced Cardiovascular Systems, Inc. Polymer composite stent with polymer particles
US20070299511A1 (en) * 2006-06-27 2007-12-27 Gale David C Thin stent coating
US8128688B2 (en) * 2006-06-27 2012-03-06 Abbott Cardiovascular Systems Inc. Carbon coating on an implantable device
US8815275B2 (en) 2006-06-28 2014-08-26 Boston Scientific Scimed, Inc. Coatings for medical devices comprising a therapeutic agent and a metallic material
CA2655793A1 (en) * 2006-06-29 2008-01-03 Boston Scientific Limited Medical devices with selective coating
US7794776B1 (en) 2006-06-29 2010-09-14 Abbott Cardiovascular Systems Inc. Modification of polymer stents with radiation
US7740791B2 (en) * 2006-06-30 2010-06-22 Advanced Cardiovascular Systems, Inc. Method of fabricating a stent with features by blow molding
US20080009938A1 (en) * 2006-07-07 2008-01-10 Bin Huang Stent with a radiopaque marker and method for making the same
US7823263B2 (en) 2006-07-11 2010-11-02 Abbott Cardiovascular Systems Inc. Method of removing stent islands from a stent
US7998404B2 (en) * 2006-07-13 2011-08-16 Advanced Cardiovascular Systems, Inc. Reduced temperature sterilization of stents
US20080014244A1 (en) * 2006-07-13 2008-01-17 Gale David C Implantable medical devices and coatings therefor comprising physically crosslinked block copolymers
WO2008008291A2 (en) * 2006-07-13 2008-01-17 Icon Medical Corp. Stent
US7757543B2 (en) 2006-07-13 2010-07-20 Advanced Cardiovascular Systems, Inc. Radio frequency identification monitoring of stents
US7794495B2 (en) * 2006-07-17 2010-09-14 Advanced Cardiovascular Systems, Inc. Controlled degradation of stents
US7886419B2 (en) * 2006-07-18 2011-02-15 Advanced Cardiovascular Systems, Inc. Stent crimping apparatus and method
US20080091262A1 (en) * 2006-10-17 2008-04-17 Gale David C Drug delivery after biodegradation of the stent scaffolding
US8016879B2 (en) * 2006-08-01 2011-09-13 Abbott Cardiovascular Systems Inc. Drug delivery after biodegradation of the stent scaffolding
US8703169B1 (en) 2006-08-15 2014-04-22 Abbott Cardiovascular Systems Inc. Implantable device having a coating comprising carrageenan and a biostable polymer
US9173733B1 (en) 2006-08-21 2015-11-03 Abbott Cardiovascular Systems Inc. Tracheobronchial implantable medical device and methods of use
US20080051881A1 (en) * 2006-08-24 2008-02-28 Feng James Q Medical devices comprising porous layers for the release of therapeutic agents
US7923022B2 (en) * 2006-09-13 2011-04-12 Advanced Cardiovascular Systems, Inc. Degradable polymeric implantable medical devices with continuous phase and discrete phase
EP2068757B1 (en) 2006-09-14 2011-05-11 Boston Scientific Limited Medical devices with drug-eluting coating
US20080086195A1 (en) * 2006-10-05 2008-04-10 Boston Scientific Scimed, Inc. Polymer-Free Coatings For Medical Devices Formed By Plasma Electrolytic Deposition
US20080103584A1 (en) 2006-10-25 2008-05-01 Biosensors International Group Temporal Intraluminal Stent, Methods of Making and Using
WO2008052179A2 (en) * 2006-10-27 2008-05-02 Medtronic, Inc. Angiotensin (1-7) eluting stent
US7981150B2 (en) 2006-11-09 2011-07-19 Boston Scientific Scimed, Inc. Endoprosthesis with coatings
US9700704B2 (en) 2006-11-20 2017-07-11 Lutonix, Inc. Drug releasing coatings for balloon catheters
US8414525B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8414909B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8414526B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Medical device rapid drug releasing coatings comprising oils, fatty acids, and/or lipids
US9737640B2 (en) 2006-11-20 2017-08-22 Lutonix, Inc. Drug releasing coatings for medical devices
US20080276935A1 (en) 2006-11-20 2008-11-13 Lixiao Wang Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US8414910B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8425459B2 (en) 2006-11-20 2013-04-23 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US8998846B2 (en) 2006-11-20 2015-04-07 Lutonix, Inc. Drug releasing coatings for balloon catheters
US8597673B2 (en) 2006-12-13 2013-12-03 Advanced Cardiovascular Systems, Inc. Coating of fast absorption or dissolution
US8099849B2 (en) 2006-12-13 2012-01-24 Abbott Cardiovascular Systems Inc. Optimizing fracture toughness of polymeric stent
US8431149B2 (en) 2007-03-01 2013-04-30 Boston Scientific Scimed, Inc. Coated medical devices for abluminal drug delivery
US8070797B2 (en) 2007-03-01 2011-12-06 Boston Scientific Scimed, Inc. Medical device with a porous surface for delivery of a therapeutic agent
US20080243228A1 (en) * 2007-03-28 2008-10-02 Yunbing Wang Implantable medical devices fabricated from block copolymers
US8067054B2 (en) 2007-04-05 2011-11-29 Boston Scientific Scimed, Inc. Stents with ceramic drug reservoir layer and methods of making and using the same
US8262723B2 (en) 2007-04-09 2012-09-11 Abbott Cardiovascular Systems Inc. Implantable medical devices fabricated from polymer blends with star-block copolymers
US8147769B1 (en) 2007-05-16 2012-04-03 Abbott Cardiovascular Systems Inc. Stent and delivery system with reduced chemical degradation
US7976915B2 (en) * 2007-05-23 2011-07-12 Boston Scientific Scimed, Inc. Endoprosthesis with select ceramic morphology
US9056155B1 (en) 2007-05-29 2015-06-16 Abbott Cardiovascular Systems Inc. Coatings having an elastic primer layer
US7829008B2 (en) * 2007-05-30 2010-11-09 Abbott Cardiovascular Systems Inc. Fabricating a stent from a blow molded tube
US7959857B2 (en) * 2007-06-01 2011-06-14 Abbott Cardiovascular Systems Inc. Radiation sterilization of medical devices
US8202528B2 (en) * 2007-06-05 2012-06-19 Abbott Cardiovascular Systems Inc. Implantable medical devices with elastomeric block copolymer coatings
US20080306582A1 (en) * 2007-06-05 2008-12-11 Yunbing Wang Implantable medical devices with elastomeric copolymer coatings
US8293260B2 (en) * 2007-06-05 2012-10-23 Abbott Cardiovascular Systems Inc. Elastomeric copolymer coatings containing poly (tetramethyl carbonate) for implantable medical devices
US8425591B1 (en) 2007-06-11 2013-04-23 Abbott Cardiovascular Systems Inc. Methods of forming polymer-bioceramic composite medical devices with bioceramic particles
US8109904B1 (en) 2007-06-25 2012-02-07 Abbott Cardiovascular Systems Inc. Drug delivery medical devices
US8048441B2 (en) 2007-06-25 2011-11-01 Abbott Cardiovascular Systems, Inc. Nanobead releasing medical devices
US7901452B2 (en) * 2007-06-27 2011-03-08 Abbott Cardiovascular Systems Inc. Method to fabricate a stent having selected morphology to reduce restenosis
US7955381B1 (en) 2007-06-29 2011-06-07 Advanced Cardiovascular Systems, Inc. Polymer-bioceramic composite implantable medical device with different types of bioceramic particles
US7942926B2 (en) * 2007-07-11 2011-05-17 Boston Scientific Scimed, Inc. Endoprosthesis coating
US8002823B2 (en) * 2007-07-11 2011-08-23 Boston Scientific Scimed, Inc. Endoprosthesis coating
JP2010533563A (ja) 2007-07-19 2010-10-28 ボストン サイエンティフィック リミテッド 吸着抑制表面を有する内部人工器官
US7931683B2 (en) 2007-07-27 2011-04-26 Boston Scientific Scimed, Inc. Articles having ceramic coated surfaces
US8815273B2 (en) * 2007-07-27 2014-08-26 Boston Scientific Scimed, Inc. Drug eluting medical devices having porous layers
WO2009018340A2 (en) * 2007-07-31 2009-02-05 Boston Scientific Scimed, Inc. Medical device coating by laser cladding
WO2009020520A1 (en) * 2007-08-03 2009-02-12 Boston Scientific Scimed, Inc. Coating for medical device having increased surface area
US20090118813A1 (en) * 2007-11-02 2009-05-07 Torsten Scheuermann Nano-patterned implant surfaces
US20090118818A1 (en) * 2007-11-02 2009-05-07 Boston Scientific Scimed, Inc. Endoprosthesis with coating
US7938855B2 (en) * 2007-11-02 2011-05-10 Boston Scientific Scimed, Inc. Deformable underlayer for stent
US8029554B2 (en) * 2007-11-02 2011-10-04 Boston Scientific Scimed, Inc. Stent with embedded material
US8216632B2 (en) 2007-11-02 2012-07-10 Boston Scientific Scimed, Inc. Endoprosthesis coating
US20090118809A1 (en) * 2007-11-02 2009-05-07 Torsten Scheuermann Endoprosthesis with porous reservoir and non-polymer diffusion layer
US8319002B2 (en) * 2007-12-06 2012-11-27 Nanosys, Inc. Nanostructure-enhanced platelet binding and hemostatic structures
WO2009073854A1 (en) 2007-12-06 2009-06-11 Nanosys, Inc. Resorbable nanoenhanced hemostatic structures and bandage materials
EP2271380B1 (en) 2008-04-22 2013-03-20 Boston Scientific Scimed, Inc. Medical devices having a coating of inorganic material
WO2009132176A2 (en) 2008-04-24 2009-10-29 Boston Scientific Scimed, Inc. Medical devices having inorganic particle layers
WO2009155328A2 (en) 2008-06-18 2009-12-23 Boston Scientific Scimed, Inc. Endoprosthesis coating
US8114429B2 (en) 2008-09-15 2012-02-14 Cv Ingenuity Corp. Local delivery of water-soluble or water-insoluble therapeutic agents to the surface of body lumens
US8257722B2 (en) 2008-09-15 2012-09-04 Cv Ingenuity Corp. Local delivery of water-soluble or water-insoluble therapeutic agents to the surface of body lumens
US8128951B2 (en) 2008-09-15 2012-03-06 Cv Ingenuity Corp. Local delivery of water-soluble or water-insoluble therapeutic agents to the surface of body lumens
US9198968B2 (en) 2008-09-15 2015-12-01 The Spectranetics Corporation Local delivery of water-soluble or water-insoluble therapeutic agents to the surface of body lumens
US9512196B2 (en) 2008-09-22 2016-12-06 Cedars-Sinai Medical Center Short-form human MD-2 as a negative regulator of toll-like receptor 4 signaling
EP2328911A4 (en) 2008-09-22 2012-03-14 Cedars Sinai Medical Center HUMAN MD-2 IN SHORT FORM AS NEGATIVE REGULATOR OF TOLL-4 TYPE RECEIVER SIGNALING
US8226603B2 (en) * 2008-09-25 2012-07-24 Abbott Cardiovascular Systems Inc. Expandable member having a covering formed of a fibrous matrix for intraluminal drug delivery
US8049061B2 (en) 2008-09-25 2011-11-01 Abbott Cardiovascular Systems, Inc. Expandable member formed of a fibrous matrix having hydrogel polymer for intraluminal drug delivery
US8076529B2 (en) * 2008-09-26 2011-12-13 Abbott Cardiovascular Systems, Inc. Expandable member formed of a fibrous matrix for intraluminal drug delivery
US8231980B2 (en) * 2008-12-03 2012-07-31 Boston Scientific Scimed, Inc. Medical implants including iridium oxide
US8071156B2 (en) * 2009-03-04 2011-12-06 Boston Scientific Scimed, Inc. Endoprostheses
UA93922C2 (ru) 2009-03-20 2011-03-25 Михаил Викторович Разуменко Способ получения пептидов, которые специфически распознают клетки определенного типа, и предназначены для терапевтических целей
US8287937B2 (en) * 2009-04-24 2012-10-16 Boston Scientific Scimed, Inc. Endoprosthese
US20100274352A1 (en) * 2009-04-24 2010-10-28 Boston Scientific Scrimed, Inc. Endoprosthesis with Selective Drug Coatings
CN105214143A (zh) 2009-04-28 2016-01-06 苏尔莫迪克斯公司 用于递送生物活性剂的装置和方法
US20100285085A1 (en) * 2009-05-07 2010-11-11 Abbott Cardiovascular Systems Inc. Balloon coating with drug transfer control via coating thickness
US8992601B2 (en) 2009-05-20 2015-03-31 480 Biomedical, Inc. Medical implants
US9309347B2 (en) 2009-05-20 2016-04-12 Biomedical, Inc. Bioresorbable thermoset polyester/urethane elastomers
US20110319987A1 (en) 2009-05-20 2011-12-29 Arsenal Medical Medical implant
AU2010249558A1 (en) * 2009-05-20 2011-12-08 Arsenal Medical, Inc. Medical implant
US9265633B2 (en) 2009-05-20 2016-02-23 480 Biomedical, Inc. Drug-eluting medical implants
US8888840B2 (en) * 2009-05-20 2014-11-18 Boston Scientific Scimed, Inc. Drug eluting medical implant
US8366763B2 (en) 2009-07-02 2013-02-05 Tryton Medical, Inc. Ostium support for treating vascular bifurcations
US8372133B2 (en) 2009-10-05 2013-02-12 480 Biomedical, Inc. Polymeric implant delivery system
US9149544B2 (en) * 2009-11-06 2015-10-06 The Penn State Research Foundation Bioconjugation of calcium phosphosilicate nanoparticles for selective targeting of cells in vivo
US8568471B2 (en) 2010-01-30 2013-10-29 Abbott Cardiovascular Systems Inc. Crush recoverable polymer scaffolds
US8808353B2 (en) 2010-01-30 2014-08-19 Abbott Cardiovascular Systems Inc. Crush recoverable polymer scaffolds having a low crossing profile
WO2011119536A1 (en) 2010-03-22 2011-09-29 Abbott Cardiovascular Systems Inc. Stent delivery system having a fibrous matrix covering with improved stent retention
MX2012011155A (es) 2010-03-29 2012-12-05 Abraxis Bioscience Llc Metodos para mejorar suministros de farmacos y efectividad de agentes terapeuticos.
NZ717490A (en) 2010-03-29 2017-12-22 Abraxis Bioscience Llc Methods of treating cancer
EP2380604A1 (en) 2010-04-19 2011-10-26 InnoRa Gmbh Improved coating formulations for scoring or cutting balloon catheters
KR20190130050A (ko) 2010-06-04 2019-11-20 아브락시스 바이오사이언스, 엘엘씨 췌장암의 치료 방법
US9757497B2 (en) 2011-05-20 2017-09-12 Surmodics, Inc. Delivery of coated hydrophobic active agent particles
US10213529B2 (en) 2011-05-20 2019-02-26 Surmodics, Inc. Delivery of coated hydrophobic active agent particles
US9861727B2 (en) 2011-05-20 2018-01-09 Surmodics, Inc. Delivery of hydrophobic active agent particles
US8726483B2 (en) 2011-07-29 2014-05-20 Abbott Cardiovascular Systems Inc. Methods for uniform crimping and deployment of a polymer scaffold
CN104168927B (zh) 2012-03-27 2016-10-05 泰尔茂株式会社 涂布组合物及医疗器械
WO2013146377A1 (ja) 2012-03-27 2013-10-03 テルモ株式会社 コーティング組成物および医療機器
US9956385B2 (en) 2012-06-28 2018-05-01 The Spectranetics Corporation Post-processing of a medical device to control morphology and mechanical properties
US10850076B2 (en) 2012-10-26 2020-12-01 Urotronic, Inc. Balloon catheters for body lumens
JP2015536709A (ja) 2012-10-26 2015-12-24 ウロトロニック・インコーポレイテッドUrotronic, Inc. 非血管狭窄のための薬物被覆バルーンカテーテル
US10881839B2 (en) 2012-10-26 2021-01-05 Urotronic, Inc. Drug-coated balloon catheters for body lumens
US11938287B2 (en) 2012-10-26 2024-03-26 Urotronic, Inc. Drug-coated balloon catheters for body lumens
US10806830B2 (en) 2012-10-26 2020-10-20 Urotronic, Inc. Drug-coated balloon catheters for body lumens
US11504450B2 (en) 2012-10-26 2022-11-22 Urotronic, Inc. Drug-coated balloon catheters for body lumens
US10898700B2 (en) 2012-10-26 2021-01-26 Urotronic, Inc. Balloon catheters for body lumens
CA2890205C (en) 2012-11-05 2020-12-22 Surmodics, Inc. Composition and method for delivery of hydrophobic active agents
US11246963B2 (en) 2012-11-05 2022-02-15 Surmodics, Inc. Compositions and methods for delivery of hydrophobic active agents
US10525171B2 (en) 2014-01-24 2020-01-07 The Spectranetics Corporation Coatings for medical devices
US9999527B2 (en) 2015-02-11 2018-06-19 Abbott Cardiovascular Systems Inc. Scaffolds having radiopaque markers
US11904072B2 (en) 2015-04-24 2024-02-20 Urotronic, Inc. Drug coated balloon catheters for nonvascular strictures
CN107635593A (zh) 2015-04-24 2018-01-26 优敦力公司 用于非血管狭窄的药物涂布的球囊导管
US9700443B2 (en) 2015-06-12 2017-07-11 Abbott Cardiovascular Systems Inc. Methods for attaching a radiopaque marker to a scaffold
EP3515517B1 (en) 2016-09-19 2022-02-16 Biotronik AG Polymer-free drug eluting vascular stents
US10898446B2 (en) 2016-12-20 2021-01-26 Surmodics, Inc. Delivery of hydrophobic active agents from hydrophilic polyether block amide copolymer surfaces
WO2018114992A1 (en) 2016-12-22 2018-06-28 Biotronik Ag Drug releasing coatings for medical devices and methods of making same
US10925632B2 (en) 2017-05-03 2021-02-23 Medtronic Vascular, Inc. Tissue-removing catheter
US11690645B2 (en) 2017-05-03 2023-07-04 Medtronic Vascular, Inc. Tissue-removing catheter
US20200190219A1 (en) * 2017-07-14 2020-06-18 Horiba, Ltd. Method of inhibiting nonspecific binding to binding proteins that bind to surface molecules of exosomes or eukaryotic cell membranes immobilized on carrier
US11648135B2 (en) 2017-09-13 2023-05-16 Boston Scientific Scimed, Inc. Coated stent
EP3866867A4 (en) * 2018-10-18 2022-07-13 The Regents of the University of California PROCESS FOR THE PREPARATION OF MODULAR HYDROGELS FROM MACROMOLECULES WITH ORTHOGONAL PHYSICOCHEMICAL REACTION
WO2020102729A1 (en) 2018-11-16 2020-05-22 Medtronic Vascular, Inc. Tissue-removing catheter
US12005206B2 (en) 2019-02-22 2024-06-11 Urotronic, Inc. Drug-coated balloon catheters for body lumens
US11819236B2 (en) 2019-05-17 2023-11-21 Medtronic Vascular, Inc. Tissue-removing catheter

