CA2536042A1 - Medical implants and anti-scarring agents - Google Patents

Medical implants and anti-scarring agents Download PDF

Info

Publication number
CA2536042A1
CA2536042A1 CA002536042A CA2536042A CA2536042A1 CA 2536042 A1 CA2536042 A1 CA 2536042A1 CA 002536042 A CA002536042 A CA 002536042A CA 2536042 A CA2536042 A CA 2536042A CA 2536042 A1 CA2536042 A1 CA 2536042A1
Authority
CA
Canada
Prior art keywords
agent
inhibitor
coating
scarring
polymeric carrier
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002536042A
Other languages
French (fr)
Inventor
David M. Gravett
William L. Hunter
Richard T. Liggins
Arpita Maiti
Pierre E. Signore
Philip M. Toleikis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Angiotech International AG
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2536042A1 publication Critical patent/CA2536042A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/11Surgical instruments, devices or methods, e.g. tourniquets for performing anastomosis; Buttons for anastomosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/12Surgical instruments, devices or methods, e.g. tourniquets for ligaturing or otherwise compressing tubular parts of the body, e.g. blood vessels, umbilical cord
    • A61B17/12022Occluding by internal devices, e.g. balloons or releasable wires
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/12Surgical instruments, devices or methods, e.g. tourniquets for ligaturing or otherwise compressing tubular parts of the body, e.g. blood vessels, umbilical cord
    • A61B17/12022Occluding by internal devices, e.g. balloons or releasable wires
    • A61B17/12131Occluding by internal devices, e.g. balloons or releasable wires characterised by the type of occluding device
    • A61B17/12181Occluding by internal devices, e.g. balloons or releasable wires characterised by the type of occluding device formed by fluidized, gelatinous or cellular remodelable materials, e.g. embolic liquids, foams or extracellular matrices
    • A61B17/1219Occluding by internal devices, e.g. balloons or releasable wires characterised by the type of occluding device formed by fluidized, gelatinous or cellular remodelable materials, e.g. embolic liquids, foams or extracellular matrices expandable in contact with liquids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/02Details
    • A61N1/04Electrodes
    • A61N1/05Electrodes for implantation or insertion into the body, e.g. heart electrode
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2250/00Special features of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2250/0058Additional features; Implant or prostheses properties not otherwise provided for
    • A61F2250/0067Means for introducing or releasing pharmaceutical products into the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/404Biocides, antimicrobial agents, antiseptic agents
    • 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
    • A61L2300/404Biocides, antimicrobial agents, antiseptic agents
    • A61L2300/406Antibiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/426Immunomodulating agents, i.e. cytokines, interleukins, interferons
    • 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
    • A61L2300/432Inhibitors, antagonists

Abstract

Published without an Abstract

Description

DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter 1e Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS
THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

MEDICAL IMPLANTS AND ANTI-SCARRING AGENTS
BACKGROUND OF THE INVENTION
Field of the Invention .
The present invention relates generally to pharmaceutical compositions, methods and devices, and more specifically, to compositions and methods for preparing and using medical implants to make them resistant to overgrowth by inflammatory and fibrous scar tissue.
Description of the Related Art The clinical function of numerous medical implants and devices is dependent upon the device being able to effectively maintain an anatomical, or surgically created, space or passageway. Unfortunately, many devices ' implanted in the body are subject to a "foreign body" response from the surrounding host tissues. In particular, injury to tubular anatomical structures (such as blood vessels, the gastrointestinal tract, the male and female reproductive tract, the urinary tract, sinuses, spinal nerve root canals, lacrimal ducts, Eustachian tubes, the auditory canal, and the respiratory tract) from surgery and/or injury created by the implantation of medical devices can lead to a well known clinical problem called "stenosis" (or narrowing). Stenosis occurs in response to trauma to the epithelial lining or the entire body tube during the procedure, including virtually any manipulation which attempts to relieve obstruction of the passageway, and is a major factor limiting the effectiveness of invasive treatments for a variety of diseases to be described later.
Stenosis (or "restenosis" if the problem recurs after an initially successful attempt to open a blocked passageway) is a form of response to injury leading to wall thickening, narrowing of the lumen, and loss of function in the tissue supplied by the particular passageway. Physical injury during an interventional procedure results in damage to epithelial lining of the tube and the smooth muscle cells (SMCs) that make up the wall. The damaged cells, particularly SMCs, release cytokines, which recruit inflammatory cells such as macrophages, lymphocytes and neutrophils (i.e., which are some of the known white blood cells) into the area. The white blood cells in turn release a variety of additional cytokines, growth factors, and tissue degrading enzymes that influence the behavior of the constituent cells of the wall (primarily epithelial cells and SMCs). Stimulation of the SMCs induces them to migrate into the inner aspect of the body passageway (often called the "intima"), proliferate and secrete an extracellar matrix - effectively filling all or parts of the lumen with reactive, fibrous scar tissue. Collectively, this creates a thickening of the intimal layer (known in some tissues as "neointimal hyperplasia") that narrows the lumen of the passageway and can be significant enough to obstruct its lumen.
The present invention discloses pharmaceutical agents which inhibit one or more aspects of the production of excessive fibrous (scar) tissue.
Furthermore, compositions and methods are described for coating medical devices and implants with drug-delivery compositions such that the pharmaceutical agent is delivered in therapeutic levels over a period sufficient to allow normal healing to occur. And finally, numerous specific implants and devices are described that produce superior clinical results as a result of being coated with agents that reduce excessive scarring and fibrous tissue accumulation as well as other related advantages.
BRIEF SUMMARY OF THE INVENTION
Briefly stated, in one aspect, the present invention provides compositions for delivery of selected therapeutic agents via medical implants or implantable medical devices, as well as methods for making and using these implants and devices. Within one aspect of the invention, drug-coated or drug-impregnated implants and medical devices are provided which reduce fibrosis in the tissue surrounding the device or implant, or inhibit scar development on the device/implant surface, thus enhancing the efficacy the procedure. Within various embodiments, fibrosis is inhibited by local or systemic release of specific pharmacological agents that become localized to the adjacent tissue.
The repair of tissues following a mechanical or surgical intervention involves two distinct processes: (1 ) regeneration (the replacement of injured cells by cells of the same type and (2) fibrosis (the replacement of injured cells by connective tissue). There are four general components to the process of fibrosis (or scarring) including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). Within one embodiment of the invention, an implant or device is adapted to release an
2 agent that inhibits fibrosis or regeneration through one or more of the mechanisms sited herein.
Within yet other aspects of the present invention, methods are provided for manufacturing a medical device or implant, comprising the step of coating (e.g., spraying, dipping, wrapping, or administering drug through) a medical device or implant. Additionally, the implant or medical device can be constructed so that the device itself is comprised of materials which inhibit fibrosis in or around the implant. A wide variety of medical devices and implants may be utilized within the context of the present invention, depending on the site and nature of treatment desired.
Within related aspects of the present invention, vascular stents, gastrointestinal stents, tracheal/bronchial stents, genital-urinary stents, ENT
stents, intraocular lenses, implants for hypertrophic scars and keloids, vascular grafts, anastomotic connector devices, surgical adhesion barriers, glaucoma drainage devices, prosthetic heart valves, tympanostomy tubes, penile implants, CVCs, ventricular assist devices (e.g., LVAD's), spinal prostheses, endotracheal and tracheostomy tubes, peritoneal dialysis catheters, intracranial pressure monitors, vena cava filters, and gastrointestinal drainage tubes are provided comprising an implant or device, wherein the implant or device is in combination with an agent which inhibits fibrosis in vivo.
Within various embodiments of the invention, the implant or device is further coated with a composition or compound, which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation. Representative examples of such agents include heparin, PLGA/MePEG, PLA, and polyethylene glycol. Within further embodiments the fibrosis-inhibiting implant or device is activated before, during, or after deployment (e.g., an inactive agent on the device is first activated to one that reduces or inhibits an in vivo fibrotic reaction).
Within various embodiments of the invention, a device or implant is coated on one aspect, portion or surface with a composition which inhibits fibrosis, as well as being coated with a composition or compound which promotes scarring on another aspect, portion or surface of the device.
Representative examples of agents that promote fibrosis and scarring include silk, wool, silica, bleomycin, neomycin, talcum powder, metallic beryllium, and copper as well as analogues and derivatives thereof.
3 Also provided by the present invention are methods for treating patients undergoing surgical, endoscopic or minimally invasive therapies where a medical device or implant is placed as part of the procedure. As utilized herein, it should be understood that "inhibits fibrosis or stenosis" refers to a statistically significant decrease in the amount of scar tissue in or around the device or an improvement in the luminal area of the device/implant, which may or may not result in a permanent prohibition of any complications or failures of the device/implant.
The pharmaceutical agents and compositions are utilized to create novel drug-coated implants and medical devices that reduce the foreign body response to implantation and limit the growth of reactive tissue on the surface of, or around in the tissue surrounding the device, such that performance is enhanced. In many instances, the devices are used to maintain body lumens or passageways such as blood vessels, the gastrointestinal tract, the male and female reproductive tract, the urinary tract, bony foramena (e.g., sinuses, spinal nerve root canals, lacrimal ducts, Eustachian tubes, the auditory canal), and the respiratory tract, where obstruction of the device by scar tissue in the post-procedural period leads to the adverse clinical sequela or failure of the intervention. Medical devices and implants coated with selected pharmaceutical agents designed to prevent scar tissue overgrowth and preserve patency can offer significant clinical advantages over uncoated devices.
For example, in one aspect the present invention is directed to devices that comprise a medical implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti-scarring agent. The agent is present so as to inhibit scarring that can otherwise occur when the implant is placed within an animal. In another aspect the present invention is directed to methods wherein both an implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti-scarring agent, are placed into an animal, and the agent inhibits scarring that can otherwise occur. These and other aspects of the invention are summarized below.
Thus, in various independent aspects, the present invention provides the following: a device, comprising a gastrointestinal implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an inferior vena cava filter implant and an anti-scarring agent or a composition comprising an anti-scarring
4
5 PCT/US2004/037426 agent, wherein the agent inhibits scarring; a device, comprising a central nervous system shunt implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a pressure monitoring implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a peritoneal dialysis catheter implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an endotracheal tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a tracheostomy tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a tympanostomy tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; device, comprising a prosthetic heart valve implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a glaucoma drainage implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an implant that provides a surgical adhesion barrier and~an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an anastomotic connector implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a sensing implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an implant for pericardial treatment of coronary artery disease and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising vascular graft implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an implant for the treatment of a hypertrophic scar and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an implant for the treatment of a keloid and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an intraocular lens implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an ENT stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an genital-urinary stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a tracheal/bronchial stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring, a device, comprising GI stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring. These and other devices are described in more detail herein.
In.each of the aforementioned devices, in separate aspects the present invention provides that: the agent is a cell cycle inhibitor; the agent is an anthracycline; the agent is a taxane; the agent is a podophyllotoxin; the agent is an immunomodulator; the agent is a heat shock protein 90 antagonist;
the agent is a HMGCoA reductase inhibitor; the agent is an inosine monophosphate dehydrogenase inhibitor; the agent is an NF kappa B inhibitor;
the agent is a p38 MAP kinase inhibitor. These and other agents are described in more detail herein.
In additional aspects, for each of the aforementioned devices combined with each of the aforementioned agents, it is, for each combination, independently disclosed that the agent may be present in a composition along with a polymer. In one embodiment of this aspect, the polymer is biodegradable. In another embodiment of this aspect, the polymer is non-biodegradable. Other features and characteristics of the polymer, which may serve to describe the present invention for every combination of device and agent described above, are set forth in greater detail herein.
In addition to devices, the present invention also provides methods. For example, in additional aspects of the present invention, for each of the aforementioned devices, and for each of the aforementioned combinations of the devices with the anti-scarring agents, the present invention provides methods whereby a specified device is implanted into an animal, and a specified agent associated with the device inhibits scarring that can otherwise occur. Each of the devices identified herein may be a "specified device", and each of the anti-scarring agents identified herein may be an "anti-scarring
6 agent", where the present invention provides, in independent embodiments, for each possible combination of the device and the agent.
The agent may be associated with the device prior to the device being placed within the animal. For example, the agent (or composition comprising the agent) may be coated onto an implant, and the resulting device then placed within the animal. In addition, or alternatively, the agent may be independently placed within the animal in the vicinity of where the device is to be, or is being, placed within the animal. For example, the agent may be sprayed or otherwise placed onto the tissue that will be contacting the medical implant or may otherwise undergo scarring. To this end, the present invention provides, in independent aspects: a method for inhibiting scarring comprising placing a gastrointestinal implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an inferior vena cava filter implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a central nervous system shunt implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a pressure monitoring implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a peritoneal dialysis catheter implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an endotracheal tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a tracheostomy tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a tympanostomy tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring
7 comprising placing a prosthetic heart valve implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a glaucoma drainage implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a pressure monitoring implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring;
a method for inhibiting scarring comprising placing a drug delivery pump implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an anastomotic connector implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a sensing implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an implant for pericardial treatment of coronary artery disease and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a vascular graft implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an implant for the treatment of a hypertrophic scar and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an implant for the treatment of a keloid and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an intraocular lens implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an ENT stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a genital-urinary stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein
8 the agent inhibits scarring; a method for inhibiting scarring comprising placing a tracheal/bronchial stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal hosts wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a GI
stent implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring In each of the aforementioned methods, in separate aspects, the present invention provides that: the agent is a cell cycle inhibitor; the agent is an anthracycline; the agent is a taxane; the agent is a podophyllotoxin; the agent is an immunomodulator; the agent is a heat shock protein 90 antagonist;
the agent is a HMGCoA reductase inhibitor; the agent is an inosine monophosphate dehydrogenase inhibitor; the agent is an NF kappa B inhibitor;
the agent is a p3i3 MAP kinase inhibitor. These and other agents are described in more detail herein.
In additional aspects, for each of the aforementioned methods used in combination with each of the aforementioned agents, it is, for each combination, independently disclosed that the agent may be present in a composition along with a polymer. In one embodiment of this aspect, the polymer is biodegradable. In another embodiment of this aspect, the polymer is non-biodegradable. Other features and characteristics of the polymer, which may serve to describe the present invention for every combination of device and agent described above, are set forth in greater detail herein.
These and other aspects of the present invention will become evident upon reference to the following detailed description and attached drawings. In addition, various references are set forth herein which describe in more detail certain procedures and/or compositions (e.g., polymers), and are therefore incorporated by reference in the entirety.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a diagram showing how a cell cycle inhibitor acts at one or more of the steps in the biological pathway.
Figure 2 is graph showing the results of a screening assay for assessing the effect of mitoxantrone (mitoxantrone ICSO=20 nM) on proliferation of human fibroblasts.
Figure 3 is a picture that shows an uninjured carotid artery from a rat balloon injury model.
9 Figure 4 is a picture that shows an injured carotid artery from a rat balloon injury model.
Figure 5 is a picture that shows a paclitaxel/mesh treated carotid artery in a rat balloon injury model (345 ~.g paclitaxel in a 50:50 PLG
coating on a 10:90 PLG mesh).
Figure 6A schematically depicts the transcriptional regulation of matrix metalloproteinases.
Figure 6B is a blot which demonstrates that IL-1 stimulates AP-1 transcriptional activity.
Figure 6C is a graph which shows that IL-1 induced binding activity decreased in lysates from chondrocytes which were pretreated with paclitaxel.
Figure 6D is a blot which shows that IL-1 induction increases collagenase and stromelysin in RNA levels in chondrocytes, and that this induction can be inhibited by pretreatment with paclitaxel.
Figures 7A-H are blots that show the effect of various anti-microtubule agents in inhibiting collagenase expression.
Figure 8 is a graph showing the results of a screening assay for assessing the effect of paclitaxel on smooth muscle cell migration (paclitaxel ICSO=0.76 nM).
Figure 9 is a graph showing the results of a screening assay for assessing the effect of geldanamycin on IL-1 (3 production by macrophages (ICSO=20 nM for IL-1 ~3 production by THP-1 cells).
Figure 10 is a graph showing the results of a screening assay for assessing the effect of geldanamycin on IL-8 production by macrophages (ICSO=27 nM for IL-8 production by THP-1 cells).
Figure 11 is a graph showing the results of a screening assay for assessing the effect of geldanamycin on MCP-1 production by macrophages (ICSO=7 nM for MCP-1 production by THP-1 cells).
Figure 12 is a graph showing the results for the screening assay for assessing the effect of mitoxantrone on nitric oxide production by macrophages.
Figure 13 is a graph showing the results for the screening assay for assessing the effect of various therapeutic agents on TNF-alpha production by macrophages.

Figure 14 is graph showing the results of a screening assay for assessing the effect of rapamycin on cell proliferation of human fibroblasts.
Figure 15 is a graph showing the results for the screening assay for assessing the effect of rapamycin concentration for TNFa production by THP-1 cells.
Figure 16 is graph showing the results of a screening assay for assessing the effect of paclitaxel on proliferation of smooth muscle cells.
Figure 17 is graph showing the results of a screening assay for assessing the effect of paclitaxel on cell proliferation of human fibroblasts.
Figure 18 is graph showing the results of a screening assay for assessing the effect of paclitaxel (ICSO=134 nM) for proliferation of the murine RAW 264.7 macrophage cell line.
DETAILED DESCRIPTION OF THE INVENTION
Definitions Prior to setting forth the invention, it may be helpful to an understanding thereof to first set forth definitions of certain terms that are used herein.
Any concentration ranges, percentage range, or ratio range recited herein are to be understood to include concentrations, percentages or ratios of any integer within that range and fractions thereof, such as one tenth and one hundredth of an integer, unless otherwise indicated. Also, any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness, are to be understood to include any integer within the recited range, unless otherwise indicated. It should be understood that the terms "a"
and "an" as used above and elsewhere herein refer to "one or more" of the enumerated components. For example, "a" polymer refers to both one polymer or a mixture comprising two or more polymers As used herein, the term "about"
means ~ 15%.
"Fibrosis," "Scarring," or "Fibrotic Response" refers to the formation of fibrous tissue in response to injury or medical intervention.
Therapeutic agents which inhibit fibrosis or scarring are referred to herein as "fibrosis-inhibiting agents", "anti-scarring agents", and the like, where these agents inhibit fibrosis through one or more mechanisms including: inhibiting angiogenesis, inhibiting migration or proliferation of connective tissue cells (such as fibroblasts, smooth muscle cells, vascular smooth muscle cells), reducing ECM production, and/or inhibiting tissue remodeling.
"Host", "Person", "Subject", "Patient" and the like are used synonymously to refer to the living being into which a device of the present invention is implanted.
"Implanted" refers to having completely or partially placed a device within a host. A device is partially implanted when some of the device reaches, or extends to the outside of, a host.
"Inhibit fibrosis", "reduce fibrosis" and the like are used synonymously to refer to the action of agents or compositions which result in a statistically significant decrease in the formation of fibrous tissue that can be expected to occur in the absence of the agent or composition.
"Inhibitor" refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence ~of a biological process. The process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
"Antagonist" refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. While the process may be a general one, typically this refers to a drug mechanism where the drug competes with a molecule for an active molecular site or prevents a molecule from interacting with the molecular site. In these situations, the effect is that the molecular process is inhibited.
"Agonist" refers to an agent which stimulates a biological process or rate or degree of occurrence of a biological process. The process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
"Anti-microtubule Agents" should be understood to include any protein, peptide, chemical, or other molecule which impairs the function of microtubules, for example, through the prevention or stabilization of polymerization. Compounds that stabilize polymerization of microtubules are referred to herein as "microtubule stabilizing agents." A wide variety of methods may be utilized to determine the anti-microtubule activity of a particular compound, including for example, assays described by Smith et al.
(Cancer Lett 79(2):213-219, 1994) and Mooberry et al., (Cancer Lett. 96(2):261-266, 1995).

"Medical Device", "Implant", "Medical Device or Implant", "implant/device" and the like are used synonymously to refer to any object that is designed to be placed partially or wholly within a patient's body for one or more therapeutic or prophylactic purposes such as for restoring physiological function, alleviating symptoms associated with disease, delivering therapeutic agents, andlor repairing or replacing or augmenting etc. damaged or diseased organs and tissues. While normally composed of biologically compatible synthetic materials (e.g., medical-grade stainless steel, titanium and other metals; polymers such as polyurethane, silicon, PLA, PLGA and other materials) that are exogenous, some medical devices and implants include materials derived from animals (e.g., "xenografts" such as whole animal organs;
animal tissues such as heart valves; naturally occurring or chemically-modified molecules such as collagen, hyaluronic acid, proteins, carbohydrates and others), human donors (e.g., "allografts" such as whole organs; tissues such as bone grafts, skin grafts and others), or from the patients themselves (e.g., "autografts" such as saphenous vein grafts, skin grafts, tendon/ligament/muscle transplants). Medical devices of particular utility in the present invention include, but are not restricted to, vascular stents, gastrointestinal stents, tracheal/bronchial stents, genital-urinary stents, ENT stents, intraocular lenses, implants for hypertrophic scars and keloids, vascular grafts, anastomotic connector devices, surgical adhesion barriers, glaucoma drainage devices, film or mesh, prosthetic heart valves, tympanostomy tubes, penile implants, endotracheal and tracheostomy tubes, peritoneal dialysis catheters, intracranial pressure monitors, vena cava filters, CVCs, ventricular assist device (e.g., LVAD), spinal prostheses, and gastrointestinal drainage tubes.
"Release of an agent" refers to a statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device.
"Biodegradable" refers to materials for which the degradation process is at least partially mediated by, and/or performed in, a biological system. "Degradation" refers to a chain scission process by which a polymer chain is cleaved into oligomers and monomers. Chain scission may occur through various mechanisms, including, for example, by chemical reaction (e.g., hydrolysis) or by a thermal or photolytic process. Polymer degradation may be characterized, for example, using gel permeation chromatography (GPC), which monitors the polymer molecular mass changes during erosion and drug release. Biodegradable also refers to materials may be degraded by an erosion process mediated by, and/or performed in, a biological system. "Erosion"
refers to a process in which material is lost from the bulk. In the case of a polymeric system, the material may be a monomer, an oligomer, a part of a polymer backbone, or a part of the polymer bulk. Erosion includes (i) surface erosion, in which erosion affects only the surface and not the inner parts of a matrix;
and (ii) bulk erosion, in which the entire system is rapidly hydrated and polymer chains are cleaved throughout the matrix. Depending on the type of polymer, erosion generally occurs by one of three basic mechanisms (see, e.g., Heller, J., CRC Critieal Review in Therapeutic Drug Carrier Systems (1984), 1 (1 ), 39-90); Siepmann, J. et al., Adv. Drug Del. Rev. (2001 ), 48, 229-247): (1 ) water-soluble polymers that have been insolubilized by covalent cross-links and that solubilize as the cross-links or the backbone undergo a hydrolytic cleavage;
(2) polymers that are initially water insoluble are solubilized by hydrolysis, ionization, or pronation of a pendant group; and (3) hydrophobic polymers are converted to small water-soluble molecules by backbone cleavage.
Techniques for characterizing erosion include thermal analysis (e.g., DSC), X-ray diffraction, scanning electron microscopy (SEM), electron paramagnetic resonance spectroscopy (EPR), NMR imaging, and recording mass loss during an erosion experiment. For microspheres, photon correlation spectroscopy (PCS) and other particles size measurement techniques may be applied to monitor the size evolution of erodible devices versus time.
As used herein, "analogue" refers to a chemical compound that is structurally similar to a parent compound, but differs slightly in composition (e.g., one atom or functional group is different, added, or removed). The analogue may or may not have different chemical or physical properties than the original compound and may or may not have improved biological and/or chemical activity. For example, the analogue may be more hydrophilic or it may have altered reactivity as compared to the parent compound. The analogue may mimic the chemical and/or biologically activity of the parent compound (i.e., it may have similar or identical activity), or, in some cases; may have increased or decreased activity. The analogue may be a naturally or non-naturally occurring (e.g., recombinant) variant of the original compound. An example of an analogue is a mutein (i.e., a protein analogue in which at least one amino acid is deleted, added, or substituted with another amino acid).
Other types of analogues include isomers (enantiomers, diasteromers, and the like) and other types of chiral variants of a compound, as well as structural isomers. The analogue may be a branched or cyclic variant of a linear compound. For example, a linear compound may have an analogue that is branched or otherwise substituted to impart certain desirable properties (e.g., improve hydrophilicity or bioavailability).
As used herein, "derivative" refers to a chemically or biologically modified version of a chemical compound that is structurally similar to a parent compound and (actually or theoretically) derivable from that parent compound.
A "derivative" differs from an "analogue" in that a parent compound may be the starting material to generate a "derivative," whereas the parent compound may not necessarily be used as the starting material to generate an "analogue." A
derivative may or may not have different chemical or physical properties of the parent compound. For example, the derivative may be more hydrophilic or it may have altered reactivity as compared to the parent compound.
Derivatization (i.e., modification) may involve substitution of one or more moieties within the molecule (e.g., a change in functional group). For example, a hydrogen may be substituted with a halogen, such as fluorine or chlorine, or a hydroxyl group (-OH) may be replaced with a carboxylic acid moiety (-COOH).
The term "derivative" also includes conjugates, and prodrugs of a parent compound (i.e., chemically modified derivatives which can be converted into the original compound under physiological conditions). For example, the prodrug may be an inactive form of an active agent. Under physiological conditions, the prodrug may be converted into the active form of the compound. Prodrugs may be formed, for example, by replacing one or two hydrogen atoms on nitrogen atoms by an acyl group (acyl prodrugs) or a carbamate group (carbamate prodrugs). More detailed information relating to prodrugs is found, for example, in Fleisher et al., Advanced Drug Delivery Reviews 19 (1996) 115; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; or H. Bundgaard, Drugs of the Future 16 (1991 ) 443. The term "derivative" is also used to describe all solvates, for example hydrates or adducts (e.g., adducts with alcohols), active metabolites, and salts of the parent compound. The type of salt that may be prepared depends on the nature of the moieties within the compound. For example, acidic groups, for example carboxylic acid groups, can form, for example, alkali metal salts or alkaline earth metal salts (e.g., sodium salts, potassium salts, magnesium salts and calcium salts, and also salts with physiologically tolerable quaternary ammonium ions and acid addition salts with ammonia and physiologically tolerable organic amines such as, for example, triethylamine, ethanolamine or tris-(2-hydroxyethyl)amine). Basic groups can form acid addition salts, for example with inorganic acids such as hydrochloric acid, sulfuric acid or phosphoric acid, or with organic carboxylic acids and sulfonic acids such as acetic acid, citric acid, benzoic acid, malefic acid, fumaric acid, tartaric acid, methanesulfonic acid or p-toluenesulfonic acid. Compounds which simultaneously contain a basic group and an acidic group, for example a carboxyl group in addition to basic nitrogen atoms, can be present as zwitterions. Salts can be obtained by customary methods known to those skilled in the art, for example by combining a compound with an inorganic or organic acid or base in a solvent or diluent, or from other salts by cation exchange or anion exchange.
As discussed above, the present invention provides compositions, methods and devices relating to medical implants, which greatly increase the ability to inhibit the formation of reactive scar tissue on, or around, the surface of the device or implant. Described in more detail below are methods for constructing medical implants, compositions and methods for generating medical implants which inhibit fibrosis, and methods for utilizing such medical implants.
A. Medicallmplants In one aspect, medical implants of the present invention are coated with, or otherwise adapted to release an agent which inhibits the formation of scar tissue. Representative examples of medical implants include:
vascular stents, gastrointestinal stents, tracheal/bronchial stents, genital-urinary stents, ENT stents, intraocular lenses, implants for hypertrophic scars and keloids, vascular grafts, anastomotic connector devices, pacemaker leads, CVCs, films and meshes, ventricular assists devices, spinal prostheses, surgical adhesion barriers, glaucoma drainage devices, prosthetic heart valves, tympanostomy tubes, penile implants, endotracheal and tracheostomy tubes, peritoneal dialysis catheters, intracranial pressure monitors, vena cava filters, and gastrointestinal drainage tubes.
B. Therapeutic Agents Suitable fibrosis or stenosis-inhibiting agents may be readily determined based upon the in vitro and in vivo (animal) models such as those provided in Examples 26-36. The assay set forth in Example 29 may be used to determine whether an agent is able to inhibit cell proliferation in fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC5° for inhibition of cell proliferation within a range of about 10-6 to about 10-0 M. The assay set forth in Example 33 may be used to determine whether an agent may inhibit migration of fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC5° for inhibition of cell migration within a range of about 10-6 to about 10'9M. Assays set forth herein may be used to determine whether an agent is able to inhibit inflammatory processes, including nitric oxide production in macrophages (Example 26), and/or TNF-alpha production by macrophages (Example 27), and/or IL-1 beta production by macrophages (Example 34), and/or IL-8 production by macrophages (Example 35), and/or inhibition of MCP-1 by macrophages (Example 36). In one aspect of the invention, the agent has an IC5° for inhibition of any one of these inflammatory processes within a range of about 10-6 to about 10-'°M.
The assay set forth in Example 31 may be used to determine whether an agent is able to inhibit MMP production. In one aspect of the invention, the agent has an IC5° for inhibition of MMP production within a range of about 10-4 to about
10-$M. The assay set forth in Example 39 (also known as the CAM assay) may be used to determine whether an agent is able to inhibit angiogenesis. In one aspect of the invention, the agent has an ICSO for inhibition of angiogenesis within a range of about 10'6 to about 10-~°M. Agents which inhibit fibrosis can also be identified through in vivo models including inhibition of intimal hyperplasia development in the rat balloon carotid artery model (Example 30) and/or a reduction of surgical adhesions formation in rabbit surgical adhesions model (Example 28).
Numerous therapeutic compounds have been identified that are of utility in the invention including:
1 ) Angioaenesis Inhibitors In one embodiment, the pharmacologically active compound is an angiogenesis inhibitor (e.g., 2-ME (NSC-659853), PI-88 (D-mannose, O-6-O-phosphono-alpha-D-mannopyranosyl-(1-3)-O-alpha-D-mannopyranosyl-(1-3)-O-alpha-D-mannopyranosyl-(1-3)-O-alpha-D-mannopyranosyl-(1-2)- hydrogen sulphate), thalidomide (1H-isoindole-1,3(2H)-dione, 2-(2,6-dioxo-3-piperidinyl)-), CDC-394, CC-5079, ENMD-0995 (S-3-amino-phthalidoglutarimide), AVE-8062A, vatalanib, SH-268, halofuginone hydrobromide, atiprimod dimaleate (2-azaspivo[4.5]decane-2-propanamine, N,N-diethyl-8,8-dipropyl, dimaleate), ATN-224, CHIR-258, combretastatin A-4 (phenol, 2-methoxy-5-[2-(3,4,5-trimethoxyphenyl)ethenyl]-, (Z)-), GCS-100LE, or an analogue or derivative thereof).
2) 5-Lipoxyaenase Inhibitors and Antagonists In another embodiment, the pharmacologically active compound is a 5-lipoxygenase inhibitor or antagonist (e.g., Wy-50295 (2-naphthaleneacetic acid, alpha-methyl-6-(2-quinolinylmethoxy)-, (S)-), ONO-LP
269 (2,11,14-eicosatrienamide, N-(4-hydroxy-2-(1H-tetrazol-5-yl)-8-quinolinyl)-, (E,Z,Z)-), licofelone (1 H-pyrrolizine-5-acetic acid, 6-(4-chlorophenyl)-2,3 dihydro-2,2-dimethyl-7-phenyl-), CMI-568 (urea, N-butyl-N-hydroxy-N'-(4-(3-(methylsulfonyl)-2-propoxy-5-(tetrahydro-5-(3,4,5-trimethoxyphenyl)-2-furanyl)phenoxy)butyl)-,trans-), IP-751 ((3R,4R)-(delta 6)-THC-DMH-11-oic acid), PF-5901 (benzenemethanol, alpha-pentyl-3-(2-quinolinylmethoxy)-), LY-293111 (benzoic acid, 2-(3-(3-((5-ethyl-4'-fluoro-2-hydroxy(1,1'-biphenyl)-4-yl)oxy)propoxy)-2-propylphenoxy)-), RG-5901-A (benzenemethanol, alpha-pentyl-3-(2-quinolinylmethoxy)-, hydrochloride), rilopirox (2(1 H)-pyridinone, ((4-(4-chlorophenoxy)phenoxy)methyl)-1-hydroxy-4-methyl-), L-674636 (acetic acid, ((4-(4-chlorophenyl)-1-(4-(2-quinolinylmethoxy)phenyl)butyl)thio)-AS)), ((3-(4-methoxy-tetrahydro-2H-pyran-4-yl)phenyl)methoxy)-4-phenylnaphtho(2,3-c)furan-1 (3H)-one, MK-886 (1 H-indole-2-propanoic acid, 1-((4-chlorophenyl)methyl)-3-((1,1-dimethylethyl)thio)-alpha, alpha-dimethyl-5-(1-methylethyl)-), quiflapon (1 H-indole-2-propanoic acid, 1-((4-chlorophenyl)methyl)-3-((1,1-dimethylethyl)thio)-alpha, alpha-dimethyl-5-(2-quinolinylmethoxy)-), quiflapon (1 H-Indole-2-propanoic acid, 1-((4-chlorophenyl)methyl)-3-((1,1-dimethylethyl)thio)-alpha, alpha-dimethyl-5-(2-quinolinylmethoxy)-), docebenone (2,5-cyclohexadiene-1,4-dione, 2-(12-hydroxy-5,10-dodecadiynyl)-3,5,6-trimethyl-), zileuton (urea, N-(1-benzo(b)thien-2-ylethyl)-N-hydroxy-), or an analogue or derivative thereof).
3) Chemokine Receptor Antagonists CCR (1, 3, and 5) In another embodiment, the pharmacologically active compound is a chemokine receptor antagonist which inhibits one or more subtypes of CCR
(1, 3, and 5) (e.g., ONO-4128 (1,4,9-triazaspiro(5.5)undecane-2,5-dione, 1-butyl-3-(cyclohexylmethyl)-9-((2,3-dihydro-1,4-benzodioxin-6-yl)methyl-), L-381, CT-112 (L-arginine, L-threonyl-L-threonyl-L-seryl-L-glutaminyl-L-valyl-L-arginyl-L-prolyl-), AS-900004, SCH-C, ZK-811752, PD-172084, UK-427857, SB-380732, vMIP II, SB-265610, DPC-168, TAK-779 (N, N-dimethyl-N-(4-(2-(4-methylphenyl)-6,7-dihydro-5H-benzocyclohepten-8-ylcarboxamido)benyl)tetrahydro-2H-pyran-4-aminium chloride), TAK-220, KRH-1120), GSK766994, SSR-150106, or an analogue or derivative thereof). Other examples of chemokine receptor antagonists include a-Immunokine-NNS03, B7C-471, CCX-282, Sch-350634; Sch-351125; Sch-417690; SCH-C, and analogues and derivatives thereof.
4) Cell Cycle Inhibitors In another embodiment, the pharmacologically active compound is a cell cycle inhibitor. Representative examples of such agents include taxanes (e.g., paclitaxel (discussed in more detail below) and docetaxel) (Schiff et al., Nature 277:665-667, 1979; Long and Fairchild, Cancer Research 54:4355-4361, 1994; Ringel and Horwitz, J. Nat'I Cancer Inst. 83(4):288-291, 1991; Pazdur et al., Caneer Treat. Rev. 79(40):351-386, 1993), etanidazole, nimorazole (B.A. Chabner and D.L. Longo. Cancer Chemotherapy and Biotherapy - Principles and Practice. Lippincott-Raven Publishers, New York, 1996, p.554), perfluorochemicals with hyperbaric oxygen, transfusion, erythropoietin, BW12C, nicotinamide, hydralazine, BSO, WR-2721, IudR, DUdR, etanidazole, WR-2721, BSO, mono-substituted keto-aldehyde compounds (L.G. Egyud. Keto-aldehyde-amine addition products and method of making same. U.S. Patent No. 4,066,650, Jan 3, 1978), nitroimidazole (K.C.
Agrawal and M. Sakaguchi. Nitroimidazole radiosensitizers for Hypoxic tumor cells and compositions thereof. U.S. Patent No. 4,462,992, Jul. 31, 1984), 5-substituted-4-nitroimidazoles (Adams et al., Int. J. Radiat. Biol. Relat.
Stud.
Phys., Chem. Med. 40(2):153-61, 1981 ), SR-2508 (Brown et al., Int. J. Radiat.
Oncol., Biol. Phys. 7(6):695-703, 1981 ), 2H-isoindolediones (J.A. Myers, 2H-Isoindolediones, the synthesis and use as radiosensitizers. Patent 4,494,547, Jan. 22, 1985), chiral (((2-bromoethyl)-amino)methyl)-nitro-1 H-imidazole-1-ethanol (V.G. Beylin, et al., Process for preparing chiral (((2-bromoethyl)-amino)methyl)-nitro-1 H-imidazole-1-ethanol and related compounds. U.S.
Patent No. 5,543,527, Aug. 6, 1996; U.S. Patent No. 4,797,397; Jan. 10, 1989;
U.S. Patent No. 5,342,959, Aug. 30, 1994), nitroaniline derivatives (W.A.

Denny, et al. Nitroaniline derivatives and the use as anti-tumor agents. U.S.
Patent No. 5,571,845, Nov. 5, 1996), DNA-affinic hypoxia selective cytotoxins (M.V. Papadopoulou-Rosenzweig. DNA-affinic hypoxia selective cytotoxins.
U.S. Patent No. 5,602,142, Feb. 11, 1997), halogenated DNA ligand (R.F.
Martin. Halogenated DNA ligand radiosensitizers for cancer therapy. U.S.
Patent No. 5,641,764, Jun 24, 1997), 1,2,4 benzotriazine oxides (W.W. Lee et al. 1,2,4-benzotriazine oxides as radiosensitizers and selective cytotoxic agents. U.S. Patent No. 5,616,584, Apr. 1, 1997; U.S. Patent No. 5,624,925, Apr. 29, 1997; Process for Preparing 1,2,4 Benzotriazine oxides. U.S. Patent No. 5,175,287, Dec. 29, 1992), nitric oxide (J.B. Mitchell et al., Use of Nitric oxide releasing compounds as hypoxic cell radiation sensitizers. U.S. Patent No. 5,650,442, Jul. 22, 1997), 2-nitroimidazole derivatives (M.J. Suto et al.