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994016706A1 (en) * 1993-01-28 1994-08-04 Neorx Corporation Therapeutic inhibitors of vascular smooth muscle cells
WO1994026291A1 (en) * 1993-05-13 1994-11-24 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
WO1995003036A1 (en) * 1993-07-19 1995-02-02 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US5429634A (en) * 1993-09-09 1995-07-04 Pdt Systems Biogenic implant for drug delivery and method
EP0691130A1 (en) * 1994-05-12 1996-01-10 American Home Products Corporation Use of rapamycin in the manufacture of a medicament for preventing and heating hyperproliferactive vascular diseases, eventually in combination with mycophenolic acid

Family Cites Families (580)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2914563A (en) 1957-08-06 1959-11-24 Wm S Merrell Co Therapeutic composition
BE564542A (US06515009-20030204-C00004.png) 1957-02-05
US3168565A (en) * 1959-11-20 1965-02-02 Richardson Merrell Inc Trifluoromethyl derivatives of amino triarylethanols, -ethanes, and -ethylenes
GB1015787A (en) 1962-08-03 1966-01-05 Giuseppe Carlo Sigurta Tris-(p-methoxyphenyl)ethylene derivatives
US3279996A (en) 1962-08-28 1966-10-18 Jr David M Long Polysiloxane carrier for controlled release of drugs and other agents
BE637389A (US06515009-20030204-C00004.png) 1962-09-13
US3288806A (en) 1964-03-23 1966-11-29 Parke Davis & Co Alpha-(aminoethoxyphenyl)-alpha-alkylstilbenes
US3445473A (en) 1965-04-10 1969-05-20 Hoechst Ag 3-anilino-thiophene-4-carboxylic acids,esters,and amides
GB1205743A (en) 1966-07-15 1970-09-16 Nat Res Dev Surgical dilator
US3526005A (en) 1967-06-29 1970-09-01 Gulf General Atomic Inc Method of preparing an intravascular defect by implanting a pyrolytic carbon coated prosthesis
US3634517A (en) * 1968-08-19 1972-01-11 Richardson Merrell Inc Triarylalkenones
US3738365A (en) 1969-07-22 1973-06-12 R Schulte Spring reinforced extensible catheter
US5082834A (en) * 1978-05-30 1992-01-21 Sorensen John R J Anti-inflammatory and anti-ulcer compounds and process
US4221785A (en) 1978-05-30 1980-09-09 Sorenson John R J Anti-inflammatory and anti-ulcer compounds and process
US5521171A (en) 1975-03-31 1996-05-28 Sorenson; John R. J. Anti-inflammatory and anti-ulcer compounds and process
US3879516A (en) 1972-12-07 1975-04-22 Technibiotics Method of constructing a catheter
JPS5640710B2 (US06515009-20030204-C00004.png) 1973-01-18 1981-09-22
US4070484A (en) 1973-01-18 1978-01-24 Kissei Pharmaceutical Co., Ltd. Antiallergic composition containing aromatic carboxylic amide derivatives and method of using the same
IT1070993B (it) 1973-12-24 1985-04-02 Bioindustria Spa Derivati del tiofene ad attivita antimicotica e tricomonicida
US3932627A (en) * 1974-02-04 1976-01-13 Rescue Products, Inc. Siver-heparin-allantoin complex
US3952334A (en) 1974-11-29 1976-04-27 General Atomic Company Biocompatible carbon prosthetic devices
US4093709A (en) 1975-01-28 1978-06-06 Alza Corporation Drug delivery devices manufactured from poly(orthoesters) and poly(orthocarbonates)
US4323707A (en) 1975-10-28 1982-04-06 Eli Lilly And Company Antifertility compounds
US4133814A (en) * 1975-10-28 1979-01-09 Eli Lilly And Company 2-Phenyl-3-aroylbenzothiophenes useful as antifertility agents
US4230862A (en) 1975-10-28 1980-10-28 Eli Lilly And Company Antifertility compounds
US4428963A (en) * 1976-08-23 1984-01-31 Hoffmann-La Roche Inc. Novel thiophene derivatives
CH628628A5 (en) 1976-08-23 1982-03-15 Hoffmann La Roche Process for the preparation of cyclic compounds
US4317915A (en) * 1976-08-23 1982-03-02 Hoffmann-La Roche Inc. Novel thiophene derivatives
JPS6012381B2 (ja) 1976-10-06 1985-04-01 久光製薬株式会社 温熱湿布剤
US4235988A (en) 1976-12-13 1980-11-25 Imperial Chemical Industries Limited Delivery means for biologically active agents
US4391797A (en) 1977-01-05 1983-07-05 The Children's Hospital Medical Center Systems for the controlled release of macromolecules
JPS5490121A (en) 1977-11-28 1979-07-17 Boettcher Barry Neutral copper bonded body and antiinflaming agent
DE2817157A1 (de) * 1978-04-17 1979-10-25 Schering Ag Verwendung von antioestrogenen und antigonadotrop wirkenden antiandrogenen zur prophylaxe und therapie der prostatahyperplasie
US4440754A (en) 1978-05-30 1984-04-03 Sorenson John R J Anti-inflammatory and anti-ulcer compounds and process
US4233968A (en) * 1978-07-26 1980-11-18 Shaw Jr Seth T IUD Arrangement
US4219520A (en) 1978-08-30 1980-08-26 Medical Evaluation Devices And Instruments Corp. Method of making thrombo-resistant non-thrombogenic objects formed from a uniform mixture of a particulate resin and colloidal graphite
US4239778A (en) 1978-09-12 1980-12-16 The University Of Illinois Foundation Azaprostanoic acid analogs and their use as inhibitors of platelet aggregation
US4732763A (en) * 1978-10-17 1988-03-22 Stolle Research And Development Corporation Active/passive immunization of the internal female reproductive organs
US4219656A (en) 1978-10-24 1980-08-26 American Cyanamid Company 3,4-Disubstituted thiophenes
US4315028A (en) * 1978-12-22 1982-02-09 Scheinberg Israel H Method of treatment of rheumatoid arthritis
US4292965A (en) 1978-12-29 1981-10-06 The Population Council, Inc. Intravaginal ring
EP0019377B1 (en) 1979-05-15 1982-08-04 Imperial Chemical Industries Plc 1-hydrocarbyloxyphenyl-1,2-diphenylalkene derivatives, their manufacture and a pharmaceutical composition containing them
US4382143A (en) 1979-07-23 1983-05-03 American Cyanamid Company Hypolipidemic and antiatherosclerotic novel (monosubstituted-amino)heteroaryl carboxylic acids and analogs
AU532174B2 (en) 1979-08-15 1983-09-22 Stephen James Beveridge Copper chelate compounds
US4487780A (en) 1979-09-18 1984-12-11 Scheinberg Israel H Method of treatment of rheumatoid arthritis
US4300244A (en) 1979-09-19 1981-11-17 Carbomedics, Inc. Cardiovascular grafts
US5658927A (en) 1980-03-07 1997-08-19 Research Corporation Technologies Diphenylcyclopropyl analogs
US5324736A (en) 1980-03-07 1994-06-28 Research Corporation Technologies, Inc. Diphenylcyclopropyl analogs as antiestrogenic and antitumor agents
US4879315A (en) 1982-03-30 1989-11-07 The Board Of Regents For The University Of Oklahoma Cyclopropyl analogs as anti-estrogenic, anti-tumor and female fertility agents
US4282246A (en) 1980-03-07 1981-08-04 Pfizer Inc. Antidiabetic furancarboxylic and thiphenecarboxylic acids
US5098903A (en) 1980-03-07 1992-03-24 Board Of Regents Of The University Of Oklahoma Diphenylcyclopropyl analogs as antiestrogenic and antitumor agents
US5658951A (en) 1980-03-07 1997-08-19 Research Corporation Technologies Diphenylcyclopropyl analogs
ATE4776T1 (de) 1980-04-29 1983-10-15 Blendax-Werke R. Schneider Gmbh & Co. Zahnpasta.
DE3065015D1 (en) 1980-04-29 1983-11-03 Blendax Werke Schneider Co Toothpaste
US4442119A (en) 1980-07-07 1984-04-10 The Board Of Regents For The University Of Oklahoma Cyclopropyl analogs as estrogenic and anti-fertility agents
JPS5737905A (en) * 1980-08-14 1982-03-02 Toshiba Corp Envelope curve wave detecting circuit
US4389330A (en) 1980-10-06 1983-06-21 Stolle Research And Development Corporation Microencapsulation process
US4956129A (en) 1984-03-30 1990-09-11 Ici Americas Inc. Microencapsulation process
US4675189A (en) 1980-11-18 1987-06-23 Syntex (U.S.A.) Inc. Microencapsulation of water soluble active polypeptides
DE3046719C2 (de) 1980-12-11 1983-02-17 Klinge Pharma GmbH, 8000 München 1,1,2-Triphenyl-but-1-en-Derivate, Verfahren zu ihrer Herstellung und Arzneimittel
US4380635A (en) 1981-04-03 1983-04-19 Eli Lilly And Company Synthesis of acylated benzothiophenes
US4418068A (en) 1981-04-03 1983-11-29 Eli Lilly And Company Antiestrogenic and antiandrugenic benzothiophenes
US4678466A (en) 1981-06-25 1987-07-07 Rosenwald Peter L Internal medication delivery method and vehicle
US4670428A (en) 1982-02-01 1987-06-02 International Copper Research Association, Inc. Method for treating convulsions and epilepsy with organic copper compounds
SE8200751L (sv) 1982-02-09 1983-08-10 Olle Larm Forfarande for kovalent koppling for framstellning av konjugat och hervid erhallna produkter
US4485096A (en) 1982-02-26 1984-11-27 Massachusetts Institute Of Technology Tissue-equivalent and method for preparation thereof
US4867973A (en) 1984-08-31 1989-09-19 Cytogen Corporation Antibody-therapeutic agent conjugates
US4389393A (en) 1982-03-26 1983-06-21 Forest Laboratories, Inc. Sustained release therapeutic compositions based on high molecular weight hydroxypropylmethylcellulose
US5015666A (en) 1982-03-30 1991-05-14 Board of Reagents of the University of Oklahoma Triarylcyclopropanes as antiestrogens and antitumor agents
SE445884B (sv) 1982-04-30 1986-07-28 Medinvent Sa Anordning for implantation av en rorformig protes
DE3245665A1 (de) 1982-05-04 1983-11-10 Boehringer Mannheim Gmbh, 6800 Mannheim Verfahren zur herstellung permanenter tierischer und humaner zellinien und deren verwendung
US4485097A (en) 1982-05-26 1984-11-27 Massachusetts Institute Of Technology Bone-equivalent and method for preparation thereof
GB2126576B (en) 1982-06-25 1985-06-19 Farmos Group Limited Alkane and alkene derivatives
US4952607A (en) 1982-05-27 1990-08-28 International Copper Research Association, Inc. Copper complex for treating cancer
US4657928A (en) 1982-05-27 1987-04-14 International Copper Research Association, Inc. Organic copper complexes as radioprotectants
FI77839C (fi) 1982-05-27 1989-05-10 Farmos Oy Foerfarande foer framstaellning av nya terapeutiskt effektiva trifenylalkan- och alkenderivat.
USRE33403E (en) 1982-06-03 1990-10-23 Stolle Research & Development Corporation Method for treating disorders of the vascular and pulmonary systems
US5491173A (en) * 1982-06-25 1996-02-13 Orion-Yhtyma Oy Tri-phenyl alkene derivatives and their preparation and use
US5656587A (en) 1982-09-24 1997-08-12 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Promotion of cell proliferation by use of transforming growth factor beta (TGF-β)
US5705477A (en) * 1982-09-24 1998-01-06 The United States Of America As Represented By The Department Of Health And Human Services Compositions of transforming growth factor β(TGF-β) which promotes wound healing and methods for their use
US4577636A (en) 1982-11-23 1986-03-25 The Beth Israel Hospital Association Method for diagnosis of atherosclerosis
US4512762A (en) 1982-11-23 1985-04-23 The Beth Israel Hospital Association Method of treatment of atherosclerosis and a balloon catheter for same
US4491574A (en) * 1983-03-02 1985-01-01 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Reduction of high dose aspirin toxicity by dietary vitamin A
US4629694A (en) 1983-07-12 1986-12-16 Cornell Research Foundation, Inc. Detecting and distinguishing between plasminogen activators
US4656083A (en) 1983-08-01 1987-04-07 Washington Research Foundation Plasma gas discharge treatment for improving the biocompatibility of biomaterials
US5034265A (en) 1983-08-01 1991-07-23 Washington Research Foundation Plasma gas discharge treatment for improving the compatibility of biomaterials
JPS6041489A (ja) 1983-08-12 1985-03-05 Kyowa Hakko Kogyo Co Ltd 新規生理活性物質k―252
IL69686A (en) * 1983-09-11 1988-03-31 Yeda Res & Dev Compositions containing cell membrane proteins and process for their preparation
US4549913A (en) 1984-01-27 1985-10-29 Sony Corporation Wafer construction for making single-crystal semiconductor device
US5197977A (en) 1984-01-30 1993-03-30 Meadox Medicals, Inc. Drug delivery collagen-impregnated synthetic vascular graft
US4687482A (en) 1984-04-27 1987-08-18 Scripps Clinic And Research Foundation Vascular prosthesis
US4753652A (en) 1984-05-04 1988-06-28 Children's Medical Center Corporation Biomaterial implants which resist calcification
US4664097A (en) 1984-05-09 1987-05-12 The Wistar Institute Of Anatomy & Biology Nuclear transplantation in the mammalian embryo by microsurgery and cell fusion
EP0170063B1 (en) 1984-07-31 1988-08-24 Mitsubishi Petrochemical Co., Ltd. Copper-type conductive coating composition
CA1289077C (en) 1984-08-13 1991-09-17 Harry H. Leveen Treatment of cancer with phlorizin and its derivatives
US4757059A (en) 1984-08-14 1988-07-12 International Copper Research Association Method for treating convulsions and epilepsy with organic copper compounds
US4758554A (en) 1984-08-14 1988-07-19 International Copper Research Association Method for treating convulsions and epilepsy with organic copper compounds
US4758555A (en) 1984-08-14 1988-07-19 International Copper Research Association Method for treating convulsions and epilepsy with organic copper compounds
US5226430A (en) 1984-10-24 1993-07-13 The Beth Israel Hospital Method for angioplasty
DE3442736A1 (de) 1984-11-23 1986-06-05 Tassilo Dr.med. 7800 Freiburg Bonzel Dilatationskatheter
IT1196390B (it) 1984-12-28 1988-11-16 Consiglio Nazionale Ricerche Uso di derivati del tiofene nel trattamento dei tumori,composizioni farmaceutiche che li contengono e composti atti allo scopo
US4897255A (en) 1985-01-14 1990-01-30 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
US4760051A (en) 1985-01-24 1988-07-26 Pickart Loren R Use of GHL-Cu as a wound-healing and anti-inflammatory agent
US4605644A (en) 1985-02-07 1986-08-12 Regents Of The University Of Minnesota Method for stimulating recovery from ischemia employing ribose and adenine
US4689046A (en) 1985-03-11 1987-08-25 Carbomedics, Inc. Heart valve prosthesis
US5284763A (en) * 1985-03-22 1994-02-08 Genentech, Inc. Nucleic acid encoding TGF-β and its uses
US4824436A (en) 1985-04-09 1989-04-25 Harvey Wolinsky Method for the prevention of restenosis
US5262319A (en) 1985-04-19 1993-11-16 Oncogene Science, Inc. Method for obtaining bone marrow free of tumor cells using transforming growth factor β3
US4826672A (en) 1985-06-07 1989-05-02 President And Fellows Of Harvard College Astatinated organic compounds
WO1986007541A1 (en) 1985-06-19 1986-12-31 Yasushi Zyo Composition which can impart antithrombotic ability and medical apparatus to be in contact with blood
US5470876A (en) 1985-07-18 1995-11-28 Proctor; Peter H. Topical sod for treating hair loss
DE3532860C1 (de) 1985-09-14 1987-03-12 Blendax Werke Schneider Co Mittel zur oralen Hygiene
US4853377A (en) 1985-10-15 1989-08-01 Pollack Robert L Method and composition for increasing production of serotonin
US4744981A (en) 1985-10-17 1988-05-17 Neorx Corporation Trichothecene antibody conjugates
US4733665C2 (en) * 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US5102417A (en) 1985-11-07 1992-04-07 Expandable Grafts Partnership Expandable intraluminal graft, and method and apparatus for implanting an expandable intraluminal graft
DE3640745A1 (de) 1985-11-30 1987-06-04 Ernst Peter Prof Dr M Strecker Katheter zum herstellen oder erweitern von verbindungen zu oder zwischen koerperhohlraeumen
DE3544663A1 (de) 1985-12-13 1987-06-19 Schering Ag Thrombosebehandlung mit fibrinolytika und prostacyclinen
US5120535A (en) 1986-11-26 1992-06-09 Oncogen Oncostatin M and novel compositions having anti-neoplastic activity
EP0556940A1 (en) 1986-02-24 1993-08-25 Robert E. Fischell Intravascular stent
DE3608158A1 (de) 1986-03-12 1987-09-17 Braun Melsungen Ag Mit vernetzter gelatine impraegnierte gefaessprothese und verfahren zu ihrer herstellung
JPS62220196A (ja) 1986-03-20 1987-09-28 Kyowa Hakko Kogyo Co Ltd 新規物質ucn―01
US5955584A (en) 1986-03-31 1999-09-21 Charter Ventures Atherosclerotic plaque specific antigens, antibodies thereto, and uses thereof
US5061273A (en) 1989-06-01 1991-10-29 Yock Paul G Angioplasty apparatus facilitating rapid exchanges
US5040548A (en) 1989-06-01 1991-08-20 Yock Paul G Angioplasty mehtod
US5350395A (en) 1986-04-15 1994-09-27 Yock Paul G Angioplasty apparatus facilitating rapid exchanges
US4859585A (en) 1986-04-17 1989-08-22 Trustees Of Tufts College In-vitro methods for identifying compositions which are agonists and antagonists of estrogens
SE453258B (sv) 1986-04-21 1988-01-25 Medinvent Sa Elastisk, sjelvexpanderande protes samt forfarande for dess framstellning
US4919939A (en) * 1986-04-29 1990-04-24 Pharmetrix Corporation Periodontal disease treatment system
US4962091A (en) 1986-05-23 1990-10-09 Syntex (U.S.A.) Inc. Controlled release of macromolecular polypeptides
US4879225A (en) 1986-06-20 1989-11-07 Neorx Corporation Enhanced production of antibodies utilizing insolubilized immune complexes
DE3621350A1 (de) 1986-06-26 1988-01-14 Bonzel Tassilo Dilatationskatheter mit einem aufweitbaren ballon
US4786500A (en) 1986-06-26 1988-11-22 Alza Corporation Programmable agent delivery system
US5264422A (en) 1986-06-30 1993-11-23 Fidia S.P.A. Esters of alginic acid with steroidal alcohols
US5314679A (en) 1986-07-03 1994-05-24 Advanced Magnetics Inc. Vascular magnetic resonance imaging agent comprising nanoparticles
US5216021A (en) 1986-08-28 1993-06-01 Sorenson John R J Analgesic method
US4999347A (en) * 1986-08-28 1991-03-12 Sorenson John R J Analgesic method
GB2196003A (en) 1986-09-11 1988-04-20 Nat Res Dev Iodo-and bromo-tamoxifen derivatives
HU199281B (en) 1986-10-17 1990-02-28 Biogal Gyogyszergyar Synergetic unsenziting face- and body-cosmetics
US4793348A (en) 1986-11-15 1988-12-27 Palmaz Julio C Balloon expandable vena cava filter to prevent migration of lower extremity venous clots into the pulmonary circulation
US4994384A (en) 1986-12-31 1991-02-19 W. R. Grace & Co.-Conn. Multiplying bovine embryos
US4748982A (en) 1987-01-06 1988-06-07 Advanced Cardiovascular Systems, Inc. Reinforced balloon dilatation catheter with slitted exchange sleeve and method