Nitroimidazole derivatives useful as radiosensitizers for hypoxic tumor cells.
U.S. Patent No. 4,797,397, Jan. 10, 1989; T. Suzuki. 2-Nitroimidazole derivative, production thereof, and radiosensitizer containing the same as active ingredient. U.S. Patent No. 5,270,330, Dec. 14, 1993; T. Suzuki et al. 2-Nitroimidazole derivative, production thereof, and radiosensitizer containing the same as active ingredient. U.S. Patent No. 5,270,330, Dec 14, 1993; T.
Suzuki. 2-Nitroimidazole derivative, production thereof and radiosensitizer containing the same as active ingredient; Patent EP 0 513 351 B1, Jan. 24, 1991 ), fluorine-containing nitroazole derivatives (T. Kagiya. Fluorine-containing nitroazole derivatives and radiosensitizer comprising the same. U.S. Patent No. 4,927,941, May 22, 1990), copper (M.J. Abrams. Copper Radiosensitizers.
' U.S. Patent No. 5,100,885, Mar. 31, 1992), combination modality cancer therapy (D.H. Picker et al. Combination modality cancer therapy. U.S. Patent No. 4,681,091, Jul. 21, 1987). 5-CIdC or (d)H4U or 5-halo-2'-halo-2'-deoxy-cytidine or -uridine derivatives (S.B. Greer. Method and Materials for sensitizing neoplastic tissue to radiation. U.S. Patent No. 4,894,364 Jan. 16, 1990), platinum complexes (K.A. Skov. Platinum Complexes with one radiosensitizing ligand. U.S. Patent No. 4,921,963. May 1, 1990; K.A. Skov.
Platinum Complexes with one radiosensitizing ligand. Patent EP 0 287 317 A3), fluorine-containing nitroazole (T. Kagiya, et al. Fluorine-containing nitroazole derivatives and radiosensitizer comprising the same. U.S. Patent No. 4,927,941. May 22,1990), benzamide (W.W. Lee. Substituted Benzamide Radiosensitizers. U.S. Patent No. 5,032,617, Jul. 16, 1991 ), autobiotics (L.G.
Egyud. Autobiotics and the use in eliminating nonself cells in vivo. U.S.
Patent No. 5,147,652. Sep. 15,1992), benzamide and nicotinamide (W.W. Lee et al.
Benzamide and Nictoinamide Radiosensitizers. U.S. Patent No. 5,215,738, Jun 1 1993), acridine-intercalator (M. Papadopoulou-Rosenzweig. Acridine Intercalator based hypoxia selective cytotoxins. U.S. Patent No. 5,294,715, Mar. 15,1994), fluorine-containing nitroimidazole (T. Kagiya et al. Fluorine containing nitroimidazole compounds. U.S. Patent No. 5,304,654, Apr. 19, 1994), hydroxylated texaphyrins (J.L. Sessler et al. Hydroxylated texaphrins.
U.S. Patent No. 5,457,183, Oct. 10, 1995), hydroxylated compound derivative (T. Suzuki et al. Heterocyclic compound derivative, production thereof and radiosensitizer and antiviral agent containing said derivative as active ingredient. Publication Number 011106775 A (Japan), Oct. 22,1987; T. Suzuki et al. Heterocyclic compound derivative, production thereof and radiosensitizer, antiviral agent and anti cancer agent containing said derivative as active ingredient. Publication Number 01139596 A (Japan), Nov. 25, 1987; S.
Sakaguchi et al. Heterocyclic compound derivative, its production and radiosensitizer containing said derivative as active ingredient; Publication Number 63170375 A (Japan), Jan. 7, 1987), fluorine containing 3-nitro-1,2,4-triazole (T. Kagitani et al. Novel fluorine-containing 3-nitro-1,2,4-triazole and radiosensitizer containing same compound. Publication Number 02076861 A
(Japan), Mar. 31, 1988), 5-thiotretrazole derivative or its salt (E. Kano et al.
Radiosensitizer for Hypoxic cell. Publication Number 61010511 A (Japan), Jun.
26, 1984), Nitrothiazole (T. Kagitani et al. Radiation-sensitizing agent.
Publication Number 61167616 A (Japan) Jan. 22, 1985), imidazole derivatives (S. Inayma et al. Imidazole derivative. Publication Number 6203767 A (Japan) Aug. 1,1985; Publication Number 62030768 A (Japan) Aug. 1, 1985;
Publication Number 62030777 A (Japan) Aug. 1, 1985), 4-nitro-1,2,3-triazole (T. Kagitani et al. Radiosensitizer. Publication Number 62039525 A (Japan), Aug. 15,1985), 3-nitro-1,2,4-triazole (T. Kagitani et al. Radiosensitizer.
Publication Number 62138427 A (Japan), Dec. 12, 1985), Carcinostatic action regulator (H. Amagase. Carcinostatic action regulator. Publication Number 63099017 A (Japan), Nov. 21, 1986), 4,5-dinitroimidazole derivative (S.
Inayama. 4,5-Dinitroimidazole derivative. Publication Number 63310873 A
(Japan) Jun. 9, 1987), nitrotriazole Compound (T. Kagitanil Nitrotriazole Compound. Publication Number 07149737 A (Japan) Jun. 22, 1993), cisplatin, doxorubin, misonidazole, mitomycin, tiripazamine, nitrosourea, mercaptopurine, methotrexate, flurouracil, bleomycin, vincristine, carboplatin, epirubicin, doxorubicin, cyclophosphamide, vindesine, etoposide (1.F. Tannock. Review Article: Treatment of Cancer with Radiation and Drugs. Journal of Clinical Oncology 14(12):3156-3174, 1996), camptothecin (Ewend M.G. et al. Local delivery of chemotherapy and concurrent external beam radiotherapy prolongs survival in metastatic brain tumor models. Cancer Research 56(22):5217-5223, 1996) and paclitaxel (Tishler R.B. et al. Taxol: a novel radiation sensitizer.
International Journal of Radiation Oncology and Biological Physics 22(3):613-617, 1992).
A number of the above-mentioned cell cycle inhibitors also have a wide variety of analogues and derivatives, including, but not limited to, cisplatin, cyclophosphamide, misonidazole, tiripazamine, nitrosourea, mercaptopurine, methotrexate, flurouracil, epirubicin, doxorubicin, vindesine and etoposide.
Analogues and derivatives include (CPA)2Pt(DOLYM) and (DACH)Pt(DOLYM) cisplatin (Choi et al., Arch. Pharmacal Res. 22(2):151-156, 1999), Cis-(PtCl2(4,7-H-5-methyl-7-oxo)1,2,4(triazolo(1,5-a)pyrimidine)2) (Navarro et al., J.
Med. Chem. 41(3):332-338, 1998), (Pt(cis-1,4-DACH)(trans-C12)(CBDCA)) ~'/2MeOH cisplatin (Shamsuddin et al., Inorg. Chem.
36(25):5969-5971, 1997), 4-pyridoxate diammine hydroxy platinum (Tokunaga et al., Pharm. Sci. 3(7):353-356, 1997), Pt(II) ~ ~ ~ Pt(II) (Pt2(NHCHN(C(CH2)(CH3)))4) (Navarro et al., Inorg. Chem. 35(26):7829-7835, 1996), 254-S cisplatin analogue (Koga et al., Neurol. Res. 18(3):244-247, 1996), o-phenylenediamine ligand bearing cisplatin analogues (Koeckerbauer &
Bednarski, J. Inorg. Biochem. 62(4):281-298, 1996), trans,cis-(Pt(OAc)212(en)) (Kratochwil et al., J. Med. Chem. 39(13):2499-2507, 1996), estrogenic 1,2-diarylethylenediamine ligand (with sulfur-containing amino acids and glutathione) bearing cisplatin analogues (Bednarski, J. Inorg. Biochem.
62(1):75, 1996), cis-1,4-diaminocyclohexane cisplatin analogues (Shamsuddin et al., J. Inorg. Biochem. 61(4):291-301, 1996), 5' orientational isomer of cis-(Pt(NH3)(4-aminoTEMP-O)~d(GpG))) (Dunham & Lippard, J. Am. Chem. Soc.
117(43):10702-12, 1995), chelating diamine-bearing cisplatin analogues (Koeckerbauer & Bednarski, J. Pharm: Sci. 84(7):819-23, 1995), 1,2-diarylethyleneamine ligand-bearing cisplatin analogues (Otto et aL, J. Cancer Res. Clin. Oncol. 121(1 ):31-8, 1995), (ethylenediamine)platinum(II) complexes (Pasini et al., J. Chem. Soc., Dalton Trans. 4:579-85, 1995), CI-973 cisplatin analogue (Yang et al., Int. J. Oncol. 5(3):597-602, 1994), cis diamminedichloroplatinum(II) and its analogues cis-1,1 cyclobutanedicarbosylato(2R)-2-methyl-1,4-butanediam-mineplatinum(11) and cis-diammine(glycolato)platinum (Claycamp & Zimbrick, J. Inorg. Biochem., 26(4):257-67, 1986; Fan et al., Cancer Res. 48(11 ):3135-9, 1988; Heiger-Bernays et al., Biochemistry 29(36):8461-6, 1990; Kikkawa et al., J. Exp.
Clin.
Cancer Res. 12(4):233-40, 1993; Murray et al., Biochemistry 31(47):11812-17, 1992; Takahashi et al., Cancer Chemother. Pharmacol. 33(1 ):31-5, 1993), cis-amine-cyclohexylamine-dichloroplatinum(II) (Yoshida et al., Biochem.
Pharmaeol. 48(4):793-9, 1994), gem-diphosphonate cisplatin analogues (FR
2683529), (meso-1,2-bis(2,6-dichloro-4-hydroxyplenyl)ethylenediamine) dichloroplatinum(II) (Bednarski et al., J. Med. Chem. 35(23):4479-85, 1992), cisplatin analogues containing a tethered dansyl group (Hartwig et al., J. Am.
Chem. Soc. 114(21 ):8292-3, 1992), platinum(II) polyamines (Siegmann et al., Inorg. Met.-Containing Polym. Mater., (Proc. Am. Chem. Soc. Int. Symp.), 335-61, 1990), cis-(3H)dichloro(ethylenediamine)platinum(II) (Eastman, Anal.
Biochem. 197(2):311-15, 1991 ), trans-diamminedichloroplatinum(II) and cis-(Pt(NH3)2(N3-cytosine)CI) (Bellon & Lippard, Biophys. Chem. 35(2-3):179-88, 1990), 3H-cis-1,2-diaminocyclohexanedichloroplatinum(II) and 3H-cis-1,2-diaminocyclohexanemalonatoplatinum (II) (Oswald et al., Res. C~mmun. Chem.
Pathol. Pharmacol. 64(1):41-58, 1989), diaminocarboxylatoplatinum (EPA
296321 ), trans-(D,1 )-1,2-diaminocyclohexane carrier ligand-bearing platinum analogues (Wyrick & Chaney, J. Labelled Compd. Radiopharm. 25(4):349-57, 1988), aminoalkylaminoanthraquinone-derived cisplatin analogues (Kitov et al., Eur. J. Med. Chem. 23(4):381-3, 1988), spiroplatin, carboplatin, iproplatin and JM40 platinum analogues (Schroyen et al., Eur. J. Caneer Clin. Oneol.
24(8):1309-12, 1988), bidentate tertiary diamine-containing cisplatinum derivatives (Orbell et al., Inorg. Chim. Acta 152(2):125-34, 1988), platinum(II), platinum(IV) (Liu & Wang, Shandong Yike Daxue ~Cuebao 24(1 ):35-41, 1986), cis-diammine(1,1-cyclobutanedicarboxylato-)platinum(II) (carboplatin, JM8) and ethylenediammine-malonatoplatinum(II) (JM40) (Begg et al., Radiother. Oncol.
9(2):157-65, 1987), JM8 and JM9 cisplatin analogues (Harstrick et al., Int. J.
Androl. 10(1 ); 139-45, 1987), (NPr4)2((PtCL4).cis-(PtCl2-(NH2Me)2)) (Brammer et al., J. Chem. Soc., Chem. Commun. 6:443-5, 1987), aliphatic tricarboxylic acid platinum complexes (EPA 185225), cis-dichloro(amino acid)(tert-butylamine)platinum(II) complexes (Pasini & Bersanetti, Inorg.
Chim.
Acta 107(4):259-67, 1985); 4-hydroperoxycylcophosphamide (Ballard et al., Cancer Chemother. Pharmacol. 26(6):397-402, 1990), acyclouridine cyclophosphamide derivatives (Zakerinia et al., Helv. Chim. Acta 73(4):912-15, 1990), 1,3,2-dioxa- and -oxa~aphosphorinane cyclophosphamide analogues (Yang et al., Tetrahedron 44(20):6305-14, 1988), C5-substituted cyclophosphamide analogues (Spada, University of Rhode Island Dissertation, 1987), tetrahydrooxazine cyclophosphamide analogues (Valente, University of Rochester Dissertation, 1988), phenyl ketone cyclophosphamide analogues (Hales et al., Teratology 39(1 ):31-7, 1989), phenylketophosphamide cyclophosphamide analogues (Ludeman et al., J. Med. Chem. 29(5):716-27, 1986), ASTA Z-7557 cyclophosphamide analogues (Evans et al., Int. J. Cancer 34(6):883-90, 1984), 3-(1-oxy-2,2,6,6-tetramethyl-4-piperidinyl)cyclophosphamide (Tsui et al., J. Med. Chem. 25(9):1106-10, 1982), 2-oxobis(2-~3-chloroethylamino)-4-,6-dimethyl-1,3,2-oxazaphosphorinane cyclophosphamide (Carpenter et al., Phosphorus Sulfur 12(3):287-93, 1982), 5-fluoro- and 5-chlorocyclophosphamide (Foster et al., J. Med. Chem.
24(12):1399-403, 1981 ), cis- and trans-4-phenylcyclophosphamide (Boyd et al., J. Med. Chem. 23(4):372-5, 1980), 5-bromocyclophosphamide, 3,5-dehydrocyclophosphamide (Ludeman et al., J. Med. Chem. 22(2):151-8, 1979), 4-ethoxycarbonyi cyclophosphamide analogues (Foster, J. Pharm. Sci.
67(5):709-10, 1978), arylaminotetrahydro-2H-1,3,2-oxazaphosphorine 2-oxide cyclophosphamide analogues (Hamacher, Arch. Pharm. (V1/einheim, Ger.) 310(5):J,428-34, 1977), NSC-26271 cyclophosphamide analogues (Montgomery & Struck, Cancer Treat. Rep. 60(4):J381-93, 1976), benzo annulated cyclophosphamide analogues (Ludeman & Zon, J. Med. Chem.
18(12):J1251-3, 1975), 6-trifluoromethylcyclophosphamide (Farmer & Cox, J.
Med. Chem. 18(11 ):J1106-10, 1975), 4-methylcyclophosphamide and 6-methycyclophosphamide analogues (Cox et al., 8iochem. Pharmacol.
24(5):J599-606, 1975); FCE 23762 doxorubicin derivative (Quaglia et al., J.
Liq.
Chromatogr. 17(18):3911-3923, 1994), annamycin (Zou et al., J. Pharm. Sci.
82(11):1151-1154, 1993), ruboxyl (Rapoport et al., J. Controlled Release 58(2):153-162, 1999), anthracycline disaccharide doxorubicin analogue (Pratesi et al., Clin. Cancer Res. 4(11 ):2833-2839, 1998), N-(trifluoroacetyl)doxorubicin and 4'-O-acetyl-N-(trifluoroacetyl)doxorubicin (Berube & Lepage, Synth.
Commun. 28(6):1109-1116, 1998), 2-pyrrolinodoxorubicin (Nagy et al., Proc.
Nat'I Acad. Sci. U.S.A. 95(4):1794-1799, 1998), disaccharide doxorubicin analogues (Arcamone et al., J. Nat'I Cancer Inst. 89(16):1217-1223, 1997), 4-d a m ethoxy-7-O-( 2, 6-d i d eoxy-4-O-(2, 3, 6-trid eoxy-3-a m i n o-a-L-I
yxo-hexopyranosyl)-a-L-lyxo-hexopyranosyl)adriamicinone doxorubicin disaccharide analogue (Monteagudo et al., Carbohydr. Res. 300(1 ):11-16, 1997), 2-pyrrolinodoxorubicin (Nagy et al., Proc. Nat'I Acad. Sci. U.S.A.
94(2):652-656, 1997), morpholinyl doxorubicin analogues (Duran et al., Cancer Chemother. Pharmacol. 38(3):210-216, 1996), enaminomalonyl-~i-alanine doxorubicin derivatives (Seitz et al., Tetrahedron Lett. 36(9):1413-16, 1995), cephalosporin doxorubicin derivatives (Vrudhula et al., J. Med. Chem.
38(8):1380-5, 1995), hydroxyrubicin (Solary et al., Int. J. Cancer 58(1 ):85-94, 1994), methoxymorpholino doxorubicin derivative (Kuhl et al., Cancer Chemother. Pharmacol. 33(1 ):10-16, 1993), (6-maleimidocaproyl)hydrazone doxorubicin derivative (Willner et al., Bioconjugate Chem. 4(6):521-7, 1993), N-(5,5-diacetoxypent-1-yl) doxorubicin (Cherif & Farquhar, J. Med. Chem.
35(17):3208-14, 1992), FCE 23762 methoxymorpholinyl doxorubicin derivative (Ripamonti et al., Br. J. Cancer 65(5):703-7, 1992), N-hydroxysuccinimide ester doxorubicin derivatives (Demant et al., Bioehim. Biophys. Acta 1118(1 ):83-90, 1991 ), polydeoxynucleotide doxorubicin derivatives (Ruggiero et al., Biochim.
Biophys. Acta 1129(3):294-302, 1991 ), morpholinyl doxorubicin derivatives (EPA 434960), mitoxantrone doxorubicin analogue (Krapcho et al., J. Med.
Chem. 34(8):2373-80. 1991 ), AD198 doxorubicin analogue (Traganos et al., Cancer Res. 51 (14):3682-9, 1991 ), 4-demethoxy-3'-N-trifluoroacetyldoxorubicin (Norton et al., Drug Des. Delivery 6(2):123-9, 1990), 4'-epidoxorubicin (Drzewoski et al., Pol. J. Pharmacol. Pharm. 40(2):159-65, 1988; Weenen et al., Eur. J. Cancer Clin. Oncol. 20(7):919-26, 1984), alkylating cyanomorpholino doxorubicin derivative (Scudder et al., J. Nat'I Cancer Inst. 80(16):1294-8, 1988), deoxydihydroiodooxorubicin (EPA 275966), adriblastin (Kalishevskaya et al., Vestn. Mosk. Univ., 16(Biol. 1 ):21-7, 1988), 4'-deoxydoxorubicin (Schoelzel et al., Leuk. Res. 10(12):1455-9, 1986), 4-demethyoxy-4'-o-methyldoxorubicin (Giuliani et al., Proc. Int. Congr. Chemother. 16:285-70-285-77, 1983), 3'-deamino-3'-hydroxydoxorubicin (Norton et al., J. Antibiot. 37(8):853-8, 1984), demethyoxy doxorubicin analogues (Barbieri et al., Drugs Exp. Clin. Res.
10(2):85-90, 1984), N-L-leucyl doxorubicin derivatives (Trouet et al., Anthracyclines (Proc. Int. Symp. TumorPharmacother.), 179-81, 1983), 3'-deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (4,314,054), 3'-deamino-3'-(4-mortholinyl) doxorubicin derivatives (4,301,277), 4'-deoxydoxorubicin and 4'-o-methyldoxorubicin (Giuliani et al., Int. J. Cancer 27(1 ):5-13, 1981 ), aglycone doxorubicin derivatives (Chan & Watson, J.
Pharm.

Sci. 67(12):1748-52, 1978), SM 5887 (Pharma Japan 1468:20, 1995), MX-2 (Pharma Japan 1420:19, 1994), 4'-deoxy-13(S)-dihydro-4'-iododoxorubicin (EP
275966), morphofinyl doxorubicin derivatives (EPA 434960), 3'-deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (4,314,054), doxorubicin-14-valerate, morpholinodoxorubicin (5,004,606), 3'-deamino-3'-(3"-cyano-4"-morpholinyl doxorubicin; 3'-deamino-3'-(3"-cyano-4"-morpholinyl)-13-dihydoxorubicin; (3'-deamino-3'-(3"-cyano-4"-morpholinyl) daunorubicin; 3'-deamino-3'-(3"-cyano-4"-morpholinyl)-3-dihydrodaunorubicin; and 3'-deamino-3'-(4"-morpholinyl-5-iminodoxorubicin and derivatives (4,585,859), 3'-deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (4,314,054) and 3-deamino-3-(4-morpholinyl) doxorubicin derivatives (4,301,277); 4,5-dimethylmisonidazole (Born et al., Biochem. Pharmacol. 43(6):1337-44, 1992), azo and azoxy misonidazole derivatives (Gattavecchia & Tonelli, Int. J. Radiat. Biol. Relat.
Stud. Phys., Chem. Med. 45(5):469-77, 1984); RB90740 (Wardman et al., Br. J.
Cancer, 74 Suppl. (27):S70-S74, 1996); 6-bromo and 6-chloro-2,3-dihydro-1,4-benzothiazines nitrosourea derivatives (Rai et al., Heterocycl. Commun.
2(6):587-592, 1996), diamino acid nitrosourea derivatives (Dulude et al., Bioorg.
Med. Chem. Lett. 4(22):2697-700, 1994; Dulude et al., Bioorg. Med. Chem.
3(2):151-60, 1995), amino acid nitrosourea derivatives (Zheleva et al., Pharmazie 50(1 ):25-6, 1995), 3',4'-didemethoxy-3',4'-dioxo-4-deoxypodophyllotoxin nitrosourea derivatives (Miyahara et al., Heterocycles 39(1 ):361-9, 1994), ACNU (Matsunaga et al., Immunopharmacology 23(3):199-204, 1992), tertiary phosphine oxide nitrosourea derivatives (Guguva et al., Pharmazie 46(8):603, 1991 ), sulfamerizine and sulfamethizole nitrosourea derivatives (Chiang et al., Zhonghua Yaozue Zazhi 43(5):401-6, 1991 ), thymidine nitrosourea analogues (Zhang et al., Cancer Commun. 3(4):119-26, 1991 ), 1,3-bis(2-chloroethyl)-1-nitrosourea (August et al., Cancer Res.
51(6):1586-90, 1991), 2,2,6,6-tetramethyl-1-oxopiperidiunium nitrosourea derivatives (U.S.S.R. 1261253), 2- and 4-deoxy sugar nitrosourea derivatives, (4,902,791 ), nitroxyl nitrosourea derivatives (U.S.S.R. 1336489), fotermstine (Boutin et al., Eur. J. Cancer Clin. Oncol. 25(9):1311-16, 1989), pyrimidine (II) nitrosourea derivatives (Wei et al., Chung-hua Yao Hsueh Tsa Chih 41(1 ):19-26, 1989), CGP 6809 (Schieweck et al., Cancer Chemother. Pharmacol.
23(6):341-7, 1989), B-3839 (Prajda et al., In Vivo 2(2):151-4, 1988), 5-halogenocytosine nitrosourea derivatives (Chiang & Tseng, T'ai-wan Yao Hsueh Tsa Chih 38(1 ):37-43, 1986), 1-(2-chloroethyl)-3-isobutyl-3-(a-maltosyl)-1-nitrosourea (Fujimoto & Ogawa, J. Pharmacobio-Dyn. 10(7):341-5, 1987), sulfur-containing nitrosoureas (Tang et al., Yaoxue ~Cuebao 21(7):502-9, 1986), sucrose, 6-((((2-chloroethyl)nitrosoamino-)carbonyl)amino)-6-deoxysucrose (NS-1 C) and 6'-((((2-chloroethyl)nitrosoamino)carbonyl)amino)-6'-deoxysucrose (NS-1 D) nitrosourea derivatives (Tanoh et al., Chemotherapy (Tokyo) 33(11 ):969-77, 1985), CNCC, RFCNU and chlorozotocin (Mena et al., Chemotherapy (Basel) 32(2):131-7, 1986), CNUA (Edanami et al., Chemotherapy (Tokyo) 33(5):455-61, 1985), 1-(2-chloroethyl)-3-isobutyl-3-(~3-maltosyl)-1-nitrosourea (Fujimoto & Ogawa, Jpn. J. Cancer Res. (Gann) 76(7):651-6, 1985), choline-like nitrosoalkylureas (Belyaev et al., Izv. Akad.
NA UK SSSR, Ser. Khim, 3:553-7, 1985), sucrose nitrosourea derivatives (JP
84219300), sulfa drug nitrosourea analogues (Chiang et al., Proc, Nat'1 Sci.
Counc., Repub. China, Part A 3(1 ):18-22, 1984), DONU (Asanuma et al., J.
Jpn. Soc. Cancer Ther. 17(8):2035-43, 1982), N,N'-bis (N-(2-chloroethyl)-N-nitrosocarbamoyl)cystamine (CNCC) (Blazsek et al., Toxicol. Appl. Pharmacol.
74(2):250-7, 1984), dimethylnitrosourea (Krutova et al., Izv. Akad. NAUK
SSSR, Ser. Biol. 3:439-45, 1984), GANU (Sava & Giraldi, Cancer Chemother.
Pharmacol. 10(3):167-9, 1983), CCNU (Capelli et al., Med., Biol., Environ.
11(1):111-16, 1983), 5-aminomethyl-2'-deoxyuridine nitrosourea analogues (Shiau, Shih Ta Hsueh Pao (Taipei) 27:681-9, 1982), TA-077 (Fujimoto &
Ogawa, Cancer Chemother. Pharmacol. 9(3):134-9, 1982), gentianose nitrosourea derivatives (JP 82 80396), CNCC, RFCNU, RPCNU AND
chlorozotocin (CZT) (Marzin et al., INSERM Symp., 19(Nitrosoureas Cancer Treat.):165-74, 1981 ), thiocolchicine nitrosourea analogues (George, Shih Ta Hsueh Pao (Taipei) 25:355-62, 1980), 2-chloroethyl-nitrosourea (teller &
Eisenbrand, Oncology 38(1 ):39-42, 1981 ), ACNU, (1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride) (Shibuya et al., Gan To Kagaku Ryoho 7(8):1393-401, 1980), N-deacetylmethyl thiocolchicine nitrosourea analogues (Lin et al., J. Med. Chem. 23(12):1440-2, 1980), pyridine and piperidine nitrosourea derivatives (Crider et al., J. Med.
Chem. 23(8):848-51, 1980), methyl-CCNU (Zimber& Perk, Refu. Vet. 35(1):28, 1978), phensuzimide nitrosourea derivatives (Crider et al., J. Med. Chem.
23(3):324-6, 1980), ergoline nitrosourea derivatives (Crider et al., J. Med.
Chem. 22(1 ):32-5, 1979), glucopyranose nitrosourea derivatives (JP 78 95917), 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (Farmer et al., J. Med. Chem.
21(6):514-20, 1978), 4-(3-(2-chloroethyl)-3-nitrosoureid-o)-cis-cyclohexanecarboxylic acid (Drewinko et al., Cancer Treat. Rep. 61(8):J1513-18, 1977), RPCNU (ICIG 1163) (Larnicol et al., Biomedicine 26(3):J176-81, 1977), IOB-252 (Sorodoc et al., Rev. Rouen. Med., Virol. 28(1 ):J 55-61, 1977), 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) (Siebert & Eisenbrand, Mutat. Res.
42(1):J45-50, 1977), 1-tetrahydroxycyclopentyl-3-nitroso-3-(2-chloroethyl)-urea (4,039,578), d-1-1-(~i-chloroethyl)-3-(2-oxo-3-hexahydroazepinyl)-1-nitrosourea (3,859,277) and gentianose nitrosourea derivatives (JP 57080396); 6-S-aminoacyloxymethyl mercaptopurine derivatives (Harada et al., Chem. Pharm.
Bull. 43(10):793-6, 1995), 6-mercaptopurine (6-MP) (Kashida et al., Biol.
Pharm. Bull. 78(11 ):1492-7, 1995), 7,8-polymethyleneimidazo-1,3,2-diazaphosphorines (Nilov et al., Mendeleev Commun. 2:67, 1995), azathioprine (Chifotides et al., J. Inorg. Biochem. 56(4):249-64, 1994), methyl-D-glucopyranoside mercaptopurine derivatives (Da Silva et al., Eur. J. Med.
Chem. 29(2):149-52, 1994) and s-alkynyl mercaptopurine derivatives (Ratsino et al., Khim.-Farm. ~h. 75(8):65-7, 1981 ); indoline ring and a modified ornithine or glutamic acid-bearing methotrexate derivatives (Matsuoka et al., Chem.
Pharm. Bull. 45(7):1146-1150, 1997), alkyl-substituted benzene ring C bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 44(12):2287-2293, 1996), benzoxazine or benzothiazine moiety-bearing methotrexate derivatives (Matsuoka et al., J. Med. Chem. 40(1):105-111, 1997), 10-deazaaminopterin analogues (DeGraw et al., J. Med. Chem. 40(3):370-376, 1997), 5-deazaaminopterin and 5,10-dideazaaminopterin methotrexate analogues (Piper et al., J. Med. Chem. 40(3):377-384, 1997), indoline moiety-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull.
44(7):1332-1337, 1996), lipophilic amide methotrexate derivatives (Pignatello et al., World Meet. Pharm., Biopharm. Pharm. Technol., 563-4, 1995), L-threo-(2S, 4S)-4-fluoroglutamic acid and DL-3,3-difluoroglutamic acid-containing methotrexate analogues (Hart et al., J. Med. Chem. 39(1 ):56-65, 1996), methotrexate tetrahydroquinazoline analogue (Gangjee, et al., J. Heterocycl.
Chem. 32(1 ):243-8, 1995), N-(a-aminoacyl) methotrexate derivatives (Cheung et al., Pteridines 3(1-2):101-2, 1992), biotin methotrexate derivatives (Fan et al., Pteridines 3(1-2):131-2, 1992), D-glutamic acid or D-erythrou, threo-4-fluoroglutamic acid methotrexate analogues (McGuire et al., Biochem.
Pharmacol. 42(12):2400-3, 1991 ), ~,y-methano methotrexate analogues (Rosowsky et al., Pteridines 2(3):133-9, 1991 ), 10-deazaaminopterin (10-EDAM) analogue (Braakhuis et al., Chem. Biol. Pteridines, Proc. Int. Symp.

Pteridines Folic Acid Deriv., 1027-30, 1989), y-tetrazole methotrexate analogue (Kalman et al., Chem. Biol. Pteridines, Proc. Int. Symp. Pteridines Folic Acid Deriv., 1154-7, 1989), N-(L-a-aminoacyl) methotrexate derivatives (Cheung et al., Heterocycles 28(2):751-8, 1989), meta and ortho isomers of aminopterin (Rosowsky et al., J. Med. Chem. 32(12):2582, 1989), hydroxymethylmethotrexate (DE 267495), y-fluoromethotrexate (McGuire et al., Cancer Res. 49(16):4517-25, 1989), polyglutamyl methotrexate derivatives (Kumar et al., Cancer Res. 46(10):5020-3, 1986), gem-diphosphonate methotrexate analogues (WO 88/06158), a- and y-substituted methotrexate analogues (Tsushima et al., Tetrahedron 44(17):5375-87, 1988), 5-methyl-5-deaza methotrexate analogues (4,725,687), N8-acyl-Na-(4-amino-4-deoxypteroyl)-L-ornithine derivatives (Rosowsky et al., J. Med. Chem.
37(7):1332-7, 1988), 8-deaza methotrexate analogues (Kuehl et al., Cancer Res. 48(6):1481-8, 1988), acivicin methotrexate analogue (Rosowsky et al., J.
Med. Chem. 30(8):1463-9, 1987), polymeric platinol methotrexate derivative (Carraher et al., Polym. Sci. Technol. (Plenum), 35(Adv. Biomed. Polym.):311-24, 1987), methotrexate-y-dimyristoylphophatidylethanolamine (Kinsky et al., Biochim. Biophys. Acta 977(2):211-18, 1987), methotrexate polyglutamate analogues (Rosowsky et al., Chem. Biol. Pteridines, Pteridines Folid Acid Deriv., Proc. Int. Symp. Pteridines Folid Acid Deriv.: Chem., Biol. Clin.
Aspects:
985-8, 1986), poly-y-glutamyl methotrexate derivatives (Kisliuk et al., Chem.
Biol. Pteridines, Pteridines Folid Acid Deriv., Proc. Int. Symp. Pteridines Folid Acid Deriv.: Chem., Biol. Clin. Aspects: 989-92, 1986), deoxyuridylate methotrexate derivatives (Webber et al., Chem. Biol. Pteridines, Pteridines Folid Acid Deriv., Proc. Int. Symp. Pteridines Folid Acid Deriv.: Chem., Biol.
Clin. Aspects: 659-62, 1986), iodoacetyl lysine methotrexate analogue (Delcamp et al., Chem. Biol. Pteridines, Pteridines Folid Acid Deriv., Proc.
Int.
Symp. Pteridines Folid Acid Deriv.: Chem., Biol. Clin. Aspects: 807-9, 1986), 2,.omega.-diaminoalkanoid acid-containing methotrexate analogues (McGuire et al., Biochem. Pharmacol. 35(15):2607-13, 1986), polyglutamate methotrexate derivatives (Kamen & Winick, Methods Enzymol. 122 (Vitam.
Coenzymes, Pt. G):339-46, 1986), 5-methyl-5-deaza analogues (Piper et al., J.
Med. Chem. 29(6):1080-7, 1986), quinazoline methotrexate analogue (Mastropaolo et al., J. Med. Chem. 29(1 ):155-8, 1986), pyrazine methotrexate analogue (Lever & Vestal, J. Heterocycl. Chem. 22(1 ):5-6, 1985), cysteic acid and homocysteic acid methotrexate analogues (4,490,529), y-tent-butyl methotrexate esters (Rosowsky et al., J. Med. Chem. 28(5):660-7, 1985), fluorinated methotrexate analogues (Tsushima et al., Heterocycles 23(1 ):45-9, 1985), folate methotrexate analogue (Trombe, J. Bacteriol. 160(3):849-53, 1984), phosphonoglutamic acid analogues (Sturtz & Guillamot, Eur. J. Med.
Chem.--Chim. Ther. 19(3):267-73, 1984), poly (L-lysine) methotrexate conjugates (Rosowsky et al., J. Med. Chem. 27(7):888-93, 1984), dilysine and trilysine methotrexate derivates (Forsch & Rosowsky, J. Org. Chem.
49(7):1305-9, 1984), 7-hydroxymethotrexate (Fabre et al., Cancer Res.
43(10):4648-52, 1983), poly-y-glutamyl methotrexate analogues (Piper &
Montgomery, Adv. Exp. Med. Biol., 163(Folyl Antifolyl Polyglutamates):95-100, 1983), 3',5'-dichloromethotrexate (Rosowsky & Yu, J. Med. Chem. 26(10):1448-52, 1983), diazoketone and chloromethylketone methotrexate analogues (Gangjee et al., J. Pharm. Sci. 71(6):717-19, 1982), 10-propargylaminopterin and alkyl methotrexate homologs (Piper et al., J. Med. Chem. 25(7):877-80, 1982), lectin derivatives of methotrexate (Lin et al., JNCI 66(3):523-8, 1981 ), polyglutamate methotrexate derivatives (Galivan, Mol. Pharmacol. 17(1 ):105-10, 1980), halogentated methotrexate derivatives (Fox, JNCI 58(4):J955-8, 1977), 8-alkyl-7,8-dihydro analogues (Chaykovsky et al., J. Med. Chem.
20(10):J1323-7, 1977), 7-methyl methotrexate derivatives and dichloromethotrexate (Rosowsky ~ Chen, J. Med. Chem. 17(12):J1308-11, 1974), lipophilic methotrexate derivatives and 3',5'-dichloromethotrexate (Rosowsky, J. Med. Chem. 16(10):J1190-3, 1973), deaza amethopterin analogues (Montgomery et al., Ann. N. Y. Acad. Sci. 186:J227-34, 1971), MX068 (Pharma Japan, 1658:18, 1999) and cysteic acid and homocysteic acid methotrexate analogues (EPA 0142220); N3-alkylated analogues of 5-fluorouracil (Kozai et al., J. Chem. Soc., Perkin Trans. 1(19):3145-3146, 1998), 5-fluorouracil derivatives with 1,4-oxaheteroepane moieties (Gomez et al., Tetrahedron 54(43):13295-13312, 1998), 5-fluorouracil and nucleoside analogues (Li, Anticancer Res. 17(1A):21-27, 1997), cis- and traps-5-fluoro-5,6-dihydro-6-alkoxyuracil (Van der Wilt et al., Br. J. Cancer 68(4):702-7, 1993), cyclopentane 5-fluorouracil analogues (Hronowski & Szarek, Can. J. Chem.
70(4):1162-9, 1992), A-OT-fluorouracil (Zhang et al., Zongguo Yiyao Gongye Zazhi 20(11 ):513-15, 1989), N4-trimethoxybenzoyl-5'-deoxy-5-fluorocytidine and 5'-deoxy-5-fluorouridine (Miwa et al., Chem. Pharm. Bull. 38(4):998-1003, 1990), 1-hexylcarbamoyl-5-fluorouracil (Hoshi et al., J. Pharmacobio-~un.
3(9):478-81, 1980; Maehara et al., Chemotherapy (Basel) 34(6):484-9, 1988), B-3839 (Prajda et al., In Vivo 2(2):151-4, 1988), uracil-1-(2-tetrahydrofuryl)-fluorouracil (Anai et al., Oncology 45(3}:144-7, 1988), 1-(2'-deoxy-2'-fluoro-a-D-arabinofuranosyl)-5-fluorouracil (Suzuko et al., Mol. Pharmacol. 31(3):301-6, 1987), doxifluridine (Matuura et al., Oyo Yakuri 29(5):803-31, 1985), 5'-deoxy-fluorouridine (Bollag & Hartmann, EUr. J. Cancer 16(4):427-32, 1980), 1-acetyl-3-O-toluyl-5-fluorouracil (Okada, Hiroshima J. Med. Sci. 28(1 ):49-66, 1979), fluorouracil-m-formylbenzene-sulfonate (JP 55059173), N'-(2-furanidyl)-5-fluorouracil (JP 53149985) and 1-(2-tetrahydrofuryl)-5-fluorouracil (JP
52089680); 4'-epidoxorubicin (Lanius, Adv. Chemother. Gastrointest. Cancer, (Int. Symp.), 159-67, 1984); N-substituted deacetylvinblastine amide (vindesine) sulfates (Conrad et al., J. Med. Chem. 22(4):391-400, 1979); and Cu(II)-VP-16 (etoposide) complex (Tawa et al., Bioorg. Med. Chem. 6(7):1003-1008, 1998), pyrrolecarboxamidino-bearing etoposide analogues (Ji et al., Bioorg. Med.
Chem. Lett. 7(5):607-612, 1997), 4~i-amino etoposide analogues (Hu, University of North Carolina Dissertation, 1992), y-lactone ring-modified arylamino etoposide analogues (~hou et al., J. Med. Chem. 37(2):287-92, 1994), N-glucosyl etoposide analogue (Allevi et al., Tetrahedron Lett.
34(45):7313-16, 1993), etoposide A-ring analogues (Kadow et al., Bioorg. Med.
Chem. Lett. 2(1 ):17-22, 1992), 4'-deshydroxy-4'-methyl etoposide (Saulnier et al., Bioorg. Med. Chem. Lett. 2(10):1213-18, 1992), pendulum ring etoposide analogues (Sinha et al., Eur. J. Cancer 26(5):590-3, 1990) and E-ring desoxy etoposide analogues (Saulnier et al., J. Med. Chem. 32(7):1418-20, 1989).
Within one preferred embodiment of the invention, the cell cycle inhibitor is paclitaxel, a compound which disrupts mitosis (M-phase) by binding to tubulin to form abnormal mitotic spindles or an analogue or derivative thereof. Briefly, paclitaxel is a highly derivatized diterpenoid (Wani et al., J. Am.
Chem. Soc. 93:2325, 1971 ) which has been obtained from the harvested and dried bark of Taxus brevifolia (Pacific Yew) and Taxomyces Andreanae and Endophytic Fungus of the Pacific Yew (Stierle et al., Science 60:214-216, 1993). "Paclitaxel" (which should be understood herein to include formulations, prodrugs, analogues and derivatives such as, for example, TAXOL (Bristol Myers Squibb, New York, NY, TAXOTERE (Aventis Pharmaceuticals, France), docetaxel, 10-desacetyl analogues of paclitaxel and 3'N-desbenzoyl-3'N-t-butoxy carbonyl analogues of paclitaxel) may be readily prepared utilizing techniques known to those skilled in the art (see, e.g., Schiff et al., Nature 277:665-667, 1979; Long and Fairchild, Cancer Research 54:4355-4361, 1994;