US4959355A (en) 1987-03-16 1990-09-25 The Trustees Of Columbia University In The City Of New York Method of inhibiting osmotic water flow
US5075321A (en) 1987-03-24 1991-12-24 University Of Pennsylvania Methods of treating diseases characterized by interactions of IgG-containing immune complexes with macrophage Fc receptors using antiestrogenic benzothiophenes
DE3877678T2 (de) 1987-04-16 1993-10-07 Christian Bindschaedler Verfahren zur herstellung eines wasserunlöslichen polymerpulvers, das in einer flüssigen phase redispergiert werden kann und verfahren zur herstellung einer dispersion des polymerpulvers.
DE3781546D1 (de) 1987-04-21 1992-10-08 Heumann Pharma Gmbh & Co Stabile loesungsmitteladdukte von z-1-(p-beta-dimethylamino-ethoxyphenyl)-1-(p-hydroxyphenyl)-2-phenylbut-1-en.
US5114719A (en) 1987-04-29 1992-05-19 Sabel Bernhard A Extended drug delivery of small, water-soluble molecules
AU1539188A (en) 1987-05-04 1988-11-10 Bristol-Myers Squibb Company TGF-B2 and novel compositions having anti-neoplastic activity
SE8702254D0 (sv) 1987-05-29 1987-05-29 Kabivitrum Ab Novel heparin derivatives
US5093330A (en) 1987-06-15 1992-03-03 Ciba-Geigy Corporation Staurosporine derivatives substituted at methylamino nitrogen
EP0297946A3 (en) 1987-06-19 1989-06-07 Syracuse University Cytochalasin purification methods and compositions
US5527337A (en) 1987-06-25 1996-06-18 Duke University Bioabsorbable stent and method of making the same
US4835002A (en) 1987-07-10 1989-05-30 Wolf Peter A Microemulsions of oil in water and alcohol
US5216024A (en) 1987-07-28 1993-06-01 Baylor College Of Medicine Cell growth inhibitors and methods of treating cancer and cell proliferative diseases
US5221620A (en) 1987-10-06 1993-06-22 Oncogen Cloning and expression of transforming growth factor β2
US4886062A (en) 1987-10-19 1989-12-12 Medtronic, Inc. Intravascular radially expandable stent and method of implant
DE3737340A1 (de) 1987-11-04 1989-05-24 Bayer Ag Neue fluormethoxyphenyl-dihydropyridine, verfahren zur herstellung und ihre verwendung in arzneimitteln
US4929602A (en) 1987-11-25 1990-05-29 Scripps Clinic And Research Foundation Method of inhibiting platelet dependent arterial thrombosis
US4916193A (en) 1987-12-17 1990-04-10 Allied-Signal Inc. Medical devices fabricated totally or in part from copolymers of recurring units derived from cyclic carbonates and lactides
US5185408A (en) * 1987-12-17 1993-02-09 Allied-Signal Inc. Medical devices fabricated totally or in part from copolymers of recurring units derived from cyclic carbonates and lactides
US4997652A (en) * 1987-12-22 1991-03-05 Visionex Biodegradable ocular implants
JP2702953B2 (ja) 1988-01-30 1998-01-26 オリンパス光学工業株式会社 薬液含浸セラミックス
US5030637A (en) 1988-02-08 1991-07-09 Regents Of The University Of Minnesota Anisodamine to prevent and treat eye disease
US5019096A (en) 1988-02-11 1991-05-28 Trustees Of Columbia University In The City Of New York Infection-resistant compositions, medical devices and surfaces and methods for preparing and using same
US5234957A (en) 1991-02-27 1993-08-10 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US5446070A (en) 1991-02-27 1995-08-29 Nover Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US4942184A (en) * 1988-03-07 1990-07-17 The United States Of America As Represented By The Department Of Health And Human Services Water soluble, antineoplastic derivatives of taxol
ATE145337T1 (de) 1988-05-02 1996-12-15 Phanos Tech Inc Verbindungen, zusammensetzungen und verfahren zum binden von bio-affektions-substanzen an oberflächenmembranen von bioteilchen
US4973601A (en) 1988-06-01 1990-11-27 The United States Of America, As Represented By The Secretary Of Agriculture Control of insects by fungal tremorgenic mycotoxins
US5338770A (en) 1988-06-08 1994-08-16 Cardiopulmonics, Inc. Gas permeable thrombo-resistant coatings and methods of manufacture
US5182317A (en) * 1988-06-08 1993-01-26 Cardiopulmonics, Inc. Multifunctional thrombo-resistant coatings and methods of manufacture
US5262451A (en) 1988-06-08 1993-11-16 Cardiopulmonics, Inc. Multifunctional thrombo-resistant coatings and methods of manufacture
DE3821544C2 (de) 1988-06-25 1994-04-28 H Prof Dr Med Just Dilatationskatheter
US5705609A (en) * 1988-06-28 1998-01-06 La Jolla Cancer Research Foundation Decorin fragments inhibiting cell regulatory factors
US5468746A (en) 1988-07-29 1995-11-21 Zambon Group S.P.A. Compounds active on the cardiovascular system
US5167960A (en) 1988-08-03 1992-12-01 New England Deaconess Hospital Corporation Hirudin-coated biocompatible substance
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
US5213580A (en) 1988-08-24 1993-05-25 Endoluminal Therapeutics, Inc. Biodegradable polymeric endoluminal sealing process
US5634946A (en) 1988-08-24 1997-06-03 Focal, Inc. Polymeric endoluminal paving process
US5092877A (en) 1988-09-01 1992-03-03 Corvita Corporation Radially expandable endoprosthesis
US5226913A (en) 1988-09-01 1993-07-13 Corvita Corporation Method of making a radially expandable prosthesis
US5019090A (en) * 1988-09-01 1991-05-28 Corvita Corporation Radially expandable endoprosthesis and the like
US5053048A (en) 1988-09-22 1991-10-01 Cordis Corporation Thromboresistant coating
US5066789A (en) 1988-09-30 1991-11-19 Neorx Corporation Targeting substance-diagnostic/therapeutic agent conjugates having Schiff base linkages
SE462364B (sv) 1988-09-30 1990-06-18 Goeran Hansson Anvaendning av gamma-interferon foer beredning av ett preparat foer behandling av vaskulaer stenos
CA1322628C (en) 1988-10-04 1993-10-05 Richard A. Schatz Expandable intraluminal graft
US4956188A (en) 1988-10-20 1990-09-11 Zinpro Corporation Copper complexes with alpha hydroxy organic acids and their use as nutritional supplements
US4886811A (en) 1988-10-24 1989-12-12 Merrell Dow Pharmaceuticals Qunolyloxazole-2-ones useful as proteinkinase C inhibitors
US5395842A (en) 1988-10-31 1995-03-07 Endorecherche Inc. Anti-estrogenic compounds and compositions
US5393785A (en) * 1988-10-31 1995-02-28 Endorecherche, Inc. Therapeutic antiestrogens
CA2002011A1 (en) 1988-11-14 1990-05-14 Anthony F. Purchio Normal human growth regulatory receptor for tgf-beta
US5162430A (en) 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
US5268358A (en) 1988-12-08 1993-12-07 Cor Therapeutics, Inc. PDGF receptor blocking peptides
US5380716A (en) 1988-12-15 1995-01-10 Glycomed, Inc. Sulfated polysaccharides as inhibitors of smooth muscle cell proliferation
CA2005120A1 (en) 1988-12-15 1990-06-15 Anthony F. Purchio Tgf-beta 1/beta 2: a novel chimeric transforming growth factor-beta
US5304541A (en) 1988-12-15 1994-04-19 Bristol-Myers Squibb Company Methods using novel chimeric transforming growth factor-β1/β2
US5032679A (en) 1988-12-15 1991-07-16 Glycomed, Inc. Heparin fragments as inhibitors of smooth muscle cell proliferation
US5571714A (en) 1988-12-22 1996-11-05 Celtrix Pharmaceuticals, Inc. Monoclonal antibodies which bind both transforming growth factors β1 and β2 and methods of use
DE3844247A1 (de) 1988-12-29 1990-07-12 Minnesota Mining & Mfg Vorrichtung, insbesondere pflaster zum transdermalen verabreichen eines medikaments
US4948594A (en) 1989-01-03 1990-08-14 Zinpro Corporation Copper complexes of alpha-amino acids that contain terminal amino groups, and their use as nutritional supplements
US4900561A (en) * 1989-01-03 1990-02-13 Zinpro Corporation Copper complexes of alpha-amino acids that contain terminal amino groups, and their use as nutritional supplements
US5284869A (en) 1991-12-17 1994-02-08 Emil Bisaccia Photophoresis methods for treating atherosclerosis and for preventing restenosis following angioplasty
US5356630A (en) 1989-02-22 1994-10-18 Massachusetts Institute Of Technology Delivery system for controlled release of bioactive factors
US6146358A (en) 1989-03-14 2000-11-14 Cordis Corporation Method and apparatus for delivery of therapeutic agent
US5015578A (en) 1989-03-23 1991-05-14 Bristol-Myers Squibb Company BMY-41950 antitumor antibiotic
US5019504A (en) 1989-03-23 1991-05-28 The United States Of America As Represented By The Secretary Of Agriculture Production of taxol or taxol-like compounds in cell culture
US5073633A (en) 1989-03-23 1991-12-17 Bristol-Myers Company BMY-41950 antitumor antibiotic
FR2645160B1 (US06515009-20030204-C00004.png) 1989-03-31 1992-10-02 Rhone Poulenc Chimie
US5364632A (en) 1989-04-05 1994-11-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Medicinal emulsions
US5026537A (en) 1989-04-06 1991-06-25 Centocor, Inc. Methods for imaging atherosclerotic plaque
WO1990013293A1 (en) 1989-04-28 1990-11-15 Syracuse University Cytochalasin compositions and therapeutic methods
US5288735A (en) 1989-05-02 1994-02-22 Trager Seymour F Treatment of glaucoma
DE69024953T3 (de) 1989-05-04 2005-01-27 Southern Research Institute, Birmingham Einkapselungsverfahren
US4990158A (en) * 1989-05-10 1991-02-05 United States Surgical Corporation Synthetic semiabsorbable tubular prosthesis
WO1990013332A1 (en) * 1989-05-11 1990-11-15 Cedars-Sinai Medical Center Stent with sustained drug delivery
US4994071A (en) * 1989-05-22 1991-02-19 Cordis Corporation Bifurcating stent apparatus and method
US4994033A (en) 1989-05-25 1991-02-19 Schneider (Usa) Inc. Intravascular drug delivery dilatation catheter
US5118791A (en) 1989-05-25 1992-06-02 Genentech, Inc. Biologically active polypeptides based on transforming growth factor-β
US5268455A (en) 1989-05-25 1993-12-07 Genentech, Inc. Process for making biologically active polypeptides based on transforming growth factor-βsequences
DE3918736C2 (de) 1989-06-08 1998-05-14 Christian Dr Vallbracht Kunststoffüberzogene Metallgitterstents
US5248764A (en) 1989-06-16 1993-09-28 Merck Frosst Canada, Inc. Chelate derivatives of atrial natriuretic factor (ANF)
GB8914061D0 (en) 1989-06-19 1989-08-09 Wellcome Found Agents for potentiating the effects of antitumour agents and combating multiple drug resistance
GB8914040D0 (en) 1989-06-19 1989-08-09 Wellcome Found Agents for potentiating the effects of antitumour agents and combating multiple drug resistance
GB8914060D0 (en) 1989-06-19 1989-08-09 Wellcome Found Agents for potentiating the effects of antitumour agents and combating multiple drug resistance
US5208238A (en) 1989-06-19 1993-05-04 Burroughs Wellcome Company Agents for potentiating the effects of antitumor agents and combating multiple drug resistance
HU212760B (en) 1989-06-20 1997-02-28 Denes Method and device for the apportion of chemical materials into the vein wall
US5242397A (en) 1989-06-20 1993-09-07 Cedars-Sinai Medical Center Catheter device and method of use for intramural delivery of protein kinase C and tyrosine protein kinase inhibitors to prevent restenosis after balloon angioplasty
AU5751290A (en) 1989-06-27 1991-01-03 C.R. Bard Inc. Coaxial ptca catheter with anchor joint
DE3924538A1 (de) 1989-07-25 1991-01-31 Goedecke Ag Indolocarbazol und dessen verwendung
US5580774A (en) 1989-07-31 1996-12-03 Eli Lilly And Company Chimeric antibodies directed against a human glycoprotein antigen
US5100885A (en) 1989-08-01 1992-03-31 Johnson Matthey, Inc. Copper radiosensitizers
US5240913A (en) 1989-08-18 1993-08-31 Biogen, Inc. Inhibitors of thrombin
US4990538A (en) * 1989-08-23 1991-02-05 Harris Adrian L Use of toremifene and its metabolites for the reversal of multidrug resistance of cancer cells against cytotoxic drugs
US5145838A (en) 1989-08-30 1992-09-08 Procyte Corporation Methods and compositions for healing ulcers
US5023237A (en) 1989-08-30 1991-06-11 Procyte Corporation Methods and compositions for healing ulcers
IL95500A (en) * 1989-09-11 1997-03-18 Matrix Pharma ANTI-PROLIFERATIVE COMPOSITIONS CONTAINING TGF-b PROTEIN IN A VISCOUS MATRIX AND THEIR USE
US5126348A (en) 1989-09-26 1992-06-30 The University Of Colorado Foundation, Inc. Bioavailability enhancers
US4984594A (en) * 1989-10-27 1991-01-15 Shell Oil Company Vacuum method for removing soil contamination utilizing surface electrical heating
US5009659A (en) 1989-10-30 1991-04-23 Schneider (Usa) Inc. Fiber tip atherectomy catheter
JP2911927B2 (ja) 1989-11-29 1999-06-28 株式会社町田製作所 可撓管の製造方法
US5059166A (en) 1989-12-11 1991-10-22 Medical Innovative Technologies R & D Limited Partnership Intra-arterial stent with the capability to inhibit intimal hyperplasia
US5176617A (en) 1989-12-11 1993-01-05 Medical Innovative Technologies R & D Limited Partnership Use of a stent with the capability to inhibit malignant growth in a vessel such as a biliary duct
KR920000459B1 (ko) 1989-12-13 1992-01-14 재단법인 한국화학연구소 다당류 유도체가 도포된 인공혈관과 그 제조방법
US5304121A (en) 1990-12-28 1994-04-19 Boston Scientific Corporation Drug delivery system making use of a hydrogel polymer coating
DE69016983T2 (de) 1989-12-29 1995-07-06 Med Inst Inc Flexibler knickbeständiger Katheter.
US5232911A (en) 1990-01-03 1993-08-03 Ventech Research Inc. Mixture of a non-covalent heterodimer complex and a basic amphiphatic peptide as cytotoxic agent
US5049132A (en) 1990-01-08 1991-09-17 Cordis Corporation Balloon catheter for delivering therapeutic agents
US5175235A (en) 1990-06-04 1992-12-29 Nova Pharmaceutical Corporation Branched polyanhydrides
US5171812A (en) 1990-01-19 1992-12-15 Nova Pharmaceutical Corporation Polyanhydrides of oligomerized unsaturated aliphatic acids
AU651084B2 (en) 1990-01-30 1994-07-14 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
DE69108423T2 (de) * 1990-02-08 1995-07-27 Howmedica Aufblasbarer Dilatator.
US5199939B1 (en) 1990-02-23 1998-08-18 Michael D Dake Radioactive catheter
US5545208A (en) 1990-02-28 1996-08-13 Medtronic, Inc. Intralumenal drug eluting prosthesis
EP0470246B1 (en) 1990-02-28 1995-06-28 Medtronic, Inc. Intralumenal drug eluting prosthesis
US5108989A (en) 1990-04-04 1992-04-28 Genentech, Inc. Method of predisposing mammals to accelerated tissue repair
WO1991015770A1 (en) 1990-04-11 1991-10-17 The General Hospital Corporation Therapeutic uses of actin-binding compounds
US5180376A (en) * 1990-05-01 1993-01-19 Cathco, Inc. Non-buckling thin-walled sheath for the percutaneous insertion of intraluminal catheters
US5290271A (en) 1990-05-14 1994-03-01 Jernberg Gary R Surgical implant and method for controlled release of chemotherapeutic agents
US5199951A (en) 1990-05-17 1993-04-06 Wayne State University Method of drug application in a transporting medium to an arterial wall injured during angioplasty
AU7998091A (en) 1990-05-17 1991-12-10 Harbor Medical Devices, Inc. Medical device polymer
US5166143A (en) 1990-05-31 1992-11-24 E. R. Squibb & Sons, Inc. Method for preventing onset of restenosis after angioplasty employing an ace inhibitor
US5140012A (en) 1990-05-31 1992-08-18 E. R. Squibb & Sons, Inc. Method for preventing onset of restenosis after angioplasty employing pravastatin
US5731144A (en) 1990-06-11 1998-03-24 Nexstar Pharmaceuticals, Inc. High affinity TGFβ nucleic acid ligands
US5731424A (en) 1990-06-11 1998-03-24 Nexstar Pharmaceuticals, Inc. High affinity TGFβ nucleic acid ligands and inhibitors
US5216115A (en) 1990-06-12 1993-06-01 Rutgers, The State University Of New Jersey Polyarylate containing derivatives of the natural amino acid L-tyrosine
AU8074591A (en) 1990-06-15 1992-01-07 Cortrak Medical, Inc. Drug delivery apparatus and method
US5064435A (en) 1990-06-28 1991-11-12 Schneider (Usa) Inc. Self-expanding prosthesis having stable axial length
US5401730A (en) 1990-07-06 1995-03-28 The Hope Heart Institute Method for reducing platelet aggregation
DE4022956A1 (de) 1990-07-19 1992-02-06 Sebastian Dr Freudenberg Endoluminalschiene
EP0470569B1 (en) 1990-08-08 1995-11-22 Takeda Chemical Industries, Ltd. Intravascular embolizing agent containing angiogenesis inhibiting substance
US5189046A (en) * 1990-08-14 1993-02-23 Nova Pharmaceutical Corporation Protein kinase C modulators
US5189212A (en) * 1990-09-07 1993-02-23 University Of Georgia Research Foundation, Inc. Triarylethylene carboxylic acids with estrogenic activity
US5258020A (en) 1990-09-14 1993-11-02 Michael Froix Method of using expandable polymeric stent with memory
US5163952A (en) 1990-09-14 1992-11-17 Michael Froix Expandable polymeric stent with memory and delivery apparatus and method
US5639738A (en) 1992-02-20 1997-06-17 Hyal Pharmaceutical Corporation Treatment of basal cell carcinoma and actinic keratosis employing hyaluronic acid and NSAIDs
US5219548A (en) 1990-10-01 1993-06-15 Board Of Regents, The University Of Texas System High affinity halogenated-tamoxifen derivatives and uses thereof
US5238714A (en) 1990-10-02 1993-08-24 Board Of Regents, The University Of Texas System Efficient microcapsule preparation and method of use
US5222971A (en) 1990-10-09 1993-06-29 Scimed Life Systems, Inc. Temporary stent and methods for use and manufacture
US5180366A (en) 1990-10-10 1993-01-19 Woods W T Apparatus and method for angioplasty and for preventing re-stenosis
US5053033A (en) 1990-10-10 1991-10-01 Boston Advanced Technologies, Inc. Inhibition of restenosis by ultraviolet radiation
KR930006431B1 (ko) 1990-10-11 1993-07-16 재단법인 한국화학연구소 약물의 미세캡슐화 방법
US5486357A (en) * 1990-11-08 1996-01-23 Cordis Corporation Radiofrequency plasma biocompatibility treatment of inside surfaces
EP0557418B1 (en) 1990-11-16 2000-06-28 Celtrix Pharmaceuticals, Inc. A beta-type transforming growth factor
US5824647A (en) 1990-12-12 1998-10-20 Postlethwaite; Arnold E. Chemotactic wound healing peptides
ATE128034T1 (de) 1991-01-03 1995-10-15 Salk Inst For Biological Studi Mitotoxin zur behandlung von gefässbeschädigung.
US5102402A (en) 1991-01-04 1992-04-07 Medtronic, Inc. Releasable coatings on balloon catheters
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
CA2060067A1 (en) 1991-01-28 1992-07-29 Lilip Lau Stent delivery system
US5378475A (en) * 1991-02-21 1995-01-03 University Of Kentucky Research Foundation Sustained release drug delivery devices
US5332576A (en) 1991-02-27 1994-07-26 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
WO1992015286A1 (en) 1991-02-27 1992-09-17 Nova Pharmaceutical Corporation Anti-infective and anti-inflammatory releasing systems for medical devices
US5171217A (en) 1991-02-28 1992-12-15 Indiana University Foundation Method for delivery of smooth muscle cell inhibitors
US5280016A (en) 1991-03-29 1994-01-18 Glycomed Incorporated Non-anticoagulant heparin derivatives
JPH04312526A (ja) 1991-04-09 1992-11-04 Fujisawa Pharmaceut Co Ltd 骨疾患治療剤
US5116864A (en) 1991-04-09 1992-05-26 Indiana University Foundation Method for preventing restenosis following reconfiguration of body vessels
IT1247527B (it) 1991-04-24 1994-12-17 Medea Res Srl Agente antiarteriosclerotico, sua preparazione ed uso
US5147332A (en) 1991-05-17 1992-09-15 C.R. Bard, Inc. Multi-valve catheter for improved reliability
US5286497A (en) 1991-05-20 1994-02-15 Carderm Capital L.P. Diltiazem formulation
US5458568A (en) 1991-05-24 1995-10-17 Cortrak Medical, Inc. Porous balloon for selective dilatation and drug delivery
GB9111439D0 (en) 1991-05-28 1991-07-17 Thrombosis Res Inst Inhibition of vascular smooth muscle cell proliferation
GB9112267D0 (en) 1991-06-07 1991-07-24 Biocompatibles Ltd Polymeric coating
US5213576A (en) 1991-06-11 1993-05-25 Cordis Corporation Therapeutic porous balloon catheter
TW257762B (US06515009-20030204-C00004.png) 1991-06-14 1995-09-21 Nippon Chemicals Pharmaceutical Co Ltd
US5229495A (en) 1991-06-18 1993-07-20 Ludwig Institute For Cancer Research Substantially pure receptor like TGF-β 1 binding molecules and uses thereof
US5216126A (en) 1991-06-19 1993-06-01 Genentech, Inc. Receptor polypeptides and their production and uses
JP3446214B2 (ja) 1991-06-21 2003-09-16 ライオン株式会社 液状透明口腔用組成物
CA2079205C (en) 1992-09-25 1998-02-10 Rudolf Edgar Falk Use of hyaluronic acid and forms to prevent arterial restenosis
US5990095A (en) 1991-07-03 1999-11-23 Hyal Pharmaceutical Corporation Use of hyaluronic acid and forms to prevent arterial restenosis
US6022866A (en) 1991-07-03 2000-02-08 Hyal Pharmaceutical Corporation Use of hyaluronic acid and forms to prevent arterial restenosis
FR2679230B1 (fr) 1991-07-16 1993-11-19 Rhone Poulenc Rorer Sa Nouveaux derives d'analogues du taxol, leur preparation et les compositions qui les contiennent.
CA2074304C (en) 1991-08-02 1996-11-26 Cyril J. Schweich, Jr. Drug delivery catheter
US5356433A (en) 1991-08-13 1994-10-18 Cordis Corporation Biocompatible metal surfaces
US5166191A (en) 1991-08-19 1992-11-24 Genentech, Inc. Use of relaxin in cardiovascular therapy
US5185260A (en) 1991-08-29 1993-02-09 The United States Of America As Represented By The United States Department Of Energy Method for distinguishing normal and transformed cells using G1 kinase inhibitors
US5811447A (en) 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6515009B1 (en) 1991-09-27 2003-02-04 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
WO1994007529A1 (en) 1992-09-25 1994-04-14 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5500013A (en) 1991-10-04 1996-03-19 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
WO1993006792A1 (en) 1991-10-04 1993-04-15 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
US5464450A (en) 1991-10-04 1995-11-07 Scimed Lifesystems Inc. Biodegradable drug delivery vascular stent
CA2380683C (en) 1991-10-28 2006-08-08 Advanced Cardiovascular Systems, Inc. Expandable stents and method for making same
AU2738392A (en) 1991-11-11 1993-05-13 Ciba-Geigy Ag Novel hybrid transforming growth factors
WO1993009790A1 (en) 1991-11-11 1993-05-27 The Trustees Of The University Of Pennsylvania Methods of inhibiting restenosis
US5304325A (en) 1991-11-13 1994-04-19 Hemagen/Pfc Emulsions containing alkyl- or alkylglycerophosphoryl choline surfactants and methods of use
US5270047A (en) 1991-11-21 1993-12-14 Kauffman Raymond F Local delivery of dipyridamole for the treatment of proliferative diseases
US5238950A (en) 1991-12-17 1993-08-24 Schering Corporation Inhibitors of platelet-derived growth factor
EP0746202A4 (en) 1992-01-06 1997-06-25 Health Maintenance Programs COMPOSITION CONTAINING A PHARMACEUTICALLY ACTIVE ANTI-OXIDANT AND METHOD FOR USE IN THE PREVENTION AND TREATMENT OF RESTENOSIS AFTER ANGIOPLASTY
CA2086642C (en) * 1992-01-09 2004-06-15 Randall E. Morris Method of treating hyperproliferative vascular disease
ATE150964T1 (de) 1992-01-17 1997-04-15 Daiichi Seiyaku Co Restenosehemmer nach perkutaner koronarer arterioplastie
DE69306755T2 (de) 1992-01-21 1997-04-10 Stanford Res Inst Int Verbessertes verfahren zur herstellung von mikronisierter polypeptidarzneimitteln
US5280109A (en) * 1992-01-27 1994-01-18 Ludwig Institute For Cancer Research Isolated, large latent complexes of TGF-β2 and TGF-β3, and new binding protein for latent form TGF-β1, TGF-β2 and TGF-β3 LTBP-2
CA2087132A1 (en) 1992-01-31 1993-08-01 Michael S. Williams Stent capable of attachment within a body lumen
US5444164A (en) 1992-02-05 1995-08-22 Bristol-Myers Squibb Company TGF-β induced gene
CA2129514A1 (en) * 1992-03-12 1993-09-16 M. Amin Khan Controlled released acth containing microspheres
WO1993019177A1 (en) 1992-03-18 1993-09-30 The General Hospital Corporation FOUR NOVEL RECEPTORS OF THE TGF-β RECEPTOR FAMILY
US5510077A (en) 1992-03-19 1996-04-23 Dinh; Thomas Q. Method of making an intraluminal stent
DE69326631T2 (de) 1992-03-19 2000-06-08 Medtronic Inc Intraluminales Erweiterungsgerät
US5282823A (en) * 1992-03-19 1994-02-01 Medtronic, Inc. Intravascular radially expandable stent
US5599352A (en) 1992-03-19 1997-02-04 Medtronic, Inc. Method of making a drug eluting stent
US5571166A (en) 1992-03-19 1996-11-05 Medtronic, Inc. Method of making an intraluminal stent
US5591224A (en) * 1992-03-19 1997-01-07 Medtronic, Inc. Bioelastomeric stent
GB9207437D0 (en) * 1992-04-03 1992-05-20 Orion Yhtymae Oy Topical administration of toremifene and its metabolites
US5288711A (en) 1992-04-28 1994-02-22 American Home Products Corporation Method of treating hyperproliferative vascular disease
AU670937B2 (en) 1992-04-28 1996-08-08 Wyeth Method of treating hyperproliferative vascular disease
DE4214215A1 (de) 1992-04-30 1993-11-04 Behringwerke Ag Verwendung von inhibitoren von plasminogenaktivatoren zur behandlung von entzuendungen
WO1993024476A1 (en) 1992-06-04 1993-12-09 Clover Consolidated, Limited Water-soluble polymeric carriers for drug delivery
US5383928A (en) * 1992-06-10 1995-01-24 Emory University Stent sheath for local drug delivery
EP0575955B1 (en) 1992-06-22 1999-09-15 Kyowa Hakko Kogyo Co., Ltd. Process for producing staurosporine derivatives
JP3297469B2 (ja) 1992-06-23 2002-07-02 出光興産株式会社 電子写真感光体
US5767079A (en) 1992-07-08 1998-06-16 Celtrix Pharmaceuticals, Inc. Method of treating ophthalmic disorders using TGF -β
DE4222380A1 (de) 1992-07-08 1994-01-13 Ernst Peter Prof Dr M Strecker In den Körper eines Patienten perkutan implantierbare Endoprothese
US5283257A (en) 1992-07-10 1994-02-01 The Board Of Trustees Of The Leland Stanford Junior University Method of treating hyperproliferative vascular disease
TW366342B (en) 1992-07-28 1999-08-11 Lilly Co Eli The use of 2-phenyl-3-aroylbenzothiophenes in inhibiting bone loss
WO1994003644A1 (en) 1992-08-10 1994-02-17 Cambridge Neuroscience, Inc. Inhibitors of cell proliferation, their preparation and use
WO1994004178A1 (en) 1992-08-12 1994-03-03 Bio-Technology General Corp. Method of inhibiting cell proliferation using apolipoprotein e
US5460807A (en) 1992-08-19 1995-10-24 Merrell Dow Pharmaceuticals Inc. Antiproliferative oligomers
US6200558B1 (en) 1993-09-14 2001-03-13 The United States Of America As Represented By The Department Of Health And Human Services Biopolymer-bound nitric oxide-releasing compositions, pharmaceutical compositions incorporating same and methods of treating biological disorders using same
US5525357A (en) 1992-08-24 1996-06-11 The United States Of America As Represented By The Department Of Health And Human Services Polymer-bound nitric oxide/nucleophile adduct compositions, pharmaceutical compositions incorporating same and methods of treating biological disorders using same
US5405919A (en) 1992-08-24 1995-04-11 The United States Of America As Represented By The Secretary Of Health And Human Services Polymer-bound nitric oxide/nucleophile adduct compositions, pharmaceutical compositions and methods of treating biological disorders
US5650447A (en) 1992-08-24 1997-07-22 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Nitric oxide-releasing polymers to treat restenosis and related disorders
US5632981A (en) 1992-08-24 1997-05-27 The United States Of America As Represented By The Department Of Health And Human Services Biopolymer-bound nitric oxide-releasing compositions, pharmaceutical compositions incorporating same and methods of treating biological disorders using same
US5821234A (en) 1992-09-10 1998-10-13 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of proliferation of vascular smooth muscle cell
JP2617407B2 (ja) 1992-09-14 1997-06-04 キッセイ薬品工業株式会社 血管内膜細胞過剰増殖疾患の予防および治療剤
US5817625A (en) 1992-09-21 1998-10-06 Oncogene Science, Inc. Methods of prevention of oral mucositis with transforming growth factor beta
US5770609A (en) 1993-01-28 1998-06-23 Neorx Corporation Prevention and treatment of cardiovascular pathologies
US6306421B1 (en) 1992-09-25 2001-10-23 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6251920B1 (en) 1993-05-13 2001-06-26 Neorx Corporation Prevention and treatment of cardiovascular pathologies
US5411967A (en) 1992-10-13 1995-05-02 American Home Products Corporation Carbamates of rapamycin
US5302584A (en) 1992-10-13 1994-04-12 American Home Products Corporation Carbamates of rapamycin
CA2146973C (en) 1992-10-29 2008-09-02 Patricia R. Segarini Uses of tgf-.beta. receptor fragment as a therapeutic agent
EP0597593A1 (en) 1992-10-30 1994-05-18 Medtronic, Inc. Thromboresistant articles
US5449382A (en) 1992-11-04 1995-09-12 Dayton; Michael P. Minimally invasive bioactivated endoprosthesis for vessel repair
US5578075B1 (en) 1992-11-04 2000-02-08 Daynke Res Inc Minimally invasive bioactivated endoprosthesis for vessel repair
US5342348A (en) * 1992-12-04 1994-08-30 Kaplan Aaron V Method and device for treating and enlarging body lumens
US5346702A (en) 1992-12-04 1994-09-13 Sterling Winthrop Inc. Use of non-ionic cloud point modifiers to minimize nanoparticle aggregation during sterilization
EP0604022A1 (en) * 1992-12-22 1994-06-29 Advanced Cardiovascular Systems, Inc. Multilayered biodegradable stent and method for its manufacture
US5443458A (en) 1992-12-22 1995-08-22 Advanced Cardiovascular Systems, Inc. Multilayered biodegradable stent and method of manufacture
GB2273873A (en) 1992-12-23 1994-07-06 Univ Sheffield Treatment of psoriasis
US5354774A (en) 1992-12-24 1994-10-11 Yale University Inhibition of smooth muscle cell proliferation by 8-methoxypsoralen photoactivated by visible light
US5393527A (en) 1993-01-04 1995-02-28 Becton, Dickinson And Company Stabilized microspheres and methods of preparation
US6159946A (en) 1993-01-07 2000-12-12 Thomas Jefferson University Antisense inhibition of c-myc to modulate the proliferation of smooth muscle cells
DE69430699D1 (de) * 1993-01-08 2002-07-04 Miravant Syst Inc Medikamente abgebende stents
US5419760A (en) 1993-01-08 1995-05-30 Pdt Systems, Inc. Medicament dispensing stent for prevention of restenosis of a blood vessel
US5420243A (en) 1993-01-26 1995-05-30 Celtrix Pharmaceuticals, Inc. Biologically active TGF-β2 peptides
US6491938B2 (en) * 1993-05-13 2002-12-10 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6663881B2 (en) 1993-01-28 2003-12-16 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5981568A (en) 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5595722A (en) * 1993-01-28 1997-01-21 Neorx Corporation Method for identifying an agent which increases TGF-beta levels
AU6171194A (en) 1993-02-05 1994-08-29 Affymax Technologies N.V. Receptor-binding antiproliferative peptides
JP3466614B2 (ja) * 1993-02-12 2003-11-17 コルバス・インターナショナル、インコーポレイテッド 血栓症のインヒビター
DE4401554A1 (de) 1993-02-16 1994-08-18 Freund Andreas Präparat zur Therapie und Prophylaxe von Erkrankungen, die bei Imbalancen von Plasmalipiden auftreten
US5252579A (en) 1993-02-16 1993-10-12 American Home Products Corporation Macrocyclic immunomodulators
US5512591A (en) 1993-02-18 1996-04-30 President And Fellows Of Harvard College Treatments for diseases characterized by neovascularization
US5358959A (en) 1993-02-18 1994-10-25 President And Fellows Of Harvard University Methods for treating arteriosclerosis
US5358844A (en) 1993-02-18 1994-10-25 Brigham And Women's Hospital, Inc. Preservation of blood platelets
US5362478A (en) 1993-03-26 1994-11-08 Vivorx Pharmaceuticals, Inc. Magnetic resonance imaging with fluorocarbons encapsulated in a cross-linked polymeric shell
US5391378A (en) * 1993-02-22 1995-02-21 Elizabeth-Hata International, Inc. Two-part medicinal tablet and method of manufacture
US5441734A (en) 1993-02-25 1995-08-15 Schering Corporation Metal-interferon-alpha crystals
WO1994020096A1 (en) 1993-03-05 1994-09-15 Boehringer Mannheim Pharmaceuticals Corporation - Smithkline Beckman Corporation Limited Method of employing carbazolyl-(4)-oxypropanolamine compounds for inhibition of smooth muscle cell proliferation
US5308862A (en) 1993-03-05 1994-05-03 Boehringer Mannheim Pharmaceuticals Corporation - Smithkline Beecham Corp., Ltd. Partnership No. 1 Use of, and method of treatment using, carbazolyl-(4)-oxypropanolamine compounds for inhibition of smooth muscle cell proliferation
US5280040A (en) * 1993-03-11 1994-01-18 Zymogenetics, Inc. Methods for reducing bone loss using centchroman derivatives
US5451603A (en) * 1993-03-11 1995-09-19 Zymogenetics, Inc. 3,4-diarylchromans for treatment of dermatitis
WO1994021308A1 (en) 1993-03-18 1994-09-29 Cedars-Sinai Medical Center Drug incorporating and releasing polymeric coating for bioprosthesis
US5482949A (en) * 1993-03-19 1996-01-09 Eli Lilly And Company Sulfonate derivatives of 3-aroylbenzo[b]thiophenes
US5474563A (en) 1993-03-25 1995-12-12 Myler; Richard Cardiovascular stent and retrieval apparatus
DE69434115T2 (de) 1993-03-25 2005-10-27 Merck & Co., Inc. Inhibitor des wachstumsfaktors für gefässendothelzellen
US5607463A (en) 1993-03-30 1997-03-04 Medtronic, Inc. Intravascular medical device
AU6525894A (en) 1993-03-31 1994-10-24 George Cooper IV A method for treating abnormal cardiac contraction
EP0619314A1 (en) * 1993-04-09 1994-10-12 Eli Lilly And Company 4-Phenyl-4H- naphtho(2,1-b)pyran derivatives and their pharmaceutical use
US5523092A (en) 1993-04-14 1996-06-04 Emory University Device for local drug delivery and methods for using the same
US5399352A (en) 1993-04-14 1995-03-21 Emory University Device for local drug delivery and methods for using the same
EP0621015B1 (en) 1993-04-23 1998-03-18 Schneider (Europe) Ag Stent with a covering layer of elastic material and method for applying the layer on the stent
US5504091A (en) 1993-04-23 1996-04-02 American Home Products Corporation Biotin esters of rapamycin
US5464650A (en) 1993-04-26 1995-11-07 Medtronic, Inc. Intravascular stent and method
US5824048A (en) 1993-04-26 1998-10-20 Medtronic, Inc. Method for delivering a therapeutic substance to a body lumen
SE9301422D0 (sv) 1993-04-28 1993-04-28 Kabi Pharmacia Ab Method and means for inhibiting posterior capsule opacification
JPH08509642A (ja) 1993-04-28 1996-10-15 フォーカル,インコーポレイテッド 管腔内フォトサーモフォーミングの装置およびその方法
ATE178797T1 (de) 1993-05-03 1999-04-15 Physion Srl Neue morphinzusammensetzungen für iontophoretische verabreichung
JPH08510451A (ja) 1993-05-13 1996-11-05 ネオルックス コーポレイション 異常増殖性平滑筋細胞に関連した病因の予防及び治療
US5478860A (en) 1993-06-04 1995-12-26 Inex Pharmaceuticals Corp. Stable microemulsions for hydrophobic compound delivery
US5342926A (en) 1993-06-08 1994-08-30 The Regents Of The University Of California Analogs of cytochalasin B as radiopharmaceuticals for nuclear imaging of trans-membrane glucose transport
EP1027834A1 (en) 1993-06-11 2000-08-16 The Board of Trustees of The Leland Stanford Junior University Food composition comprising L-arginine
US5445941A (en) 1993-06-21 1995-08-29 Eli Lilly And Company Method for screening anti-osteoporosis agents
IL109990A (en) 1993-06-21 1999-06-20 Lilly Co Eli Materials and methods for screening anti-osteoporosis agents
DE4320898A1 (de) 1993-06-24 1995-01-05 Denecke Rainer Dr Med Vet Präparat zur Therapie und Prophylaxe von Erkrankungen, die bei Imbalancen von Plasmalipiden auftreten
DE4320896A1 (de) 1993-06-24 1995-01-05 Denecke Rainer Dr Med Vet Präparat zur Therapie und Prophylaxe von Demenz-Erkrankungen
DE69405491T2 (de) 1993-06-24 1998-02-19 Lilly Co Eli Antiöstrogene 2-Phenyl-3-Aroylbenzothiophene als hypoglykämische Mittel
US5994341A (en) 1993-07-19 1999-11-30 Angiogenesis Technologies, Inc. Anti-angiogenic Compositions and methods for the treatment of arthritis
EG20321A (en) * 1993-07-21 1998-10-31 Otsuka Pharma Co Ltd Medical material and process for producing the same
TW303299B (US06515009-20030204-C00004.png) 1993-07-22 1997-04-21 Lilly Co Eli