Ringel and Horwitz, J. Nat'I Cancer Inst. 33(4):288-291, 1991; Pazdur et al., Cancer Treat. Rev. 79(4):351-386, 1993; WO 94107882; WO 94/07881; WO
94/07880; WO 94/07876; WO 93/23555; WO 93/10076; W094/00156;
WO 93/24476; EP 590267; WO 94120089; U.S. Patent Nos. 5,294,637;
5,283,253; 5,279,949; 5,274,137; 5,202,448; 5,200,534; 5,229,529; 5,254,580;
5,412,092; 5,395,850; 5,380,751; 5,350,866; 4,857,653; 5,272,171; 5,411,984;
5,248,796; 5,248,796; 5,422,364; 5,300,638; 5,294,637; 5,362,831; 5,440,056;
4,814,470; 5,278,324; 5,352,805; 5,411,984; 5,059,699; 4,942,184;
Tetrahedron Letters 35(52):9709-9712, 1994; J. Med. Ghem. 35:4230-4237, 1992; J. Med. Chem. 34:992-998, 1991; J. Natural Prod. 57( 10):1404-1410, 1994; J. Natural Prod. 57(11 ):1580-1583, 1994; J. Am. Chem. Soc. 110:6558-6560, 1988), or obtained from a variety of commercial sources, including for example, Sigma Chemical Co., St. Louis, Missouri (T7402 - from Taxus brevifolia).
Representative examples of paclitaxel derivatives or analogues include 7-deoxy-docetaxol, 7,8-cyclopropataxanes, N-substituted 2-azetidones, 6,7-epoxy paclitaxels, 6,7-modified paclitaxels, 10-desacetoxytaxol, 10-deacetyltaxol (from 10-deacetylbaccatin III), phosphonooxy and carbonate derivatives of taxol, taxol 2',7-di(sodium 1,2-benzenedicarboxylate, 10-desacetoxy-11,12-dihydrotaxol-10,12(18)-diene derivatives, 10-desacetoxytaxol, Protaxol (2'-and/or 7-O-ester derivatives), (2'-and/or 7-O-carbonate derivatives), asymmetric synthesis of taxol side chain, fluoro taxols, 9-deoxotaxane, (13-acetyl-9-deoxobaccatine III, 9-deoxotaxol, 7-deoxy-9-deoxotaxol, 10-desacetoxy-7-deoxy-9-deoxotaxol, Derivatives containing hydrogen or acetyl group and a hydroxy and tert-butoxycarbonylamino, sulfonated 2'-acryloyltaxol and sulfonated 2'-O-acyl acid taxol derivatives, succinyltaxol, 2'-y-aminobutyryltaxol formate, 2'-acetyl taxol, 7-acetyl taxol, 7-glycine carbamate taxol, 2'-OH-7-PEG(5000) carbamate taxol, 2'-benzoyl and 2',7-dibenzoyl taxol derivatives, other prodrugs (2'-acetyltaxol; 2',7-diacetyltaxol;
2'succinyltaxol; 2'-(beta-alanyl)-taxol); 2'gamma-aminobutyryltaxol formate;
ethylene glycol derivatives of 2'-succinyltaxol; 2'-glutaryltaxol; 2'-(N,N-dimethylglycyl) taxol; 2'-(2-(N,N-dimethylamino)propionyl)taxol;
2'orthocarboxybenzoyl taxol; 2'aliphatic carboxylic acid derivatives of taxol, Prodrugs ~2'(N,N-diethylaminopropionyl)taxol, 2'(N,N-dimethylglycyl)taxol, 7(N,N-dimethylglycyl)taxol, 2',7-di-(N,N-dimethylglycyl)taxol, 7(N,N-diethylaminopropionyl)taxol, 2',7-di(N,N-diethylaminopropionyl)taxol, 2'-(L-glycyl)taxol, 7-(L-glycyl)taxol, 2',7-di(L-glycyl)taxol, 2'-(L-alanyl)taxol, 7-(L-alanyl)taxol, 2',7-di(L-alanyl)taxol, 2'-(L-leucyl)taxol, 7-(L-leucyl)taxol, 2',7-di(L-leucyl)taxol, 2'-(L-isoleucyl)taxol, 7-(L-isoleucyl)taxol, 2',7-di(L-isoleucyl)taxol, 2'-(L-valyl)taxol, 7-(L-valyl)taxol, 2'7-di(L-valyl)taxol, 2'-(L-phenylalanyl)taxol, 7-(L-phenylalanyl)taxol, 2',7-di(L-phenylalanyl)taxol, 2'-(L-prolyl)taxol, 7-(L-prolyl)taxol, 2',7-di(L-prolyl)taxol, 2'-(L-lysyl)taxol, 7-(L-lysyl)taxol, 2',7-di(L-lysyl)taxol, 2'-(L-glutamyl)taxol, 7-(L-glutamyl)taxol, 2',7-di(L-glutamyl)taxol, 2'-(L-arginyl)taxol, 7-(L-arginyl)taxol, 2',7-di(L-arginyl)taxol}, taxol analogues with modified phenylisoserine side chains, TAXOTERE, (N-debenzoyl-N-tert-(butoxycaronyl)-10-deacetyltaxol, and taxanes (e.g., baccatin III, cephalomannine, 10-deacetylbaccatin III, brevifoliol, yunantaxusin and taxusin);
and other taxane analogues and derivatives, including 14-beta-hydroxy-10 deacetybaccatin III, debenzoyl-2-acyl paclitaxel derivatives, benzoate paclitaxel derivatives, phosphonooxy and carbonate paclitaxel derivatives, sulfonated 2'-acryloyltaxol; sulfonated 2'-O-acyl acid paclitaxel derivatives, 13-site-substituted paclitaxel derivatives, chlorinated paclitaxel analogues, C4 methoxy ether paclitaxel derivatives, sulfonamide taxane derivatives, brominated paclitaxel analogues, Girard taxane derivatives, nitrophenyl paclitaxel, 10-deacetylated substituted paclitaxel derivatives, 14- beta -hydroxy-10 deacetylbaccatin III
taxane derivatives, C7 taxane derivatives, C10 taxane derivatives, 2-debenzoyl-2-acyl taxane derivatives, 2-debenzoyl and -2-acyl paclitaxel derivatives, taxane and baccatin III analogues bearing new CZ and C4 functional groups, n-acyl paclitaxel analogues, 10-deacetylbaccatin III and 7-protected-10-deacetylbaccatin III derivatives from 10-deacetyl taxol A, 10-deacetyl taxol B, and 10-deacetyl taxol, benzoate derivatives of taxol, 2-aroyl-4-acyl paclitaxel analogues, orthro-ester paclitaxel analogues, 2-aroyl-4-acyl paclitaxel analogues and 1-deoxy paclitaxel and 1-deoxy paclitaxel analogues.
In one aspect, the cell cycle inhibitor is a taxane having the formula (C1 ):

(C 1 ), where the gray-highlighted portions may be substituted and the non-highlighted portion is the taxane core. A side-chain (labeled "A" in the diagram) is desirably present in order for the compound to have good activity as a cell cycle inhibitor.
Examples of compounds having this structure include paclitaxel (Merck Index entry 7117), docetaxol (TAXOTERE, Merck Index entry 3458), and 3'-desphenyl-3'-(4-ntirophenyl)-N-debenzoyl-N-(t-butoxycarbonyl)-10-deacetyltaxol.
In one aspect, suitable taxanes such as paclitaxel and its analogues and derivatives are disclosed in U.S. Patent No. 5,440,056 as having the structure (C2):

H C~ w RIO'S _ ~ O
R6 _- 1~
R$O
R~° (C2) wherein X may be oxygen (paclitaxel), hydrogen (9-deoxy derivatives), thioacyl, or dihydroxyl precursors; R~ is selected from paclitaxel or TAXOTERE side chains or alkanoyl of the formula (C3) O
R
(C3) wherein R7 is selected from hydrogen, alkyl, phenyl, alkoxy, amino, phenoxy (substituted or unsubstituted); R$ is selected from hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, aminoalkyl, phenyl (substituted or unsubstituted), alpha or beta-naphthyl; and R9 is selected from hydrogen, alkanoyl, substituted alkanoyl, and aminoalkanoyl; where substitutions refer to hydroxyl, sulfhydryl, allalkoxyl, carboxyl, halogen, thioalkoxyl, N,N-dimethylamino, alkylamino, dialkylamino, nitro, and -OS03H, and/or may refer to groups containing such substitutions;

is selected from hydrogen or oxygen-containing groups, such as hydrogen, hydroxyl, alkoyl, alkanoyloxy, aminoalkanoyloxy, and peptidyalkanoyloxy; R3 is selected from hydrogen or oxygen-containing groups, such as hydrogen, hydroxyl, alkoyl, alkanoyloxy, aminoalkanoyloxy, and peptidyalkanoyloxy, and may further be a silyl containing group or a sulphur containing group; R4 is selected from acyl, alkyl, alkanoyl, aminoalkanoyl, peptidylalkanoyl and aroyl;
R5 is selected from acyl, alkyl, alkanoyl, aminoalkanoyl, peptidylalkanoyl and aroyl; R6 is selected from hydrogen or oxygen-containing groups, such as hydrogen, hydroxyl alkoyl, alkanoyloxy, aminoalkanoyloxy, and peptidyalkanoyloxy.
In one aspect, the paclitaxel analogues and derivatives useful as cell cycle inhibitors are disclosed in PCT International Patent Application No.
WO 93/10076. As disclosed in this publication, the analogue or derivative should have a side chain attached to the taxane nucleus at C~3, as shown in the structure below (formula C4), in order to confer antitumor activity to the taxane.

(C4) WO 93/10076 discloses that the taxane nucleus may be substituted at any position with the exception of the existing methyl groups.
The substitutions may include, for example, hydrogen, alkanoyloxy, alkenoyloxy, aryloyloxy. In addition, oxo groups may be attached to carbons labeled 2, 4, 9, and/or 10. As well, an oxetane ring may be attached at carbons 4 and 5. As well, an oxirane ring may be attached to the carbon labeled 4.

In one aspect, the taxane-based cell cycle inhibitor useful in the present invention is disclosed in U.S. Patent 5,440,056, which discloses 9-deoxo taxanes. These are compounds lacking an oxo group at the carbon labeled 9 in the taxane structure shown above (formula C4). The taxane ring may be substituted at the carbons labeled 1, 7 and 10 (independently) with H, OH, O-R, or O-CO-R where R is an alkyl or an aminoalkyl. As well, it may be substituted at carbons labeled 2 and 4 (independently) with aryol, alkanoyl, aminoalkanoyl or alkyl groups. The side chain of formula (C3) may be substituted at R~ and R$ (independently) with phenyl rings, substituted phenyl rings, linear alkanes/alkenes, and groups containing H, O or N. R9 may be substituted with H, or a substituted or unsubstituted alkanoyl group.
Taxanes in general, and paclitaxel is particular, is considered to function as a cell cycle inhibitor by acting as an anti-microtubule agent, and more specifically as a stabilizer. These compounds have been shown useful in the treatment of proliferative disorders, including: non-small cell (NSC) lung;
small cell lung; breast; prostate; cervical; endometrial; head and neck cancers.
In another aspect, the anti-microtuble agent (microtubule inhibitor) is albendazole (carbamic acid, [5-(propylthio)-1 H-benzimidazol-2-yl]-, methyl ester), LY-355703 (1,4-dioxa-8,11-diazacyclohexadec-13-ene-2,5,9,12-tetrone, 10-[(3-chloro-4-methoxyphenyl)methyl]-6,6-dimethyl-3-(2-methylpropyl)-16-[(1S)-1-[(2S,3R)-3-phenyloxiranyl]ethyl]-, (3S,10R,13E,16S)-), vindesine (vincaleukoblastine, 3-(aminocarbonyl)-04-deacetyl-3-de(methoxycarbonyl)-), or WAY-174286 In another aspect, the cell cycle inhibitor is a vinca alkaloid. Vinca alkaloids have the following general structure. They are indole-dihydroindole dimers.
1e dihydroindole As disclosed in U.S. Patent Nos. 4,841,045 and 5,030,620, R~ can be a formyl or methyl group or alternately H. R~ can also be an alkyl group or an aldehyde-substituted alkyl (e.g., CH2CH0). R2 is typically a CH3 or NH2 group. However it can be alternately substituted with a lower alkyl ester or the ester linking to the dihydroindole core may be substituted with C(O)-R where R
is NH2, an amino acid ester or a peptide ester. R3 is typically C(O)CH3, CH3 or H. Alternately, a protein fragment may be linked by a bifunctional group, such as maleoyl amino acid. R3 can also be substituted to form an alkyl ester which may be further substituted. R4 may be -CH2- or a single bond. R5 and R6 may be H, OH or a lower alkyl, typically -CH2CH3. Alternatively R6 and R~ may together form an oxetane ring. R7 may alternately be H. Further substitutions include molecules wherein methyl groups are substituted with other alkyl groups, and whereby unsaturated rings may be derivatized by the addition of a side group such as an alkane, alkene, alkyne, halogen, ester, amide or amino group.
Exemplary vinca alkaloids are vinblastine, vincristine, vincristine sulfate, vindesine, and vinorelbine, having the structures:
H
Ri Rz R3 Ra R5 Vinblastine:CH3 C(O)CH3 CHz Vincristine:CH3 C(O)CH3 CHz Vindesine: NHz H OH CHz Vinorelbine:CH3 CH3 H single CH3 bond Analogues typically require the side group (shaded area) in order to have activity. These compounds are thought to act as cell cycle inhibitors by functioning as anti-microtubule agents, and more specifically to inhibit polymerization. These compounds have been shown useful in treating proliferative disorders, including NSC lung; small cell lung; breast;
prostate;

brain; head and neck; retinoblastoma; bladder; and penile cancers; and soft tissue sarcoma.
In another aspect, the cell cycle inhibitor is a camptothecin, or an analog or derivative thereof. Camptothecins have the following general structure.
In this structure, X is typically O, but can be other groups, e.g., NH
in the case of 21-lactam derivatives. R~ is typically H or OH, but may be other groups, e.g., a terminally hydroxylated C~_3 alkane. R2 is typically H or an amino containing group such as (CH3)2NHCH2, but may be other groups e.g., N02, NH2, halogen (as disclosed in, e.g., U.S. Patent 5,552,156) or a short alkane containing these groups. R3 is typically H or a short alkyl such as C2H5.
R4 is typically H but may be other groups, e.g., a methylenedioxy group with R~
Exemplary camptothecin compounds include topotecan, irinotecan (CPT-11 ), 9-aminocamptothecin, 21-lactam-20(S)-camptothecin, 10,11-methylenedioxycamptothecin, SN-38, 9-nitrocamptothecin, 10-hydroxycamptothecin. Exemplary compounds have the structures:
R~ R~ R3 Camptothecin: H H H
Topotecan: OH (CH3)2NHCH2 H
SN-38: OH H C~HS
X: O for most analogs, NH for 21-lactam analogs Camptothecins have the five rings shown here. The ring labeled E must be intact (the lactone rather than carboxylate form) for maximum activity and minimum toxicity. These compounds are useful to as cell cycle inhibitors, where they can function as topoisomerase I inhibitors and/or DNA cleavage agents. They have been shown useful in the treatment of proliferative disorders, including, for example, NSC lung; small cell lung; and cervical cancers.
In another aspect, the cell cycle inhibitor is a podophyllotoxin, or a derivative or an analogue thereof. Exemplary compounds of this type are etoposide or teniposide, which have the following structures:
K
Etoposide CH3 Teniposide s OH
These compounds are thought to function as cell cycle inhibitors by being topoisomerase II inhibitors and/or by DNA cleaving agents. They have been shown useful as antiproliferative agents in, e.g., small cell lung, prostate, and brain cancers, and in retinoblastoma.
Another example of a DNA topoisomerase inhibitor is lurtotecan dihydrochloride (11H-1,4-dioxino[2,3-g]pyrano[3',4':6,7]indolizino[1,2-b]quinoline-9,12(8H,14H)-dione, 8-ethyl-2,3-dihydro-8-hydroxy-15-[(4-methyl-1-piperazinyl)methyl]-, dihydrochloride, (S)-).
In another aspect, the cell cycle inhibitor is an anthracycline.
Anthracyclines have the following general structure, where the R groups may be a variety of organic groups:
According to U.S. Patent 5,594,158, suitable R groups are: R~ is CH3 or CH20H; R2 is daunosamine or H; R3 and R4 are independently one of OH, N02, NH2, F, CI, Br, I, CN, H or groups derived from these; R5_~ are all H
or R5 and R6 are H and R~ and R$ are alkyl or halogen, or vice versa: R~ and R$
are H and R5 and R6 are alkyl or halogen.
According to U.S. Patent 5,843,903, R2 may be a conjugated peptide. According to U.S. Patent Nos. 4,215,062 and 4,296,105, R5 may be OH or an ether linked alkyl group. R~ may also be linked to the anthracycline ring by a group other than C(O), such as an alkyl or branched alkyl group having the C(O) linking moiety at its end, such as -CH2CH(CH2-?C)C(O)-R~, wherein X is H or an alkyl group (see, e.g., U.S. Patent 4,215,062). R2 may alternately be a group linked by the functional group =N-NHC(O)-Y, where Y is a group such as a phenyl or substituted phenyl ring. Alternately R3 may have the following structure:
H3C p NH

Rio in which R9 is OH either in or out of the plane of the ring, or is a second sugar moiety such as R3. Rio may be H or form a secondary amine with a group such as an aromatic group, saturated or partially saturated 5 or 6 membered heterocyclic having at least one ring nitrogen (see U.S. Patent 5,843,903).
Alternately, Rio may be derived from an amino acid, having the structure -C(O)CH(NHR~~)(R~2), in which R~~ is H, or forms a C3_4 membered alkylene with R~2. R~2 may be H, alkyl, aminoalkyl, amino, hydroxy, mercapto, phenyl, benzyl or methylthio (see U.S. Patent 4,296,105).
Exemplary anthracyclines are doxorubicin, daunorubicin, idarubicin, epirubicin, pirarubicin, zorubicin, and carubicin. Suitable compounds have the structures:

p O OH
Ri / r"'OH
R, O OH O
H C O
a NHz Ra R, Rz Ra Doxorobicin:OCHaCH,OHOH out of ring plane Epirubicin:OCHaCHzOHOH in ring plane ~

(4' oxorubicin) eplmar of d Daunorubicin: CHa OH out OCHa of ring plane Idarobicin:H CHa OH out of ring plane PirarubicinOCHaOH A

ZorubicinOCHa=N-NHC(O)CaHa B

CarubicinOH CHa B

A: ~ / 3: O /
O CHa O
OH
NH, Other suitable anthracyclines are anthramycin, mitoxantrone, menogaril, nogalamycin, aclacinomycin A, olivomycin A, chromomycin A3, and plicamycin having the structures:
off OH
H
Ha0 ~ N Anthramycin N ~ ~ NHz O
O
R, Rz Ra Menogaril H OCHa H
off o HN~ ~oH Nogalamycin O-sugar H COOCHa NH
sugar: CHa HaC O
O
OH O HN OH HaCO cH3 OCHa ~NH~
Mitoxantrone HO O v HO "''C~ O

O ~/
~.

CHa HaC O
R,O

HO
R~ Rz Ra RQ

Olivomycin A COCH(CHa)zCHaCOCHaH

Chromomycin Az COCHa CHaCOCHaCHa Plicamycin H H H CHa O OCHa O
CHa rr~.pH

These compounds are thought to function as cell cycle inhibitors by being topoisomerase inhibitors and/or by DNA cleaving agents. They have been shown useful in the treatment of proliferative disorders, including small cell lung; breast; endometrial; head and neck; retinoblastoma; liver; bile duct;
islet cell; and bladder cancers; and soft tissue sarcoma.
In another aspect, the cell cycle inhibitor is a platinum compound.
In general, suitable platinum complexes may be of Pt(II) or Pt(IV) and have this basic structure:

X
R1\Pt-R~ ~Y

wherein X and Y are anionic leaving groups such as sulfate, phosphate, carboxylate, and halogen; R1 and R2 are alkyl, amine, amino alkyl any may be further substituted, and are basically inert or bridging groups. For Pt(II) complexes Z1 and Z2 are non-existent. For Pt(IV) Z1 and Z2 may be anionic groups such as halogen, hydroxy, carboxylate, ester, sulfate or phosphate.
See, e.g., U.S. Patent Nos. 4,588,831 and 4,250,189.
Suitable platinum complexes may contain multiple Pt atoms. See, e.g., U.S. Patent Nos. 5,409,915 and 5,380,897. For example bisplatinum and triplatinum complexes of the type:

Z~ Zt R X R
X\ Pt/ ' \ ~t~ z y/ I ~A~ I \Y
Zz Zz X\ I / R t X\ Z/ A\ } / X
y/ It~ ~ It\Y R~ It\Y

Zz Zz Zz Z1 Zt X\ I / Rz Rz~ / X
Pt\ /Pt\
Y/ I A I Y
z2 I z2 Zz~ ~ / R3 Pt y/I\Z
X
Exemplary platinum compounds are cisplatin, carboplatin, oxaliplatin, and miboplatin having the structures:

IHs O O\
Pt CIIt-NH3 I ~NH3 O
CI
O
Cisplatin Carboplatin O o NHZ O NHz ~Pt H
O NHS"" O HN
z O O
Oxaliplatin Miboplatin These compounds are thought to function as cell cycle inhibitors by binding to DNA, i.e., acting as alkylating agents of DNA. These compounds have been shown useful in the treatment of cell proliferative disorders, including, e.g., NSC lung; small cell lung; breast; cervical; brain; head and neck;
esophageal; retinoblastom; liver; bile duct; bladder; penile; and vulvar cancers;
and soft tissue sarcoma.

In another aspect, the cell cycle inhibitor is a nitrosourea.
Nitrosourease have the following general structure (C5), where typical R
groups are shown below.

R'~ ,R
N NH
i N~
Wo (C5) R Group:
HZC
O
OH
OH
Carmustine OH O-CHs Ranimustine Lomustine NHZ OH
\CH3 ~ O
OH
O /~
~CHa ~ ~CH3 OH OH
Fotemustine Nimustine Chlorozotocin Streptozocin Other suitable R groups include cyclic alkanes, alkanes, halogen substituted groups, sugars, aryl and heteroaryl groups, phosphonyl and sulfonyl groups. As disclosed in U.S. Patent No. 4,367,239, R may suitably be CH2-C(X)(Y)(Z), wherein X and Y may be the same or different members of the following groups: phenyl, cyclyhexyl, or a phenyl or cyclohexyl group substituted with groups such as halogen, lower alkyl (C~_4), trifluore methyl, cyano, phenyl, cyclohexyl, lower alkyloxy (C~_4). Z has the following structure:
-alkylene-N-R~R2, where R~ and R2 may be the same or different members of the following group: lower alkyl (C~_4) and benzyl, or together R~ and R2 may form a saturated 5 or 6 membered heterocyclic such as pyrrolidine, piperidine, morfoline, thiomorfoline, N-lower alkyl piperazine, where the heterocyclic may be optionally substituted with lower alkyl groups.
As disclosed in U.S. Patent No. 6,096,923, R and R' of formula (C5) may be the same or different, where each may be a substituted or unsubstituted hydrocarbon having 1-10 carbons. Substitutions may include hydrocarbyl, halo, ester, amide, carboxylic acid, ether, thioether and alcohol groups. As disclosed in U.S. Patent No. 4,472,379, R of formula (C5) may be an amide bond and a pyranose structure (e.g., methyl 2'-(N-(N-(2-chloroethyl)-N-nitroso-carbamoyl)-glycyl)amino-2'-deoxy-a-D-glucopyranoside). As disclosed in U.S. Patent No. 4,150,146, R of formula (C5) may be an alkyl group of 2 to 6 carbons and may be substituted with an ester, sulfonyl, or hydroxyl group. It may also be substituted with a carboxylic acid or CONH2 group.
Exemplary nitrosoureas are BCNU (carmustine), methyl-CCNU
(semustine), CCNU (lomustine), ranimustine, nimustine, chlorozotocin, fotemustine, and streptozocin, having the structures:
O
CI' ~ ~ ,R
i NH R Group:
N
O
'CI
Carmustine HZC OH
O O
OH OH
OH OH O-CH OH OH
Ranimustine Lomustine O

NHZ OH \N~NH
H N~O
N CH3 OH ~ DH
Nimustine Chlorozotocin \CH3 ~~ \O~CH3 O
Fotemustine These nitrosourea compounds are thought to function as cell cycle inhibitors by binding to DNA, that is, by functioning as DNA alkylating agents. These cell cycle inhibitors have been shown useful in treating cell proliferative disorders such as, for example, islet cell; small cell lung;
melanoma; and brain cancers.
In another aspect, the cell cycle inhibitor is a nitroimidazole, where exemplary nitroimidazoles are metronidazole, benznidazole, etanidazole, and misonidazole, having the structures:

~R~

R3~
R~ R~ R3 Metronidazole OH CH3 NOz Benznidazole C(O)NHCHZ benzyl NOZ H
Etanidazole CONHCHZCH20H NO~ H
Suitable nitroimidazole compounds are disclosed in, e.g., U.S.
Patent Nos. 4,371,540 and 4,462,992.
In another aspect, the cell cycle inhibitor is a folic acid antagonist, such as methotrexate or derivatives or analogues thereof, including edatrexate, trimetrexate, raltitrexed, piritrexim, denopterin, tomudex, and pteropterin.
Methotrexate analogues have the following general structure:
Rg The identity of the R group may be selected from organic groups, particularly those groups set forth in U.S. Patent Nos. 5,166,149 and 5,382,582.
For example, R~ may be N, R2 may be N or C(CH3), R3 and R3' may H or alkyl, e.g., CH3, R4 may be a single bond or NR, where R is H or alkyl group. 85,6,8 may be H, OCH3, or alternately they can be halogens or hydro groups. R~ is a side chain of the general structure:
HO

wherein n = 1 for methotrexate, n = 3 for pteropterin. The carboxyl groups in the side chain may be esterified or form a salt such as a Zn2+ salt. R9 and Rio can be NH2 or may be alkyl substituted.
Exemplary folic acid antagonist compounds have the structures:
Rp R,R, R, R, Rs Rg Ra R, MethotrexaleNH,N N H N(CH,)H H A(n=i)H

EdatrexataNH,N N H N(CH,CH,)H H A(n=1)H

TrimetrexateNH,N C(CH,)H NH H OCH, OCH, OCH, PteroptednNH,N N H N(CH,)H H A H
(n~3) DenoptednOH N N CH, N(CH,)H H A(n=1)H

PiritreximNH,N C(CH,)singleOCH,H H OCH,H
H

bond A~ o NH
H0~

o off n HOOC~ O ~ H3 S N ~ ~ NH
HOOC~NH
O
Tomudex These compounds are thought to function as cell cycle inhibitors by serving as antimetabolites of folic acid. They have been shown useful in the treatment of cell proliferative disorders including, for example, soft tissue sarcoma, small cell lung, breast, brain, head and neck, bladder, and penile cancers.
In another aspect, the cell cycle inhibitor is a cytidine analogue, such as cytarabine or derivatives or analogues thereof, including enocitabine, FMdC ((E(-2'-deoxy-2'-(fluoromethylene)cytidine), gemcitabine, 5-azacitidine, ancitabine, and 6-azauridine. Exemplary compounds have the structures:

R, RZ R3 RQ

Cytarabine OH H CH
H

Enocitabine OH H CH
C(O)(CHz)zoCH3 Gemcitabine F F CH
H

Azacitidine H OH N
H

FMdC H CHZFH CH

Ancitabine 6-Azauridine These compounds are thought to function as cell cycle inhibitors as acting as antimetabolites of pyrimidine. These compounds have been shown useful in the treatment of cell proliferative disorders including, for example, pancreatic, breast, cervical, NSC lung, and bile duct cancers.
In another aspect, the cell cycle inhibitor is a pyrimidine analogue.
In one aspect, the pyrimidine analogues have the general structure:
wherein positions 2', 3' and 5' on the sugar ring (R2, R3 and R4, respectively) can be H, hydroxyl, phosphoryl (see, e.g., U.S. Patent 4,086,417) or ester (see, e.g., U.S. Patent 3,894,000). Esters can be of alkyl, cycloalkyl, aryl or heterocyclo/aryl types. The 2' carbon can be hydroxylated at either R2 or R2', the other group is H. Alternately, the 2' carbon can be substituted with halogens e.g., fluoro or difluoro cytidines such as Gemcytabine. Alternately, the sugar can be substituted for another heterocyclic group such as a furyl group or for an alkane, an alkyl ether or an amide linked alkane such as C(O)NH(CH2)5CH3. The 2° amine can be substituted with an aliphatic acyl (R~) linked with an amide (see, e.g., U.S. Patent 3,991,045) or urethane (see, e.g., U.S. Patent 3,894,000) bond. It can also be further substituted to form a quaternary ammonium salt. R5 in the pyrimidine ring may be N or CR, where R
is H, halogen containing groups, or alkyl (see, e.g., U.S. Patent No.
4,086,417).
R6 and R~ can together can form an oxo group or R6 = -NH-R~ and R~ = H. R$
is H or R~ and R$ together can form a double bond or R$ can be X, where X is:
CN
O ~ ~ O O

Specific pyrimidine analogues are disclosed in U.S. Patent No.
3,894,000 (see, e.g., 2'-O-palmityl-ara-cytidine, 3'-O-benzoyl-ara-cytidine, and more than 10 other examples); U.S. Patent No. 3,991,045 (see, e.g., N4-acyl-1-~i-D-arabinofuranosylcytosine, and numerous acyl groups derivatives as listed therein, such as palmitoyl.
In another aspect, the cell cycle inhibitor is a fluoropyrimidine analogue, such as 5-fluorouracil, or an analogue or derivative thereof, including carmofur, doxifluridine, emitefur, tegafur, and floxuridine. Exemplary compounds have the structures:

Rz F
O \~N~
Ri Ri R2 5-FluorouracilH H

CarmofurC(O)NH(CHZ)SCH3 H

DoxifluridineA, H

FloxuridineAZ H

EmitefurCHZOCH2CH3 B

TegafurC H

Ai HO > HO
p O CF
OH OH OH
B / ~ CN O O
O N O
O
Other suitable fluoropyrimidine analogues include 5-FudR (5-fluoro-deoxyuridine), or an analogue or derivative thereof, including 5-iododeoxyuridine (5-IudR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP).
Exemplary compounds have the structures:

R
'NH
HO ~
N' 'O
O
OH
5-Fluoro-2'-deoxyuridine: R = F
5-Bromo-2'-deoxyuridine: R = Br 5-lodoo-2'-deoxyuridine: R = I
These compounds are thought to function as cell cycle inhibitors by serving as antimetabolites of pyrimidine. These compounds have been shown useful in the treatment of cell proliferative disorders such as breast, cervical, non-melanoma skin, head and neck, esophageal, bile duct, pancreatic, islet cell, penile, and vulvar cancers.
In another aspect, the cell cycle inhibitor is a purine analogue.
Purine analogues have the following general structure.
R
wherein X is typically carbon; R~ is H, halogen, amine or a substituted phenyl;
R2 is H, a primary, secondary or tertiary amine, a sulfur containing group, typically-SH, an alkane, a cyclic alkane, a heterocyclic or a sugar; R3 is H, a sugar (typically a furanose or pyranose structure), a substituted sugar or a cyclic or heterocyclic alkane or aryl group. See, e.g., U.S. Patent No.
5,602,140 for compounds of this type.
In the case of pentostatin, X-R2 is -CH~CH(OH)-. In this case a second carbon atom is inserted in the ring between X and the adjacent nitrogen atom. The X-N double bond becomes a single bond.
U.S. Patent No. 5,446,139 describes suitable purine analogues of the type shown in the formula.
R\3 \Z'sV
B
R~
~A
X /w R~
~Y
wherein N signifies nitrogen and V, W, X, Z can be either carbon or nitrogen with the following provisos. Ring A may have 0 to 3 nitrogen atoms in its structure. If two nitrogens are present in ring A, one must be in the W
position.
If only one is present, it must not be in the Q position. V and Q must not be simultaneously nitrogen. Z and Q must not be simultaneously nitrogen. If Z is nitrogen, R3 is not present. Furthermore, R~_3 are independently one of H, halogen, C~_7 alkyl, C~_~ alkenyl, hydroxyl, mercapto, C~_~ alkylthio, C~_~
alkoxy, C~_~ alkenyloxy, aryl oxy, vitro, primary, secondary or tertiary amine containing group. R5_8 are H or up to two of the positions may contain independently one of OH, halogen, cyano, azido, substituted amino, R5 and R7 can together form a double bond. Y is H, a C~_7 alkylcarbonyl, or a mono- di or tri phosphate.
Exemplary suitable purine analogues include 6-mercaptopurine, thiguanosine, thiamiprine, cladribine, fludaribine, tubercidin, puromycin, pentoxyfilline; where these compounds may optionally be phosphorylated.
Exemplary compounds have the structures: .
Ra N~ N
R~N I y R~
R, Ra R~ A: o~ BaHo 6-Mercaptopurine H SN H
Thioguanosine NHa SH B, j CHI OH OH
Thiamiprine NHa A H
Ba: B3:
Cladribine CI NHa Ba HO HO
Fludarabine F NH, B, H O OH
Puromycin H N(CH,)a Bq off off HO
Tubercidin H NHa B, B4' H
NH OH

O N

N

Pentoxyfilline These compounds are thought to function as cell cycle inhibitors by serving as antimetabolites of purine.
In another aspect, the cell cycle inhibitor is a nitrogen mustard.
Many suitable nitrogen mustards are known and are suitably used as a cell cycle inhibitor in the present invention. Suitable nitrogen mustards are also known as cyclophosphamides.
A preferred nitrogen mustard has the general structure:

R~
N
A~ ~CI
Where A is:
~~ i P\0 N~
Rz or -CH3 or other alkane, or chloronated alkane, typically CH2CH(CH3)CI, or a polycyclic group such as B, or a substituted phenyl such as C or a heterocyclic group such as D.

H ~~~'H
H0~--~~H
HOOC
NHZ
H
N
O
H

(IV) Examples of suitable nitrogen mustards are disclosed in U.S.
Patent No. 3,808,297, wherein A is:
P\
O
N~

R~_2 are H or CH2CH2C1; R3 is H or oxygen-containing groups such as hydroperoxy; and R4 can be alkyl, aryl, heterocyclic.
The cyclic moiety need not be intact. See, e.g., U.S. Patent Nos.
5,472,956, 4,908,356, 4,841,085 that describe the following type of structure:
wherein R~ is H or CH2CH2C1, and R2_6 are various substituent groups.
Exemplary nitrogen mustards include methylchloroethamine, and analogues or derivatives thereof, including methylchloroethamine oxide hydrohchloride, novembichin, and mannomustine (a halogenated sugar).
Exemplary compounds have the structures:
CI
CI
R
I HCI
R

Mechlorethanime CH3 Mechiorethanime Oxide HCI
Novembichin CHZCH(CH3)CI
The nitrogen mustard may be cyclophosphamide, ifosfamide, perfosfamide, or torofosfamide, where these compounds have the structures:

R~ R2 R3 Cyclophosphamide H CHZCHZCIH

Ifosfamide CH2CH~CI H H

Perfosfamide CH2CHZCI H OOH

Torofosfamide CHZCHZCI H
CH~CHZCI

The nitrogen mustard may be estramustine, or an analogue or derivative thereof, including phenesterine, prednimustine, and estramustine P04. Thus, suitable nitrogen mustard type cell cycle inhibitors of the present invention have the structures:
The nitrogen mustard may be chlorambucil, or an analogue or derivative thereof, including melphalan and chlormaphazine. Thus, suitable nitrogen mustard type cell cycle inhibitors of the present invention have the structures:
R
Estramustine OH
Phenesterine C(CH3)(CHZ)3CH(CH3)z N I
R~
Rz R I

R~ Rz R3 Chlorambucil H H
CHZCOOH

Melphalan COOH NHZ H

Chlornaphazine together H ~ forms a benzene ring The nitrogen mustard may be uracil mustard, which has the structure:
H
O ~ ~ I
H
CI
The nitrogen mustards are thought to function as cell cycle inhibitors by serving as alkylating agents for DNA. Nitrogen mustards have been shown useful in the treatment of cell proliferative disorders including, for example, small cell lung, breast, cervical, head and neck, prostate, retinoblastoma, and soft tissue sarcoma.
The cell cycle inhibitor of the present invention may be a hydroxyurea. Hydroxyureas have the following general structure:

~N N~
R2 R~
Suitable hydroxyureas are disclosed in, for example, U.S. Patent No. 6,080,874, wherein R~ is:
/ s R2 and R2 is an alkyl group having 1-4 carbons and R3 is one of H, acyl, methyl, ethyl, and mixtures thereof, such as a methylether.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,665,768, wherein R~ is a cycloalkenyl group, for example N-(3-(5-(4-fluorophenylthio)-furyl)-2-cyclopenten-1-yl)N-hydroxyurea; R2 is H or an alkyl group having 1 to 4 carbons and R3 is H; X is H or a cation.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 4,299,778, wherein R~ is a phenyl group substituted with on or more fluorine atoms; R2 is a cyclopropyl group; and R3 and X is H.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,066,658, wherein R2 and R3 together with the adjacent nitrogen form:
(~~zm Y . N-(CHz)m wherein m is 1 or 2, n is 0-2 and Y is an alkyl group.
In one aspect, the hydroxy urea has the structure:

~oH

Hydroxyurea Hydroxyureas are thought to function as cell cycle inhibitors by serving to inhibit DNA synthesis.
In another aspect, the cell cycle inhibitor is a mytomicin, such as mitomycin C, or an analogue or derivative thereof, such as porphyromycin.
Exemplary compounds have the structures:
R
Mitomycin C H
Porphyromycin CH3 (N-methyl Mitomycin C) These compounds are thought to function as cell cycle inhibitors by serving as DNA alkylating agents. Mitomycins have been shown useful in the treatment of cell proliferative disorders such as, for example, esophageal, liver, bladder, and breast cancers.
In another aspect, the cell cycle inhibitor is an alkyl sulfonate, such as busulfan, or an analogue or derivative thereof, such as treosulfan, improsulfan, piposulfan, and pipobroman. Exemplary compounds have the structures:

H ~O CHa ~

() R
~~

O O

R

Busulfan single bond Improsulfan -CHZ NH-CHZ

Piposulfan ~ O
N~

~N~
// ~ ~

O

//~~ O
B~N~ ~Br ~~ ~ \~/O
1 O Pipobroman These compounds are thought to function as cell cycle inhibitors by serving as DNA alkylating agents.
In another aspect, the cell cycle inhibitor is a benzamide. In yet another aspect, the cell cycle inhibitor is a nicotinamide. These compounds have the basic structure:
A
wherein X is either O or S; A is commonly NHa or it can be OH or an alkoxy group; B is N or C-R4, where R4 is H or an ether-linked hydroxylated alkane such as OCH~CH20H, the alkane may be linear or branched and may contain 5~

one or more hydroxyl groups. Alternately, B may be N-R5 in which case the double bond in the ring involving B is a single bond. R5 may be H, and alkyl or an aryl group (see, e.g., U.S. Patent No. 4,258,052); R2 is H, OR6, SR6 or NHR6, where R6 is an alkyl group; and R3 is H, a lower alkyl, an ether linked lower alkyl such as -O-Me or-O-ethyl (see, e.g., U.S. Patent No. 5,215,738).
Suitable benzamide compounds have the structures:
x z \NH2 Y
N
Benzamides X=OorS
Y = H, OR, CH3, or acetoxy Z = H, OR, SR, or NHR
R = alkyl group where additional compounds are disclosed in U.S. Patent No. 5,215,738, (listing some 32 compounds).
Suitable nicotinamide compounds have the structures:
X
Z
\NHz N
Nicotinamides X=OorS
Z = H, OR, SR, NHR
R = alkyl group where additional compounds are disclosed in U.S. Patent No.
5,215,738, R O
z O II
Rz j~
N-~ ~-NH~O~R, Rz O
N
Rz H3C N
~ O
Rz I I Rz Rz Rz ~N O NHz R, Rz Benzodepa phenyl H ~ ~~CH3 Meturedepa CH3 CH3 Carboquone Uredepa CH3 H

In another aspect, the cell cycle inhibitor is a halogenated sugar, such as mitolactol, or an analogue or derivative thereof, including mitobronitol and mannomustine. Exemplary compounds have the structures:
CHyBr CHaBr CH~NHZ CHaCH2Cl H OH HO H HO H
HO H HO H HO H
HO H H OH H OH
H OH H OH H OH
CHZBr CHzBr CHaNHp+CH2CHZC1 Mitolactol Mitobronitol Mannomustine In another aspect, the cell cycle inhibitor is a diazo compound, such as azaserine, or an analogue or derivative thereof, including 6-diazo-5-oxo-L-norleucine and 5-diazouracil (also a pyrimidine analog). Exemplary compounds have the structures:

N=N~ R~~R~
OH
O NHZ
R~ R2 Azaserine O single bond 6-d i azo-5-oxo-L-norleucine single bond CH2 Other compounds that may serve as cell cycle inhibitors according to the present invention are pazelliptine; wortmannin;
metoclopramide; RSU; buthionine sulfoxime; tumeric; curcumin; AG337, a thymidylate synthase inhibitor; levamisole; lentinan, a polysaccharide;
razoxane, an EDTA analogue; indomethacin; chlorpromazine; oc and (i interferon; MnBOPP; gadolinium texaphyrin; 4-amino-1,8-naphthalimide;
staurosporine derivative of CGP; and SR-2508.
Thus, in one aspect, the cell cycle inhibitor is a DNA alylating agent. In another aspect, the cell cycle inhibitor is an anti-microtubule agent.
In another aspect, the cell cycle inhibitor is a topoisomerase inhibitor. In another aspect, the cell cycle inhibitor is a DNA cleaving agent. In another aspect, the cell cycle inhibitor is an antimetabolite. In another aspect, the cell cycle inhibitor functions by inhibiting adenosine deaminase (e.g., as a purine analogue). In another aspect, the cell cycle inhibitor functions by inhibiting purine ring synthesis and/or as a nucleotide interconversion inhibitor (e.g., as a . purine analogue such as mercaptopurine). In another aspect, the cell cycle inhibitor functions by inhibiting dihydrofolate reduction and/or as a thymidine monophosphate block (e.g., methotrexate). In another aspect, the cell cycle inhibitor functions by causing DNA damage (e.g., bleomycin). In another aspect, the cell cycle inhibitor functions as a DNA intercalation agent and/or RNA synthesis inhibition (e.g., doxorubicin, aclarubicin, or detorubicin (acetic acid, diethoxy-, 2-[4-[(3-amino-2,3,6-trideoxy-alpha-L-lyxo-hexopyranosyl)oxy]-1,2,3,4,6,11-hexahydro-2, 5,12-trihyd roxy-7-methoxy-6,11-d ioxo-2-naphthacenyl]-2-oxoethyl ester, (2S-cis)-)). In another aspect, the cell cycle inhibitor functions by inhibiting pyrimidine synthesis (e.g., N-phosphonoacetyl-L-aspartate). In another aspect, the cell cycle inhibitor functions by inhibiting ribonucleotides (e.g., hydroxyurea). In another aspect, the cell cycle inhibitor functions by inhibiting thymidine monophosphate (e.g., 5-fluorouracil). In another aspect, the cell cycle inhibitor functions by inhibiting DNA synthesis (e.g., cytarabine). In another aspect, the cell cycle inhibitor functions by causing DNA adduct formation (e.g., platinum compounds). In another aspect, the cell cycle inhibitor functions by inhibiting protein synthesis (e.g., L-asparginase). In another aspect, the cell cycle inhibitor functions by inhibiting microtubule function (e.g., taxanes). In another aspect, the cell cycle inhibitor acts at one or more of the steps in the biological pathway shown in FIG. 1.
Additional cell cycle inhibitor s useful in the present invention, as well as a discussion of the mechanisms of action, may be found in Hardman J.G., Limbird L.E. Molinoff R.B., Ruddon R W., Gilman A.G. editors, Chemotherapy of Neoplastic Diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics Ninth Edition, McGraw-Hill Health Professions Division, New York, 1996, pages 1225-1287. See also U.S. Patent Nos. 3,387,001; 3,808,297; 3,894,000; 3,991,045; 4,012,390; 4,057,548;
4,086,417; 4,144,237; 4,150,146; 4,210,584; 4,215,062; 4,250,189; 4,258,052;
4,259,242; 4,296,105; 4,299,778; 4,367,239; 4,374,414; 4,375,432; 4,472,379;
4,588,831; 4,639,456; 4,767,855; 4,828,831; 4,841,045; 4,841,085; 4,908,356;
4,923,876; 5,030,620; 5,034,320; 5,047,528; 5,066,658; 5,166,149; 5,190,929;
5,215,738; 5,292,731; 5,380,897; 5,382,582; 5,409,915; 5,440,056; 5,446,139;
5,472,956; 5,527,905; 5,552,156; 5,594,158; 5,602,140; 5,665,768; 5,843,903;
6,080,874; 6,096,923; and RE030561.