US5453492A (en) 1993-07-28 1995-09-26 La Jolla Cancer Research Foundation 60 kDa transforming growth factor-β-binding protein and its use to detect or purify TGF-β
US5422362A (en) 1993-07-29 1995-06-06 Quadra Logic Technologies, Inc. Method to inhibit restenosis
AU7476894A (en) 1993-07-29 1995-02-28 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Method of treating atherosclerosis or restenosis using microtubule stabilizing agent
US5387680A (en) * 1993-08-10 1995-02-07 American Home Products Corporation C-22 ring stabilized rapamycin derivatives
US5354801A (en) 1993-08-12 1994-10-11 Cytec Technology Corp. Process for producing small polymer phase droplet microemulsions by multistep aqueous phase addition
CA2129288C (en) 1993-08-17 2000-05-16 Jerzy Golik Phosphonooxymethyl esters of taxane derivatives
US5441947A (en) 1993-08-25 1995-08-15 Eli Lilly And Company Methods of inhibiting vascular restenosis
US5380299A (en) * 1993-08-30 1995-01-10 Med Institute, Inc. Thrombolytic treated intravascular medical device
GB9318844D0 (en) 1993-09-10 1993-10-27 Esselte Letraset Ltd Nibb units for pens
US6087479A (en) 1993-09-17 2000-07-11 Nitromed, Inc. Localized use of nitric oxide-adducts to prevent internal tissue damage
US5324739A (en) 1993-10-07 1994-06-28 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Oregon State University Compound exhibiting antiproliferative activity against cells
US6133242A (en) 1993-10-15 2000-10-17 Thomas Jefferson Univerisity Inhibition of extracellular matrix synthesis by antisense compounds directed to nuclear proto-oncogenes
US5391557A (en) * 1993-10-15 1995-02-21 Eli Lilly And Company Methods for the treatment of peri-menopausal syndrome
US5457113A (en) 1993-10-15 1995-10-10 Eli Lilly And Company Methods for inhibiting vascular smooth muscle cell proliferation and restinosis
US5461065A (en) 1993-10-15 1995-10-24 Eli Lilly And Company Methods for inhibiting endometriosis
US5462966A (en) 1993-10-15 1995-10-31 Carter-Wallace Inc. Methods for the prevention and control of cellular damage resulting from coronary artery occlusion-reperfusion
US5457116A (en) 1993-10-15 1995-10-10 Eli Lilly And Company Methods of inhibiting uterine fibrosis
CN1058390C (zh) 1993-10-15 2000-11-15 伊莱利利公司 苯并噻吩类化合物的用途
US5482950A (en) 1993-10-15 1996-01-09 Eli Lilly And Company Methods for lowering serum cholesterol
US5418252A (en) 1993-10-15 1995-05-23 Eli Lilly And Company Method for inhibiting cartilage degradation
US5441964A (en) 1993-10-15 1995-08-15 Eli Lilly And Company Methods for inhibiting bone loss using substituted benzothiophene
WO1995010989A1 (en) 1993-10-19 1995-04-27 Scimed Life Systems, Inc. Intravascular stent pump
CA2117967A1 (en) 1993-10-27 1995-04-28 Thomas W. Sander Tissue repair device and apparatus and method for fabricating same
US5411988A (en) 1993-10-27 1995-05-02 Bockow; Barry I. Compositions and methods for inhibiting inflammation and adhesion formation
US5480904A (en) * 1993-10-28 1996-01-02 Eli Lilly And Company Methods for inhibiting uterine fibrosis
JPH09508892A (ja) 1993-11-17 1997-09-09 マサチューセッツ インスティテュート オブ テクノロジー ヘパリナーゼを用いる血管形成の阻害方法
US5436243A (en) 1993-11-17 1995-07-25 Research Triangle Institute Duke University Aminoanthraquinone derivatives to combat multidrug resistance
US5393772A (en) 1993-11-24 1995-02-28 Boehringer Mannheim Pharmaceuticals Corporation Use of, and method of treatment using, hydroxycarbazole compounds for inhibition of smooth muscle migration and proliferation
HU213407B (en) 1993-12-09 1997-06-30 Egyt Gyogyszervegyeszeti Gyar Process for producing tablet with diffusive-osmotic release
US5552415A (en) 1993-12-21 1996-09-03 Eli Lilly And Company Method of inhibiting Alzheimer's Disease
US5462949A (en) 1993-12-21 1995-10-31 Eli Lilly And Company Methods of inhibiting fertility in women
US5492927A (en) 1993-12-21 1996-02-20 Eli Lilly And Company Non-peptide tachykinin receptor antagonists to treat allergy
US5593987A (en) 1993-12-21 1997-01-14 Eli Lilly And Company Methods of inhibiting breast disorders
US5461064A (en) 1993-12-21 1995-10-24 Eli Lilly And Company Methods of inhibiting atrophy of the skin and vagina
US5521198A (en) 1993-12-21 1996-05-28 Eli Lilly And Company Methods of inhibiting autoimmune diseases
US5441966A (en) 1993-12-21 1995-08-15 Eli Lilly And Company Methods of inhibiting Turner's syndrome
US5389670A (en) * 1993-12-21 1995-02-14 Eli Lilly Company Methods of inhibiting the symptoms of premenstrual syndrome/late luteal phase dysphoric disorder
US6417198B1 (en) 1993-12-21 2002-07-09 Eli Lilly And Company Methods of inhibiting CNS problems in post-menopausal women
US5574047A (en) 1993-12-21 1996-11-12 Eli Lilly And Company Methods of inhibiting imperfect tissue repair
US5441965A (en) 1993-12-21 1995-08-15 Eli Lilly And Company Methods of inhibiting thrombin
US5708009A (en) 1993-12-21 1998-01-13 Eli Lilly And Company Methods of inhibiting myeloperoxidase activity
US5451590A (en) 1993-12-21 1995-09-19 Eli Lilly & Co. Methods of inhibiting sexual precocity
US5447941A (en) 1993-12-21 1995-09-05 Eli Lilly And Company Methods of inhibiting pulmonary hypertensive diseases with raloxifene and related benzothiophenes
US5451589A (en) 1993-12-21 1995-09-19 Eli Lilly And Company Methods of inhibiting ovarian dysgenesis, delayed puberty, or sexual infantilism
EP0963756B1 (en) 1993-12-21 2004-02-25 Eli Lilly And Company Inhibition of advanced glycosylation end products
US5439923A (en) 1993-12-21 1995-08-08 Eli Lilly And Company Method of inhibiting seborrhea and acne
US5534526A (en) 1993-12-21 1996-07-09 Eli Lilly And Company Methods for inhibiting vasomotor symptoms and attending psychological disturbances surrounding post-menopausal syndrome
US5462950A (en) 1993-12-21 1995-10-31 Eli Lilly And Company Methods of treating menstrual symptoms and compositions therefore
US5439931A (en) 1993-12-21 1995-08-08 Eli Lilly And Company Method for increasing libido in post-menopausal women
US5446053A (en) 1993-12-21 1995-08-29 Eli Lilly And Company Methods of inhibiting dysfunctional uterine bleeding
US5519042A (en) 1994-01-13 1996-05-21 Hoechst Aktiengesellschaft Method of treating hyperproliferative vascular disease
US5591753A (en) 1994-01-28 1997-01-07 Eli Lilly And Company Combination treatment for osteoporosis
US5407955A (en) 1994-02-18 1995-04-18 Eli Lilly And Company Methods for lowering serum cholesterol and inhibiting smooth muscle cell proliferation, restenosis, endometriosis, and uterine fibroid disease
ZA951497B (en) 1994-03-02 1996-08-23 Lilly Co Eli Orally administerable pharmaceutical formulations
US5478847A (en) 1994-03-02 1995-12-26 Eli Lilly And Company Methods of use for inhibiting bone loss and lowering serum cholesterol
US5484772A (en) 1994-03-04 1996-01-16 Eli Lilly And Company Antithrombotic agents
US5451414A (en) 1994-03-15 1995-09-19 Heritage Environmental Services, Inc. Micronutrient supplement
US6461381B2 (en) 1994-03-17 2002-10-08 Medinol, Ltd. Flexible expandable stent
US5843120A (en) 1994-03-17 1998-12-01 Medinol Ltd. Flexible-expandable stent
US5733303A (en) 1994-03-17 1998-03-31 Medinol Ltd. Flexible expandable stent
US5525610A (en) 1994-03-31 1996-06-11 American Home Products Corporation 42-Epi-rapamycin and pharmaceutical compositions thereof
CA2145614A1 (en) 1994-03-31 1995-10-01 Jeffrey A. Dodge Intermediates and processes for preparing benzothiophene compounds
US5362718A (en) 1994-04-18 1994-11-08 American Home Products Corporation Rapamycin hydroxyesters
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US5384332A (en) * 1994-05-11 1995-01-24 Eli Lilly And Company Methods for inhibiting aortal smooth muscle cell proliferation and restenosis with 1,1,2-triphenylbut-1-ene derivatives
US5455275A (en) 1994-05-11 1995-10-03 Eli Lilly And Company Methods for inhibiting endometriosis and uterine fibroid disease with 1,1,2-triphenylbut-1-ene derivatives
US5426123A (en) 1994-05-11 1995-06-20 Eli Lilly And Company Method for lowering serum cholesterol with 1,1,2-triphenylbut-1-ene derivatives
US5470883A (en) 1994-05-23 1995-11-28 Stromberg; Brent V. Method of treating peripheral vasoconstriction with tamoxifen citrate
US5580898A (en) 1994-05-24 1996-12-03 The Trustees Of The University Of Pennsylvania Method of stabilizing microtubules
JPH07316192A (ja) 1994-05-27 1995-12-05 Hoechst Japan Ltd L−リシル−グリシル−l−ヒスチジンおよびこれを含有する創傷治療剤
US5656450A (en) 1994-05-27 1997-08-12 Board Of Regents, The University Of Texas System Activation of latent transforming growth factor β by matrix vesicles
US5466810A (en) 1994-06-10 1995-11-14 Eli Lilly And Company 2-amino-3-aroyl-benzo[β]thiophenes and methods for preparing and using same to produce 6-hydroxy-2-(4-hydroxyphenyl)-3-[4-(2-aminoethoxy)-benzoyl]benzo[β]thiophenes
US5424331A (en) 1994-06-10 1995-06-13 Bio-Virus Research Incorporated Pharmaceutical compositions and dietary soybean food products for the prevention of osteoporosis
US6492106B1 (en) 1994-06-27 2002-12-10 The Johns Hopkins University Mammalian proteins that bind to FKBP12 in a rapamycin-dependent fashion
US5629077A (en) 1994-06-27 1997-05-13 Advanced Cardiovascular Systems, Inc. Biodegradable mesh and film stent
US6476200B1 (en) 1994-06-27 2002-11-05 The Johns Hopkins University Mammalian proteins that bind to FKBP12 in a rapamycin-dependent fashion
US5441986A (en) 1994-07-19 1995-08-15 Pfizer Inc. Estrogen agonists as remedies for prostate and cardiovascular diseases
US5434166A (en) 1994-08-22 1995-07-18 Eli Lilly And Company Methods of inhibiting demyelinating and desmyelinating diseases
US5496828A (en) 1994-08-22 1996-03-05 Eli Lilly And Company Methods of inhibiting ulcerative mucositis
US5491159A (en) * 1994-08-30 1996-02-13 American Home Products Corporation 2-(3,5-di-tert-butyl-4-hydroxy-phenyl)-oxazoles as anti-atherosclerotic agents
US5731436A (en) 1994-08-31 1998-03-24 Eli Lilly And Company Process for preparing benzoic acid derivative intermediates and benzothiophene pharmaceutical agents
US5660873A (en) 1994-09-09 1997-08-26 Bioseal, Limited Liability Corporaton Coating intraluminal stents
US7501441B1 (en) * 1994-09-20 2009-03-10 Eli Lilly And Company Naphthyl compounds, intermediates, processes, compositions, and methods
US6399634B1 (en) * 1994-09-20 2002-06-04 Eli Lilly And Company Benzothiophene compounds, compositions, and methods
US5649977A (en) 1994-09-22 1997-07-22 Advanced Cardiovascular Systems, Inc. Metal reinforced polymer stent
US5583153A (en) 1994-10-06 1996-12-10 Regents Of The University Of California Use of taxol in the treatment of rheumatoid arthritis
US5521191A (en) 1994-10-17 1996-05-28 Washington University Method for treatment of arterial stenosis
US5643580A (en) 1994-10-17 1997-07-01 Surface Genesis, Inc. Biocompatible coating, medical device using the same and methods
US5516807A (en) 1994-10-25 1996-05-14 Warner-Lambert Company Method for treating vascular proliferative disorders following balloon angioplasty
US5498775A (en) 1994-11-07 1996-03-12 American Home Products Corporation Polyanionic benzylglycosides as inhibitors of smooth muscle cell proliferation
US5563145A (en) 1994-12-07 1996-10-08 American Home Products Corporation Rapamycin 42-oximes and hydroxylamines
CA2163837C (en) 1994-12-13 1999-07-20 Robert K. Perrone Crystalline paclitaxel hydrates
US5637113A (en) 1994-12-13 1997-06-10 Advanced Cardiovascular Systems, Inc. Polymer film for wrapping a stent structure
US5700559A (en) 1994-12-16 1997-12-23 Advanced Surface Technology Durable hydrophilic surface coatings
US5489587A (en) * 1995-01-20 1996-02-06 Eli Lilly And Company Benzofurans used to inhibit bone loss
US5571808A (en) 1995-01-31 1996-11-05 Eli Lilly And Company Method for treating smoking-related bone loss
US5484808A (en) 1995-02-09 1996-01-16 Eli Lilly And Company Methods of inhibiting cell-cell adhesion
US5510357A (en) * 1995-02-28 1996-04-23 Eli Lilly And Company Benzothiophene compounds as anti-estrogenic agents
US5605696A (en) * 1995-03-30 1997-02-25 Advanced Cardiovascular Systems, Inc. Drug loaded polymeric material and method of manufacture
US5563054A (en) 1995-03-31 1996-10-08 Eli Lilly And Company Process for preparation of benzo[B]thiophene glucuronides
US6120536A (en) 1995-04-19 2000-09-19 Schneider (Usa) Inc. Medical devices with long term non-thrombogenic coatings
US20020091433A1 (en) 1995-04-19 2002-07-11 Ni Ding Drug release coated stent
US5837313A (en) 1995-04-19 1998-11-17 Schneider (Usa) Inc Drug release stent coating process
US6099562A (en) 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
KR19990007865A (ko) 1995-04-19 1999-01-25 스피겔알렌제이 약물 방출용 피복 스텐트
US5688855A (en) 1995-05-01 1997-11-18 S.K.Y. Polymers, Inc. Thin film hydrophilic coatings
US5622975A (en) 1995-06-01 1997-04-22 Eli Lilly And Company Methods for inhibiting vascular smooth muscle cell migration
US5639274A (en) 1995-06-02 1997-06-17 Fischell; Robert E. Integrated catheter system for balloon angioplasty and stent delivery
US5609629A (en) * 1995-06-07 1997-03-11 Med Institute, Inc. Coated implantable medical device
CA2178541C (en) 1995-06-07 2009-11-24 Neal E. Fearnot Implantable medical device
US5877224A (en) 1995-07-28 1999-03-02 Rutgers, The State University Of New Jersey Polymeric drug formulations
US5726186A (en) 1995-09-08 1998-03-10 Eli Lilly And Company Pentacyclic compounds, intermediates, processes, compositions, and methods
US6001622A (en) 1995-12-21 1999-12-14 Sunnybrook Health Science Centre Integrin-linked kinase and its use
US5722984A (en) 1996-01-16 1998-03-03 Iso Stent, Inc. Antithrombogenic radioactive coating for an intravascular stent
US6783543B2 (en) 2000-06-05 2004-08-31 Scimed Life Systems, Inc. Intravascular stent with increasing coating retaining capacity
US5797898A (en) 1996-07-02 1998-08-25 Massachusetts Institute Of Technology Microchip drug delivery devices
US6086910A (en) 1997-09-19 2000-07-11 The Howard Foundation Food supplements
US5980972A (en) 1996-12-20 1999-11-09 Schneider (Usa) Inc Method of applying drug-release coatings
US5863285A (en) * 1997-01-30 1999-01-26 Cordis Corporation Balloon catheter with radioactive means
US5994388A (en) 1997-03-18 1999-11-30 The Children's Medical Center Corporation Cytochalasin and isoindolinone derivatives as inhibitors of angiogenesis
US5824054A (en) 1997-03-18 1998-10-20 Endotex Interventional Systems, Inc. Coiled sheet graft stent and methods of making and use
US5779732A (en) 1997-03-31 1998-07-14 Medtronic, Inc. Method and apparatus for implanting a film with an exandable stent
EP2292225B9 (en) 1997-03-31 2012-06-27 Boston Scientific Scimed Limited Dosage form comprising taxol in crystalline form
US5948639A (en) 1997-04-10 1999-09-07 Millennium Pharmaceuticals, Inc. TGF-β pathway genes
US5879697A (en) 1997-04-30 1999-03-09 Schneider Usa Inc Drug-releasing coatings for medical devices
US5976067A (en) 1997-05-28 1999-11-02 Ablation Technologies, Inc. Combination radioactive and temperature self-regulating thermal seed implant for treating tumors
US6106454A (en) 1997-06-17 2000-08-22 Medtronic, Inc. Medical device for delivering localized radiation
US6203536B1 (en) 1997-06-17 2001-03-20 Medtronic, Inc. Medical device for delivering a therapeutic substance and method therefor
US6110483A (en) 1997-06-23 2000-08-29 Sts Biopolymers, Inc. Adherent, flexible hydrogel and medicated coatings
US6309414B1 (en) 1997-11-04 2001-10-30 Sorin Biomedica Cardio S.P.A. Angioplasty stents
SE9704401D0 (sv) 1997-11-28 1997-11-28 Astra Ab Matrix pellets for greasy, oily or sticky drug substances
KR100228188B1 (ko) 1997-12-24 1999-11-01 김성년 방사성 스텐트 및 그의 제조방법
EP1043949A2 (en) 1997-12-31 2000-10-18 Pharmasonics, Inc. Methods and systems for the inhibition of vascular hyperplasia
US6241762B1 (en) 1998-03-30 2001-06-05 Conor Medsystems, Inc. Expandable medical device with ductile hinges
US6099499A (en) 1998-04-28 2000-08-08 Medtronic, Inc. Device for in vivo radiation delivery and method for delivery
US6013099A (en) * 1998-04-29 2000-01-11 Medtronic, Inc. Medical device for delivering a water-insoluble therapeutic salt or substance
US6093142A (en) 1998-04-30 2000-07-25 Medtronic Inc. Device for in vivo radiation delivery and method for delivery
US6129757A (en) 1998-05-18 2000-10-10 Scimed Life Systems Implantable members for receiving therapeutically useful compositions
US6168619B1 (en) * 1998-10-16 2001-01-02 Quanam Medical Corporation Intravascular stent having a coaxial polymer member and end sleeves
US6198016B1 (en) 1998-12-01 2001-03-06 3M Innovative Properties Company Wet skin adhesive article
ATE346156T1 (de) 1999-09-17 2006-12-15 Whitehead Biomedical Inst Umkehrtransfektionsverfahren
AU770395B2 (en) 1999-11-17 2004-02-19 Boston Scientific Limited Microfabricated devices for the delivery of molecules into a carrier fluid
US6491617B1 (en) 1999-12-30 2002-12-10 St. Jude Medical, Inc. Medical devices that resist restenosis
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
US6764507B2 (en) 2000-10-16 2004-07-20 Conor Medsystems, Inc. Expandable medical device with improved spatial distribution
US6656216B1 (en) 2001-06-29 2003-12-02 Advanced Cardiovascular Systems, Inc. Composite stent with regioselective material
US6497647B1 (en) 2001-07-18 2002-12-24 Ati Medical, Inc. Radiation and thermal energy source
US20030065377A1 (en) 2001-09-28 2003-04-03 Davila Luis A. Coated medical devices
US20040236416A1 (en) 2003-05-20 2004-11-25 Robert Falotico Increased biocompatibility of implantable medical devices

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994016706A1 (en) * 1993-01-28 1994-08-04 Neorx Corporation Therapeutic inhibitors of vascular smooth muscle cells
WO1994026291A1 (en) * 1993-05-13 1994-11-24 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
WO1995003036A1 (en) * 1993-07-19 1995-02-02 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US5429634A (en) * 1993-09-09 1995-07-04 Pdt Systems Biogenic implant for drug delivery and method
EP0691130A1 (en) * 1994-05-12 1996-01-10 American Home Products Corporation Use of rapamycin in the manufacture of a medicament for preventing and heating hyperproliferactive vascular diseases, eventually in combination with mycophenolic acid

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BIOTECHNOL. BIOACT. POLYM., [PROC. AM. CHEM. SOC. SYMP.] (1994), MEETING DATE 1992, 259-68. EDITOR(S): GEBELEIN, CHARLES G.;CARRAHER, CHARLES E., JR. PUBLISHER: PLENUM, NEW YORK, N. Y. CODEN: 60QOAU, 1994 *
CARDIOVASC. RES. (1993), 27(12), 2200-4 CODEN: CVREAU;ISSN: 0008-6363, 1993 *
CHEMICAL ABSTRACTS, vol. 120, no. 22, 30 May 1994, Columbus, Ohio, US; abstract no. 279947, MORE, RANJIT S ET AL: "A targeted antithrombotic conjugate with antiplatelet and fibrinolytic properties which reduces in vivo thrombus formation" XP002008097 *
CHEMICAL ABSTRACTS, vol. 121, no. 22, 28 November 1994, Columbus, Ohio, US; abstract no. 263625, LEVY, ROBERT J. ET AL: "Strategies for treating arterial restenosis using polymeric controlled release implants" XP002008098 *