In another embodiment, the cell-cycle inhibitor is camptothecin, mitoxantrone, etoposide, 5-fluorouracil, doxorubicin, methotrexate, peloruside A, mitomycin C, or a CDK-2 inhibitor or an analogue or derivative of any member of the class of listed compounds.
In another embodiment, the cell-cycle inhibitor is HTI-286, plicamycin; or mithramycin, or an analogue or derivative thereof.
Other examples of cell cycle inhibitors also include, e.g., 7-hexanoyltaxol (QP-2), cytochalasin A, lantrunculin D, actinomycin-D, Ro-31 7453 (3-(6-vitro-1-methyl-3-indolyl)-4-(1-methyl-3-indolyl)pyrrole-2,5-dione), PNU-151807, brostallicin, C2-ceramide, cytarabine ocfosfate (2(1 H) pyrimidinone, 4-amino-1-(5-O-(hydroxy(octadecyloxy)phosphinyl)-f~-D-arabinofuranosyl)-, monosodium salt), paclitaxel (5f5,20-epoxy-1,2 alpha,4,7(3,10f3,13 alpha-hexahydroxytax-11-en-9-one-4,10-diacetate-2-benzoate-13-(alpha-phenylhippurate)), doxorubicin (5,12-naphthacenedione, 10-((3-amino-2,3,6-trideoxy-alpha-L-lyxo-hexopyranosyl)oxy)-7,8,9,10-tetrahydro-6,8,11-trihydroxy-8-(hydroxyacetyl)-1-methoxy-, (8S)-cis-), daunorubicin (5,12-naphthacenedione, 8-acetyl-10-((3-amino-2,3,6-trideoxy-alpha-L-lyxo-hexopyranosyl)oxy)-7,8,9,10-tetrahydro-6,8,11-trihydroxy-1-methoxy-, (8S-cis)-), gemcitabine hydrochloride (cytidine, 2'-deoxy-2', 2'-difluoro-,monohydrochloride), nitacrine (1,3-propanediamine, N,N-dimethyl-N'-(1-vitro-9-acridinyl)-), carboplatin (platinum, diammine(1,1-cyclobutanedicarboxylato(2-))-, (SP-4-2)-), altretamine (1,3,5-triazine-2,4,6-triamine, N,N,N',N',N",N"-hexamethyl-), teniposide (furo(3',4':6,7)naphtho(2,3-d)-1,3-dioxol-6(5aH)-one, 5,8,8a,9-tetrahydro-5-(4-hydroxy-3,5-dimethoxyphenyl)-9-((4,6-O-(2-thienylmethylene)-f~-D-glucopyranosyl)oxy)-, (5R-(5alpha,5a(3,8aAlpha,9f3(R*)))-), eptaplatin (platinum, ((4R,5R)-2-(1-methylethyl)-1,3-dioxolane-4,5-dimethanamine-kappa N4,kappa N5)(propanedioato(2-)-kappa 01, kappa 03)-, (SP-4-2)-), amrubicin hydrochloride (5,12-naphfhacenedione, 9-acetyl-9-amino-7-((2-deoxy-f3-D-erythro-pentopyranosyl)oxy)-7,8,9,10-tetrahydro-6,11-dihydroxy-, hydrochloride, (7S-cis)-), ifosfamide (2H-1,3,2-oxazaphosphorin-2-amine, N,3-bis(2-chloroethyl)tetrahydro-,2-oxide), cladribine (adenosine, 2-chloro-2'-deoxy-), mitobronitol (D-mannitol, 1,6-dibromo-1,6-dideoxy-), fludaribine phosphate (9H-purin-6-amine, 2-fluoro-9-(5-O-phosphono-(3-D-arabinofuranosyl)-), enocitabine (docosanamide, N-(1-(3-D-arabinofuranosyl-1,2-dihydro-2-oxo-4-pyrimidinyl)-), vindesine (vincaleukoblastine, 3-(aminocarbonyl)-04-deacetyl-3-de(methoxycarbonyl)-), idarubicin (5,12-naphthacenedione, 9-acetyl-7-((3-amino-2,3,6-trideoxy-alpha-L-lyxo-hexopyranosyl)oxy)-7,8,9,10-tetrahyd ro-6,9,11-trihydroxy-, (7S-cis)-), zinostatin (neocarzinostatin), vincristine (vincaleukoblastine, 22-oxo-), tegafur (2,4(1 H,3H)-pyrimidinedione, 5-fluoro-(tetrahydro-2-furanyl)-), razoxane (2,6-piperazinedione, 4,4'-(1-methyl-1,2-ethanediyl)bis-), methotrexate (L-glutamic acid, N-(4-(((2,4-diamino-6-pteridinyl)methyl)methylamino)benzoyl)-), raltitrexed (L-glutamic acid, N-((5-(((1,4-dihydro-2-methyl-4-oxo-6-quinazolinyl)methyl)methylamino)-2=
thienyl)carbonyl)-), oxaliplatin (platinum, (1,2-cyclohexanediamine-N,N')(ethanedioato(2-)-O,O')-, (SP-4-2-(1 R-trans))-), doxifluridine (uridine, 5'-deoxy-5-fluoro-), mitolactol (galactitol, 1,6-dibromo-1,6-dideoxy-), piraubicin (5,12-naphthacenedione, 10-((3-amino-2,3,6-trideoxy-4-O-(tetrahydro-2H-pyran-2-yl)-alpha-L-lyxo-hexopyranosyl)oxy)-7,8,9,10-tetrahydro-6,8,11-trihydroxy-8-(hydroxyacetyl)-1-methoxy-, (8S-(8 alpha, 10 alpha(S*)))-), docetaxel ((2R,3S)-N-carboxy-3-phenylisoserine, N-tert-butyl ester, 13-ester with 5f~,20-epoxy-1,2 alpha,4,7f3,10f3,13 alpha-hexahydroxytax-11-en-9-one 4-acetate 2-benzoate-), capecitabine (cytidine, 5-deoxy-5-fluoro-N-((pentyloxy)carbonyl)-), cytarabine (2(1 H)-pyrimidone, 4-amino-1-f3-D-arabino furanosyl-), valrubicin (pentanoic acid, 2-(1,2,3,4,6,11-hexahydro-2,5,12-trihydroxy-7-methoxy-6,11-dioxo-4-((2,3,6-trideoxy-3-((trifluoroacetyl)amino)-alpha-L-lyxo-hexopyranosyl)oxy)-2-naphthacenyl)-2-oxoethyl ester (2S-cis)-), trofosfamide (3-2-(chloroethyl)-2-(bis(2-chloroethyl)amino)tetrahydro-2H-1,3,2-oxazaphosphorin 2-oxide), prednimustine (pregna-1,4-diene-3,20-dione, 21-(4-(4-(bis(2-chloroethyl)amino)phenyl)-1-oxobutoxy)-11,17-dihydrbxy-, (11f~)-), lomustine (Urea, N-(2-chloroethyl)-N'-cyclohexyl-N-nitroso-), epirubicin (5,12-naphthacenedione, 10-((3-amino-2,3,6-trideoxy-alpha-L-arabino-hexopyranosyl)oxy)-7,8,9,10-tetrahydro-6,8,11-trihydroxy-8-(hydroxyacetyl)-1-methoxy-, (8S-cis)-), or an analogue or derivative thereof).
5) Cyclin Dependent Protein Kinase Inhibitors In another embodiment, the pharmacologically active compound is a cyclin dependent protein kinase inhibitor (e.g., R-roscovitine, CYC-101, CYC-103, CYC-400, MX-7065, alvocidib (4H-1-Benzopyran-4-one, 2-(2-chlorophenyl)-5,7-dihydroxy-8-(3-hydroxy-1-methyl-4-piperidinyl)-, cis-(-)-), SU-9516, AG-12275, PD-0166285, CGP-79807, fascaplysin, GW-8510 (benzenesulfonamide, 4-(((~)-(6,7-dihydro-7-oxo-8H-pyrrolo(2,3-g)benzothiazol-8-ylidene)methyl)amino)-N-(3-hydroxy-2,2-dimethylpropyl)-), GW-491619, Indirubin 3' monoxime, GW8510, AZD-5438, ZK-CDK or an analogue or derivative thereof).
6) EGF (Epidermal Growth Factor) Receptor Kinase Inhibitors In another embodiment, the pharmacologically active compound is an EGF (epidermal growth factor) kinase inhibitor (e.g., erlotinib (4-quinazolinamine, N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-, monohydrochloride), erbstatin, BIBX-1382, gefitinib (4-quinazolinamine, N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-(4-morpholinyl)propoxy)), or an analogue or derivative thereof).
7) Elastase Inhibitors In another embodiment, the pharmacologically active compound is ~an elastase inhibitor (e.g., ONO-6818, sivelestat sodium hydrate (glycine, N-(2-(((4-(2,2-dimethyl-1-oxopropoxy)phenyl)sulfonyl)amino)benzoy1)-), erdosteine (acetic acid, ((2-oxo-2-((tetrahydro-2-oxo-3-thienyl)amino)ethyl)thio)-), MDL-100948A, MDL-104238 (N-(4-(4-morpholinylcarbonyl)benzoyl)-L-valyl-N'-(3,3,4,4,4-pentafluoro-1-(1-methylethyl)-2-oxobutyl)-L-2-azetamide), MDL-27324 (L-prolinamide, N-((5-(dimethylamino)-1-naphthalenyl)sulfonyl)-L-alanyl-L-alanyl-N-(3,3,3-trifluoro-1-(1-methylethyl)-2-oxopropyl)-, (S)-), SR-26831 (thieno(3,2-c)pyridinium, 5-((2-chlorophenyl)methyl)-2-(2,2-dimethyl-1-oxopropoxy)-4,5,6,7-tetrahydro-5-hydroxy-), Win-68794, Win-63110, SSR-69071 (2-(9(2-piperidinoethoxy)-4-oxo-4H-pyrido(1,2-a)pyrimidin-2-yloxymethyl)-4-( 1-methylethyl)-6-methyoxy-1,2-benzisothiazol-3(2H)-one-1,1-dioxide), (N(Alpha)-(1-adamantylsulfonyl)N(epsilon)-succinyl-L-lysyl-L-prolyl-L-valinal), Ro-31-3537 (N alpha-(1-adamantanesulphonyl)-N-(4-carboxybenzoyl)-L-lysyl-alanyl-L-valinal), R-665, FCE-28204, ((6R,7R)-2-(benzoyloxy)-7-methoxy-3-methyl-4-pivaloyl-3-cephem 1,1-dioxide), 1,2-benzisothiazol-3(2H)-one, 2-(2,4-dinitrophenyl)-, 1,1-dioxide, L-658758 (L-proline, 1-((3-((acetyloxy)methyl)-7-methoxy-8-oxo-5-thia-1-azabicyclo(4.2.0)oct-2-en-2-yl)carbonyl)-, S,S-dioxide, (6R-cis)-), L-659286 (pyrrolidine, 1-((7-methoxy-8-oxo-3-(((1,2,5,6-tetrahydro-2-methyl-5,6-dioxo-1,2,4-triazin-3-yl)thio)methyl)-thia-1-azabicyclo(4.2.0)oct-2-en-2-yl)carbonyl)-, S,S-dioxide, (6R-cis)-), L-680833 (benzeneacetic acid, 4-((3,3-diethyl-1-(((1-(4-methylphenyl)butyl)amino)carbonyl)-4-oxo-2-azetidinyl)oxy)-, (S-(R*,S*))-), FK-706 (L-prolinamide, N-[4-[[(carboxymethyl)amino]carbonylJbenzoyl]-L-valyl-N-[3,3,3-trifluoro-1-(1-methylethyl)-2-oxopropyl]-, monosodium salt), Roche 8-665, or an analogue or derivative thereof).
8) Factor Xa Inhibitors In another embodiment, the pharmacologically active compound is a factor Xa inhibitor (e.g., CY-222, fondaparinux sodium (alpha-D-glucopyranoside, methyl O-2-deoxy-6-O-sulfo-2-(sulfoamino)-alpha-D-glucopyranosyl-( 1-4)-O-f3-D-glucopyranuronosyl-(1-4)-O-2-deoxy-3, 6-d i-O-sulfo-2-(sulfoamino)-alpha-D-glucopyranosyl-(1-4)-O-2-O-sulfo-alpha-L-idopyranuronosyl-(1-4)-2-deoxy-2-(sulfoamino)-, 6-(hydrogen sulfate)), danaparoid sodium, or an analogue or derivative thereof).
9) Farnesyltransferase Inhibitors In another embodiment, the pharmacologically active compound is a farnesyltransferase inhibitor (e.g., dichlorobenzoprim (2,4-diamino-5-(4-(3,4-dichlorobenzylamino)-3-nitrophenyl)-6-ethylpyrimidine), B-581, B-956 (N-(8(R)-amino-2(S)-benzyl-5(S)-isopropyl-9-sulfanyl-3(Z),6(E)-nonadienoyl)-L-methionine), OSI-754, perillyl alcohol (1-cyclohexene-1-methanol, 4-(1-methylethenyl)-, RPR-114334, lonafarnib (1-piperidinecarboxamide, 4-(2-(4-((11 R)-3,10-dibromo-8-chloro-6,11-dihydro-5H-benzo(5,6)cyclohepta(1,2-b)pyridin-11-yl)-1-piperidinyl)-2-oxoethyl)-), Sch-48755, Sch-226374, (7,8-dichloro-5H-dibenzo(b,e)(1,4)diazepin-11-y1)-pyridin-3-ylmethylamine, J-104126, L-639749, L-731734 (pentanamide, 2-((2-((2-amino-3-mercaptopropyl)amino)-3-methylpentyl)amino)-3-methyl-N-(tetrahydro-2-oxo-3-furanyl)-, (3S-(3R*(2R*(2R*(S*),3S*),3R*)))-), L-744832 (butanoic acid, 2-((2-((2-((2-amino-3-mercaptopropyl)amino)-3-methylpentyl)oxy)-1-oxo-3-phenylpropyl)amino)-4-(methylsulfonyl)-, 1-methylethyl ester, (2S-(1(R*(R*)),2R*(S*),3R*))-), L-745631 (1-piperazinepropanethiol, (3-amino-2-(2-methoxyethyl)-4-(1-naphthalenylcarbonyl)-, (f3R,2S)-), N-acetyl-N-naphthylmethyl-2(S)-((1-(4-cyanobenzyl)-1 H-imidazol-5-yl)acetyl)amino-3(S)-methylpentamine, (2alpha)-2-hydroxy-24,25-dihydroxylanost-8-en-3-one, BMS-316810, UCF-1-C (2,4-decadienamide, N-(5-hydroxy-5-(7-((2-hydroxy-5-oxo-1-cyclopenten-I-yl)amino-oxo-1,3,5-heptatrienyl)-2-oxo-7-oxabicyclo(4.1.0)hept-3-en-3-yl)-2,4,6-trimethyl-, (1S-(1alpha,3(2E,4E,6S*),5 alpha, 5(1E,3E,5E), 6 alpha))-), UCF-116-B, ARGLABIN (3H-oxireno[8,8a]azuleno[4,5-b]furan-8(4aH)-one, 5,6,6a,7,9a,9b-hexahydro-1,4a-dimethyl-7-methylene-, (3aR,4aS,6aS,9aS,9bR)-) from ARGLABIN - Paracure, Inc. (Virginia Beach, VA), or an analogue or derivative thereof).
10) Fibrinogen Antagonists In another embodiment, the pharmacologically active compound is a fibrinogen antagonist (e.g., 2(S)-((p-toluenesulfonyl)amino)-3-(((5,6,7,8,-tetrahydro-4-oxo-5-(2-(piperidin-4-yl)ethyl)-4H-pyrazolo-( 1,5-a)( 1,4)diazepin-2-yl)carbonyl)-amino)propionic acid, streptokinase (kinase (enzyme-activating), strepto-), urokinase (kinase (enzyme-activating), uro-), plasminogen activator, pamiteplase, monteplase, heberkinase, anistreplase, alteplase, pro-urokinase, picotamide (1,3-benzenedicarboxamide, 4-methoxy-N,N'-bis(3-pyridinylmethyl)-), or an analogue or derivative thereof).
11 ) Guanylate Cyclase Stimulants In another embodiment, the pharmacologically active compound is a guanylate cyclase stimulant (e.g., isosorbide-5-mononitrate (D-glucitol, 1,4:3,6-dianhydro-, 5-nitrate), or an analogue or derivative thereof).
12) Heat Shock Protein 90 Antagonists In another embodiment, the pharmacologically active compound is a heat shock protein 90 antagonist (e.g., geldanamycin; NSC-33050 (17-allylaminogeldanamycin), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), 17AAG, or an analogue or derivative thereof).
13) HMGCoA Reductase Inhibitors In another embodiment, the pharmacologically active compound is an HMGCoA reductase inhibitor (e.g., BCP-671, BB-476, fluvastatin (6-heptenoic acid, 7-(3-(4-fluorophenyl)-1-(1-methylethyl)-1 H-indol-2-yl)-3,5-dihydroxy-, monosodium salt, (R*,S*-(E))-(~)-), dalvastatin (2H-pyran-2-one, 6-(2-(2-(2-(4-fluoro-3-methylphenyl)-4,4,6,6-tetramethyl-1-cyclohexen-1-yl)ethenyl)tetrahydro)-4-hydroxy-, (4alpha,6f3(E))-(+/-)-), glenvastatin (2H-pyran-2-one, 6-(2-(4-(4-fluorophenyl)-2-(1-methylethyl')-6-phenyl-3-pyridinyl)ethenyl)tetrahydro-4-hydroxy-, (4R-(4alpha,6(3(E)))-), S-2468, N-(1 oxododecyl)-4Alpha,10-dimethyl-8-aza-trans-decal-3f~-ol, atorvastatin calcium (1 H-Pyrrole-1-heptanoic acid, 2-(4-fluorophenyl)-f3,delta-dihydroxy-5-(1-methylethyl)-3-phenyl-4-((phenylamino)carbonyl)-, calcium salt (R-(R*,R*))-), CP-83101 (6,8-nonadienoic acid, 3,5-dihydroxy-9,9-diphenyl-, methyl ester, (R*,S*-(E))-(+/-)-), pravastatin (1-naphthaleneheptanoic acid, 1,2,6,7,8,8a-hexahydro-f~,delta,6-trihydroxy-2-methyl-8-(2-methyl-1-oxobutoxy)-, monosodium salt, (1S-(1 alpha(f3S*,deltaS*),2 alpha,6 alpha,8f3(R*),8a alpha))-), U-20685, pitavastatin (6-heptenoic acid, 7-(2-cyclopropyl-4-(4-fluorophenyl)-3-quinolinyl)-3,5-dihydroxy-, calcium salt (2:1), (S-(R*,S*-(E)))-), N-((1-methylpropyl)carbonyl)-8-(2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl)-perhydro-isoquinoline, dihydromevinolin (butanoic acid, 2-methyl-, 1, 2,3, 4, 4a, 7, 8, 8a-octa hyd ro-3, 7-d i m ethyl-8-(2-(tetra hyd ro-4-hyd roxy-6-oxo-2 H-pyran-2-yl)ethyl)-1-naphthalenyl ester(1 alpha(R*), 3 alpha, 4a alpha,7(3,8f3(2S*,4S*),8a(3))-), HBS-107, dihydromevinolin (butanoic acid, 2-methyl-, 1,2,3,4,4a,7,8,8a-octahydro-3,7-dimethyl-8-(2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl)-1-naphthalenyl ester(1 alpha(R*), 3 alpha,4a alpha,7f3,8f3(2S*,4S*),8af3))-), L-669262 (butanoic acid, 2,2-dimethyl-, 1,2,6,7,8,8a-hexahydro-3,7-dimethyl-6-oxo-8-(2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl)-1-naphthalenyl(1 S-(1 Alpha,7f3,8f3(2S*,4S*),8af3))-), simvastatin (butanoic acid, 2,2-dimethyl-, 1,2,3,7,8,8a-hexahydro-3,7-dimethyl-8-(2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl)-1-naphthalenyl ester, , (1 S-(1 alpha, 3alpha,7f3,8f3(2S*,4S*),8af3))-), rosuvastatin calcium (6-heptenoic acid, 7-(4-(4-fluorophenyl)-6-(1-methylethyl)-2-(methyl(methylsulfonyl)amino)-pyrimdinyl)-3,5-dihydroxy- calcium salt (2:1 ) (S-(R*, S*-(E)))), meglutol (2-hydroxy-2-methyl-1,3-propandicarboxylic acid), lovastatin (butanoic acid, 2-methyl-, 1,2,3,7,8,8a-hexahydro-3,7-dimethyl-8-(2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl)-1-naphthalenyl ester, (1S-(1 alpha.(R*),3 alpha,7f3,8(~(2S*,4S*),8a(3))-), or an analogue or derivative thereof).
14) Hydroorotate Dehydrogenase Inhibitors In another embodiment, the pharmacologically active compound is a hydroorotate dehydrogenase inhibitor (e.g., leflunomide (4-isoxazolecarboxamide, 5-methyl-N-(4-(trifluoromethyl)phenyl)-), laflunimus (2-propenamide, 2-cyano-3-cyclopropyl-3-hydroxy-N-(3-methyl-4(trifluoromethyl)phenyl)-, (Z)-), or atovaquone (1,4-naphthalenedione, 2-[4-(4-chlorophenyl)cyclohexyl]-3-hydroxy-, trans-, or an analogue or derivative thereof).
15) IKK2lnhibitors In another embodiment, the pharmacologically active compound is an IKK2 inhibitor (e.g., MLN-120B, SPC-839, or an analogue or derivative thereof).
16) IL-1, ICE and IRAK Antactonists In another embodiment, the pharmacologically active compound is an IL-1, ICE or an IRAK antagonist (e.g., E-5090 (2-propenoic acid, 3-(5-ethyl-4-hydroxy-3-methoxy-1-naphthalenyl)-2-methyl-, (Z)-), CH-164, CH-172, CH-490, AMG-719, iguratimod (N-(3-(formylamino)-4-oxo-6-phenoxy-4H-chromen-7-yl) methanesulfonamide), AV94-88, pralnacasan (6H-pyridazino(1,2-a)(1,2)diazepine-1-carboxamide, N-((2R,3S)-2-ethoxytetrahydro-5-oxo-3-furanyl)octahydro-9-((1-isoquinolinylcarbonyl)amino)-6,10-dioxo-, (1S,9S)-), (2S-cis)-5-(benzyloxycarbonylamino-1,2,4,5,6,7-hexahydro-4-(oxoazepino(3,2,1-hi)indole-2-carbonyl)-amino)-4-oxobutanoic acid, AVE-9488, esonarimod (benzenebutanoic acid, alpha-((acetylthio)methyl)-4-methyl-gamma-oxo-), pralnacasan (6H-pyridazino(1,2-a)(1,2)diazepine-1-carboxamide, N-((2 R,3S)-2-ethoxytetrahydro-5-oxo-3-furanyl)octahydro-9-(( 1-isoquinolinylcarbonyl)amino)-6,10-dioxo-, (1S,9S)-), tranexamic acid (cyclohexanecarboxylic acid, 4-(aminomethyl)-, trans-), Win-72052, romazarit (Ro-31-3948) (propanoic acid, 2-((2-(4-chlorophenyl)-4-methyl-5-oxazolyl)methoxy)-2-methyl-), PD-163594, SDZ-224-015 (L-alaninamide N-((phenylmethoxy)carbonyl)-L-valyl-N-((1 S)-3-((2,6-dichlorobenzoyl)oxy)-1-(2-ethoxy-2-oxoethyl)-2-oxopropyl)-), L-709049 (L-alaninamide, N-acetyl-L-tyrosyl-L-valyl-N-(2-carboxy-1-formylethyl)-, (S)-), TA-383 (1 H-imidazole, 2-(4-chlorophenyl)-4,5-dihydro-4,5-diphenyl-, monohydrochloride, cis-), EI-1507-1 (6a,12a-epoxybenz(a)anthracen-1,12(2H,7H)-dione, 3,4-dihydro-3,7-dihydroxy-8-methoxy-3-methyl-), ethyl 4-(3,4-dimethoxyphenyl)-6,7-dimethoxy-2-(1,2,4-triazol-1-yl methyl)quinoline-3-carboxylate, EI-1941-1, TJ-114, anakinra (interleukin 1 receptor antagonist (human isoform x reduced), N2-L-methionyl-), IX-207-887 (acetic acid, (10-methoxy-4H-benzo[4,5]cyclohepta[1,2-b]thien-4-ylidene)-), K-832, or an analogue or derivative thereof).
17) IL-4 Aaonists In another embodiment, the pharmacologically active compound is an IL-4 agonist (e.g., glatiramir acetate (L-glutamic acid, polymer with L-alanine, L-lysine and L-tyrosine, acetate (salt)), or an analogue or derivative thereof).
18) Immunomodulatory Ae~ents In another embodiment, the pharmacologically active compound is an immunomodulatory agent (e.g., biolimus, ABT-578, methylsulfamic acid 3-(2-methoxyphenoxy)-2-(((methylamino)sulfonyl)oxy)propyl ester, sirolimus (also referred to as rapamycin or RAPAMUNE (American Home Products, Inc., Madison, NJ)), CCI-779 (rapamycin 42-(3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate)), LF-15-0195, NPC15669 (L-leucine, N-(((2,7-dimethyl-9H-fluoren-9-yl)methoxy)carbonyl)-), NPC-15670 (L-leucine, N-(((4,5-dimethyl-9H-fluoren-9-yl)methoxy)carbonyl)-), NPC-16570 (4-(2-(fluoren-9-yl)ethyloxy-carbonyl)aminobenzoic acid), sufosfamide (ethanol, 2-((3-(2-chloroethyl)tetrahydro-2H-1,3,2-oxazaphosphorin-2-yl)amino)-, methanesulfonate (ester), P-oxide), tresperimus (2-(N-(4-(3-aminopropylamino)butyl)carbamoyloxy)-N-(6-guanidinohexyl)acetamide), 4-(2-(fluoren-9-yl)ethoxycarbonylamino)-benzo-hydroxamic acid, iaquinimod, PBI-1411, azathioprine (6-((1-Methyl-4-vitro-1 H-imidazol-5-yl)thio)-1 H-purine), PB10032, beclometasone, MDL-28842 (9H-purin-6-amine, 9-(5-deoxy-5-fluoro-(3-D-threo-pent-4-enofuranosyl)-, (~)-), FK-788, AVE-1726, ZK-90695, ~K-90695, Ro-54864, didemnin-B, Illinois (didemnin A, N-(1-(2-hydroxy-1-oxopropyl)-L-prolyl)-, (S)-), SDI-62-826 (ethanaminium, 2-((hydroxy((1-((octadecyloxy)carbonyl)-3-piperidinyl)methoxy)phosphinyl)oxy)-N,N,N-trimethyl-, inner salt), argyrin B ((4S,7S,13R,22R)-13-Ethyl-4-(1 H-indol-3-ylmethyl)-7-(4-methoxy-1 H-indol-3-ylmethyl)18,22-dimethyl-16-methyl-eve-24-this-3,6,9,12,15,18,21,26-octaazabicyclo(21.2.1)-hexacosa-1(25),23(26)-diene-2,5,8,11,14,17,20-heptaone), everolimus (rapamycin, 42-O-(2-hydroxyethyl)-), SAR-943, L-687795, 6-((4-chlorophenyl)sulfinyl)-2,3-dihydro-2-(4-methoxy-phenyl)-5-methyl-3-oxo-4-pyridazinecarbonitrile, 91Y78 (1H-imidazo(4,5-c)pyridin-4-amine, 1-f3-D-ribofuranosyl-), auranofin (gold, (1-thio-f3-D-glucopyranose 2,3,4,6-tetraacetato-S)(triethylphosphine)-), 27-0-demethylrapamycin, tipredane (androsta-1,4-dien-3-one, 17-(ethylthio)-9-fluoro-11-hydroxy-17-(methylthio)-, (1113,17 alpha)-), AI-402, LY-178002 (4-thiazolidinone, 5-((3,5-bis(1,1-dimethylethyl)-4-hydroxyphenyl)methylene)-), SM-8849 (2-thiazolamine, 4-(1-(2-fluoro(1,1'-biphenyl)-4-yl)ethyl)-N-methyl-), piceatannol, resveratrol, triamcinolone acetonide (pregna-1,4-diene-3,20-dione, 9-fluoro-11,21-dihydroxy-16,17-((1-methylethylidene)bis(oxy))-, (11f~,16 alpha)-), ciclosporin (cyclosporin A), tacrolimus (15,19-epoxy-3H-pyrido(2,1-c)(1,4)oxaazacyclotricosine-1,7,20,21 (4H,23H)-tetrone, 5,6,8,11,12,13,14,15,16,17,18,19,24,25,26,26a-hexadecahydro-5,19-dihydroxy-3-(2-(4-hydroxy-3-methoxycyclohexyl)-1-methylethenyl)-14,16-dimethoxy-4,10,12,18-tetramethyl-8-(2-propenyl)-, (3S-(3R*(E( 1 S*,3S*,4S*)),4S*,5R*,8S*,9E,12R*,14R*,15S*,16R*,18S*,19S*,26aR*)) -), gusperimus (heptanamide, 7-((aminoiminomethyl)amino)-N-(2-((4-((3-aminopropyl)amino)butyl)amino)-1-hydroxy-2-oxoethyl)-, (+/-)-), tixocortol pivalate (pregn-4-ene-3,20-dione, 21-((2,2-dimethyl-1-oxopropyl)thio)-11,17-dihydroxy-, (11 (3)-), alefacept (1-92 LFA-3 (antigen) (human) fusion protein with immunoglobulin G1 (human hinge-CH2=CH3 gammal-chain), dimer), halobetasol propionate (pregna-1,4-diene-3,20-dione, 21-chloro-6,9-difluoro-11 hydroxy-16-methyl-17-(1-oxopropoxy)-, (6Alpha,11 f3,16f3)-), iloprost trometamol (pentanoic acid, 5-(hexahydro-5-hydroxy-4-(3-hydroxy-4-methyl-1-octen-6 ynyl)-2(1 H)-pentalenylidene)-), beraprost (1 H-cyclopenta(b)benzofuran-5-butanoic acid, 2,3,3a,8b-tetrahydro-2-hydroxy-1-(3-hydroxy-4-methyl-1-octen-6-ynyl)-), rimexolone (androsta-1,4-dien-3-one,11-hydroxy-16,17-dimethyl-17-(1-oxopropy1)-, (11f~,16Alpha,17f3)-), dexamethasone (pregna-1,4-diene-3,20-dione,9-fluoro-11,17,21-trihydroxy-16-methyl-, (11f~,16alpha)-), sulindac (cis-fluoro-2-methyl-1-((p-methylsulfinyl)benzylidene)indene-3-acetic acid), proglumetacin (1 H-Indole-3-acetic acid, 1-(4-chlorobenzoyl)-5-methoxy-2-methyl-, 2-(4-(3-((4-(benzoylamino)-5-(dipropylamino)-1,5-dioxopentyl)oxy)propyl)-1-piperazinyl)ethylester, (+/-)-), alclometasone dipropionate (pregna-1,4-diene-3,20-dione, 7-chloro-11-hydroxy-16-methyl-17,21-bis(1-oxopropoxy)-, (7alpha,l1 f3,16alpha)-), pimecrolimus (15,19-epoxy-3H-pyrido(2,1-c)(1,4)oxaazacyclotricosine-1,7,20,21 (4H,23H)-tetrone, 3-(2-(4-chloro-3-methoxycyclohexyl)-1-methyletheny)-8-ethyl-5,6,8,11,12,13,14,15,16,17,18,19,24,25,26,26a-hexadecahydro-5,19-dihydroxy-14,16-d imethoxy-4,10,12,18-tetramethyl-, (3S-(3R*(E(1 S*,3S*,4R*)),4S*,5R*,8S*,9E,12R*,14R*,15S*,16R*,18S*,19S*,26aR*)) -), hydrocortisone-17-butyrate (pregn-4-ene-3,20-dione, 11,21-dihydroxy-17-(1-oxobutoxy)-, (11f3)-), mitoxantrone (9,10-anthracenedione, 1,4-dihydroxy-5,8-bis((2-((2-hydroxyethyl)amino)ethyl)amino)-), mizoribine (1 H-imidazole-4-carboxamide, 5-hydroxy-1-f3-D-ribofuranosyl-), prednicarbate (pregna-1,4-diene-3,20-dione, 17-((ethoxycarbonyl)oxy)-11-hydroxy-21-(1-oxopropoxy)-, (11 f3)-), iobenzarit (benzoic acid, 2-((2-carboxypheny!)amino)-4-chloro-), glucametacin (D-glucose, 2-(((1-(4-chloroberrzoyl)-5-methoxy-2-methyl-1 H-indol-3-yl)acetyl)amino)-2-deoxy-), fluocortolone monohydrate ((6 alpha)-fluoro-16alpha-methylpregna-1,4-dien-11(3,21-diol-3,20-dione), fluocortin butyl (pregna-1,4-dien-21-oic acid, 6-fluoro-11-hydroxy-16-methyl-3,20-dioxo-, butyl ester, (6alpha,11f5,16alpha)-), difluprednate (pregna-1,4-diene-3,20-dione, 21-(acetyloxy)-6,9-difluoro-11-hydroxy-17-(1-oxobutoxy)-, (6 alpha,11f~)-), diflorasone diacetate (pregna-1,4-diene-3,20-dione, 17,21-bis(acetyloxy)-6,9-difluoro-11-hydroxy-16-methyl-, (6Alpha,11f5,16f3)-), dexamethasone valerate (pregna-1,4-diene-3,20-dione, 9-fluoro-11,21-dihydroxy-16-methyl-17-((1-oxopentyl)oxy)-, (11(3,16AIpha)-), methylprednisolone, deprodone propionate (pregna-1,4-diene-3,20-dione, 11-hydroxy-17-(1-oxopropoxy)-, (l1.beta.)-), bucillamine (L-cysteine, N-(2-mercapto-2-methyl-1-oxopropyl)-), amcinonide (benzeneacetic acid, 2-amino-3-benzoyl-, monosodium salt, monohydrate), acemetacin (1 H-indole-3-acetic acid, 1-(4-chlorobenzoyl)-5-methoxy-2-methyl-, carboxymethyl ester), or an analogue or derivative thereof).
Further, analogues of rapamycin include tacrolimus and derivatives thereof (e.g., EP0184162B1 and U.S. Patent No. 6,258,823) everolimus and derivatives thereof (e.g., U.S. Patent No. 5,665,772). Further representative examples of sirolimus analogues and derivatives can be found in PCT Publication Nos. WO 97/10502, WO 96/41807, WO 96/35423, WO
96103430, WO 96/00282, WO 95116691, WO 95/15328, WO 95/07468, WO
95!04738, WO 95/04060, WO 94/25022, WO 94/21644, WO 94/18207, WO
94/10843, WO 94/09010, WO 94/04540, WO 94/02485, WO 94/02137, WO
94/02136, WO 93/25533, WO 93/18043, WO 93/13663, WO 93/11130, WO
93/10122, WO 93/04680, WO 92/14737, and WO 92/05179. Representative U.S. patents include U.S. Patent Nos. 6,342,507; 5,985,890; 5,604,234;
5,597,715; 5,583,139; 5,563,172; 5,561,228; 5,561,137; 5,541,193; 5,541,189;
5,534,632; 5,527,907; 5,484,799; 5,457,194; 5,457,182; 5,362,735; 5,324,644;
5,318,895; 5,310,903; 5,310,901; 5,258,389; 5,252,732; 5,247,076; 5,225,403;
5,221,625; 5,210,030; 5,208,241; 5,200,411; 5,198,421; 5,147,877; 5,140,018;
5,116,756; 5,109,112; 5,093,338; and 5,091,389.
The structures of sirolimus, everolimus, and tacrolimus are provided below:

Name Code Name Company Structure Everolimus SAR-943 Novartis See below Sirolimus AY-22989 Wyeth See below Rapamycin Tacrolimus FK506 Fujusawa See below as ''a Everolimus .., o Tacrolimus ~"0 0 o ~ tr c~ 0 0 0~
wa ,"' Sirolimus Further sirolimus analogues and derivatives include tacrolimus and derivatives thereof (e.g., EP0184162B1 and U.S. Patent No. 6,258,823) everolimus and derivatives thereof (e.g., US Patent No. 5,665,772). Further representative examples of sirolimus analogues and derivatives include ABT-578 and others may be found in PCT Publication Nos. WO 97/10502, WO
96/41807, WO 96/35423, WO 96/03430, WO 9600282, WO 95/16691, WO
9515328, WO 95/07468, WO 95/04738, WO 95/04060, WO 94/25022, WO
94/21644, WO 94/18207, WO 94/10843, WO 94/09010, WO 94/04540, WO
94/02485, WO 94/02137, WO 94/02136, WO 93/25533, WO 93/18043, WO
93/13663, WO 93/11130, WO 93/10122, WO 93/04680, WO 92/14737, and WO
92/05179. Representative U.S. patents include U.S. Patent Nos. 6,342,507;
5,985,890; 5,604,234; 5,597,715; 5,583,139; 5,563,172; 5,561,228; 5,561,137;
5,541,193; 5,541,189; 5,534,632; 5,527,907; 5,484,799; 5,457,194; 5,457,182;
5,362,735; 5,324,644; 5,318,895; 5,310,903; 5,310,901; 5,258,389; 5,252,732;
5,247,076; 5,225,403; 5,221,625; 5,210,030; 5,208,241, 5,200,411; 5,198,421;
5,147,877; 5,140,018; 5,116,756; 5,109,112; 5,093,338; and 5,091,389.
In one aspect, the fibrosis-inhibiting agent may be, e.g., rapamycin (sirolimus), everolimus, biolimus, tresperimus, auranofin, 27-0-demethylrapamycin, tacrolimus, gusperimus, pimecrolimus, or ABT-578.
19) Inosine monophosphate dehydrogenase inhibitors In another embodiment, the pharmacologically active compound is an inosine monophosphate dehydrogenase (IMPDH) inhibitor (e.g., mycophenolic acid, mycophenolate mofetil (4-hexenoic acid, 6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-, 2-(4-morpholinyl)ethyl ester, (E)-), ribavirin (1 H-1,2,4-triazole-3-carboxamide, 1-(3-D-ribofuranosyl-), tiazofurin (4-thiazolecarboxamide, 2-f3-D-ribofuranosyl-), viramidine, aminothiadiazole, thiophenfurin, tiazofurin) or an analogue or derivative thereof. Additional representative examples are included in U.S.
Patent Nos. 5,536,747, 5,807,876, 5,932,600, 6,054,472, 6,128,582, 6,344,465, 6,395,763, 6,399,773, 6,420,403, 6,479,628, 6,498,178, 6,514,979, 6,518,291, 6,541,496, 6,596,747, 6,617,323, 6,624,184, Patent Application Publication Nos. 2002/0040022A1, 2002/0052513A1, 2002/0055483A1, 2002/0068346A1, 2002/0111378A1, 2002/0111495A1, 2002/0123520A1, 2002/0143176A1, 2002/0147160A1, 2002/0161038A1, 2002/0173491 A1, 2002/0183315A1, 2002/0193612A1, 2003/0027845A1, 2003/0068302A1, 2003/0105073A1, 2003/0130254A1, 2003/0143197A1, 2003/0144300A1, 2003/0166201 A1, 2003/0181497A1, 2003/0186974A1, 2003/0186989A1, 2003/0195202A1, and PCT Publication Nos. WO 0024725A1, WO 00/25780A1, WO 00/26197A1, WO
00/51615A1, WO 00/56331A1, WO 00/73288A1, WO 01/00622A1, WO
01/66706A1, WO 01/79246A2, WO 01/81340A2, WO 01/85952A2, WO
02/16382A1, WO 02/18369A2, WO 2051814A1, WO 2057287A2, WO2057425A2, WO 2060875A1, WO 2060896A1, WO 2060898A1, WO
2068058A2, WO 3020298A1, WO 3037349A1, WO 3039548A1, WO
3045901 A2, WO 3047512A2, WO 3053958A1, WO 3055447A2, WO
3059269A2, WO 3063573A2, WO 3087071 A1, WO 90/01545A1, WO
97/40028A1, WO 97/41211A1, WO 98/40381 A1, and WO 99/55663A1).
20) Leukotriene Inhibitors In another embodiment, the pharmacologically active compound is a leukotreine inhibitor (e.g., ONO-4057(benzenepropanoic acid, 2-(4-carboxybutoxy)-6-((6-(4-methoxyphenyl)-5-hexenyl)oxy)-, (E)-), ONO-LB-448, pirodomast 1,8-naphthyridin-2(1 H)-one, 4-hydroxy-1-phenyl-3-(1-pyrrolidinyl)-, Sch-40120 (benzo(b)(1,8)naphthyridin-5(7H)-one, 10-(3-chlorophenyl)-6,8,9,10-tetrahydro-), L-656224 (4-benzofuranol, 7-chloro-2-((4-methoxyphenyl)methyl)-3-methyl-5-propyl-), MAFP (methyl arachidonyl fluorophosphonate), ontazolast (2-benzoxazolamine, N-(2-cyclohexyl-1-(2-pyridinyl)ethyl)-5-methyl-, (S)-), amelubant (carbamic acid, ((4-((3-((4-(1-(4-hydroxyphenyl)-1-methylethyl)phenoxy)methyl)phenyl)methoxy)phenyl)iminomethyl)- ethyl ester), SB-201993 (benzoic acid, 3-((((6-((1 E)-2-carboxyethenyl)-5-((8-(4-methoxyphenyl)octyl)oxy)-2-pyridinyl)methyl)thio)methyl)-), LY-203647 (ethanone, 1-(2-hydroxy-3-propyl-4-(4-(2-(4-(1H-tetrazol-5-yl)butyl)-2H-tetrazol-5-yl)butoxy)phenyl)-), LY-210073, LY-223982 (benzenepropanoic acid, 5-(3-carboxybenzoyl)-2-((6-(4-methoxyphenyl)-5-hexenyl)oxy)-, (E)-), LY-293111 (benzoic acid, 2-(3-(3-((5-ethyl-4'-fluoro-2-hydroxy(1,1'-biphenyl)-4-yl)oxy)propoxy)-2-propylphenoxy)-), SM-9064 (pyrrolidine, 1-(4,11-dihydroxy-13-(4-methoxyphenyl)-1-oxo-5,7,9-tridecatrienyl)-, (E,E,E)-), T-0757 (2,6-octadienamide, N-(4-hydroxy-3,5-dimethylphenyl)-3,7-dimethyl-, (2E)-), or an analogue or derivative thereof).
21 ) MCP-1 Antagonists In another embodiment, the pharmacologically active compound is a MCP-1 antagonist (e.g., nitronaproxen (2-napthaleneacetic acid, 6-methoxy-alpha-methyl 4-(nitrooxy)butyl ester (alpha S)-), bindarit (2-(1-benzylindazol-3-ylmethoxy)-2-methylpropanoic acid), 1-alpha-25 dihydroxy vitamin D3, or an analogue or derivative thereof).
22) MMP Inhibitors In another embodiment, the pharmacologically active compound is a matrix metalloproteinase (MMP) inhibitor (e.g., D-9120, doxycycline (2-naphthacenecarboxamide, 4-(dimethylamino)-1,4,4a,5,5a,6,11,12a-octahydro-3,5,10,12,12a-pentahydroxy-6-methyl-1,11-dioxo- (4S-(4 alpha, 4a alpha, 5 Ipha, 5a alpha, 6 alpha, 12a alpha))-), BB-2827, BB-1101 (2S-allyl-N1-hydroxy-3R-isobutyl-N4-(1 S-methylcarbamoyl-2-phenylethyl)-succinamide), BB-2983, solimastat (N'-(2,2-dimethyl-1 (S)-(N-(2-pyridyl)carbamoyl)propyl)-N4-hydroxy-2(R)-isobutyl-3(S)-methoxysuccinamide), batimastat (butanediamide, N4-hydroxy-N 1-(2-(methylamino)-2-oxo-1-(phenylmethyl)ethyl)-2-(2-methylpropyl)-3-((2-thienylthio)methyl)-, (2R-(1(S*),2R*,3S*))-), CH-138, CH-5902, D-1927, D-5410, EF-13 (gamma-linolenic acid lithium salt),CMT-3 (2-naphthacenecarboxamide, 1,4,4a,5,5a,6,11,12a-octahydro-3,10,12,12a-tetrahydroxy-1,11-dioxo-, (4aS,5aR,12aS)-), marimastat (N-(2,2-dimethyl-1(S)-(N-methylcarbamoyl)propyl)-N,3(S)-dihydroxy-2(R)-isobutylsuccinamide), TIMP'S,ONO-4817, rebimastat (L-Valinamide, N-((2S)-2-mercapto-1-oxo-4-(3,4,4-trimethyl-2,5-dioxo-1-imidazolidinyl)butyl)-L-leucyl-N,3-dimethyl-), PS-508, CH-715, nimesulide (methanesulfonamide, N-(4-nitro-2-phenoxyphenyl)-), hexahydro-2-(2(R)-(1 (RS)-(hydroxycarbamoyl)-4-phenylbutyl)nonanoyl)-N-(2,2,6,6-etramethyl-4-piperidinyl)-3(S)-pyridazine carboxamide, Rs-113-080, Ro-1130830, cipemastat (1-piperidinebutanamide, f3-(cyclopentylmethyl)-N-hydroxy-gamma-oxo-alpha-((3,4,4-trimethyl-2,5-dioxo-1-imidazolidinyl)methyl)-,(alpha R,f3R)-), 5-(4'-biphenyl)-5-(N-(4-nitrophenyl)piperazinyl)barbituric acid, 6-methoxy-1,2,3,4-tetrahydro-norharman-1-carboxylic acid, Ro-31-4724 (L-alanine, N-(2-(2-(hydroxyamino)-2-oxoethyl)-4-methyl-1-oxopentyl)-L-leucyl-, ethyl ester), prinomastat (3-thiomorpholinecarboxamide, N-hydroxy-2,2-dimethyl-4-((4-(4-pyridinyloxy) phenyl)sulfonyl)-, (3R)-), AG-3433 (1 H-pyrrole-3-propanic acid, 1-(4'-cyano(1,1'-biphenyl)-4-yl)-b-((((3S)-tetrahydro-4,4-dimethyl-2-oxo-3-furanyl)amino)carbonyl)-, phenylmethyl ester, (bS)-), PNU-142769 (2H-Isoindole-2-butanamide, 1,3-dihydro-N-hydroxy-alpha-((3S)-3-(2-methylpropyl)-2-oxo-1-(2-phenylethyl)-3-pyrrolidinyl)-1,3-dioxo-, (alpha R)-), (S)-1-(2-((((4,5-dihydro-5-thioxo-1,3,4-thiadiazol-2-yl)amino)-carbonyl)amino)-1-oxo-3-(pentafluorophenyl)propyl)-4-(2-pyridinyl)piperazine, SU-5402 (1 H-pyrrole-3-propanoic acid, 2-((1,2-dihydro-2-oxo-3H-indol-3-ylidene)methyl)-4-methyl-), SC-77964, PNU-171829, CGS-27023A, N-hydroxy-2(R)-((4-methoxybenzene-sulfonyl)(4-picolyl)amino)-2-(2-tetrahydrofuranyl)-acetamide, L-758354 ((1,1'-biphenyl)-4-hexanoic acid, alpha-butyl-gamma-(((2,2-dimethyl-1-((methylamino)carbonyl)propyl)amino)carbonyl)-4'-fluoro-, (alpha S-(alpha R*, gammaS*(R*)))-, GI-155704A, CPA-926, TMI-005, XL-784, or an analogue or derivative thereof). Additional representative examples are included in U.S.
Patent Nos. 5,665,777; 5,985,911; 6,288,261; 5,952,320; 6,441,189; 6,235,786;
6,294,573; 6,294,539; 6,563,002; 6,071,903; 6,358,980; 5,852,213; 6,124,502;
6,160,132; 6,197,791; 6,172,057; 6,288,086; 6,342,508; 6,228,869; 5,977,408;
5,929,097; 6,498,167; 6,534,491; 6,548,524; 5,962,481; 6,197,795; 6,162,814;
6,441,023; 6,444,704; 6,462,073; 6,162,821; 6,444,639; 6,262,080; 6,486,193;
6,329,550; 6,544,980; 6,352,976; 5,968,795; 5,789,434; 5,932,763; 6,500,847;
5,925,637; 6,225,314; 5,804,581; 5,863,915; 5,859,047; 5,861,428; 5,886,043;
6,288,063; 5,939,583; 6,166,082; 5,874,473; 5,886,022; 5,932,577; 5,854,277;
5,886,024; 6,495,565; 6,642,255; 6,495,548; 6,479,502; 5,696,082; 5,700,838;
6,444,639; 6,262,080; 6,486,193; 6,329,550; 6,544,980; 6,352,976; 5,968,795;
5,789,434; 5,932,763; 6,500,847; 5,925,637; 6,225,314; 5,804,581; 5,863,915;
5,859,047; 5,861,428; 5,886,043; 6,288,063; 5,939,583; 6,166,082; 5,874,473;
5,886,022; 5,932,577; 5,854,277; 5,886,024; 6,495,565; 6,642,255; 6,495,548;
6,479,502; 5,696,082; 5,700,838; 5,861,436; 5,691,382; 5,763,621; 5,866,717;
5,902,791; 5,962,529; 6,017,889; 6,022,873; 6,022,898; 6,103,739; 6,127,427;
6,258,851; 6,310,084; 6,358,987; 5,872,152; 5,917,090; 6,124,329; 6,329,373;