Cited By (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7862605B2 (en) 1995-06-07 2011-01-04 Med Institute, Inc. Coated implantable medical device
US8945206B2 (en) 1995-06-07 2015-02-03 Cook Medical Technologies Llc Methods for making implantable medical devices
US6774278B1 (en) 1995-06-07 2004-08-10 Cook Incorporated Coated implantable medical device
US20110046723A1 (en) * 1995-06-07 2011-02-24 Bates Brian L Coated implantable medical device
US8469943B2 (en) * 1995-06-07 2013-06-25 Cook Medical Technologies Llc Coated implantable medical device
US5977163A (en) * 1996-03-12 1999-11-02 Pg-Txl Company, L. P. Water soluble paclitaxel prodrugs
US7384977B2 (en) 1996-03-12 2008-06-10 Pg-Txl Company, L.P. Water soluble paclitaxel prodrugs
US6441025B2 (en) 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US6884817B2 (en) 1996-03-12 2005-04-26 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US7060724B2 (en) 1996-03-12 2006-06-13 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US7135496B2 (en) 1996-03-12 2006-11-14 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
EP2098230A1 (en) * 1997-03-31 2009-09-09 Boston Scientific Scimed Limited Use of cytoskeletal inhibitors in crystalline form for the inhibition or prevention of restenosis
WO1998043618A3 (en) * 1997-03-31 1998-11-05 Neorx Corp Use of cytoskeletal inhibitors for the prevention of restenosis
EP2292225A1 (en) * 1997-03-31 2011-03-09 Boston Scientific Scimed Limited Dosage form comprising taxol in crystalline form
EP1512398A1 (en) * 1997-03-31 2005-03-09 Boston Scientific Limited Intravascular stent with cytoskeletal inhibitors for the prevention of restenosis
WO1998043618A2 (en) * 1997-03-31 1998-10-08 Neorx Corporation Use of cytoskeletal inhibitors for the prevention of restenosis
WO1998056312A1 (en) * 1997-06-13 1998-12-17 Scimed Life Systems, Inc. Stents having multiple layers of biodegradable polymeric composition
SG165156A1 (en) * 1997-06-27 2010-10-28 Abraxis Bioscience Llc Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
WO1999008729A1 (en) * 1997-08-13 1999-02-25 Boston Scientific Limited Loading and release of water-insoluble drugs
US6306166B1 (en) 1997-08-13 2001-10-23 Scimed Life Systems, Inc. Loading and release of water-insoluble drugs
US6485514B1 (en) 1997-12-12 2002-11-26 Supergen, Inc. Local delivery of therapeutic agents
US8052734B2 (en) 1998-03-30 2011-11-08 Innovational Holdings, Llc Expandable medical device with beneficial agent delivery mechanism
US8623068B2 (en) 1998-03-30 2014-01-07 Conor Medsystems, Inc. Expandable medical device with ductile hinges
US8439968B2 (en) 1998-03-30 2013-05-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US8052735B2 (en) 1998-03-30 2011-11-08 Innovational Holdings, Llc Expandable medical device with ductile hinges
US8361537B2 (en) 1998-03-30 2013-01-29 Innovational Holdings, Llc Expandable medical device with beneficial agent concentration gradient
WO2000006244A3 (en) * 1998-07-30 2000-06-15 James F Hainfeld Loading metal particles into cell membrane vesicles and metal particle use for imaging and therapy
US6730064B2 (en) 1998-08-20 2004-05-04 Cook Incorporated Coated implantable medical device
WO2000012147A1 (en) * 1998-09-02 2000-03-09 Scimed Life Systems, Inc. Drug delivery device for stent
WO2000032238A1 (en) * 1998-12-03 2000-06-08 Scimed Life Systems, Inc. Stent having drug crystals thereon
US6120847A (en) * 1999-01-08 2000-09-19 Scimed Life Systems, Inc. Surface treatment method for stent coating
US6333347B1 (en) 1999-01-29 2001-12-25 Angiotech Pharmaceuticals & Advanced Research Tech Intrapericardial delivery of anti-microtubule agents
WO2000044443A3 (en) * 1999-01-29 2001-08-16 Angiotech Pharm Inc Intra-pericardial delivery of anti-microtubule agents
WO2000044443A2 (en) * 1999-01-29 2000-08-03 Angiotech Pharmaceuticals Inc. Intra-pericardial delivery of anti-microtubule agents
US6419692B1 (en) 1999-02-03 2002-07-16 Scimed Life Systems, Inc. Surface protection method for stents and balloon catheters for drug delivery
US6656156B2 (en) 1999-02-03 2003-12-02 Scimed Life Systems, Inc. Dual surface protection coating for drug delivery balloon catheters and stents
US6322847B1 (en) 1999-05-03 2001-11-27 Boston Scientific, Inc. Medical device coating methods and devices
US6156373A (en) * 1999-05-03 2000-12-05 Scimed Life Systems, Inc. Medical device coating methods and devices
EP1221997A4 (en) * 1999-10-06 2006-05-31 Penn State Res Found SYSTEM AND DEVICE FOR PREVENTING RESTENOSIS IN BLOOD VESSELS
EP1221997A1 (en) * 1999-10-06 2002-07-17 The Penn State Research Foundation System and device for preventing restenosis in body vessels
WO2001036007A2 (en) * 1999-11-12 2001-05-25 Angiotech Pharmaceuticals, Inc. Compositions of a combination of radioactive therapy and cell-cycle inhibitors
WO2001036007A3 (en) * 1999-11-12 2002-07-04 Angiotech Pharm Inc Compositions of a combination of radioactive therapy and cell-cycle inhibitors
US7173041B2 (en) 2000-03-17 2007-02-06 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US7153864B2 (en) 2000-03-17 2006-12-26 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
WO2001070295A1 (en) * 2000-03-22 2001-09-27 Zenon Kyriakides Coronary artery stent covered with endothelin receptor antagonist
US7265199B2 (en) 2000-04-11 2007-09-04 Celonova Biosciences Germany Gmbh Poly-tri-fluoro-ethoxypolyphosphazene coverings and films
US9694162B2 (en) 2000-10-31 2017-07-04 Cook Medical Technologies Llc Coated medical device
US8974522B2 (en) 2000-10-31 2015-03-10 Cook Medical Technologies Llc Coated medical device
US9814865B2 (en) 2000-10-31 2017-11-14 Cook Medical Technologies Llc Coated medical device
US9080146B2 (en) 2001-01-11 2015-07-14 Celonova Biosciences, Inc. Substrates containing polyphosphazene as matrices and substrates containing polyphosphazene with a micro-structured surface
US8097236B2 (en) 2001-02-22 2012-01-17 Psimedica Limited Devices and methods for the treatment of cancer
US9066990B2 (en) 2001-03-26 2015-06-30 Bayer Intellectual Property Gmbh Preparation for restenosis prevention
US8389043B2 (en) 2001-03-26 2013-03-05 Bayer Pharma Aktiengesellschaft Preparation for restenosis prevention
US8101275B2 (en) 2001-08-17 2012-01-24 Celonova Biosciences, Inc. Device based on nitinol, a process for its production, and its use
US7842083B2 (en) 2001-08-20 2010-11-30 Innovational Holdings, Llc. Expandable medical device with improved spatial distribution
US7850727B2 (en) 2001-08-20 2010-12-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
WO2003041756A1 (en) * 2001-11-08 2003-05-22 Dsb Invest Holding Sa Endoluminal devices coated with latrunculin to prevent ingrowth of cells
US10532190B2 (en) 2002-07-12 2020-01-14 Cook Medical Technologies Llc Coated medical device
US9302080B2 (en) 2002-09-20 2016-04-05 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US9216272B2 (en) 2002-09-20 2015-12-22 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US9687635B2 (en) 2002-09-20 2017-06-27 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US9649476B2 (en) 2002-09-20 2017-05-16 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US8349390B2 (en) 2002-09-20 2013-01-08 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US9254202B2 (en) 2002-09-20 2016-02-09 Innovational Holdings Llc Method and apparatus for loading a beneficial agent into an expandable medical device
US9216273B2 (en) 2002-09-20 2015-12-22 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
EP2216055B1 (de) 2002-09-20 2017-07-26 Bayer Intellectual Property GmbH Medizinische Vorrichtung zur Arnzeimittelabgabe
US8197881B2 (en) 2003-09-22 2012-06-12 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US10973770B2 (en) 2004-10-25 2021-04-13 Varian Medical Systems, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US9114162B2 (en) 2004-10-25 2015-08-25 Celonova Biosciences, Inc. Loadable polymeric particles for enhanced imaging in clinical applications and methods of preparing and using the same
US9597419B2 (en) 2004-10-25 2017-03-21 Boston Scientific Limited Loadable polymeric particles for enhanced imaging in clinical applications and methods of preparing and using the same
US9107850B2 (en) 2004-10-25 2015-08-18 Celonova Biosciences, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US9107899B2 (en) 2005-03-03 2015-08-18 Icon Medical Corporation Metal alloys for medical devices
US7922764B2 (en) 2006-10-10 2011-04-12 Celonova Bioscience, Inc. Bioprosthetic heart valve with polyphosphazene
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
US9724497B2 (en) 2008-05-01 2017-08-08 Bayer Intellectual Property Gmbh Method of coating a catheter balloon having a fold
US9126025B2 (en) 2008-05-01 2015-09-08 Bayer Intellectual Property Gmbh Method of coating a folded catheter 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
US10080821B2 (en) 2009-07-17 2018-09-25 Boston Scientific Scimed, Inc. Nucleation of drug delivery balloons to provide improved crystal size and density
US9034245B2 (en) 2010-03-04 2015-05-19 Icon Medical Corp. Method for forming a tubular medical device
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
US11779685B2 (en) 2014-06-24 2023-10-10 Mirus Llc Metal alloys for medical devices
US11766506B2 (en) 2016-03-04 2023-09-26 Mirus Llc Stent device for spinal fusion

Also Published As

Publication number Publication date
CA2559392A1 (en) 1996-08-22
DE69632310T2 (de) 2004-09-09
US20120195951A1 (en) 2012-08-02
US8097642B2 (en) 2012-01-17
US20070148207A1 (en) 2007-06-28
JP2007075623A (ja) 2007-03-29
ATE265233T1 (de) 2004-05-15
EP1669091A2 (en) 2006-06-14
US6599928B2 (en) 2003-07-29
US20020040064A1 (en) 2002-04-04
EP0809515B1 (en) 2004-04-28
US8158670B2 (en) 2012-04-17
DE69635968T2 (de) 2007-01-25
ES2217306T3 (es) 2004-11-01
JPH11500635A (ja) 1999-01-19
DE69632310D1 (de) 2004-06-03
US20110300221A1 (en) 2011-12-08
AU4985196A (en) 1996-09-04
US6171609B1 (en) 2001-01-09
ATE321573T1 (de) 2006-04-15
US20070134314A1 (en) 2007-06-14
CA2212537A1 (en) 1996-08-22
DE69635968D1 (de) 2006-05-18
EP1407786B1 (en) 2006-03-29
EP0809515A1 (en) 1997-12-03
EP1669091A3 (en) 2009-06-17
US6515009B1 (en) 2003-02-04
EP1407786A1 (en) 2004-04-14
CA2212537C (en) 2006-12-19

Similar Documents

Publication Publication Date Title
US6171609B1 (en) Therapeutic inhibitor of vascular smooth muscle cells
US8067022B2 (en) Therapeutic inhibitor of vascular smooth muscle cells
EP0752885B1 (en) Therapeutic inhibitor of vascular smooth muscle cells
US6358989B1 (en) Therapeutic inhibitor of vascular smooth muscle cells
US6663881B2 (en) Therapeutic inhibitor of vascular smooth muscle cells
US6569441B2 (en) Therapeutic inhibitor of vascular smooth muscle cells
US6306421B1 (en) Therapeutic inhibitor of vascular smooth muscle cells
CA2154698C (en) Therapeutic inhibitor of vascular smooth muscle cells
US20030083733A1 (en) Therapeutic inhibitor of vascular smooth muscle cells
EP1350523B1 (en) Therapeutic conjugates inhibiting vascular smooth muscle cells
EP1826280A2 (en) Therapeutic conjugates inhibiting vascular smooth muscle cells

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AZ BY KG KZ RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE

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

Ref document number: 2212537

Country of ref document: CA

Ref country code: CA

Ref document number: 2212537

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1996 525163

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996906490

Country of ref document: EP

ENP Entry into the national phase

Ref country code: US

Ref document number: 1997 894350

Date of ref document: 19971010

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1996906490

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 1996906490

Country of ref document: EP