6,344,457; 5,698,706; 5,872,146; 5,853,623; 6,624,144; 6,462,042; 5,981,491;
5,955,435; 6,090,840; 6,114,372; 6,566,384; 5,994,293; 6,063,786; 6,469,020;
6,118,001; 6,187,924; 6,310,088; 5,994,312; 6,180,611; 6,110,896; 6,380,253;
5,455,262; 5,470,834; 6,147,114; 6,333,324; 6,489,324; 6,362,183; 6,372,758;
6,448,250; 6,492,367; 6,380,258; 6,583,299; 5,239,078; 5,892,112; 5,773,438;
5,696,147; 6,066,662; 6,600,057; 5,990,158; 5,731,293; 6,277,876; 6,521,606;
6,168,807; 6,506,414; 6,620,813; 5,684,152; 6,451,791; 6,476,027; 6,013,649;
6,503,892; 6,420,427; 6,300,514; 6,403,644; 6,177,466; 6,569,899; 5,594,006;
6,417,229; 5,861,510; 6,156,798; 6,387,931; 6,350,907; 6,090,852; 6,458,822;
6,509,337; 6,147,061; 6,114,568; 6,118,016; 5,804,593; 5,847,153; 5,859,061;
6,194,451; 6,482,827; 6,638,952; 5,677,282; 6,365,630; 6,130,254; 6,455,569;
6,057,369; 6,576,628; 6,110,924; 6,472,396; 6,548,667; 5,618,844; 6,495,578;
6,627,411; 5,514,716; 5,256,657; 5,773,428; 6,037,472; 6,579,890; 5,932,595;
6,013,792; 6,420,415; 5,532,265; 5,691,381; 5,639,746; 5,672,598; 5,830,915;
6,630,516; 5,324,634; 6,277,061; 6,140,099; 6,455,570; 5,595,885; 6,093,398;
6,379,667; 5,641,636; 5,698,404; 6,448,058; 6,008,220; 6,265,432; 6,169,103;
6,133,304; 6,541,521; 6,624,196; 6,307,089; 6,239,288; 5,756,545; 6,020,366;
6,117,869; 6,294,674; 6,037,361; 6,399,612; 6,495,568; 6,624,177; 5,948,780;
6,620,835; 6,284,513; 5,977,141; 6,153,612; 6,297,247; 6,559,142; 6,555,535;
6,350,885; 5,627,206; 5,665,764; 5,958,972; 6,420,408; 6,492,422; 6,340,709;
6,022,948; 6,274,703; 6,294,694; 6,531,499; 6,465,508; 6,437,177; 6,376,665;
5,268,384; 5,183,900; 5,189,178; 6,511,993; 6,617,354; 6,331,563; 5,962,466;
5,861,427; 5,830,869; and 6,087,359.
23) NF kappa B Inhibitors In another embodiment, the pharmacologically active compound is a NF kappa B (NFKB) inhibitor (e.g., AVE-0545, Oxi-104 (benzamide, 4-amino-3-chloro-N-(2-(diethylamino)ethyl)-), dexlipotam, R-flurbiprofen ((1,1'-biphenyl)-4-acetic acid, 2-fluoro-alpha-methyl), SP100030 (2-chloro-N-(3,5-di(trifluoromethyl)phenyl)-4-(trifluoromethyl)pyrimidine-5-carboxamide), AVE-0545, Viatris, AVE-0547, Bay 11-7082, Bay 11-7085, 15 deoxy-prostaylandin J2, bortezomib (boronic acid, ((1 R)-3-methyl-1-(((2S)-1-oxo-3-phenyl-2-((pyrazinylcarbonyl)amino)propyl)amino)butyl)-, benzamide an d nicotinamide derivatives that inhibit NF-kappaB, such as those described in U.S. Patent Nos.
5,561,161 and 5,340,565 (OxiGene), PG490-88Na, or an analogue or derivative thereof).
24) NO Aaonists In another embodiment, the pharmacologically active compound is a NO antagonist (e.g., NCX-4016 (benzoic acid, 2-(acetyloxy)-, 3-((nitrooxy)methyl)phenyl ester, NCX-2216, L-arginine or an analogue or derivative thereof).
25) P38 MAP Kinase Inhibitors In another embodiment, the pharmacologically active compound is a p38 MAP kinase inhibitor (e.g., GW-2286, CGP-52411, BIRB-798, SB220025, RO-320-1195, RWJ-67657, RWJ-68354, SCIO-469, SCIO-323, AMG-548, CMC-146, SD-31145, CC-8866, Ro-320-1195, PD-98059 (4H-1-benzopyran-4-one, 2-(2-amino-3-methoxyphenyl)-), CGH-2466, doramapimod, SB-203580 (pyridine, 4-(5-(4-fluorophenyl)-2-(4-(methylsulfinyl)phenyl)-1 H-imidazol-4-yl)-), SB-220025 ((5-(2-amino-4-pyrimidinyl)-4-(4-fluorophenyl)-1-(4-piperidinyl)imidazole), SB-281832, PD169316, SB202190, GSK-681323, EO-1606, GSK-681323, or an analogue or derivative thereof). Additional representative examples are included in U.S. Patent Nos. 6,300,347;
6,316,464; 6,316,466; 6,376,527; 6,444,696; 6,479,507; 6,509,361; 6,579,874;
6,630,485, U.S. Patent Application Publication Nos. 2001 /0044538A1;
2002/0013354A1; 2002/0049220A1; 2002/0103245A1; 2002/0151491 A1;
2002/0156114A1; 2003/0018051 A1; 2003/0073832A1; 2003/0130257A1;
2003/0130273A1; 200310130319A1; 2003/0139388A1; 20030139462A1;
2003/0149031 A1; 2003/0166647A1; 2003/0181411 A1; and PCT Publication Nos. WO 00/63204A2; WO 01 /21591 A1; WO 01 /35959A1; W O 01 /74811 A2;
WO 02/18379A2; WO 2064594A2; WO 2083622A2; WO 2094842A2; WO
2096426A1; WO 2101015A2; WO 2103000A2; WO 3008413A1; WO
3016248A2; WO 3020715A1; WO 3024899A2; WO 3031431A1;
W03040103A1; WO 3053940A1; WO 3053941A2; WO 3063799A2; WO
3079986A2; WO 3080024A2; WO 3082287A1; WO 97/44467A1; WO
99/01449A1; and WO 99/58523A1.
26) Phosphodiesterase Inhibitors In another embodiment, the pharmacologically active compound is a phosphodiesterase inhibitor (e.g., CDP-840 (pyridine, 4-((2R)-2-(3-(cyclopentyloxy)-4-methoxyphenyl)-2-phenylethyl)-), CH-3697, CT-2820, D-22888 (imidazo(1,5-a)pyrido(3,2-e)pyrazin-6(5H)-one, 9-ethyl-2-methoxy-7-methyl-5-propyl-), D-4418 (8-methoxyquinoline-5-(N-(2,5-dichloropyridin-3-yl))carboxamide), 1-(3-cyclopentyloxy-4-methoxyphenyl)-2-(2,6-dichloro-4-pyridyl) ethanone oxime, D-4396, ONO-6126, CDC-998, CDC-801, V-11294A
(3-(3-(cyclopentyloxy)-4-methoxybenzyl)-6-(ethylamino)-8-isopropyl-3H-purine hydrochloride), S,S'-methylene-bis(2-(8-cyclopropyl-3-propyl-6-(4-pyridylmethylamino)-2-thio-3H-purine)) tetrahyrochloride, rolipram (2-pyrrolidinone, 4-(3-(cyclopentyloxy)-4-methoxyphenyl)-), CP-293121, CP-353164 (5-(3-cyclopentyloxy-4-methoxyphenyl)pyridine-2-carboxamide), oxagrelate (6-phthalazinecarboxylic acid, 3,4-dihydro-1-(hydroxymethyl)-5,7-dimethyl-4-oxo-, ethyl ester), PD-168787, ibudilast (1-propanone, 2-methyl-1-(2-(1-methylethyl)pyrazolo(1,5-a)pyridin-3-yl)-), oxagrelate (6-phthalazinecarboxylic acid, 3,4-dihydro-1-(hydroxymethyl)-5,7-dimethyl-4-oxo-, ethyl ester), griseolic acid (alpha-L-talo-oct-4-enofuranuronic acid, 1-(6-amino-9H-purin-9-yl)-3,6-anhydro-6-C-carboxy-1,5-dideoxy-), KW-4490, KS-506, T-440, roflumilast (benzamide, 3-(cyclopropylmethoxy)-N-(3,5-dichloro-4-pyridinyl)-4-(difluoromethoxy)-), rolipram, milrinone, triflusinal (benzoic acid, 2-(acetyloxy)-4-(trifluoromethyl)-), anagrelide hydrochloride (imidazo(2,1-b)quinazolin-2(3H)-one, 6,7-dichloro-1,5-dihydro-, monohydrochloride), cilostazol (2(1 H)-quinolinone, 6-(4-(1-cyclohexyl-1 H-tetrazol-5-yl)butoxy)-3,4-dihydro-), propentofylline (1 H-purine-2,6-dione, 3,7-dihydro-3-methyl-1-(5-oxohexyl)-7-propyl-), sildenafil citrate (piperazine, 1-((3-(4,7-dihydro-1-methyl-7-oxo-3-propyl-1 H-pyrazolo(4,3-d)pyrimidin-5-yl)-4-ethoxyphenyl)sulfonyl)-4-methyl, 2-hydroxy-1,2,3-propanetricarboxylate- (1:1)), tadalafil (pyrazino(1',2':1,6)pyrido(3,4-b)indole1,4-dione, 6-(1,3-benzodioxol-5-yl)-2,3,6,7,12,12a-hexahydro-2-methyl-, (6R-trans)), vardenafil (piperazine, 1-(3-(1,4-dihydro-5-methyl(-4-oxo-7-propylimidazo(5,1-f)(1,2,4)-triazin-2-yl)-4-ethoxyphenyl)sulfonyl)-4-ethyl-), milrinone ((3,4'-bipyridine)-5-carbonitrile, 1,6-dihydro-2-methyl-6-oxo-), enoximone (2H-imidazol-2-one, 1,3-dihydro-4-methyl-5-(4-(methylthio)benzoyl)-), theophylline (1H-purine-2,6-dione, 3,7-dihydro-1,3-dimethyl-), ibudilast (1-propanone, 2-methyl-1-(2-(1-methylethyl)pyrazolo(1,5-a)pyridin-3-yl)-), aminophylline (1H-purine-2,6-dione, 3,7-dihydro-1,3-dimethyl-, compound with 1,2-ethanediamine (2:1 )-), acebrophylline (7H-purine-7-acetic acid, 1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-, compd. with trans-4-(((2-amino-3,5-dibromophenyl)methyl)amino)cyclohexanol (1:1 )), plafibride (propanamide, 2-(4-chlorophenoxy)-2-methyl-N-(((4-morpholinylmethyl)amino)carbonyl)-), ioprinone hydrochloride (3-pyridinecarbonitrile, 1,2-dihydro-5-imidazo(1,2-a)pyridin-6-yl-6-methyl-2-oxo-, monohydrochloride-), fosfosal (benzoic acid, 2-(phosphonooxy)-), amrinone ((3,4'-bipyridin)-6(1 H)-one, 5-amino-, or an analogue or derivative thereof).
Other examples of phosphodiesterase inhibitors include denbufylline (1H-purine-2,6-dione, 1,3-dibutyl-3,7-dihydro-7-(2-oxopropyl)-), propentofylline (1 H-purine-2,6-dione, 3,7-dihydro-3-methyl-1-(5-oxohexyl)-7-propyl-) and pelrinone (5-pyrimidinecarbonitrile, 1,4-dihydro-2-methyl-4-oxo-6-[(3-pyridinylmethyl)amino]-).
Other examples of phosphodiesterase III inhibitors include enoximone (2H-imidazol-2-one, 1,3-dihyd;ro-4-methyl-5-[4-(methylthio)benzoyl]-), and saterinone (3-pyridinecarbonitrile, 1,2-dihydro-5-[4-[2-hydroxy-3-[4-(2-methoxyphenyl)-1-piperazinyl]propoxy]phenyl]-6-methyl-2-oxo-) Other examples of phosphodiesterase IV inhibitors include AWD-12-281, 3-auinolinecarboxylic acid, 1-ethyl-6-fluoro-1,4-dihydro-7-(4-methyl-1-piperazinyl)-4-oxo-), tadalafil (pyrazino(1',2':1,6)pyrido(3,4-b)indole1,4-dione, 6-(1,3-benzodioxol-5-yl)-2,3,6,7,12,12a-hexahydro-2-methyl-, (6R-trans)), and filaminast (ethanone, 1-[3-(cyclopentyloxy)-4-methoxyphenyl]-, O-(aminocarbonyl)oxime, (1 E)-) Another example of a phosphodiesterase V inhibitor is vardenafil (piperazine, (3-(1,4-dihydro-5-methyl(-4-oxo-7-propylimidazo(5,1-f)(1,2,4)-triazin-2-yl)-4-ethoxyphenyl)sulfonyl)-4-ethyl-).
27) TGF beta Inhibitors In another embodiment, the pharmacologically active compound is a TGF beta Inhibitor (e.g., mannose-6-phosphate, LF-984, tamoxifen (ethanamine, 2-(4-(1,2-diphenyl-1-butenyl)phenoxy)-N,N-dimethyl-, (Z)-), tranilast, or an analogue or derivative thereof).
28) Thromboxane A2 Antagonists In another embodiment, the pharmacologically active compound is a thromboxane A2 antagonist (e.g., CGS-22652 (3-pyridineheptanoic acid, y-(4-(((4-chlorophenyl)sulfonyl)amino)butyl)-, (±)-), ozagrel (2-propenoic acid, 3-(4-(1 H-imidazol-1-ylmethyl)phenyl)-, (E)-), argatroban (2-piperidinecarboxylic acid, 1-(5-((aminoiminomethyl)amino)-1-oxo-2-(((1,2,3,4-tetrahydro-3-methyl-8-quinolinyl)sulfonyl)amino)pentyl)-4-methyl-), ramatroban (9H-carbazole-9-propanoic acid, 3-(((4-fluorophenyl)sulfonyl)amino)-1,2,3,4-tetrahydro-, (R)-), torasemide (3-pyridinesulfonamide, N-(((1-methylethyl)amino)carbonyl)-4-((3-methylphenyl)amino)-), gamma linoleic acid ((Z,Z,Z)-6,9,12-octadecatrienoic acid), seratrodast (benzeneheptanoic acid, zeta-(2,4,5-trimethyl-3,6-dioxo-1,4-cyclohexadien-1-yl)-, (+/-)-, or an analogue or derivative thereof).
29) TNFa Antagonists and TACE Inhibitors In another embodiment, the pharmacologically active compound is a TNFa antagonist or TACE inhibitor (e.g., E-5531 (2-deoxy-6-0-(2-deoxy-3-0-(3(R)-(5(Z)-dodecenoyloxy)-decyl)-6-0-methyl-2-(3-oxotetradecanamido)-4-O-phosphono-f3-D-glucopyranosyl)-3-0-(3(R)-hydroxydecyl)-2-(3-oxotetradecanamido)-alpha-D-glucopyranose-1-O-phosphate), AZD-4717, glycophosphopeptical, UR-12715 (B=benzoic acid, 2-hydroxy-5-((4-(3-(4-(2-methyl-1 H-imidazol(4,5-c)pyridin-1-yl)methyl)-1-piperidinyl)-3-oxo-1-phenyl-1-propenyl)phenyl)azo) (Z)), PMS-601, AM-87, xyloadenosine (9H-purin-6-amine, 9-f3-D-xylofuranosyl-), RDP-58, RDP-59, BB2275, benzydamine, E-3330 (undecanoic acid, 2-((4,5-dimethoxy-2-methyl-3,6-dioxo-1,4-cyclohexadien-1-yl)methylene)-, (E)-), N-(D,L-2-(hydroxyaminocarbonyl)methyl-4-methylpentanoyl)-L-3-(2'-naphthyl)alanyl-L-alanine, 2-aminoethyl amide, CP-564959, MLN-608, SPC-839, ENMD-0997, Sch-23863 ((2-(10,11-dihydro-5-ethoxy-5H-dibenzo (a,d) cyclohepten-S-yl)-N, N-dimethyl-ethanamine), SH-636, PKF-241-466, PKF-242-484, TNF-484A, cilomilast (cis-4-cyano-4-(3-(cyclopentyloxy)-4-methoxyphenyl)cyclohexane-1-carboxylic acid), GW-3333, GW-4459, BMS-561392, AM-87, cloricromene (acetic acid, ((8-chloro-3-(2-(diethylamino)ethyl)-4-methyl-2-oxo-2H-1-benzopyran-7-yl)oxy)-, ethyl ester), thalidomide (1H-Isoindole-1,3(2H)-dione, 2-(2,6-dioxo-3-piperidinyl)-), vesnarinone (piperazine, 1-(3,4-dimethoxybenzoyl)-4-(1,2,3,4-tetrahydro-2-oxo-6-quinolinyl)-), infliximab, lentinan, etanercept (1-235-tumor necrosis factor receptor (human) fusion protein with 236-467-immunoglobulin G1 (human gamma1-chain Fc fragment)), diacerein (2-anthracenecarboxylic acid, 4,5-bis(acetyloxy)-9,10-dihydro-9,10-dioxo-, or an analogue or derivative thereof).
30) Tyrosine Kinase Inhibitors In another embodiment, the pharmacologically active compound is a tyrosine kinase inhibitor (e.g., SKI-606, ER-068224, SD-208, N-(6-benzothiazolyl)-4-(2-(1-piperazinyl)pyrid-5-yl)-2-pyrimidineamine, celastrol (24,25,26-trinoroleana-1(10),3,5,7-tetraen-29-oic acid, 3-hydroxy-9,13-dimethyl-2-oxo-, (9 beta.,13alpha,14f3,20 alpha)-), GP-127374 (geldanamycin, 17-demethoxy-17-(2-propenylamino)-), CP-564959, PD-171026, CGP-52411 (1 H-Isoindole-1,3(2H)-dione, 4,5-bis(phenylamino)-), CGP-53716 (benzamide, N-(4-methyl-3-((4-(3-pyridinyl)-2-pyrimidinyl)amino)phenyl)-), imatinib (4-((methyl-piperazinyl)methyl)-N-(4-methyl-3-((4-(3-pyridinyl)-2-pyrimidinyl)amino)-phenyl)benzamide methanesulfonate), NVP-AAK980-NX, KF-250706 (13-chloro,5(R),6(S)-epoxy-14,16-dihydroxy-11-(hydroyimino)-3(R)-methyl-3,4,5,6,11,12-hexahydro-1H-2-benzoxacyclotetradecin-1-one), 5-(3-(3-methoxy-4-(2-((E)-2-phenylethenyl)-4-oxazolylmethoxy)phenyl)propyl)-3-(2-((E)-2-phenylethenyl)-4-oxazolylmethyl)-2,4-oxazolidinedione, genistein, Nv-06, or an analogue or derivative thereof).
31 ) Vitronectin Inhibitors In another embodiment, the pharmacologically active compound is a vitronectin inhibitor (e.g., O-(9,10-dimethoxy-1,2,3,4,5,6-hexahydro-4-((1,4,5,6-tetrahydro-2-pyrimidinyl)hydrazono)-8-benz(e)azulenyl)-N-((phenylmethoxy)carbonyl)-DL-homoserine 2,3-dihydroxypropyl ester, (2S)-benzoylcarbonylamino-3-(2-((4S)-(3-(4,5-dihydro-1 H-imidazol-2-ylamino)-propyl)-2,5-dioxo-imidazolidin-1-yl)-acetylamino)-propionate, Sch-221153, S-836, SC-68448 (f3-((2-2-(((3-((aminoiminomethyl)amino)-phenyl)carbonyl)amino)acetyl)amino)-3,5-dichlorobenzenepropanoic acid), SD-7784, S-247, or an analogue or derivative thereof).
32) Fibroblast Growth Factor Inhibitors In another embodiment, the pharmacologically active compound is a fibroblast growth factor inhibitor (e.g., CT-052923 (((2H-benzo(d)1,3-dioxalan-5-methyl)amino)(4-(6,7-dimethoxyquinazolin-4-yl)piperazinyl)methane-1-thione), or an analogue or derivative thereof).
33) Protein Kinase Inhibitors In another embodiment, the pharmacologically active compound is a protein kinase inhibitor (e.g., KP-0201448, NPC15437 (hexanamide, 2,6-diamino-N-((1-(1-oxotridecyl)-2-piperidinyl)methyl)-), fasudil (1H-1,4-diazepine, hexahydro-1-(5-isoquinolinylsulfonyi)-), midostaurin (benzamide, N-(2,3,10,11,12,13-hexahyd ro-10-methoxy-9-methyl-1-oxo-9,13-epoxy-1 H,9H-diindolo(1,2,3-gh:3',2',1'-Im)pyrrolo(3,4-j)( 1,7)benzodiazonin-11-yl)-N-methyl-, (9Alpha,10(3,11(3,13AIpha)-),fasudil (1H-1,4-diazepine, hexahydro-1-(5-isoquinolinylsulfonyl)-, dexniguldipine (3,5-pyridinedicarboxylic acid, 1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-, 3-(4,4-diphenyl-1-piperidinyl)propyl methyl ester, monohydrochloride, (R)-), LY-317615 (1 H-pyrole-2,5-dione, 3-(1-methyl-1 H-indol-3-yl)-4-[1-[1-(2-pyridinylmethyl)-4-piperidinyl]-1 H-indol-3-yl]-, monohydrochloride), perifosine (piperidinium, 4-[[hydroxyloctadecyloxy)phosphinyl]oxy]-1,1-dimethyl-, inner salt), LY-333531 (9H,18H-5,21:12,17-d imethenodibenzo(e,lc)pyrrolo(3,4-h)(1,4,13)oxadiazacyclohexadecine-18,20(19H)-dione,9-((dimethylamino)methyl)-6,7,10,11-tetrahydro-, (S)-), Kynac; SPC-100270 (1,3-octadecanediol, 2-amino-, [S-(R*,R*)]-), Kynacyte, or an analogue or derivative thereof).
34) PDGF Receptor Kinase Inhibitors In another embodiment, the pharmacologically active compound is a PDGF receptor kinase inhibitor (e.g., RPR-127963E, or an analogue or derivative thereof).
35) Endothelial Growth Factor Receptor Kinase Inhibitors In another embodiment, the pharmacologically active compound is an endothelial growth factor receptor kinase inhibitor (e.g., CEP-7055, SU-0879 ((E)-3-(3,5-di-tert-butyl-4-hydroxyphenyl)-2-(aminothiocarbonyl)acrylonitrile), BIBF-1000, AG-013736 (CP-868596), AMG-706, AVE-0005, NM-3 (3-(2-methylcarboxymethyl)-6-methoxy-8-hydroxy-isocoumarin), Bay-43-9006, SU-011248,or an analogue or derivative thereof).
36) Retinoic Acid Receptor Antagonists In another embodiment, the pharmacologically active compound is a retinoic acid receptor antagonist (e.g., etarotene (Ro-15-1570) (naphthalene, 6-(2-(4-(ethylsulfonyl)phenyl)-1-methylethenyl)-1,2,3,4-tetrahydro-1,1,4,4-tetramethyl-, (E)-), (2E,4E)-3-methyl-5-(2-((E)-2-(2,6,6-trimethyl-1-cyclohexen-1-yl)ethenyl)-1-cyclohexen-1-yl)-2,4-pentadienoic acid, tocoretinate (retinoic acid, 3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyltridecyl)-2H-1-benzopyran-6-yl ester, (2R*(4R*,8R*))-(~)-), aliretinoin (retinoic acid, cis-9, trans-13-), bexarotene (benzoic acid, 4-(1-(5,6,7,8-tetrahydro-3,5,5,8,8-pentamethyl-2-naphthalenyl)ethenyl)-), tocoretinate (retinoic acid, 3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyltridecyl)-benzopyran-6-yl ester, [2R*(4R*,8R*)]-(~)-, or an analogue or derivative thereof).
37) Platelet Derived Growth Factor Receptor Kinase Inhibitors In another embodiment, the pharmacologically active compound is a platelet derived growth factor receptor kinase inhibitor (e.g., leflunomide (4-isoxazolecarboxamide, 5-methyl-N-(4-(trifluoromethyl)phenyl)-, or an analogue or derivative thereof).
38) Fibronogin Antagonists In another embodiment, the pharmacologically active compound is a fibrinogin antagonist (e.g., picotamide (1,3-benzenedicarboxamide, 4-methoxy-N,N'-bis(3-pyridinylmethyl)-, or an analogue or derivative thereof).
39) Antimycotic Agents In another embodiment, the pharmacologically active compound is an antimycotic agent (e.g., miconazole, sulconizole; parthenolide, rosconitine, nystatin, isoconazole, fluconazole, ketoconasole, imidazole, itraconazole, terpinafine, elonazole, bifonazole, clotrimazole, conazole, terconazole (piperazine, 1-(4-((2-(2,4-dichlorophenyl)-2-(1 H-1,2,4-triazol-1-ylmethyl)-1,3-dioxolan-4-yl)methoxy)phenyl)-4-(1-methylethyl)-, cis-), isoconazole (1-(2-(2-dichlorobenzyloxy)-2-(2-,4-dichlorophenyl)ethyl)), griseofulvin (spiro(benzofuran-2(3H),1'-(2)cyclohexane)-3,4'-dione, 7-chloro-2',4,6-trimeth-oxy-6'methyl-, (1'S-trans)-), bifonazole (1H-imidazole, 1-((1,1'-biphenyl)-4-ylphenylmethyl)-), econazole nitrate (1-(2-((4-chlorophenyl)methoxy)-2-(2,4-dichlorophenyl)ethyl)-1 H-imidazole nitrate), croconazole (1 H-imidazole, 1-(1-(2-((3-chlorophenyl)methoxy)phenyl)ethenyl)-), sertaconazole (1 H-Imidazole, 1-(2-((7-chlorobenzo(b)thien-3-yl)methoxy)-2-(2,4-dichlorophenyl)ethyl)-), omoconazole (1 H-imidazole, 1-(2-(2-(4-chlorophenoxy)ethoxy)-2-(2,4-dichlorophenyl)-1-methylethenyl)-, (Z)-), flutrimazole (1 H-imidazole, 1-((2-fluorophenyl)(4-fluorophenyl)phenylmethyl)-), fluconazole (1H-1,2,4-triazole-1-ethanol, alpha-(2,4-difluorophenyl)-alpha-(1H-1,2,4-triazol-1-ylmethyl)-), neticonazole (1 H-Imidazole, 1-(2-(methylthio)-1-(2-(pentyloxy)phenyl)ethenyl)-, monohydrochloride, (E)-), butoconazole (1 H-imidazole, 1-(4-(4-chlorophenyl)-2-((2,6-dichlorophenyl)thio)butyl)-, (+/-)-), clotrimazole (1-((2-chlorophenyl)diphenylmethyl)-1 H-imidazole, or an analogue or derivative thereof).
40) Bisphosphonates In another embodiment, the pharmacologically active compound is a bisphosphonate (e.g., clodronate, alendronate, pamidronate, zoledronate, or an analogue or derivative thereof).
41 ) Phospholipase A1 Inhibitors In another embodiment, the pharmacologically active compound is a phospholipase A1 inhibitor (e.g., ioteprednol etabonate (androsta-1,4-diene-17-carboxylic acid, 17-((ethoxycarbonyl)oxy)-11-hydroxy-3-oxo-, chloromethyl ester, (11 f~,17 alpha)-, or an analogue or derivative thereof).
42) Histamine H11H2/H3 Receptor Antagonists In another embodiment, the pharmacologically active compound is a histamine H1, H2, or H3 receptor antagonist (e.g., ranitidine (1,1-ethenediamine, N-(2-(((5-((dimethylamino)methyl)-2-furanyl)methyl)thio)ethyl)-N'-methyl-2-nitro-), niperotidine (N-(2-((5-((dimethylamino)methyl)furfuryl)thio)ethyl)-2-nitro-N'-piperonyl-1,1-ethenediamine), famotidine (propanimidamide, 3-(((2-((aminoiminomethyl)amino)-4-thiazolyl)methyl)thio)-N-(aminosulfonyl)-), roxitadine acetate HCI (acetamide, 2-(acetyloxy)-N-(3-(3-(1-piperidinylmethyl)phenoxy)propyl)-, monohydrochloride), lafutidine (acetamide, 2-((2-furanylmethyl)sulfinyl)-N-(4-((4-(1-piperidinylmethyl)-2-pyridinyl)oxy)-butenyl)-, (Z)-), nizatadine (1,1-ethenediamine, N-(2-(((2-((dimethylamino)methyl)-4-thiazolyl)methyl)thio)ethyl)-N'-methyl-2-nitro-), ebrotidine (benzenesulfonamide, N-(((2-(((2-((aminoiminomethyl)amino)-4-thiazofy)methyl)thio)ethyl)amino)methylene)-4-bromo-), rupatadine (5H-benzo(5,6)cyclohepta(1,2-b)pyridine, 8-chloro-6,11-dihydro-11-(1-((5-methyl-3-pyridinyl)methyl)-4-piperidinylidene)-, trihydrochloride-), fexofenadine HCI
(benzeneacetic acid, 4-(1-hydroxy-4-(4(hydroxydiphenylmethyl)-1-piperidinyl)butyl)-alpha, alpha-dimethyl-, hydrochloride, or an analogue or derivative thereof).
43) Macrolide Antibiotics In another embodiment, the pharmacologically active compound is a macrolide antibiotic (e.g., dirithromycin (erythromycin, 9-deoxo-11-deoxy-9,11-(imino(2-(2-methoxyethoxy)ethylidene)oxy)-, (9S(R))-), flurithromycin ethylsuccinate (erythromycin, 8-fluoro-mono(ethyl butanedioate) (ester)-), erythromycin stinoprate (erythromycin, 2'-propanoate, compound with N-acetyl-L-cysteine (1:1 )), clarithromycin (erythromycin, 6-O-methyl-), azithromycin (9-deoxo-9a-aza-9a-methyl-9a-homoerythromycin-A), telithromycin (3-de((2,6-dideoxy-3-C-methyl-3-O-methyl-alpha-L-ribo-hexopyranosyl)oxy)-11,12-dideoxy-6-O-methyl-3-oxo-12,11-(oxycarbonyl((4-(4-(3-pyridinyl)-1 H-imidazol-1-yl)butyl)imino))-), roxithromycin (erythromycin, 9-(O-((2-methoxyethoxy)methyl)oxime)), rokitamycin (leucomycin V, 4B-butanoate 3B-propanoate), RV-11 (erythromycin monopropionate mercaptosuccinate), midecamycin acetate (leucomycin V, 3B,9-diacetate 3,4B-dipropanoate), midecamycin (leucomycin V, 3,4B-dipropanoate), josamycin (leucomycin V, 3-acetate 4B-(3-methylbutanoate), or an analogue or derivative thereof).
44) GPllb Illa Receptor Antagonists In another embodiment, the pharmacologically active compound is a GPllb Ills receptor antagonist (e.g., tirofiban hydrochloride (L-tyrosine, N
(butylsulfonyl)-O-(4-(4-piperidinyl)butyl)-, monohydrochloride-), eptifibatide (L
cysteinamide, N6-(aminoiminomethyl)-N2-(3-mercapto-1-oxopropyl)-L
lysylglycyl-L-alpha-aspartyl-L-tryptophyl-L-prolyl-, cyclic(1->6)-disulfide), xemilofiban hydrochloride, or an analogue or derivative thereof).
45) Endothelia Receptor Antagonists In another embodiment, the pharmacologically active compound is an endothelia receptor antagonist (e.g., bosentan (benzenesulfonamide, 4-(1,1-dimethylethyl)-N-(6-(2-hydroxyethoxy)-5-(2-methoxyphenoxy)(2,2'-bipyrimidin)-4-yl)-, or an analogue or derivative thereof).
46) Peroxisome Proliferator-Activated Receptor Aaonists In another embodiment, the pharmacologically active compound is a peroxisome proliferator-activated receptor agonist (e.g., gemfibrozil (pentanoic acid, 5-(2,5-dimethylphenoxy)-2,2-dimethyl-), fenofibrate (propanoic acid, 2-(4-(4-chlorobenzoyl)phenoxy)-2-methyl-, 1-methylethyl ester), ciprofibrate (propanoic acid, 2-(4-(2,2-dichlorocyclopropyl)phenoxy)-2-methyl-), rosiglitazone maleate (2,4-thiazolidinedione, 5-((4-(2-(methyl-2-pyridinylamino)ethoxy)phenyl)methyl)-, (Z)-2-butenedioate (1:1 )), pioglitazone hydrochloride (2,4-thiazolidinedione, 5-((4-(2-(5-ethyl-2-pyridinyl)ethoxy)phenyl)methyl)-, monohydrochloride (+/-)-), etofylline clofibrate (propanoic acid, 2-(4-chlorophenoxy)-2-methyl-, 2-(1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-7H-purin-7-yl)ethyl ester), etofibrate (3-pyridinecarboxylic acid, 2-(2-(4-chlorophenoxy)-2-methyl-1-oxopropoxy)ethyl ester), clinofibrate (butanoic acid, 2,2'-(cyclohexylidenebis(4,1-phenyleneoxy))bis(2-methyl-)), bezafibrate (propanoic acid, 2-(4-(2-((4-chlorobenzoyl)amino)ethyl)phenoxy)-2-methyl-), binifibrate (3-pyridinecarboxylic acid, 2-(2-(4-chlorophenoxy)-2-methyl-1-oxopropoxy)-1,3-propanediyl ester), or an analogue or derivative thereof).
In one aspect, the pharmacologically active compound is a peroxisome proliferator-activated receptor alpha agonist, such as GW-590735, GSK-677954, GSK501516, pioglitazone hydrochloride (2,4-thiazolidinedione, 5-[[4-[2-(5-ethyl-2-pyridinyl)ethoxy]phenyl]methyl]-, monohydrochloride (+/-)-, or an analogue or derivative thereof).
47) Estroe~en Receptor Agents In another embodiment, the pharmacologically active compound is an estrogen receptor agent (e.g., estradiol, 17-(3-estradiol, or an analogue or derivative thereof).
48) Somatostatin Analogues In another embodiment, the pharmacologically active compound is a somatostatin analogue (e.g., angiopeptin, or an analogue or derivative thereof).
49) Neurokinin 1 Antagonists In another embodiment, the pharmacologically active compound is a neurokinin 1 antagonist (e.g., GW-597599, lanepitant ((1,4'-bipiperidine)-1'-acetamide, N-(2-(acetyl((2-methoxyphenyl)methyl)amino)-1-(1 H-indol-3-ylmethyl)ethyl)- (R)-), nolpitantium chloride (1-azoniabicyclo[2.2.2]octane, 1-[2-[3-(3,4-dichlorophenyl)-1-[[3-(1-methylethoxy)phenyl]acetyl)-3-piperidinyl]ethyl]-4-phenyl-, chloride, (S)-), or saredutant (benzamide, N-[4-[4-(acetylamino)-4-phenyl-1-piperidinyl]-2-(3,4-dichlorophenyl)butyl]-N-methyl-, (S)-), or vofopitant (3-piperidinamine, N-[[2-methoxy-5-[5-(trifluoromethyl)-1 H-tetrazol-1-yl]phenyl]methyl]-2-phenyl-, (2S,3S)-, or an analogue or derivative thereof).
50) Neurokinin 3 Antagonist In another embodiment, the pharmacologically active compound is a neurokinin 3 antagonist (e.g., talnetant (4-quinolinecarboxamide, 3-hydroxy-2-phenyl-N-[(1S)-1-phenylpropyl]-, or an analogue or derivative thereof).
51 ) Neurokinin Antagonist In another embodiment, the pharmacologically active compound is a neurokinin antagonist (e.g., GSK-679769, GSK-823296, SR-489686 (benzamide, N-[4-[4-(acetylamino)-4-phenyl-1-piperidinyl]-2-(3,4-dichlorophenyl)butyl]-N-methyl-, (S)-), SB-223412; SB-235375 (4-quinolinecarboxamide, 3-hydroxy-2-phenyl-N-[(1S)-1-phenylpropyl]-), UK-226471, or an analogue or derivative thereof).
52) VLA-4 Antagonist In another embodiment, the pharmacologically active compound is a VLA-4 antagonist (e.g., GSK683699, or an analogue or derivative thereof).
53) Osteoclast Inhibitor In another embodiment, the pharmacologically active compound is a osteoclast inhibitor (e.g., ibandronic acid (phosphonic acid, [1-hydroxy-(methylpentylamino)propylidene] bis-), alendronate sodium, or an analogue or derivative thereof).
54) DNA topoisomerase ATP Hydrolysing Inhibitor In another embodiment, the pharmacologically active compound is a DNA topoisomerase ATP hydrolysing inhibitor (e.g., enoxacin (1,8-naphthyridine-3-carboxylic acid, 1-ethyl-6-fluoro-1,4-dihydro-4-oxo-7-(1-piperazinyl)-), levofloxacin (7H-Pyrido[1,2,3-de]-1,4-benzoxazine-6-carboxylic acid, 9-fluoro-2,3-dihydro-3-methyl-10-(4-methyl-1-piperazinyl)-7-oxo-, (S)-), ofloxacin (7H-pyrido[1,2,3-de]-1,4-benzoxazine-6-carboxylic acid, 9-fluoro-2,3-dihydro-3-methyl-10-(4-methyl-1-piperazinyl)-7-oxo-, (+/-)-), pefloxacin (3-quinolinecarboxylic acid, 1-ethyl-6-fluoro-1,4-dihydro-7-(4-methyl-1-piperazinyl)-4-oxo-), pipemidic acid (pyrido[2,3-d]pyrimidine-6-carboxylic acid, 8-ethyl-5,8-dihydro-5-oxo-2-(1-piperazinyl)-), pirarubicin (5,12-naphthacenedione, 10-[[3-amino-2,3,6-trideoxy-4-O-(tetrahydro-2H-pyran-2-yl)-alpha-L-lyxo-hexopyranosyl]oxy]-7,8,9,10-tetrahyd ro-6,8,11-trihydroxy-8-(hydroxyacetyl)-1-methoxy-, [8S-[8 alpha,10 alpha(S*)]]-), sparfloxacin (3-quinolinecarboxylic acid, 5-amino-1-cyclopropyl-7-(3,5-dimethyl-1-piperazinyl)-6,8-difluoro-1,4-dihydro-4-oxo-, cis-), AVE-6971, cinoxacin ([1,3]dioxolo[4,5-g]cinnoline-3-carboxylic acid, 1-ethyl-1,4-dihydro-4-oxo-), or an analogue or derivative thereof).
55) Anaiotensin I Converting Enzyme Inhibitor In another embodiment, the pharmacologically active compound is an angiotensin I converting enzyme inhibitor (e.g., ramipril (cyclopenta[b]pyrrole-2-carboxylic acid, 1-[2-[[1-(ethoxycarbonyl)-3-phenylpropyl]amino]-1-oxopropyl]octahydro-, [2S-[1 [R*(R*)],2 alpha, 3af3, 6a(3]]-), trandolapril (1 H-indole-2-carboxylic acid, 1-[2-[(1-carboxy-3-phenylpropyl)amino]-1-oxopropyl]octahydro-, [2S-[1 [R*(R*)],2 alpha,3a alpha,7af~]]-), fasidotril (L-alanine, N-[(2S)-3-(acetylthio)-2-(1,3-benzodioxol-5-ylmethyl)-1-oxopropyl]-, phenylmethyl ester), cilazapril (6H-pyridazino[1,2-a][1,2]diazepine-1-carboxylic acid, 9-[[1-(ethoxycarbonyl)-3-phenylpropyl]amino]octahydro-10-oxo-, [1 S-[1 alpha, 9 alpha(R*)]]-), ramipril (cyclopenta[b]pyrrole-2-carboxylic acid, 1-[2-[[1-(ethoxycarbonyl)-3-phenylpropyl]amino]-1-oxopropyl]octahydro-, [2S-[1 [R*(R*)], 2 alpha,3a(3,6a(3]]-, or an analogue or derivative thereof).
56) Ane~iotensin II Antagonist In another embodiment, the pharmacologically active compound is an angiotensin II antagonist (e.g., HR-720 (1 H-imidazole-5-carboxylic acid, 2-butyl-4-(methylthio)-1-[[2'-[[[(propylamino)carbonyl]amino]sulfonyl][1,1'-biphenyl]-4-yl]methyl]-, dipotassium salt, or an analogue or derivative thereof).
57) Enkephalinase Inhibitor In another embodiment, the pharmacologically active compound is an enkephalinase inhibitor (e.g., Aventis 100240 (pyrido[2,1-a][2]benzazepine-4-carboxylic acid, 7-[[2-(acetylthio)-1-oxo-3-phenylpropyl]amino]-1,2,3,4,6,7,8,12b-octahydro-6-oxo-, [4S-[4 alpha, 7 alpha(R*),12bf~]]-), AVE-7688, or an analogue or derivative thereof).
58) Peroxisome Proliferator-Activated Receptor Gamma Aaonist Insulin Sensitizer In another embodiment, the pharmacologically active compound is peroxisome proliferator-activated receptor gamma agonist insulin sensitizer (e.g., rosiglitazone maleate (2,4-thiazolidinedione, 5-((4-(2-(methyl-2-pyridinylamino)ethoxy)phenyl)methyl)-, (Z)-2-butenedioate (1:1 ), farglitazar (GI-262570, GW-2570, GW-3995, GW-5393, GW-9765), LY-929, LY-519818, LY-674, or LSN-862), or an analogue or derivative thereof).
59) Protein Kinase C Inhibitor In another embodiment, the pharmacologically active compound is a protein kinase C inhibitor, such as ruboxistaurin mesylate (9H,18H-5,21:12,17-dimethenodibenzo(e,k)pyrrolo(3,4-h)(1,4,13)oxadiazacyclohexadecine-18,20(19H)-dione,9-((dimethylamino)methyl)-6,7,10,11-tetrahydro-, (S)-), safingol (1,3-octadecanediol, 2-amino-, [S-(R*,R*)]-}, or enzastaurin hydrochloride (1 H-pyrole-2,5-dione, 3-(1-methyl-1 H-indol-3-yl)-4-[1-[1-(2-pyridinylmethyl)-4-piperidinyl]-1 H-indol-3-yl]-, monohydrochloride), or an analogue or derivative thereof.
60) ROCK (rho-associated kinase) Inhibitors In another embodiment, the pharmacologically active compound is a ROCK (rho-associated kinase) inhibitor, such as Y-27632, HA-1077, H-1152 and 4-1-(aminoalkyl)-N-(4-pyridyl) cyclohexanecarboxamide or an analogue or derivative thereof.
61 ) CXCR3 Inhibitors In another embodiment, the pharmacologically active compound is a CXCR3 inhibitor such as T-487, T0906487 or analogue or derivative thereof.
62) Itk Inhibitors In another embodiment, the pharmacologically active compound is an Itk inhibitor such as BMS-509744 or an analogue or derivative thereof.
63) Cytosolic phospholipase A~-alpha Inhibitors In another embodiment, the pharmacologically active compound is a cytosolic phospholipase A2-alpha inhibitor such as efipladib (PLA-902) or analogue or derivative thereof.
64) PPAR Aaonist In another embodiment, the pharmacologically active compound is a PPAR Agonist (e.g., Metabolex ((-)-benzeneacetic acid, 4-chloro-alpha-[3-(trifluoromethyl)-phenoxy]-, 2-(acetylamino)ethyl ester), balaglitazone (5-(4-(3-methyl-4-oxo-3,4-dihydro-quinazolin-2-yl-methoxy)-benzyl)-thiazolidine-2,4-dione), ciglitazone (2,4-thiazolidinedione, 5-[[4-[(1-methylcyclohexyl)methoxy]phenyl]methyl]-), DRF-10945, farglitazar, GSK-677954, GW-409544, GW-501516, GW-590735, GW-590735, K-111, KRP-101, LSN-862, LY-519818, LY-674, LY-929, muraglitazar; BMS-298585 (Glycine, N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5-methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]-), netoglitazone; isaglitazone (2,4-thiazolidinedione, 5-[[6-[(2-fluorophenyl)methoxy]-2-naphthalenyl]methyl]-), Actos AD-4833; U-72107A (2,4-thiazolidinedione, 5-[[4-[2-(5-ethyl-2-pyridinyl)ethoxy]phenyl]methyl]-, monohydrochloride (+l-)-), JTT-501; PNU-182716 (3,5-Isoxazolidinedione, 4-[[4-[2-(5-methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]-), AVANDIA (from SB Pharmco Puerto Rico, Inc. (Puerto Rico); BRL-48482;BRL-49653;BRL-49653c; NYRACTA and Venvia (both from (SmithKline Beecham (United Kingdom)); tesaglitazar ((2S)-2-ethoxy-3-[4-[2-[4-[(methylsulfonyl)oxy]phenyl]ethoxy]phenyl] propanoic acid), troglitazone (2,4-Thiazolidinedione, 5-[[4-[(3,4-dihydro-6-hydroxy-2,5,7,8-tetramethyl-2H-1-benzopyran-2-yl)methoxy]phenyl]methyl]-), and analogues and derivatives thereof).
65) Immunosuppressants In another embodiment, the pharmacologically active compound is an immunosuppressant (e.g., batebulast (cyclohexanecarboxylic acid, 4-[[(aminoiminomethyl)amino]methyl]-, 4-(1,1-dimethylethyl)phenyl ester, trans-), cyclomunine, exalamide (benzamide, 2-(hexyloxy)-), LYN-001, CCI-779 (rapamycin 42-(3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate)), 1726;
1726-D; AVE-1726, or an analogue or derivative thereof).
66) Erb Inhibitor In another embodiment, the pharmacologically active compound is an Erb inhibitor (e.g., canertinib dihydrochloride (N-[4-(3-(chloro-4-fluoro-phenylamino)-7-(3-morpholin-4-yl-propoxy)-quinazolin-6-yl]-acrylamide dihydrochloride), CP-724714, or an analogue or derivative thereof).
67) Apoptosis Agonist In another embodiment, the pharmacologically active compound is an apoptosis agonist (e.g., CEFLATONIN (CGX-635) (from Chemgenex Therapeutics, Inc., Menlo Park, CA), CHML, LBH-589, metoclopramide (benzamide, 4-amino-5-chloro-N-[2-(diethylamino)ethyl]-2-methoxy-), patupilone (4,17-dioxabicyclo(14.1.0)heptadecane-5,9-dione, 7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-(1-methyl-2-(2-methyl-4-thiazolyl)ethenyl, (1R,3S,7S,10R,11S,12S,16R)), AN-9; pivanex (butanoic acid, (2,2-dimethyl-1-oxopropoxy)methyl ester), SL-100; SL-102; SL-11093; SL-11098; SL-11099;
SL-93; SL-98; SL-99, or an analogue or derivative thereof).
68) Lipocortin Ae~onist In another embodiment, the pharmacologically active compound is an lipocortin agonist (e.g., CGP-13774 (9Alpha-chloro-6Alpha-fluoro 11 f~,17alpha-dihydroxy-16AIpha-methyl-3-oxo-1,4-androstadiene-17f3-carboxylic acid-methylester-17-propionate), or analogue or derivative thereof).
69) VCAM-1 antaetonist In another embodiment, the pharmacologically active compound is a VCAM-1 antagonist (e.g., DW-908e, or an analogue or derivative thereof).
70) Collagen Antagonist In another embodiment, the pharmacologically active compound is a collagen antagonist (e.g., E-5050 (Benzenepropanamide, 4-(2,6-dimethylheptyl)-N-(2-hydroxyethyl)-f3-methyl-), lufironil (2,4-Pyridinedicarboxamide, N,N'-bis(2-methoxyethyl)-), or an analogue or derivative thereof).
71 ) Alpha 2 Intearin Antagonist In another embodiment, the pharmacologically active compound is an alpha 2 integrin antagonist (e.g., E-7820, or an analogue or derivative thereof).
72) TNF Alpha Inhibitor In another embodiment, the pharmacologically active compound is a TNF alpha inhibitor (e.g., ethyl pyruvate, Genz-29155, lentinan (Ajinomoto Co., Inc. (Japan)), linomide (3-quinolinecarboxamide, 1,2-dihydro-4-hydroxy N,1-dimethyl-2-oxo-N-phenyl-), UR-1505, or an analogue or derivative thereof).
73) Nitric Oxide Inhibitor In another embodiment, the pharmacologically active compound is a nitric oxide inhibitor (e.g., guanidioethyldisulfide, or an analogue or derivative thereof).
74) . Cathepsin Inhibitor In another embodiment, the pharmacologically active compound is a cathepsin inhibitor (e.g., SB-462795 or an analogue or derivative therof).
Within various embodiments of the invention, a device incorporates or is coated on one aspect, portion or surface with a composition which inhibits fibrosis (and/or restenosis), as well as with a composition or compound which promotes fibrosis on another aspect, portion or surface of the device. Representative examples of agents that promote fibrosis include silk and other irritants (e.g., talc, wool (including animal wool, wood wool, and synthetic wool), talcum powder, copper, metallic beryllium (or its oxides), quartz dust, silica, crystalline silicates), polymers (e.g., polylysine, polyurethanes, polyethylene terephthalate), PTFE, poly(alkylcyanoacrylates), and polyethylene-co-vinylacetate); vinyl chloride and polymers of vinyl chloride;
peptides with high lysine content; growth factors and inflammatory cytokines involved in angiogenesis, fibroblast migration, fibroblast proliferation, ECM
synthesis and tissue remodeling, such as epidermal growth factor (EGF) family, transforming growth factor-a (TGF- a), transforming growth factor-~ (TGF-9-1, TGF-9-2, TGF-9-3, platelet-derived growth factor (PDGF), fibroblast growth factor (acidic - aFGF; and basic - bFGF), fibroblast stimulating factor-1, activins, vascular endothelial growth factor (including VEGF-2, VEGF-3, VEGF-A, VEGF-B, VEGF-C, placental growth factor- PIGF), angiopoietins, insulin-like growth factors (IGF), hepatocyte growth factor (HGF), connective tissue growth factor (CTGF), myeloid colony-stimulating factors (CSFs), monocyte chemotactic protein, granulocyte-macrophage colony-stimulating factors (GM-CSF), granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), erythropoietin, interleukins (particularly IL-1, IL-8, and IL-6), tumor necrosis factor-a (TNF9), nerve growth factor (NGF), interferon-a, interferon-(3, histamine, endothelin-1, angiotensin II, growth hormone (GH), and synthetic peptides, analogues or derivatives of these factors are also suitable for release from specific implants and devices to be described later. Other examples include CTGF (connective tissue growth factor); inflammatory microcrystals (e.g., crystalline minerals such as crystalline silicates); bromocriptine, methylsergide, methotrexate, chitosan, N-carboxybutyl chitosan, carbon tetrachloride, thioacetamide, fibrosin, ethanol, bleomycin, naturally occurring or synthetic peptides containing the Arg-Gly-Asp (RGD) sequence, generally at one or both termini (see, e.g., U.S. Patent No.
5,997,895), and tissue adhesives, such as cyanoacrylate and crosslinked polyethylene glycol) - methylated collagen compositions. Other examples of fibrosis-inducing agents include bone morphogenic proteins (e.g., BMP-2, BMP-3, BMP-4, BMP-5, BMP-6 (Vgr-1 ), BMP-7 (OP-1 ), BMP-8, BMP-9, BMP-10, BMP-11, BMP-12, BMP-13, BMP-14, BMP-15, and BMP-16. Of these, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, and BMP-7 are of particular utility. Bone morphogenic proteins are described, for example, in U.S. Patent Nos.
4,877,864; 5,013,649; 5,661,007; 5,688,678; 6,177,406; 6,432,919; and 6,534,268 and Wozney, J.M., et al. (1988) Science: 242(4885); 1528-1534.
Other representative examples of fibrosis-inducing agents include components of extracellular matrix (e.g., fibronectin, fibrin, fibrinogen, collagen (e.g., bovine collagen), including fibrillar and non-fibrillar collagen, adhesive glycoproteins, proteoglycans (e.g., heparin sulfate, chondroitin sulfate, dermatan sulfate), hyaluronan, secreted protein acidic and rich in cysteine (SPARC), thrombospondins, tenacin, and cell adhesion molecules (including integrins, vitronectin, fibronectin, laminin, hyaluronic acid, elastin, bitronectin), proteins found in basement membranes, and fibrosin) and inhibitors of matrix metalloproteinases, such as TIMPs (tissue inhibitors of matrix metalloproteinases) and synthetic TIMPs, such as, e.g., marimistat, batimistat, doxycycline, tetracycline, minocycline, TROCADE, Ro-1130830, CGS 27023A, and BMS-275291 and analogues and derivatives thereof.
The medical implant may include a fibrosis-inhibiting agent and an anti-thrombotic agent and/or antiplatelet agent and/or a thrombolytic agent, which reduces the likelihood of thrombotic events upon implantation of a medical implant. Within various embodiments of the invention, a device is coated on one aspect with a composition which inhibits fibrosis (and/or restenosis), as well as being coated with a composition or compound which prevents thrombosis on another aspect of the device. Representative examples of anti-thrombotic and/or antiplatelet and/or thrombolytic agents include heparin, heparin fragments, organic salts of heparin, heparin complexes (e.g., benzalkonium heparinate, tridodecylammonium heparinate), dextran, sulfonated carbohydrates such as dextran sulphate, coumadin, coumarin, heparinoid, danaparoid, argatroban chitosan sulfate, chondroitin sulfate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, acetylsalicylic acid, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, streptokinase, factor Xa inhibitors, such as DX9065a, magnesium, and tissue plasminogen activator. Further examples include plasminogen, lys-plasminogen, alpha-2-antiplasmin, urokinase, aminocaproic acid, ticlopidine, clopidogrel, trapidil (triazolopyrimidine), naftidrofuryi, auriritricarboxylic acid and gfycoprotein Ifb/Illa inhibitors such as abcixamab, eptifibatide, and tirogiban. Other agents capable of affecting the rate of clotting include glycosaminoglycans, danaparoid, 4 hydroxycourmarin, warfarin sodium, dicumarol, phenprocoumon, indan-1,3-dione, acenocoumarol, anisindione, and rodenticides including bromadiolone, brodifacoum, diphenadione, chlorophacinone, and pidnone.
The thrombogenicity of a medical implant may be reduced by coating the implant with a polymeric formulation that has anti-thrombogenic properties. For example, a medical device may be coated with a hydrophilic polymer gel. The polymer gel can comprise a hydrophilic, biodegradable polymer that is physically removed from the surface of the device over time, thus reducing adhesion of platelets to the device surface. The gel composition can include a polymer or a blend of polymers. Representative examples include alginates, chitosan and chitosan sulfate, hyaluronic acid, dextran sulfate, PLURONIC polymers (e.g., F-127 or F87), chain extended PLURONIC
polymers, various polyester-polyether block copolymers of various configurations (e.g., AB, ABA, or BAB, where A is a polyester such as PLA, PGA, PLGA, PCL or the like), examples of which include MePEG-PLA, PLA-PEG-PLA, and the like). In one embodiment, the anti-thrombotic composition can include a crosslinked gel formed from a combination of molecules (e.g., PEG) having two or more terminal electrophilic groups and two or more nucleophilic groups.
In one aspect, the present invention also provides for the combination of a medical implant (as well as compositions and methods for making medical implants) that includes an anti-fibrosing agent and an anti-infective agent, which reduces the likelihood of infections in medical implants.
Infection is a common complication of the implantation of foreign bodies such as medical devices. Foreign materials provide an ideal site for micro-organisms to attach and colonize. It is also hypothesized that there is an impairment of host defenses to infection in the microenvironment surrounding a foreign material. These factors make medical implants particularly susceptible to infection and make eradication of such an infection difficult, if not impossible, in most cases.
The present invention provides agents (e.g., chemotherapeutic agents) that can be released from an implantable device, and which have potent antimicrobial activity at extremely low doses. A wide variety of anti-infective agents can be utilized in combination with a fibrosing agent according to the invention. Discussed in more detail below are several representative examples of agents that can be used: (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin).
(A) Anthracyclines Anthracyclines have the following general structure, where the R
groups may be a variety of organic groups:

According to U.S. Patent 5,594,158, suitable R groups are as follows: R~ is CH3 or CH~OH; R2 is daunosamine or H; R3 and R4 are independently one of OH, N02, NH2, F, CI, Br, I, CN, H or groups derived from these; R5 is hydrogen, hydroxyl, or methoxy; and R6_$ are all hydrogen.
Alternatively, R5 and R6 are hydrogen and R~ and R$ are alkyl or halogen, or vice versa.
According to U.S. Patent 5,843,903, R~ may be a conjugated peptide. According to U.S. Patent 4,296,105, R5 may be an ether linked alkyl group. According to U.S. Patent 4,215,062, R5 may be OH or an ether linked alkyl group. R~ may also be linked to the anthracycline ring by a group other than C(O), such as an alkyl or branched alkyl group having the C(O) linking moiety at its end, such as -CH2CH(CH2-X)C(O)-R~, wherein X is H or an alkyl group (see, e.g., U.S. Patent 4,215,062). R2 may alternately be a group linked by the functional group =N-NHC(O)-Y, where Y is a group such as a phenyl or substituted phenyl ring. Alternately R3 may have the following structure:
HaC O
~NH

Rio in which R9 is OH either in or out of the plane of the ring, or is a second sugar moiety such as R3. Rio may be H or form a secondary amine with a group such as an aromatic group, saturated or partially saturated 5 or 6 membered heterocyclic having at least one ring nitrogen (see U.S. Patent 5,843,903).
Alternately, Rio may be derived from an amino acid, having the structure -C(O)CH(NHR~~)(R~~), in which R~~ is H, or forms a C3_4 membered alkylene with R~2. R~2 may be H, alkyl, aminoalkyl, amino, hydroxyl, mercapto, phenyl, benzyl or methylthio (see U.S. Patent 4,296,105).

Exemplary anthracyclines are doxorubicin, daunorubicin, idarubicin, epirubicin, pirarubicin, zorubicin, and carubicin. Suitable compounds have the structures:
O OH
R~ R2 R3 Doxorubicin: OCH3 C(O)CH20H OH out of ring plane Epirubicin:

(4' epimer of OCH3 C(O)CH2OH OH in ring plane doxorubicin) Daunorubicin: OCH3 C(O)CH3 OH out of ring plane Idarubicin: H C(O)CH3 OH out of ring plane Pirarubicin: OCH3 C(O)CH20H

Zorubicin: OCH3 C(CH3)(=N)NHC(O)C6H5OH

Carubicin: OH C(O)CH3 OH out of ring plane Other suitable anthracyclines are anthramycin, mitoxantrone, menogaril, nogalamycin, aclacinomycin A, olivomycin A, chromomycin A3, and plicamycin having the structures:

'NHZ
s Menogaril H OCH3 H
OH O HN' /~ OH
~NH~ Nogalarrycin O-sugar H COOCH3 sugar ~C o OH O HN\ /~ OH Fi3C0 ~H3 OCI-l~
~NH~
Mtoxantrone 0 ocH, cH, ",oH
n i HO

O

~CH~3~
R~O,C
~ ~

HO R, Ra Pa Ra OliwmycinCOCH(CH3)zCOGH3H

ChromomycinCOCH3 COCH3CH3 PlicamycinH H H CH3 Other representative anthracyclines include, FCE 23762, a doxorubicin derivative (Quaglia et al., J. Liq. Chromatogr. 77(18):3911-3923, 1994), annamycin (Zoo et al., J. Pharm. Sci. 82(11 ):1151-1154, 1993), ruboxyl (Rapoport et al., J. Controlled Release 58(2):153-162, 1999), anthracycline disaccharide doxorubicin analogue (Pratesi et al., Clin. Cancer Res.
4(11 ):2833-2839, 1998), N-(trifluoroacetyl)doxorubicin and 4'-O-acetyl-N-(trifluoroacetyl)doxorubicin (Berube ~t Lepage, Synth. Common. 28(6):1109-1116, 1998), 2-pyrrolinodoxorubicin (Nagy et al., Proc. Nat'I Acad. Sci.
U.S.A.
95(4):1794-1799, 1998), disaccharide doxorubicin analogues (Arcamone et al., J. Nat'I Cancer Inst. 89(16):1217-1223, 1997), 4-demethoxy-7-O-[2,6-dideoxy-4-O-(2,3,6-trideoxy-3-amino-a-L-lyxo-hexopyranosyl)-a-L-lyxo-hexopyranosyl]-adriamicinone doxorubicin disaccharide analogue (Monteagudo et al., Carbohydr. Res. 300(1 ):11-16, 1997), 2-pyrrolinodoxorubicin (Nagy et al., Proc.
Nat'I Acad. Sci. U.S.A. 94(2):652-656, 1997), morpholinyl doxorubicin analogues (Duran et al., Cancer Chemother. Pharmacol. 38(3):210-216, 1996), enaminomalonyl-~-alanine doxorubicin derivatives (Seitz et al., Tetrahedron Lett. 36(9):1413-16, 1995), cephalosporin doxorubicin derivatives (Vrudhula et al., J. Med. Chem. 38(8):1380-5, 1995), hydroxyrubicin (Solary et al., Int. J.

Cancer 58(1 ):85-94, 1994), methoxymorpholino doxorubicin derivative (Kuhl et al., Cancer Chemother. Pharmacol. 33(1 ):10-16, 1993), (6-maleimidocaproyl)hydrazone doxorubicin derivative (Willner et al., Bioconjugate Chem. 4(6):521-7, 1993), N-(5,5-diacetoxypent-1-yl) doxorubicin (Cherif &
Farquhar, J. Med. Chem. 35(17):3208-14, 1992), FCE 23762 methoxymorpholinyl doxorubicin derivative (Ripamonti et al., Br. J. Cancer 65(5):703-7, 1992), N-hydroxysuccinimide ester doxorubicin derivatives (Demant et al., Biochim. Biophys. Acta 1118(1 ):83-90, 1991 ), polydeoxynucleotide doxorubicin derivatives (Ruggiero et al., Biochim.
Biophys.
Acta 1129(3):294-302, 1991 ), morpholinyl doxorubicin derivatives (EPA
434960), mitoxantrone doxorubicin analogue (Krapcho et al., J. Med. Chem.
34(8):2373-80. 1991 ), AD198 doxorubicin analogue (Traganos et al., Cancer Res. 51(14):3682-9, 1991 ), 4-demethoxy-3'-N-trifluoroacetyldoxorubicin (Norton et al., Drug Des. Delivery 6(2):123-9, 1990), 4'-epidoxorubicin (Drzewoski et al., Pol. J. Pharmacol. Pharm. 40(2):159-65, 1988; Weenen et al., Eur. J. Cancer Clin. Oncol. 20(7):919-26, 1984), alkylating cyanomorpholino doxorubicin derivative (Scudder et al., J. Nat'I Cancer Inst. 8Q(16):1294-8, 1988), deoxydihydroiodooxorubicin (EPA 275966), adriblastin (Kalishevskaya et al., Vestn. Mosk. Univ., 16(Biol. 1 ):21-7, 1988), 4'-deoxydoxorubicin (Schoelzel et al., Leuk. Res. 10(12):1455-9, 1986), 4-demethyoxy-4'-o-methyldoxorubicin (Giuliani et al., Proc. Int. Congr. Chemother. 16:285-70-285-77, 1983), 3'-deamino-3'-hydroxydoxorubicin (Norton et al., J. Antibiot. 37(8):853-8, 1984), demethyoxy doxorubicin analogues (Barbieri et al., Drugs Exp. Clin. Res.
10(2):85-90, 1984), N-L-leucyl doxorubicin derivatives (Trouet et al., Anthracyclines (Proc. lnt. Symp. Tumor Pharmacother.), 179-81, 1983), 3'-deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054), 3'-deamino-3'-(4-mortholinyl) doxorubicin derivatives (U.S. 4,301,277), 4'-deoxydoxorubicin and 4'-o-methyldoxorubicin (Giuliani et al., Int. J. Cancer 27(1 ):5-13, 1981 ), aglycone doxorubicin derivatives (Chan & Watson, J.
Pharm.
Sci. 67(12):1748-52, 1978), SM 5887 (Pharma Japan 1468:20, 1995), MX-2 (Pharma Japan 1420:19, 1994), 4'-deoxy-13(S)-dihydro-4'-iododoxorubicin (EP
275966), morpholinyl doxorubicin derivatives (EPA 434960), 3'-deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054), doxorubicin-14-valerate, morpholinodoxorubicin (U.S. 5,004,606), 3'-deamino-3'-(3"-cyano-4"-morpholinyl doxorubicin; 3'-deamino-3'-(3"-cyano-4"-morpholinyl)-13-dihydoxorubicin; (3'-deamino-3'-(3"-cyano-4"-morpholinyl) daunorubicin; 3'-deamino-3'-(3"-cyano-4"-morpholinyl)-3-dihydrodaunorubicin; and 3'-deamino-3'-(4"-morpholinyl-5-iminodoxorubicin and derivatives (U.S. 4,585,859), 3'-deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054) and 3-deamino-3-(4-morpholinyl) doxorubicin derivatives (U.S. 4,301,277).
(B) Fluoropyrimidine analogues In another aspect, the therapeutic agent is a fluoropyrimidine analog, such as 5-fluorouracil, or an analogue or derivative thereof, including carmofur, doxifluridine, emitefur, tegafur, and floxuridine. Exemplary compounds have the structures:

R~ \ F
N I
O N
R' R~ R2 5-FluorouracilH H

Carmofur C(O)NH(CH2)5CH3 H

DoxifluridineA~ H

Floxuridine A2 H

Emitefur CH20CH2CH3 B

Tegafur C H

B CN
O ~ ~ O O
O N O
C
O

Other suitable fluoropyrimidine analogues include 5-FudR (5-fluoro-deoxyuridine), or an analogue or derivative thereof, including 5-iododeoxyuridine (5-IudR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP).
Exemplary compounds have the structures:
5-Fluoro-2'-deoxyuridine: R = F
5-Bromo-2'-deoxyuridine: R = Br 5-lodo-2'-deoxyuridine: R = I
Other representative examples of fluoropyrimidine analogues include N3-alkylated analogues of 5-fluorouracil (Kozai et al., J. Chem. Soc., Perkin Trans. 7(19):3145-3146, 1998), 5-fluorouracil derivatives with 1,4-oxaheteroepane moieties (Gomez et al., Tetrahedron 54(43):13295-13312, 1998), 5-fluorouracil and nucleoside analogues (Li, Anticancer Res. 17(1A):21-27, 1997), cis- and trans-5-fluoro-5,6-dihydro-6-alkoxyuracil (Van der Wilt et al., Br. J. Cancer 68(4):702-7, 1993), cyclopentane 5-fluorouracil analogues (Hronowski & Szarek, Can. J. Chem. 70(4):1162-9, 1992), A-OT-fluorouracil (Zhang et al., Zongguo Yiyao Gongye Zazhi 20(11 ):513-15, 1989), N4-trimethoxybenzoyl-5'-deoxy-5-fluorocytidine and 5'-deoxy-5-fluorouridine (Miwa et al., Chem. Pharm. Bull. 38(4):998-1003, 1990), 1-hexylcarbamoyl-5-fluorouracil (Hoshi et al., J. Pharmacobio-Dun. 3(9):478-81, 1980; Maehara et al., Chemotherapy (Basel) 34(6):484-9, 1988), B-3839 (Prajda et al., In Vivo 2(2):151-4, 1988), uracil-1-(2-tetrahydrofuryl)-5-fluorouracil (Anai et al., Oncology 45(3):144-7, 1988), 1-(2'-deoxy-2'-fluoro-(3-D-arabinofuranosyl)-5-fluorouracil (Suzuko et al., Mol. Pharmacol. 37(3):301-6, 1987), doxifluridine (Matuura et al., Oyo Yakuri 29(5):803-31, 1985), 5'-deoxy-5-fluorouridine (Bollag & Hartmann, Eur. J. Cancer 76(4):427-32, 1980), 1-acetyl-3-O-toluyl-5-fluorouracil (Okada, Hiroshima J. Med. Sci. 28(1):49-66, 1979), 5-fluorouracil-m-formylbenzene-sulfonate (JP 55059173), N'-(2-furanidyl)-5=fluorouracil (JP
53149985) and 1-(2-tetrahydrofuryl)-5-fluorouracil (JP 52089680).
These compounds are believed to function as therapeutic agents by serving as antimetabolites of pyrimidine.
(C) Folic acid antagonists In another aspect, the therapeutic agent is a folic acid antagonist, such as methotrexate or derivatives or analogues thereof, including edatrexate, trimetrexate, raltitrexed, piritrexim, denopterin, tomudex, and pteropterin.
Methotrexate analogues have the following general structure:

The identity of the R group may be selected from organic groups, particularly those groups set forth in U.S. Patent Nos. 5,166,149 and 5,382,582. For example, R~ may be N, R2 may be N or C(CH3), R3 and R3' may H or alkyl, e.g., CH3, R4 may be a single bond or NR, where R is H or alkyl group. R5,6,$ may be H, OCH3, or alternately they can be halogens or hydro groups. R~ is a side chain of the general structure:
HO
wherein n = 1 for methotrexate, n = 3 for pteropterin. The carboxyl groups in the side chain may be esterified or form a salt such as a Zn2+ salt. R9 and Rio can be NH2 or may be alkyl substituted.

Exemplary folic acid antagonist compounds have the structures:
RT N N

R6 \ 'R2 Ra Ro R~
R$
Ha Ro R~ Rz R3 Ra R5 Rs R~ Rs MethotrexateNHz N N H N(CH3) H H A (n=1H
) EdatrexateNHz N N H CH(CHzCH3)H H A (n=1H
) TrimetrexateNHz CH C(CH3)H NH H OCH3 OCH3 OCH3 PteropterinOH N N H NH H H A (n=3)H

DenopterinOH N N CH3 N(CH3) H H A (n=1H
) PeritreximNHz N C(CH3)H single OCH3 H H OCH3 bond A: p NH
HO
O
O off HOOC~ O ~ H3 S N ~ ~ NH
HOOC N
O
Tomudex Other representative examples include 6-S-aminoacyloxymethyl mercaptopurine derivatives (Harada et al., Chem. Pharm. Bull. 43(10):793-6, 1995), 6-mercaptopurine (6-MP) (Kashida et al., Biol. Pharm. Bull. 78(11 ):1492-7, 1995), 7,8-polymethyleneimidazo-1,3,2-diazaphosphorines (Nilov et al., Mendeleev Commun. 2:67, 1995), azathioprine (Chifotides et al., J. Inorg.
Biochem. 56(4):249-64, 1994), methyl-D-glucopyranoside mercaptopurine derivatives (Da Silva et al., Eur. J. Med. Chem. 29(2):149-52, 1994) and s-alkynyl mercaptopurine derivatives (Ratsino et al., Khim.-Farm. Zh. 15(8):65-7, 1981 ); indoline ring and a modified ornithine or glutamic acid-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 45(7):1146-1150, 1997), alkyl-substituted benzene ring C bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 44(12):2287-2293, 1996), benzoxazine or benzothiazine moiety-bearing methotrexate derivatives (Matsuoka et al., J.
Med. Chem. 40(1):105-111, 1997), 10-deazaaminopterin analogues (DeGraw et al., J. Med. Chem. 40(3):370-376, 1997), 5-deazaaminopterin and 5,10-dideazaaminopterin methotrexate analogues (Piper et al., J. Med. Chem.
40(3):377-384, 1997), indoline moiety-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 44(7):1332-1337, 1996), lipophilic amide methotrexate derivatives (Pignatello et al., VIlorld Meet. Pharm. Biopharm.
Pharm. Technol., 563-4, 1995), L-threo-(2S,4S)-4-fluoroglutamic acid and DL-3,3-difluoroglutamic acid-containing methotrexate analogues (Hart et al., J.
Med. Chem. 39(1 ):56-65, 1996), methotrexate tetrahydroquinazoline analogue (Gangjee, et al., J. Heterocycl. Chem. 32(1 ):243-8, 1995), N-(a-aminoacyl) methotrexate derivatives (Cheung et al., Pteridines 3(1-2):101-2, 1992), biotin methotrexate derivatives (Fan et al., Pteridines 3(1-2):131-2, 1992), D-glutamic acid or D-erythrou, threo-4-fluoroglutamic acid methotrexate analogues (McGuire et al., Biochem. Pharmacol. 42(12):2400-3, 1991 ), ~3,y-methano methotrexate analogues (Rosowsky et al., Pteridines 2(3):133-9, 1991 ), 10-deazaaminopterin (10-EDAM) analogue (Braakhuis et al., Chem. Biol.
Pteridines, Proc. Int Symp. Pteridines Folic Acid Deriv., 1027-30, 1989), y-tetrazole methotrexate analogue (Kalman et al., Chem. Biol. Pteridines, Proc.
Int. Symp. Pteridines Folic Acid Deriv., 1154-7, 1989), N-(L-a-aminoacyl) methotrexate derivatives (Cheung et al., Heterocycles 28(2):751-8, 1989), meta and ortho isomers of aminopterin (Rosowsky et al., J. Med. Chem. 32(12):2582, 1989), hydroxymethylmethotrexate (DE 267495), y-fluoromethotrexate (McGuire et al., Cancer Res. 49(16):4517-25, 1989), polyglutamyl methotrexate derivatives (Kumar et al., Cancer Res. 46(10):5020-3, 1986), gem-diphosphonate methotrexate analogues (WO 88/06158), a- and y-substituted methotrexate analogues (Tsushima et al., Tetrahedr~n 44(17):5375-87, 1988), 5-methyl-5-deaza methotrexate analogues (4,725,687), N8-acyl-Na-(4-amino-4-deoxypteroyl)-L-ornithine derivatives (Rosowsky et al., J. Med. Chem.
37(7):1332-7, 1988), 8-deaza methotrexate analogues (Kuehl et al., Cancer Res. 48(6):1481-8, 1988), acivicin methotrexate analogue (Rosowsky et al., J.
Med: Chem. 30(8):1463-9, 1987), polymeric platinol methotrexate derivative (Carraher et al., Polym. Sci. Technol. (Plenum), 35(Adv. Biomed. Polym.):311-24, 1987), methotrexate-y-dimyristoylphophatidylethanolamine (Kinsky et al., Biochim. Biophys. Acta 917(2):211-18, 1987), methotrexate polyglutamate analogues (Rosowsky et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin.
Aspects:
985-8, 1986), poly-y-glutamyl methotrexate derivatives (Kisliuk et al., Chem.
Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 989-92, 1986), deoxyuridylate methotrexate derivatives (Webber et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin.
Aspects: 659-62, 1986), iodoacetyl lysine methotrexate analogue (Delcamp et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp.
Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 807-9, 1986), 2,.omega.-diaminoalkanoid acid-containing methotrexate analogues (McGuire et al., Biochem. Pharmacol. 35(15):2607-13, 1986), polyglutamate methotrexate derivatives (Kamen & Winick, Methods Enzymol. 122(Vitam.
Coenzymes, Pt. G):339-46, 1986), 5-methyl-5-deaza analogues (Piper et al., J.
Med. Chem. 29(6):1080-7, 1986), quinazoline methotrexate analogue (Mastropaolo et al., J. Med. Chem. 29(1 ):155-8, 1986), pyrazine methotrexate analogue (Lever & Vestal, J. Heterocycl. Chem. 22(1 ):5-6, 1985), cysteic acid and homocysteic acid methotrexate analogues (4,490,529), y-tert-butyl methotrexate esters (Rosowsky et al., J. Med. Chem. 28(5):660-7, 1985), fluorinated methotrexate analogues (Tsushima et al., Heterocycles 23(1 ):45-9, 1985), folate methotrexate analogue (Trombe, J. Bacteriol. 160(3):849-53, 1984), phosphonoglutamic acid analogues (Sturtz & Guillamot, Eur. J. Med.
Chem.--Chim. Ther. 19(3):267-73, 1984), poly (L-lysine) methotrexate conjugates (Rosowsky et al., J. Med. Chem. 27(7):888-93, 1984), dilysine and trilysine methotrexate derivates (Forsch & Rosowsky, J. Org. Chem.
49(7):1305-9, 1984), 7-hydroxymethotrexate (Fabre et al., Cancer Res.
43(10):4648-52, 1983), poly-y-glutamyl methotrexate analogues (Piper &
Montgomery, Adv. Exp. Med. Biol., 163(Folyl Antif~lyl Polyglutamates):95-100, 1983), 3',5'-dichloromethotrexate (Rosowsky & Yu, J. Med. Chem. 26(10):1448-52, 1983), diazoketone and chloromethylketone methotrexate analogues (Gangjee et al., J. Pharm. Sci. 71(6):717-19, 1982), 10-propargylaminopterin and alkyl methotrexate homologs (Piper et al., J. Med. Chem. 25(7):877-80, 1982), lectin derivatives of methotrexate (Lin et al., JNCI 66(3):523-8, 1981 ), polyglutamate methotrexate derivatives (Galivan, Mol. Pharmacol. 17(1 ):105-10, 1980), halogentated methotrexate derivatives (Fox, JNCI 58(4):J955-8, 1977), 8-alkyl-7,8-dihydro analogues (Chaykovsky et al., J. Med. Chem.
20(10):J1323-7, 1977), 7-methyl methotrexate derivatives and dichloromethotrexate (Rosowsky & Chen, J. Med. Chem. 17(12):J1308-11, 1974), lipophilic methotrexate derivatives and 3',5'-dichloromethotrexate (Rosowsky, J. Med. Chem. 16(10):J1190-3, 1973), deaza amethopterin analogues (Montgomery et al., Ann. N. Y. Acad. Sci. 186:J227-34, 1971), MX068 (Pharma Japan, 1658:18, 1999) and cysteic.acid and homocysteic acid methotrexate analogues (EPA 0142220);
These compounds are believed to act as antimetabolites of folic acid.
(D) Podophyllotoxins In another aspect, the therapeutic agent is a Podophyllotoxin, or a derivative or an analogue thereof. Exemplary compounds of this type are etoposide or teniposide, which have the following structures:
Etoposide CH3 Teniposide s OH
Other representative examples of podophyllotoxins include Cu(II)-VP-16 (etoposide) complex (Tawa et al., Bioorg. Med. Chem. 6(7):1003-1008, 1998), pyrrolecarboxamidino-bearing etoposide analogues (Ji et al., Bioorg.
Med. Chem. Lett. 7(5):607-612, 1997), 4a-amino etoposide analogues (Hu, University of North Carolina Dissertation, 1992), y-lactone ring-modified arylamino etoposide analogues (Zhou et al., J. Med. Chem. 37(2):287-92, 1994), N-glucosyl etoposide analogue (Allevi et al., Tetrahedron Lett.
34(45):7313-16, 1993), etoposide A-ring analogues (Kadow et al., Bioorg. Med.
Chem. Lett. 2(1 ):17-22, 1992), 4'-deshydroxy-4'-methyl etoposide (Saulnier et al., Bioorg. Med. Chem. Lett. 2(10):1213-18, 1992), pendulum ring etoposide analogues (Sinha et al., Eur. J. Cancer 26(5):590-3, 1990) and E-ring desoxy etoposide analogues (Saulnier et al., J. Med. Chem. 32(7):1418-20, 1989).
These compounds are believed to act as topoisomerase II
inhibitors and/or DNA cleaving agents.
(E) Camptothecins In another aspect, the therapeutic agent is camptothecin, or an analogue or derivative thereof. Camptothecins have the following general structure.
In this structure, X is typically O, but can be other groups, e.g., NH
in the case of 21-lactam derivatives. R1 is typically H or OH, but may be other groups, e.g., a terminally hydroxylated C1_3 alkane. R2 is typically H or an amino containing group such as (CH3)2NHCH2, but may be other groups e.g., N02, NH2, halogen (as disclosed in, e.g., U.S. Patent 5,552,156) or a short alkane containing these groups. R3 is typically H or a short alkyl such as C2H5.
R4 is typically H but may be other groups, e.g., a methylenedioxy group with R1, Exemplary camptothecin compounds include topotecan, irinotecan (CPT-11), 9-aminocamptothecin, 21-lactam-20(S)-camptothecin, 10,11-methylenedioxycamptothecin, SN-38, 9-nitrocamptothecin, 10-hydroxycamptothecin. Exemplary compounds have the structures:
R~ Rz R3 Camptothecin: H H H
Topotecan: OH (CH3)zNHCHz H
SN-38: OH H CZHS
x: O for most analogs, NH for 21-lactam analogs Camptothecins have the five rings shown here. The ring labeled E must be intact (the lactone rather than carboxylate form) for maximum activity and minimum toxicity.
Camptothecins are believed to function as topoisomerase I
inhibitors and/or DNA cleavage agents.
(F) Hydroxyureas The therapeutic agent of the present invention may be a hydroxyurea. Hydroxyureas have the following general structure:

~N N\
R2 R~
Suitable hydroxyureas are disclosed in, for example, U.S. Patent No. 6,080,874, wherein R~ is:
OJ '% -~:
and R2 is an alkyl group having 1-4 carbons and R3 is one of H, acyl, methyl, ethyl, and mixtures thereof, such as a methylether.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,665,768, wherein R~ is a cycloalkenyl group, for exz;mple N-[3-[5-(4-fluorophenylthio)-furyl]-2-cyclopenten-1-yIJN-hydroxyurea; R2 is H or an alkyl group having 1 to 4 carbons and R3 is H; X is H or a cation.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 4,299,778, wherein R~ is a phenyl group substituted with one or more fluorine atoms; R2 is a cyclopropyl group; and R3 and X is H.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,066,658, wherein R2 and R3 together with the adjacent nitrogen form:
cc~2)~
Y . N-(C a)m wherein m is 1 or 2, n is 0-2 and Y is an alkyl group.
In one aspect, the hydroxyurea has the structure:
O
,OH

Hydroxyurea These compounds are thought to function by inhibiting DNA
synthesis.
(G) Platinum complexes In another aspect, the therapeutic agent is a platinum compound.
In general, suitable platinum complexes may be of Pt(II) or Pt(IV) and have this basic structure:
z1 R1 x ~~t~
R~ ~Y

wherein X and Y are anionic leaving groups such as sulfate, phosphate, carboxylate, and halogen; R1 and R2 are alkyl, amine, amino alkyl any may be further substituted, and are basically inert or bridging groups. For Pt(II) complexes Z1 and Z2 are non-existent. For Pt(IV) Z1 and Z2 may be anionic groups such as halogen, hydroxy, carboxylate, ester, sulfate or phosphate.
See, e.g., U.S. Patent Nos. 4,588,831 and 4,250,189.
Suitable platinum complexes may contain multiple Pt atoms. See, e.g., U.S. Patent Nos. 5,409,915 and 5,380,897. For example bisplatinum and triplatinum complexes of the type:

R X R
X\Pt/ I \ ~t~ z y~ I \A~ I \Y
Zz Zz ZI ZI ZI
X\ I / RI X\ /A / X
Pt It/ \ It Y~ I ~ A~ I \ Y Rz~ ~ \ Y
Zz Zz Zz ZI ZI
X~ I ~ R2 Rz\ I / X
y/ ~t~A~ It\Y
Zz ~ Zz Zz\Pt/ R3 Y/I\ZI
X
Exemplary platinum compounds are cisplatin, carboplatin, oxaliplatin, and miboplatin having the structures:

NH3 O O~
Pt CIIt-NH3 I ~NH3 O
CI
O
Cisplatin Carboplatin p H H
O \/
O N
O NHz \ / Pt Pt / '>. H
O \NH ~~~'\ O ~N
z O H
O
CJ Oxaliplatin Miboplatin Other representative platinum compounds include (CPA)2Pt[DOLYM] and (DACH)Pt[DOLYM] cisplatin (Choi et al., Arch.
Pharmacal Res. 22(2):151-156, 1999), Cis-[PtCl2(4,7-H-5-methyl-7-oxo]1,2,4[triazolo[1,5-a]pyrimidine)2] (Navarro et al., J. Med. Chem.
41(3):332-338, 1998), [Pt(cis-1,4-DACH)(trans-CI2)(CBDCA)] ~'/ZMeOH cisplatin (Shamsuddin et al., Inorg. Cherri. 36(25):5969-5971, 1997), 4-pyridoxate diammine hydroxy platinum (Tokunaga et al., Pharm. Sci. 3(7):353-356, 1997), Pt(II) ... Pt(II) (Pty[NHCHN(C(CH2)(CH3))]4) (Navarro et al., Inorg. Chem.
35(26):7829-7835, 1996), 254-S cisplatin analogue (Koga et al., Neurol. Res.
18(3):244-247, 1996), o-phenylenediamine ligand bearing cisplatin analogues (Koeckerbauer & Bednarski, J. Inorg. Biochem. 62(4):281-298, 1996), trans, cis-[Pt(OAc)21~(en)] (Kratochwil et al., J. Med. Chem. 39(13):2499-2507, 1996), estrogenic 1,2-diarylethylenediamine ligand (with sulfur-containing amino acids and glutathione) bearing cisplatin analogues (Bednarski, J. Inorg. Biochem.
62(1):75, 1996), cis-1,4-diaminocyclohexane cisplatin analogues (Shamsuddin et al., J. Inorg. Biochem. 61 (4):291-301, 1996), 5' orientational isomer of cis-[Pt(NH3)(4-aminoTEMP-O){d(GpG)}] (Dunham & Lippard, J. Am. Chem. Soc.
117(43):10702-12, 1995), chelating diamine-bearing cisplatin analogues (Koeckerbauer& Bednarski, J. Pharm. Sci. 84(7):819-23, 1995), 1,2-diarylethyleneamine ligand-bearing cisplatin analogues (Otto et al., J. Cancer Res. Clin. Oncol. 121(1 ):31-8, 1995), (ethylenediamine)platinum(II) complexes (Pasini et al., J. Chem. Soc., Dalton Trans. 4:579-85, 1995), CI-973 cisplatin analogue (Yang et al., Int. J. Oncol. 5(3):597-602, 1994), cis-diaminedichloroplatinum(II) and its analogues cis-1,1-cyclobutanedicarbosylato(2R)-2-methyl-1,4-butanediamineplatinum(II) and cis-diammine(glycolato)platinum (Claycamp & Zimbrick, J. Inorg. Biochem.
26(4):257-67, 1986; Fan et al., Cancer Res. 48(11 ):3135-9, 1988; Heiger-Bernays et al., Biochemistry 29(36):8461-6, 1990; Kikkawa et al., J. Exp.
Clin.
Cancer Res. 12(4):233-40, 1993; Murray et al., Biochemistry 31 (47):11812-17, 1992; Takahashi et al., Cancer Chemother. Pharmacol. 33(1 ):31-5, 1993), cis-amine-cyclohexylari~ine-dichloroplatinum(II) (Yoshida et al., Biochem.
Pharmacol. 48(4):793-9, 1994), gem-diphosphonate cisplatin analogues (FR
2683529), (meso-1,2-bis(2,6-dichloro-4-hydroxyplenyl)ethylenediamine) dichloroplatinum(II) (Bednarski et al., J. Med. Chem. 35(23):4479-85, 1992), cisplatin analogues containing a tethered dansyl group (Hartwig et al., J. Am.
Chem. Soc. 114(21 ):8292-3, 1992), platinum(II) polyamines (Siegmann et al., Inorg. Met.-Containing Polym. Mater., (Proc. Am. Chem. Soc. Int. Symp.), 335-61, 1990), cis-(3H)dichloro(ethylenediamine)platinum(II) (Eastman, Anal.
Biochem. 197(2):311-15, 1991), trans-diamminedichloroplatinum(II) and cis-(Pt(NH3)2(N3-cytosine)CI) (Bellon & Lippard, Biophys. Chem. 35(2-3):179-88, 1990), 3H-cis-1,2-diaminocyclohexanedichloroplatinum(II) and 3H-cis-1,2-diaminocyclohexanemalonatoplatinum (II) (Oswald et al., Res. Commun. Chem.
Pathol. Pharmacol. 64(1 ):41-58, 1989), diaminocarboxylatoplatinum (EPA
296321 ), trans-(D,1 )-1,2-diaminocyclohexane carrier ligand-bearing platinum analogues (Wyrick & Chaney, J. Labelled Compd. Radiopharm. 25(4):349-57, 1988), aminoalkylaminoanthraquinone-derived cisplatin analogues (Kitov et al., Eur. J. Med. Chem. 23(4):381-3, 1988), spiroplatin, carboplatin, iproplatin and JM40 platinum analogues (Schroyen et al., Eur. J. Cancer Clin. Oneol.
24(8):1309-12, 1988), bidentate tertiary diamine-containing cisplatinum derivatives (Orbell et al., Inorg. Chim. Acta 752(2):125-34, 1988), platinum(II), platinum(IV) (Liu & Wang, Shandong Yike Daxue Xuebao 24(1 ):35-41, 1986), cis-diammine(1,1-cyclobutanedicarboxylato-)platinum(II) (carboplatin, JM8) and ethylenediammine-malonatoplatinum(II) (JM40) (Begg et al., Radiother. Oncol.
9(2):157-65, 1987), JM8 and JM9 cisplatin analogues (Harstrick et al., Int. J.
Androl. 70(1 ); 139-45, 1987), (NPr4)2((PtCL4).cis-(PtCl2-(NH2Me)2)) (Brammer et al., J. Chem. Soc., Chem. Commun. 6:443-5, 1987), aliphatic tricarboxylic acid platinum complexes (EPA 185225), and cis-dichloro(amino acid)(tert-butylamine)platinum(II) complexes (Pasini & Bersanetti, Inorg.
Chim.
Acta 707(4):259-67, 1985). These compounds are thought to function by binding to DNA, i.e., acting as alkylating agents of DNA.
As medical implants are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area, design and portions of the implant coated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Regardless of the method of application of the drug to the cardiac implant, the preferred anticancer agents, used alone or in combination, should be administered under the following dosing guidelines:
(a) Anthracyclines. Utilizing the anthracycline doxorubicin as an example, whether applied as a polymer coating, incorporated into the polymers which make up the implant components, or applied without a carrier polymer, the total dose of doxorubicin applied to the implant should not exceed 25 mg (range of 0.1 p,g to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 ~.g to 5 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied andlor incorporated) should fall within the range of 0.01 ~g - 100 g,g per mm2 of surface area. In a particularly preferred embodiment, doxorubicin should be applied to the implant surface at a dose of 0.1 p.g/mm2 -10 pg/mm2. As different polymer and non-polymer coatings will release doxorubicin at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10-x- 10~ M of doxorubicin is maintained on the surface. It is necessary to insure that surface drug concentrations exceed concentrations of doxorubicin known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10-4 M;
although for some embodiments lower concentrations are sufficient). In a preferred embodiment, doxorubicin is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of doxorubicin (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as doxorubicin is administered at half the above parameters, a compound half as potent as doxorubicin is administered at twice the above parameters, etc.).
Utilizing mitoxantrone as another example of an anthracycline, whether applied as a polymer coating, incorporated into the polymers which make up the implant, or applied without a carrier polymer, the total dose of mitoxantrone applied should not exceed 5 mg (range of 0.01 p,g to 5 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 0.1 p.g to 1 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 ~g - 20 p,g per mm2 of surface area. In a particularly preferred embodiment, mitoxantrone should be applied to the implant surface at a dose of 0.05 p,g/mm2 - 3 p,g/mm2.
As different polymer and non-polymer coatings will release mitoxantrone at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10-5 - 10-6 M of mitoxantrone is maintained. It is necessary to insure that drug concentrations on the implant surface exceed concentrations of mitoxantrone known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10-5 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, mitoxantrone is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of mitoxantrone (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as mitoxantrone is administered at half the above parameters, a compound half as potent as mitoxantrone is administered at twice the above parameters, etc.).
(b) Fluoropyrimidines Utilizing the fluoropyrimidine 5-fluorouracil as an example, whether applied as a polymer coating, incorporated into the polymers which make up the implant, or applied without a carrier polymer, the total dose of 5-fluorouracil applied should not exceed 250 mg (range of 1.0 p.g to 250 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 10 ~g to 25 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.1 ~,g - 1 mg per mm2 of surface area. In a particularly preferred embodiment, 5-fluorouracil should be applied to the implant surface at a dose of 1.0 p,g/mm2 - 50 ~.g/mm2. As different polymer and non-polymer coatings will release 5-fluorouracil at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10-4- 10-~ M of 5-fluorouracil is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of 5-fluorouracil known to be lethal to numerous species of bacteria and fungi (i.e., are in excess of 10-4 M;
although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, 5-fluorouracil is released from the implant surface such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of 5-fluorouracil (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as 5-fluorouracil is administered at half the above parameters, a compound half as potent as 5-fluorouracil is administered at twice the above parameters, etc.).
(c) Podophylotoxins Utilizing the podophylotoxin etoposide as an example, whether applied as a polymer coating, incorporated into the polymers which make up the cardiac implant, or applied without a carrier polymer, the total dose of etoposide applied should not exceed 25 mg (range of 0.1 ~g to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 pg to 5 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 ~g - 100 ~,g per mm2 of surface area. In a particularly preferred embodiment, etoposide should be applied to the implant surface at a dose of 0.1 ~g/mm2 - 10 ~,g/mm2. As different polymer and non-polymer coatings will release etoposide at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a concentration of 10-5 - 10-6 M of etoposide is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of etoposide known to be lethal to a variety of bacteria and fungi (i.e., are in excess of 10-5 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, etoposide is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of etoposide (as described previously) with similar functional activity can be utilized for the purposes of this invention;
the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as etoposide is administered at half the above parameters, a compound half as potent as etoposide is administered at twice the above parameters, etc.).
(d) Combination therapy. It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate and/or podophylotoxins (e.g., etoposide) can be utilized to enhance the antibacterial activity of the implant coating.
Similarly anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate and/or podophylotoxins (e.g., etoposide) can be combined with traditional antibiotic and/or antifungal agents to enhance efficacy. The anti-infective agent may be further combined with anti-thrombotic and/or antiplatelet agents (for example, heparin, dextran sulphate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, aspirin, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, ticlopidine, clopidogrel, abcixamab, eptifibatide, tirofiban, streptokinase, and/or tissue plasminogen activator) to enhance efficacy. In certain embodiments, the fibrosis-inhibiting agent is combined with an agent that can modify metabolism of the agent in vivo to enhance efficacy of the fibrosis-inhibiting agent. One class of therapeutic agents that can be used to alter drug metabolism includes agents capable of inhibiting oxidation of the anti-scarring agent by cytochrome P450 (CYP). In one embodiment, compositions are provided that include a fibrosis-inhibiting agent (e.g., paclitaxel, rapamycin, everolimus) and a CYP inhibitor, which may be combined (e.g., coated) with any of the devices described herein, including, without limitation, stents, grafts, patches, valves, wraps, and films. Representative examples of CYP inhibitors include flavones, azole antifungals, macrolide antibiotics, HIV protease inhibitors, and anti-sense oligomers. Devices comprising a combination of a fibrosis-inhibiting agent and a CYP inhibitor may be used to treat a variety of proliferative conditions that can lead to undesired scarring of tissue, including intimal hyperplasia, surgical adhesions, and tumor growth.
Although the above therapeutic agents have been provided for the purposes of illustration, it should be understood that the present invention is not so limited. For example, although agents are specifically referred to above, the present invention should be understood to include analogues, derivatives and conjugates of such agents. As an illustration, paclitaxel should be understood to refer to not only the common chemically available form of paciitaxel, but analogues (e.g., taxotere, as noted above) and paclitaxel conjugates (e.g_, paclitaxel-PEG, paclitaxel-dextran, or paclitaxel-xylos). In addition, to the individual compounds listed above, specif agents that are covalently bound to each other or to another of the described therapeutic agents can also be used for the applications described below. In addition, as will be evident to one of skill in the art, although the agents set forth above may be noted within the context of one class, many of the agents listed in fact have multiple biological activities. Further, more than one therapeutic agent may be utilized at a time (i.e., in combination), or delivered sequentially.
C. Methods for Generating Medical Devices Which Include or Release a Fibrosis-Inhibiting Agent In the practice of this invention, drug-coated or drug-impregnated implants and medical devices are provided which inhibit fibrosis in and around the device, or prevent "stenosis" of the device/implant in situ, thus enhancing the efficacy. Within various embodiments, fibrosis is inhibited by local or systemic release of specific pharmacological agents that become localized to the tissue adjacent to the device or implant. There are numerous methods available for optimizing delivery of the fibrosis-inhibiting agent to the site of the intervention and several of these are described below.
1 ) Devices and Implants That Include or Release Fibrosis-Inhibiting Agents Medical devices or implants of the present invention are coated with, or otherwise adapted to release an agent which inhibits fibrosis on the surface of, or around, the medical device or implant. Medical devices or implants may be adapted to release a fibrosis-inhibiting agent by (a) directly affixing to the implant or device a desired therapeutic agent or composition containing the therapeutic agent (e.g., by either spraying or electrospraying the medical implant with a drug and/or carrier (polymeric or non-polymeric)-drug composition to create a film and/or coating on all, or parts of the internal or external surface of the device; by dipping the implant or device into a drug and/or carrier (polymeric or non-polymeric)-drug solution to coat all or parts of the device or implant; or by other covalent or noncovalent attachment of the therapeutic agent to the device or implant surface); (b) by coating the medical device or implant with a substance such as a hydrogel which either contains or which will in turn absorb the desired fibrosis-inhibiting agent or composition; (c) by interweaving a "thread" composed of, or coated with, the fibrosis-inhibiting agent into the medical implant or device (e.g., a polymeric strand composed of materials that inhibit fibrosis (e.g., paclitaxel, mitoxantrone, doxorubicin, epithilone B, etoposide, TAXOTERE, Tubercidin, vinblastine, geldanamycin, simvastatin, halifuginone, sirolimus, everolimus, mycophenolic acid, 1-alpha-dihydroxy vitamin D3, Bay 11-7082, SB202190, sulconizole polymerized drug compositions) or polymers which release a fibrosis-inhibiting agent from the ' thread); (d) by covering all, or portions of the device or implant with a sleeve, cover, electrospun fabric or mesh containing a fibrosis-inhibiting agent (i.e., a covering comprised of a fibrosis-inhibiting agent - paclitaxel, mitoxantrone, doxorubicin, epithilone B, etoposide, TAXOTERE, tubercidin, vinblastine, geldanamycin, simvastatin, halifuginone, sirolimus, everolimus, mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3, Bay 11-7082, SB202190, sulconizole or polymerized compositions containing fibrosis-inhibiting agents); (e) constructing all, or parts of the device or implant itself with the desired agent or composition (e.g., paclitaxel, mitoxantrone, doxorubicin, epithilone B, etoposide, TAXOTERE, tubercidin, vinblastine, geldanamycin, simvastatin, halifuginone, sirolimus, everolimus, mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3, Bay 11-7082, SB202190, sulconizole or polymerized compositions of fibrosis-inhibiting agents); (f) otherwise impregnating the device or implant with the desired fibrosis-inhibiting agent or composition; (g) composing all, or parts, of the device or implant from metal alloys that inhibit fibrosis; (h) constructing all, or parts of the device or implant itself from a degradable or non-degradable polymer that releases one or more fibrosis-inhibiting agents; (i) utilizing specialized multi-drug releasing medical device systems (for example, U.S.
Patent. Nos. 6,527,799; '6,293,967; 6,290,673; 6241762, U.S. Application Publication Nos. 2003/0199970A1 and 2003/0167085A1, and PCT Publication WO 03/015664) to deliver fibrosis-inhibiting agents alone or in combination.
2) Systemic, Regional and Local Delivery of Fibrosis-Inhibiting Agents A variety of drug-delivery technologies are available for systemic, regional and local delivery of therapeutic agents. Several of these techniques can be suitable to achieve preferentially elevated levels of fibrosis-inhibiting agents in the vicinity of the medical device or implant, including: (a) using drug-delivery catheters for local, regional or systemic delivery of fibrosis inhibiting agents to the tissue surrounding the device or implant (typically, drug delivery catheters are advanced through the circulation or inserted directly into tissues under radiological guidance until they reach the desired anatomical location;
the fibrosis inhibiting agent can then be released from the catheter lumen in high local concentrations in order to deliver therapeutic doses of the drug to the tissue surrounding the device or implant); (b) drug localization techniques such as magnetic, ultrasonic or MRI-guided drug delivery; (c) chemical modification of the fibrosis-inhibiting drug or formulation designed to increase uptake of the agent into damaged tissues (e.g., antibodies directed against damaged or healing tissue components such as macrophages, neutrophils, smooth muscle cells, fibroblasts, extracellular matrix components, neovascular tissue); (d) chemical modification of the fibrosis-inhibiting drug or formulation designed to localize the drug to areas of bleeding or disrupted vasculature; and/or (e) direct injection of the fibrosis-inhibiting agent, for example, under endoscopic vision.
3) Infiltration of Fibrosis-Inhibiting Agents into the Tissue Surrounding a Device or Implant Alternatively, the tissue cavity into which the device or implant is placed can be treated with a fibrosis-inhibiting agent prior to, during, or after the procedure. This can be accomplished in several ways including: (a) topical application of the fibrosis-inhibiting agent into the anatomical space where the device will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the anti-fibrosing agent over a period ranging from several hours to several weeks. Compositions that can be used for this application include, e.g., fluids, microspheres, pastes, gels, hydrogels, crosslinked gels, microparticulates, sprays, aerosols, solid implants and other formulations which release a fibrosis inhibiting agent into the region where the device or implant will be implanted); (b) microparticulate forms of the therapeutic agent are also useful for directed delivery into the implantation site;
(c) sprayable collagen-containing formulations such as COSTASIS (from Angiotech Pharmaceuticals, Inc., Canada), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the implant/device surface); (d) sprayable PEG-containing formulations such as COSEAL
(Angiotech Pharmaceuticals, Inc.), SPRAYGEL or DURASEAL (both from Confluent Surgical, Inc., Boston, MA), FOCALSEAL (Genzyme Corporation, Cambridge, MA), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the implant/device surface); (e) fibrin-containing formulations such as FLOSEAL or TISSEEL (both from Baxter Healthcare Corporation, Fremont, CA), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the implant/device surface); (f) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE
(both from Q-Med AB, Sweden), HYLAFORM (Inamed Corporation (Santa Barbara, CA)), SYNVISC (Biomatrix, Inc., Ridgefield, NJ), SEPRAFILM or SEPRACOAT (both from Genzyme Corporation, Cambridge, MA), INTERGEL
(Lifecore Biomedical) loaded with a fibrosis-inhibiting agent applied to the implantation site (or the implant/device surface); (g) polymeric gels for surgical implantation such as REPEL (Life Medical Sciences, Inc., Princeton, NJ) or FLOGEL (Baxter Healthcare Corporation) loaded with a fibrosis-inhibiting agent applied to the implantation site (or the implant/device surface); (h) orthopedic "cements" used to hold prostheses and tissues in place with a fibrosis-inhibiting agent applied to the implantation site (or the implant/device surface); (i) surgical adhesives containing cyanoacrylates such as DERMABOND (Johnson &
Johnson, Inc., New Brunswick, NJ), INDERMIL (U.S. Surgical Company, Norwalk, CT), GLUSTITCH (Blacklock Medical Products Inc., Canada), TISSUMEND II (Veterniary Products Laboratories" Phoenix, AZ), VETBOND
(3M Company, St. Paul, MN), HISTOACRYL BLUE (Davis & Geck, St. Louis, MO) and ORABASE SMOOTHE-N-SEAL Liquid Protectant (Colgate-Palmolive Company, New York, NY), either alone, or loaded with a fibrosis-inhibiting agent, applied to the implantation site (or the implant/device surface); (k) surgical implants containing hydroxyapatite, calcium sulfate, tricalcium phosphate, demineralized bone loaded with a fibrosis-inhibiting agent applied to the implantation site (or the implant/device surface);
4) Sustained-Release Preparations of Fibrosis-Inhibiting Agents As described previously desired fibrosis-inhibiting agents may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymer composition (which may be either biodegradable or non-biodegradable) or a non-polymeric composition in order to release the therapeutic agent over a prolonged period of time. For many of the aforementioned embodiments, localized delivery as well as localized sustained delivery of the fibrosis inhibiting agent may be required. For example, a desired fibrosis-inhibiting agent may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymeric composition (which may be either biodegradable or non-biodegradable) or non-polymeric composition in order to release the fibrosis-inhibiting agent over a period of time.
Representative examples of biodegradable polymers suitable for the delivery of fibrosis-inhibiting agents include albumin, collagen, gelatin, hyaluronic acid, starch, cellulose and cellulose derivatives (e.g., methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, hydroxypropylmethylcellulose phthalate), casein, dextrans, polysaccharides, fibrinogen, poly(ether ester) multiblock copolymers, based on polyethylene glycol) and poly(butylene terephthalate), tyrosine-derived polycarbonates (e.g., U.S. Patent No. 6,120,491 ), poly(hydroxyl acids), poly(D,L-lactide), poly(D,L-lactide-co-glycolide), poly(glycolide), poly(hydroxybutyrate), polydioxanone, poly(alkylcarbonate) and poly(orthoesters), polyesters, poly(hydroxyvaleric acid), polydioxanone, degradable polyesters, poly(malic acid), poly(tartronic acid), poly(acrylamides), polyanhydrides, polyphosphazenes, poly(amino acids), poly(alkylene oxide)-poly(ester) block copolymers (e.g., X-Y, X-Y-X or Y-X-Y, R-(Y-X)", R-(X-Y)"
where X is a polyalkylene oxide and Y is a polyester (e.g., polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, ~-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y-decanolactone, ~-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-2one.), R is a multifunctional initiator and copolymers as well as blends thereof) and the copolymers as well as blends thereof (see generally, Illum, L., Davids, S.S.
(eds.) "Polymers in Controlled Drug Delivery" Wright, Bristol, 1987; Arshady, J.
Controlled Release 77:1-22, 1991; Pitt, Int. J. Phar. 59:173-196, 1990;
Holland et al., J. Controlled Release 4:155-0180, 1986).
Representative examples of non-degradable polymers suitable for the delivery of fibrosis-inhibiting agents include polyethylene-co-vinyl acetate) ("EVA") copolymers, non-degradable polyesters, such as polyethylene terephthalate), silicone rubber, acrylic polymers (polyacrylate, polyacrylic acid, polymethylacrylic acid, polymethylmethacrylate, poly(butyl methacrylate)), poly(alkylcynoacrylate) (e.g., poly(ethylcyanoacrylate), poly(butylcyanoacrylate) pbly(hexylcyanoacrylate) poly(octylcyanoacrylate)), acrylic resin, polyethylene, polypropylene, polyamides (nylon 6,6), polyurethanes (e.g., CHRONOFLEX
AR, CHRONOFLEX AL, BIONATE, and PELLETHANE), polyester urethanes), poly(ether urethanes), polyester-urea), cellulose esters (e.g., nitrocellulose), polyethers (poly(ethylene oxide), polypropylene oxide), polyoxyalkylene ether block copolymers based on ethylene oxide and propylene oxide such as the PLURONIC polymers (e.g., F-127 or F87) from BASF Corporation (Mount Olive, NJ), and poly(tetramethylene glycol), styrene-based polymers (polystyrene, polystyrene sulfonic acid), poly(styrene)-block-poly(isobutylene)-block-poly(styrene), poly(styrene)-poly(isoprene) block copolymers), and vinyl polymers (polyvinylpyrrolidone, polyvinyl alcohol), polyvinyl acetate phthalate) as well as copolymers and blends thereof. Polymers may also be developed which are either anionic (e.g., alginate, carrageenan, carboxymethyl cellulose, poly(acrylamido-2-methyl propane sulfonic acid) and copolymers thereof, poly(methacrylic acid and copolymers thereof and poly(acrylic acid) and copolymers thereof, as well as blends thereof, or cationic (e.g., chitosan, poly-L-lysine, polyethylenimine, and poly(allyl amine)) and blends, copolymers and branched polymers thereof (see generally, Dunn et al., J. Applied Polymer Sci.
50:353-365, 1993; Cascone et al., J. Materials Sci.: Materials in Medicine 5:770-774, 1994; Shiraishi et al., Biol. Pharm. Bull. 16(11 ):1164-1168, 1993;
Thacharodi and Rao, Int'I J. Pharm. 120:115-118, 1995; Miyazaki et al., Int'I
J.
Pharm. 118:257-263, 1995).
Particularly preferred polymeric carriers include polyethylene-co-vinyl acetate), polyurethanes (e.g., CHRONOFLEX AR, CHRONOFLEX AL, BIONATE, and PELLETHANE), poly (D,L-lactic acid) oligomers and polymers, poly (L-lactic acid) oligomers and polymers, poly (glycolic acid), copolymers of lactic acid and glycolic acid, poly (caprolactone), poly (valerolactone), polyanhydrides, copolymers of poly (caprolactone) or poly (lactic acid) with a polyethylene glycol (e.g., MePEG), poly(alkylene oxide)-polyester) block copolymers (e.g., X-Y, X-Y-X or Y-X-Y, R-(Y-X)", R-(X-Y)~ where X is a polyalkylene oxide and Y is a polyester (e.g., polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y-decanolactone, ~-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-Zone.), R is a multifunctional initiator and copolymers as well as blends thereof), nitrocellulose, silicone rubbers, poly(styrene)block-poly(isobutylene)-block-poly(styrene), poly(acrylate) polymers and blends, admixtures, or co-polymers of any of the above. Other preferred polymers include collagen, poly(alkylene oxide)-based polymers, polysaccharides such as hyaluronic acid, chitosan and fucans, and copolymers of polysaccharides with degradable polymers, as well as blends thereof.
Other representative polymers capable of sustained localized delivery of fibrosis-inhibiting agents include carboxylic polymers, polyacetates, polycarbonates, polyethers, polyethylenes, polyvinylbutyrals, polysilanes, polyureas, polyoxides, polystyrenes, polysulfides, polysulfones, polysulfonides, polyvinylhalides, pyrrolidones, rubbers, thermal-setting polymers, cross-linkable acrylic and methacrylic polymers, ethylene acrylic acid copolymers, styrene acrylic copolymers, vinyl acetate polymers and copolymers, vinyl acetal polymers and copolymers, epoxies, melamines, other amino resins, phenolic polymers, and copolymers thereof, water-insoluble cellulose ester polymers (including cellulose acetate propionate, cellulose acetate, cellulose acetate butyrate, cellulose nitrate, cellulose acetate phthalate, and mixtures thereof), polyvinylpyrrolidone, polyethylene glycols, polyethylene oxide, polyvinyl alcohol, polyethers, polysaccharides, hydrophilic polyurethane, polyhydroxyacrylate, dextran, xanthan, hydroxypropyl cellulose, and homopolymers and copolymers of N-vinylpyrrolidone, N-vinyllactam, N-vinyl butyrolactam, N-vinyl caprolactam, other vinyl compounds having polar pendant groups, acrylate and methacrylate having hydrophilic esterifying groups, hydroxyacrylate, and acrylic acid, and combinations thereof; cellulose esters and ethers, ethyl cellulose, hydroxyethyl cellulose, cellulose nitrate, cellulose acetate, cellulose acetate butyrate, cellulose acetate propionate, natural and synthetic elastomers, rubber, acetal, styrene polybutadiene, acrylic resin, polyvinylidene chloride, polycarbonate, homopolymers and copolymers of vinyl compounds, polyvinylchloride, and polyvinylchloride acetate.
Representative examples of patents relating to drug-delivery polymers and the preparation include PCT Publication Nos. WO 98/19713, WO
01/17575, WO 01/41821, WO 01/41822, and WO 01/15526 (as well as the corresponding U.S. applications); U.S. Patent Nos. 4,500,676, 4,582,865, 4,629,623, 4,636,524, 4,713,448, 4,795,741, 4,913,743, 5,069,899, 5,099,013, 5,128,326, 5,143,724, 5,153,174, 5,246,698, 5,266,563, 5,399,351, 5,525,348, 5,800,412, 5,837,226, 5,942,555, 5,997,517, 6,007,833, 6,071,447, 6,090,995, 6,106,473, 6,110,483, 6,121,027, 6,156,345, 6,214,901, 6,368,611 6,630,155, 6,528,080, RE37,950, 6,46,1631, 6,143,314, 5,990,194, 5,792,469, 5,780,044, 5,759,563, 5,744,153, 5,739,176, 5,733,950, 5,681,873, 5,599,552, 5,340,849, 5,278,202, 5,278,201, 6,589,549, 6,287,588, 6,201,072, 6,117,949, 6,004,573, 5,702,717, 6,413,539, 5,714,159, 5,612,052; and U.S. Patent Application Publication Nos. 2003/0068377, 2002/0192286, 2002/0076441, and 2002/0090398.
In one embodiment, all or a portion of the device is coated with a primer (bonding) layer and a drug release layer, as described in U.S. Patent application entitled, "Stent with Medicated Multi-Layer Hybrid Polymer Coating,"
filed September 16, 2003 (U.S. Serial No. 10/662,877).
In order to develop a hybrid polymer delivery system for targeted therapy, it is desirable to be able to control and manipulate the properties of the system both in terms of physical and drug release characteristics. The active agents can be imbibed into a surface hybrid polymer layer, or incorporated directly into the hybrid polymer coating solutions. Imbibing drugs into surface polymer layers is an efficient method for evaluating polymer-drug performance in the laboratory, but for commercial production it may be preferred for the polymer and drug to be premixed in the casting mixture. Greater efficacy can be achieved by combining the two elements in the coating mixtures in order to control the ratio of active agent to polymer in the coatings. Such ratios are important parameters to the final properties of the medicated layers, i.e., they allow for better control of active agent concentration and duration of pharmacological activity.
Typical polymers used in the drug-release system can include water-insoluble cellulose esters, various polyurethane polymers including hydrophilic and hydrophobic versions, hydrophilic polymers such as polyethylene glycol (PEG), polyethylene oxide (PEO), polyvinylpyrrolidone (PVP), PVP copolymers such as vinyl acetate, hydroxyethyl methacrylate (HEMA) and copolymers such as methylmethacrylate (PMMA-HEMA), and other hydrophilic and hydrophobic acrylate polymers and copolymers containing functional groups such as carboxyl and/or hydroxyl.
Cellulose esters such as cellulose acetate, cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, and cellulose nitrate may be used. In one aspect of the invention, the therapeutic agent is formulated with a cellulose ester. Cellulose nitrate is a preferred cellulose ester because of its compatibility with the active agents and its ability to impart non-tackiness and cohesiveness to the coatings. Cellulose nitrate has been shown to stabilize entrapped drugs in ambient and processing conditions.
Various grades of cellulose nitrate are available and may be used in a coating on a device, including cellulose nitrate having a nitrogen content = 11.8-12.2%.
Various viscosity grades, including 3.5, 0.5 or 0.25 seconds, may be used in order to provide proper rheological properties when combined with the coating solids used in these formulations. Higher or lower viscosity grades can be used. However, the higher viscosity grades can be more difficult to use because of their higher viscosities. Thus, the lower viscosity grades, such as 3.5, 0.5 or 0.25 seconds, are generally preferred. Physical properties such as tensile strength, elongation, flexibility, and softening point are related to viscosity (molecular weight) and can decrease with the lower molecular weight species, especially below the 0.25 second grades.
The cellulose derivatives comprise hydroglucose structures.
Cellulose nitrate is a hydrophobic, water-insoluble polymer, and has high water resistance properties. This structure leads to high compatibility with many active agents, accounting for the high degree of stabilization provided to drugs entrapped in cellulose nitrate. The structure of nitrocellulose is given below:
~01~
~..
I~.O~~SY irr~onR
~r~'OF3, GJ-1~0 E~ y R~Mt3~ qr ~E
nit~~o~ellu3ose Cellulose nitrate is a hard, relatively inflexible polymer, and has limited adhesion to many polymers that are typically used to make medical devices. Also, control of drug elution dynamics is limited if only one polymer is used in the binding matrix. Accordingly, in one embodiment of the invention, the therapeutic agent is formulated with two or more polymers before being associated with the device. In one aspect, the agent is formulated with both polyurethane ((e.g., CHRONOFLEX AR, CHRONOFLEX AL, and BIONATE, PELLETHANE) and cellulose nitrate to provide a hybrid polymer drug loaded matrix. Polyurethanes provide the hybrid polymer matrix with greater flexibility and adhesion to the device, particularly when the device has been pre-coated with a primer. Polyurethanes can also be used to slow or hasten the drug elution from coatings. Aliphatic, aromatic, polytetramethylene ether glycol, and polycarbonate are among the types of polyurethanes, which can be used in the coatings. In one aspect, an anti-scarring agent (e.g., paclitaxel) may be incorporated into a carrier that includes a polyurethane and a cellulose derivative. A heparin complex, such as benzalkonium heparinate or tridodecylammonium heparinate), may optionally be included in the formulation.
From the structure below, it is possible to see how more or less hydrophilic polyurethane polymers may be created based on the number of hydrophilic groups contained in the polymer structures. In one aspect of the invention, the device is associated with a formulation that includes therapeutic agent, cellulose ester, and a polyurethane that is water-insoluble, flexible, and compatible with the cellulose ester.
O O
n po~y~r~et~anes , R=ps~lyelh~r or polyester R'=aCip~~atic or aromatic Polyvinylpyrrolidone (PVP) is a polyamide that possesses unusual complexing and colloidal properties and is essentially physiologically inert.
PVP
and other hydrophilic polymers are typically biocompatible. PVP may be incorporated into drug loaded hybrid polymer compositions in order to increase drug release rates. In one embodiment, the concentration of PVP that is used in drug loaded hybrid polymer compositions can be less than 20%. This concentration can not make the layers bioerodable or lubricious. In general, PVP concentrations from <1 % to greater than ~0% are deemed workable. In one aspect of the invention, the therapeutic agent that is associated with an device is formulated with a PVP polymer.
6-l~~ ~,~, j,.,', ~~=
~I~-~Ha - y ~~~yvir~~lpyrro~i~~~

Acrylate polymers and copolymers including polymethylmethacrylate (PMMA) and polymethylmethacrylate hydroxyethyl methacrylate (PMMA/HEMA) are known for their biocompatibility as a result of their widespread use in contact and intraocular lens applications. This class of polymer generally provokes very little smooth muscle and endothelial cell growth, and very low inflammatory response (Bar). These polymersicopolymers are compatible with drugs and the other polymers and layers of the instant invention. Thus, in one aspect, the device is associated with a composition that comprises an anti-scarring agent as described above, and an acrylate polymer or copolymer.
CH3 ~ H3 I n I
C=O C=
I I

Methylmethacrylate hydroxyethylmethacrylate copolymer It should be obvious to one of skill in the art that the polymers as described herein can also be blended or copolymerized in various compositions as required to deliver therapeutic doses of fibrosis-inhibiting agents.
Polymeric carriers for fibrosis-inhibiting agents can be fashioned in a variety of forms, with desired release characteristics and/or with specific properties depending upon the device, composition or implant being utilized.
For example, polymeric carriers may be fashioned to release a fibrosis-inhibiting agent upon exposure to a specific triggering event such as pH (see, e.g., Heller et al., "Chemically Self-Regulated Drug Delivery Systems," in Polymers in Medicine III, Elsevier Science Publishers B.V., Amsterdam, 1988, pp. 175-188; Kang et al., J. Applied Polymer Sci. 48:343-354, 1993; Dong et al., J. Controlled Release 19:171-178, 1992; Dong and Hoffman, J. Controlled Release 15:141-152, 1991; Kim et al., J. Controlled Release 28:143-152, 1994;
cornejo-Bravo et al., J. Controlled Release 33:223-229, 1995; Wu and Lee, Pharm. Res. 10(10):1544-1547, 1993; Serres et al., Pharm. Res. 13(2):196-201, 1996; Peppas, "Fundamentals of pH- and Temperature-Sensitive Delivery Systems," in Gurny et al. (eds.), Pulsatile Drug Delivery, Wissenschaftliche Verlagsgesellschaft mbH, Stuttgart, 1993, pp. 41-55; Doelker, "cellulose Derivatives," 1993, in Peppas and Langer (eds.), Biopolymers I, Springer-Verlag, Berlin). Representative examples of pH-sensitive polymers include poly (acrylic acid) and its derivatives (including for example, homopolymers such as poly(aminocarboxylic acid); poly(acrylic acid); poly(methyl acrylic acid), copolymers of such homopolymers, and copolymers of poly(acrylic acid) and/or acrylate or acrylamide (monomers such as those discussed above. Other pH
sensitive polymers include polysaccharides such as cellulose acetate phthalate;
hydroxypropylmethylcellulose phthalate; hydroxypropylmethylcellulose acetate succinate; cellulose acetate trimellilate; and chitosan. Yet other pH
sensitive polymers include any mixture of a pH sensitive polymer and a water-soluble polymer.
Likewise, fibrosis-inhibiting agents can be delivered via polymeric carriers which are temperature sensitive (see, e.g., Chen et al., "Novel Hydrogels of a Temperature-Sensitive PLURONIC Grafted to a Bioadhesive Polyacrylic Acid Backbone for Vaginal Drug Delivery," in Proceed. Intern.
Symp.
Control. Rel. Bioact. Mater. 22:167-168, Controlled Release Society, Inc., 1995;
Okano, "Molecular Design of Stimuli-Responsive Hydrogels for Temporal Controlled Drug Delivery," in Proceed. Intern. Symp. Control. Rel. Bioact.
Mater. 22:111-112, Controlled Release Society, Inc., 1995; Johnston et al., Pharm. Res. 9(3):425-433, 1992; Tung, Int'1 J. Pharm. 707:85-90, 1994; Harsh and Gehrke, J. Controlled Release 77:175-186, 1991; Bae et al., Pharm. Res.
8(4):531-537, 1991; Dinarvand and D'Emanuele, J. Controlled Release 36:221-227, 1995; Yu and Grainger, "Novel Thermo-sensitive Amphiphilic Gels: Poly N-isopropylacrylamide-co-sodium acrylate-co-n-N-alkylacrylamide Network Synthesis and Physicochemical Characterization," Dept. of Chemical &
Biological Sci., Oregon Graduate Institute of Science & Technology, Beaverton, OR, pp. 820-821; ~hou and Smid, "Physical Hydrogels of Associative Star Polymers," Polymer Research Institute, Dept. of Chemistry, College of Environmental Science and Forestry, State Univ. of New York, Syracuse, NY, pp. 822-823; Hoffman et al., "Characterizing Pore Sizes and Water 'Structure' in Stimuli-Responsive Hydrogels," Center for Bioengineering, Univ. of Washington, Seattle, WA, p. 828; Yu and Grainger, "Thermo-sensitive Swelling Behavior in Crosslinked N-isopropylacrylamide Networks: Cationic, Anionic and Ampholytic Hydrogels," Dept. of Chemical & Biological Sci., Oregon Graduate Institute of Science & Technology, Beaverton, OR, pp. 829-830; Kim et al., Pharm. Res. 9(3):283-290, 1992; Bae et al., Pharm. Res. 8(5):624-628, 1991;

Kono et al., J. Controlled Release 30:69-75, 1994; Yoshida et al., J.
Controlled Release 32:97-102, 1994; Okano et al., J. Controlled Release 36:125-133, 1995; Chun and Kim, J. Controlled Release 38:39-47, 1996; D'Emanuele and Dinarvand, Int'1 J. Pharm. 118:237-242, 1995; Katono et al., J. Controlled Release 16:215-228, 1991; Hoffman, "Thermally Reversible Hydrogels Containing Biologically Active Species," in Migliaresi et al. (eds.), Polymers in Medicine III, Elsevier Science Publishers B.V., Amsterdam, 1988, pp. 161-167;
Hoffman, "Applications of Thermally Reversible Polymers and Hydrogels in Therapeutics and Diagnostics," in Third International Symposium on Recent Advances in Drug Delivery Systems, Salt Lake City, UT, Feb. 24-27, 1987, pp.
297-305; Gutowska et al., J. Controlled Release 22:95-104, 1992; Palasis and Gehrke, J. Controlled Release 18:1-12, 1992; Paavola et al., Pharm. Res.
12(12):1997-2002, 1995).
Representative examples of thermogelling polymers, and the gelatin temperature (LCST (°C)) include homopolymers such as poly(N-methyl-N-n-propylacrylamide), 19.8; poly(N-n-propylacrylamide), 21.5;
poly(N-methyl-N-isopropylacrylamide), 22.3; poly(N-n-propylmethacrylamide), 28.0; poly(N-isopropylacrylamide), 30.9; poly(N, n-diethylacrylamide), 32.0;
poly(N-isopropylmethacrylamide), 44.0; poly(N-cyclopropylacrylamide), 45.5;
poly(N-ethylmethyacrylamide), 50.0; poly(N-methyl-N-ethylacrylamide), 56.0;
poly(N-cyclopropylmethacrylamide), 59.0; poly(N-ethylacrylamide), 72Ø
Moreover thermogelling polymers may be made by preparing copolymers between (among) monomers of the above, or by combining such homopolymers with other water-soluble polymers such as acrylmonomers (e.g., acrylic acid and derivatives thereof, such as methylacrylic acid, acrylate monomers and derivatives thereof, such as butyl methacrylate, butyl acrylate, lauryl acrylate, and acrylamide monomers and derivatives thereof, such as N-butyl acrylamide and acrylamide).
Other representative examples of thermogelling polymers include cellulose ether derivatives such as hydroxypropyl cellulose, 41 °C;
methyl cellulose, 55°C; hydroxypropylmethyl cellulose, 66°C; and ethylhydroxyethyl cellulose, polyalkylene oxide-polyester block copolymers of the structure X-Y, Y-X-Y and X-Y-X where X in a polyalkylene oxide and Y is a biodegradable polyester (e.g., PLG-PEG-PLG) and PLURONICs such as F-127, 10 - 15°C;
L-122, 19°C; L-92, 26°C; L-81, 20°C; and L-61, 24°C.

Representative examples of patents relating to thermally gelling polymers and the preparation include U.S. Patent Nos. 6,451,346; 6,201,072;
6,117,949; 6,004,573; 5,702,717; and 5,484,610; and PCT Publication Nos.
WO 99/07343; WO 99/18142; WO 03/17972; WO 01/82970; WO 00/18821;
WO 97/15287; WO 01/41735; WO 00/00222 and WO 00/38651.
Fibrosis-inhibiting agents may be linked by occlusion in the matrices of the polymer, bound by covalent linkages, or encapsulated in microcapsules. Within certain embodiments of the invention, therapeutic compositions are provided in non-capsular formulations such as microspheres (ranging from nanometers to micrometers in size), pastes, threads of various size, films, or sprays. In one aspect, the anti-scarring agent may be incorporated into biodegradable magnetic nanospheres. The nanospheres may be used, for example, to replenish an anti-scarring agent into an implanted intravascular device, such as a stent containing a weak magnetic alloy (see, e.g., Z. Forbes, B.B. Yellen, G. Friedman, K. Barbee. "An approach to targeted drug delivery based on uniform magnetic fields," IEEE Trans. Magn. 39(5):
3372-3377 (2003)).
Within certain aspects of the present invention, therapeutic compositions may be fashioned in the form of microspheres, microparticles and/or nanoparticles having any size ranging from about 30 nm to 500 ~,m, depending upon the particular use. These compositions can be formed by spray-drying methods, milling methods, coacervation methods, W/O emulsion methods, W/O/W emulsion methods, and solvent evaporation methods. In other aspects, these compositions can include microemulsions, emulsions, liposomes and micelles. Alternatively, such compositions may also be readily applied as a "spray", which solidifies into a film or coating for use as a device/implant surface coating or to line the tissues of the implantation site.
Such sprays may be prepared from microspheres of a wide array of sizes, including for example, from 0.1 ~m to 3 pm, from 10 ~m to 30 pm, and from 30 pm to 100 ~,m.
Therapeutic compositions of the present invention may also be prepared in a variety of "paste" or gel forms. For example, within one embodiment of the invention, therapeutic compositions are provided which are liquid at one temperature (e.g., temperature greater than 37°C, such as 40°C, 45°C, 50°C, 55°C or 60°C), and solid or semi-solid at another temperature (e.g., ambient body temperature, or any temperature lower than 37°C). Such "thermopastes" may be readily made utilizing a variety of techniques (see, e.g., PCT Publication WO 98/24427). Other pastes may be applied as a liquid, which solidify in vivo due to dissolution of a water-soluble component of the paste and precipitation of encapsulated drug into the aqueous body environment. These "pastes" and "gels" containing fibrosis-inhibiting agents are particularly useful for application to the surface of tissues that will be in contact with the implant or device.
Within yet other aspects of the invention, the therapeutic compositions of the present invention may be formed as a film or tube. These films or tubes can be porous or non-porous. Preferably, such films or tubes are generally less than 5, 4, 3, 2, or 1 mm thick, more preferably less than 0.75 mm, 0.5 mm, 0.25 mm, or, 0.10 mm thick. Films or tubes can also be generated of thicknesses less than 50 ~.m, 25 pm or 10 Vim. Such films are preferably flexible with a good tensile strength (e.g., greater than 50, preferably greater than 100, and more preferably greater than 150 or 200 N/cm2), good adhesive properties (i.e., adheres to moist or wet surfaces), and have controlled permeability. Fibrosis-inhibiting agents contained in polymeric films are particularly useful for application to the surface of a device or implant as well as to the surface of tissue, cavity or an organ.
Within further aspects of the present invention, polymeric carriers are provided which are adapted to contain and release a hydrophobic fibrosis-inhibiting compound, and/or the carrier containing the hydrophobic compound in combination with a carbohydrate, protein or polypeptide. Within certain embodiments, the polymeric carrier contains or comprises regions, pockets, or granules of one or more hydrophobic compounds. For example, within one embodiment of the invention, hydrophobic compounds may be incorporated within a matrix which contains the hydrophobic fibrosis-inhibiting compound, followed by incorporation of the matrix within the polymeric carrier. A
variety of matrices can be utilized in this regard, including for example, carbohydrates and polysaccharides such as starch, cellulose, dextran, methylcellulose, sodium alginate, heparin, chitosan and hyaluronic acid, proteins or polypeptides such as albumin, collagen and gelatin. Within alternative embodiments, hydrophobic compounds may be contained within a hydrophobic core, and this core contained within a hydrophilic shell.
Other carriers that may likewise be utilized to contain and deliver fibrosis-inhibiting fibrosis-inhibiting agents described herein include:

hydroxypropyl cyclodextrin (Cserhati and Hollo, Int. J. Pharm. 108:69-75, 1994), liposomes (see, e.g., Sharma et al., Cancer Res. 53:5877-5881, 1993; Sharma and Straubinger, Pharm. Res. 11(60):889-896, 1994; WO 93/18751; U.S.
Patent No. 5,242,073), liposome/gel (WO 94/26254), nanocapsules (Bartoli et al., J. Microencapsulation 7(2):191-197, 1990), micelles (Alkan-Onyuksel et al., Pharm. Res. 11(2):206-212, 1994), implants (Jampel et al., Invest. Ophthalm.
Vis. Science 34(11 ):3076-3083, 1993; Walter et al., Cancer Res. 54:22017-2212, 1994), nanoparticles (Violante and Lanzafame PAACR), nanoparticles -modified (U.S. Patent No. 5,145,684), nanoparticles (surface modified) (U.S. -Patent No. 5,399,363), micelle (surfactant) (U.S. Patent No. 5,403,858), synthetic phospholipid compounds (U.S. Patent No. 4,534,899), gas borne dispersion (U.S. Patent No. 5,301,664), liquid emulsions, foam, spray, gel, lotion, cream, ointment, dispersed vesicles, particles or droplets solid- or liquid-aerosols, microemulsions (U.S. Patent No. 5,330,756), polymeric shell (nano-and micro- capsule) (U.S. Patent No~. 5,439,686), emulsion (Tarr et al., Pharm Res. 4: 62-165, 1987), nanospheres (Hagan et al., Proc. Intern. Symp. Control Rel. Bioact. Mater. 22, 1995; Kwon et al., Pharm Res. 12(2):192-195; Kwon et al., Pharm Res. 10(7):970-974; Yokoyama et al., J. Contr. Rel. 32:269-277, 1994; Gref et al., Science 263:1600-1603, 1994; Bazile et al., J. Pharm. Sci.
84:493-498, 1994) and implants (U.S. Patent No. 4,882,168).
Within another aspect of the present invention, polymeric carriers can be materials that are formed in situ. In one embodiment, the precursors can be monomers or macromers that contain unsaturated groups that can be polymerized and/or cross-linkeds. The monomers or macromers can then, for example, be injected into the treatment area or onto the surface of the treatment area and polymerized in situ using a radiation source (e.g., visible or UV light) or a free radical system (e.g., potassium persulfate and ascorbic acid or iron and hydrogen peroxide). The polymerization step can be performed immediately prior to, simultaneously to or post injection of the reagents into the treatment site. Representative examples of compositions that undergo free radical polymerization reactions are described in WO 01/44307, WO 01/68720, WO 02/072166, WO 03/043552, WO 93/17669, WO 00/64977; U.S. Patent Nos. 5,900,245, 6,051,248, 6,083,524, 6,177,095, 6,201,065, 6,217,894, 6,639,014, 6,352,710, 6,410,645, 6,531,147, 5,567,435, 5,986,043, 6,602,975;
U.S. Patent Application Publication Nos. 2002/012796A1, 2002/0127266A1, 2002/0151650A1, 2003/0104032A1, 2002/0091229A1, and 2003/0059906A1.

In another embodiment, the reagents can undergo an electrophilic-nucleophilic reaction to produce a crosslinked matrix. For example, a 4-armed thiol derivatized polyethylene glycol can be reacted with a 4 armed NHS-derivatized polyethylene glycol under basic conditions (pH >
about 8). Representative examples of compositions that undergo electrophilic-nucleophilic crosslinking reactions are described in U.S. Patent. Nos.
5,752,974; 5,807,581; 5,874,500; 5,936,035; 6,051,648; 6,165,489; 6,312,725;
6,458,889; 6,495,127; 6,534,591; 6,624,245; 6,566,406; 6,610,033; 6,632,457;
PCT Application Published Nos. WO 04/060405 and WO 04/060346. Other examples of in situ forming materials that can be used include those based on the crosslinking of proteins (described in U.S. Patent Nos. RE38158;
4,839,345;
5,514,379, 5,583,114; 6,458,147; 6,371,975; U.S. Publication Nos 200210161399; 2001!0018598 and PCT Publication Nos. WO 03/090683; WO
01/45761; WO 99/66964 and WO 96/03159).
As described above, the anti-fibrosing agent can be associated with a medical device using the polymeric carriers or coatings described above.
In addition to the compositions and methods described above, there are various other compositions and methods that are known in the art. Representative examples of these compositions and methods for applying (e.g., coating) these compositons to devices are described in U.S. Patent. Nos. 6,610,016;
6,358,557; 6,306,176; 6,110,483; 6,106,473; 5,997,517; 5,800,412; 5,525,348;
5,331,027; 5,001,009; 6,562,136; 6,406,754; 6,344,035; 6,254,921; 6,214,901;
6,077,698; 6,603,040; 6,278,018; 6,238,799; 6,096,726, 5,766,158, 5,599,576, 4,119,094; 4,100,309; 6,599,558; 6,369,168; 6,521,283; 6,497,916; 6,251,964;
6,225,431; 6,087,462; 6,083,257; 5,739,237; 5,739,236; 5,705,583; 5,648,442;
5,645,883; 5,556,710; 5,496,581; 4,689,386; 6,214,115; 6,090,901; 6,599,448;
6,054,504; 4,987,182; 4,847,324; and 4,642,267; U.S. Patent Application Publication Nos. 2002/0146581, 2003/0129130, 2003/0129130, 2001 /0026834;
2003/0190420; 2001 /0000785; 2003/0059631; 2003/0190405; 2002/0146581;
2003/020399; 2001 /0026834; 2003/0190420; 2001 /0000785; 2003/0059631;
2003/0190405; and 2003/020399; and PCT Publication Nos. WO 02/055121;
WO 01/57048; WO 01/52915; and WO 01/01957.
Within another aspect of the invention, the biologically active agent can be delivered with a non-polymeric agent. These non-polymeric carriers can include sucrose derivatives (e.g., sucrose acetate isobutyrate, sucrose oleate), sterols such as cholesterol, stigmasterol, ~3-sitosterol, and estradiol; cholesteryl esters such as cholesteryl stearate; CT2 -C~~ fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid; C~$ -C36 mono-, di- and triacylglycerides such as glyceryl monooleate, glyceryl monolinoleate, glyceryl monolaurate, glyceryl monodocosanoate, glyceryl monomyristate, glyceryl monodicenoate, glyceryl dipalmitate, glyceryl didocosanoate, glyceryl dimyristate, glyceryl didecenoate, glyceryl tridocosanoate, glyceryl trimyristate, glyceryl tridecenoate, glycerol tristearate and mixtures thereof; sucrose fatty acid esters such as sucrose distearate and sucrose palmitate; sorbitan fatty acid esters such as sorbitan monostearate, sorbitan monopalmitate and sorbitan tristearate; C~6 -C~$ fatty alcohols such as cetyl alcohol, myristyl alcohol, stearyl alcohol, and cetostearyl alcohol; esters of fatty alcohols and fatty acids such as cetyl palmitate and cetearyl palmitate; anhydrides of fatty acids such as stearic anhydride;
phospholipids including phosphatidylcholine (lecithin), phosphatidylserine, phosphatidylethanolamine, phosphatidylinositol, and lysoderivatives thereof;
sphingosine and derivatives thereof; spingomyelins such as stearyl, palmitoyl, and tricosanyl spingomyelins; ceramides such as stearyl and palmitoyl ceramides; glycosphingolipids; lanolin and lanolin alcohols, calcium phosphate, sintered and unscintered hydoxyapatite, zeolites; and combinations and mixtures thereof.
Representative examples of patents relating to non-polymeric delivery systems and the preparation include U.S. Patent Nos. 5,736,152;
5,888,533; 6,120,789; 5,968,542; and 5,747,058.
The fibrosis-inhibiting agent may be delivered as a solution. The fibrosis-inhibiting agent can be incorporated directly into the solution to provide a homogeneous solution or dispersion. In certain embodiments, the solution is an aqueous solution. The aqueous solution may futher include buffer salts, as well as viscosity modifying agents (e.g., hyaluronic acid, alginates, carboxymethylcelluloe (CMC), and the like). In another aspect of the invention, the solution can include a biocompatible solvent, such as ethanol, DMSO, glycerol, PEG-200, PEG-300 or NMP.
Within another aspect of the invention, the fibrosis-inhibiting agent can further comprise a secondary carrier. The secondary carrier can be in the form of microspheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), nanospheres (PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), liposomes, emulsions, microemulsions, micelles (SDS, block copolymers of the form X-Y, X-Y-X or Y-X-Y, R-(Y-X)", R-(X-Y)" where X is a polyalkylene oxide (e.g., poly(ethylene oxide, polypropylene oxide, block copolymers of polyethylene oxide) and polypropylene oxide) and Y is a polyester (e.g., polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y-decanolactone, ~-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-Zone.), R is a multifunctional initiator and copolymers as well as blends thereof.), zeolites or cyclodextrins.
Within another aspect of the invention, these fibrosis-inhibiting agent/secondary carrier compositions can be a) incorporated directly into or onto the device, b) incorporated into a solution, c) incorporated into a gel or viscous solution, d) incorporated into the composition used for coating the device or e) incorporated into or onto the device following coating of the device with a coating composition.
For example, fibrosis-inhibiting agent loaded PLGA microspheres can be incorporated into a polyurethane coating solution which is then coated onto the device.
In yet another example, the device can be coated with a polyurethane and then allowed to partially dry such that the surface is still tacky.
A particulate form of the fibrosis-inhibiting agent or fibrosis-inhibiting agent/secondary carrier can then be applied to all or a portion of the tacky coating after which the device is dried.
In yet another example, the device can be coated with one of the coatings described above. A thermal treatment process can then be used to soften the coating, afterwhich the fibrosis-inhibiting agent or the fibrosis-inhibiting agent/secondary carrier is applied to the entire device or to a portion of the device (e.g., outer surface) Within another aspect of the invention, the coated device which inhibits or reduces an in vivo fibrotic reaction is further coated with a compound or compositions which delay the release of and/or activity of the fibrosis-inhibiting agent. Representative examples of such agents include biologically inert materials such as gelatin, PLGA/MePEG film, PLA, polyurethanes, silicone rubbers, surfactants, lipids, or polyethylene glycol, as well as biologically active materials such as heparin (e.g., to induce coagulation).

For example, in one embodiment of the invention, the active agent on the device is top-coated with a physical barrier. Such barriers can include non-degradable materials or biodegradable materials such as gelatin, PLGA/MePEG film, PLA, or polyethylene glycol among others. In one embodiment, the rate of diffusion of the therapeutic agent in the barrier coat is slower that the rate of diffusion of the therapeutic agent in the coating layer. In the case of PLGA/ MePEG, once the PLGAi MePEG becomes exposed to the bloodstream, the MePEG can dissolve out of the PLGA, leaving channels through the PLGA layer to an underlying layer containing the fibrosis-inhibiting agent, which then can then diffuse into the vessel wall and initiate its biological activity.
In another embodiment of the invention, a particulate form of the active agent may be coated onto the stent (or any of the devices described below) using a polymer (e.g., PLG, PLA, aor a polyurethane). A second polymer, that dissolves slowly or degrades (e.g., MePEG-PLGA or PLG) and that does not contain the active agent, may be coated over the first layer.
Once the top layer dissolves or degrades, it exposes the under coating which allows the active agent to be exposed to the treatment site or to be released from the coating.
Within another aspect of the invention, the outer layer of the coating of a coated device, which inhibits an in vivo fibrotic response, is further treated to crosslinlc the outer layer of the coating. This can be accomplished by subjecting the coated device to a plasma treatment process. The degree of crosslinking and nature of the surface modification can be altered by changing the RF power setting, the location with respect to the plasma, the duration of treatment as well as the gas composition introduced into the plasma chamber.
Protection of a biologically active surface can also be utilized by coating the device surface with an inert molecule that prevents access to the active site through steric hindrance, or by coating the surface with an inactive form of the fibrosis-inhibiting agent, which is later activated. ' For example, the device can be coated with an enzyme, which causes either release of the fibrosis-inhibiting agent or activates the fibrosis-inhibiting agent.
In another embodiment, the device is coated with a fibrosis-inhibiting agent and then further coated with a composition that comprises an anticoagulant such as heparin. As the anticoagulant dissolves away, the anticoagulant activity slows or stops, and the newly exposed fibrosis-inhibiting agent is available to inhibit or reduce fibrosis from occurring in the adjacent tissue.
The device can be coated with an inactive form of the fibrosis-inhibiting agent, which is then activated once the device is deployed. Such activation can be achieved by injecting another material into the treatment area after the device (as desribed below) is deployed or after the fibrosis-inhibiting agent has been administered to the treatment area (via, e.g., injections, spray, wash, drug delivery catheters or balloons). For example, the device can be coated with an inactive form of the fibrosis-inhibiting agent. Once the device is deployed, the activating substance is injected or applied into or onto the treatment site where the inactive form of the fibrosis-inhibiting agent has been applied. For example, a device can be coated with a biologically active fibrosis-inhibiting agent and a first substance having moieties that capable of forming an ester bond with another material. The coating can be covered with a second substance such as polyethylene glycol. The first and second substances can react to form an ester bond via, e.g., a condensation reaction. Prior to the deployment of the device, an esterase is injected into the treatment site around the outside of the device, which can cleave the bond between the ester and the fibrosis-inhibiting agent, allowing the agent to initiate fibrosis-inhibition.
In another aspect, a medical device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug. The reservoirs may be formed from divets in the device surface or micropores or channels in the device body. In one aspect, the reservoirs are formed from voids in the structure of the device. The reservoirs may house a single type of drug or more than one type of drug. The drugs) may be formulated with a carrier (e.g., a polymeric or non-polymeric material) that is loaded into the reservoirs. The filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier. In certain embodiments, the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount of drug that is released from the substrate. The multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void.

Within certain embodiments of the invention, the therapeutic compositions may also comprise additional ingredients such as surfactants (e.g., PLURONICS, such as F-127, L-122, L-101, L-92, L-81, and L-61 ), anti-inflammatory agents (e.g., dexamethasone or asprin), anti-thrombotic agents (e.g., heparin, high activity heparin, heparin quaternary amine complexes (e.g., heparin benzalkonium chloride complex)), anti-infective agents (e.g., 5-fluorouracil, triclosan, rifamycim, and silver compounds), preservatives, anti-oxidants and/ or anti-platelet agents.
Within certain embodiments of the invention, the therapeutic agent or carrier can also comprise radio-opaque, echogenic materials and magnetic resonance imaging (MRI) responsive materials (i.e., MRI contrast agents) to aid in visualization of the device under ultrasound, fluoroscopy and/or MRI. For example, a device may be made with or coated with a composition which is echogenic or radiopaque (e.g., made with echogenic or radiopaque with materials such as powdered tantalum, tungsten, barium carbonate, bismuth oxide, barium sulfate, metrazimide, iopamidol, iohexol, iopromide, iobitridol, iomeprol, iopentol, ioversol, ioxilan, iodixanol, iotrolan, acetrizoic acid derivatives, diatrizoic acid derivatives, iothalamic acid derivatives, ioxithalamic acid derivatives, metrizoic acid derivatives, iodamide, lypophylic agents, iodipamide and ioglycamic acid or, by the addition of microspheres or bubbles which present an acoustic interface). Visualization of a device by ultrasonic imaging may be achieved using an echogenic coating.
Echogenic coatings are described in, e.g., U.S. Patent Nos. 6,106,473 and 6,610,016. For visualization under MRI, contrast agents (e.g., gadolinium (III) chelates or iron oxide compounds) may be incorporated into or onto the device, such as, for example, as a component in a coating or within the void volume of the device (e.g., within a lumen, reservoir, or within the structural material used to form the device). In some embodiments, a medical device may include radio-opaque or MRI visible markers (e.g., bands) that may be used to orient and guide the device during the implantation procedure.
In another embodiment, these agents can be contained within the same coating layer as the therapeutic agent or they may be contained in a coating layer (as described above) that is either applied before or after the therapeutic agent containing layer.
Medical implants may, alternatively, or in addition, be visualized under visible light, using fluorescence, or by other spectroscopic means.

Visualization agents that can be included for this purpose include dyes, pigments, and other colored agents. In one aspect, the medical implant may further include a colorant to improve visualization of the implant in vivo and/or ex vivo. Frequently, implants can be difficult to visualize upon insertion, especially at the margins of implant. A coloring agent can be incorporated into a medical implant to reduce or eliminate the incidence or severity of this problem. The coloring agent provides a unique color, increased contrast, or unique fluorescence characteristics to the device. In one aspect, a solid implant is provided that includes a colorant such that it is readily visible (under visible light or using a fluorescence technique) and easily differentiated from its implant site. In another aspect, a colorant can be included in a liquid or semi-solid composition. For example, a single component of a two component mixture may be colored, such that when combined ex-vivo or in-vivo, the mixture is sufficiently colored.
The coloring agent may be, for example, an endogenous compound (e.g., an amino acid or vitamin) or a nutrient or food material and may be a hydrophobic or a hydrophilic compound. Preferably, the colorant has a very low or no toxicity at the concentration used. Also preferred are colorants that are safe and normally enter the body through absorption such as (i-carotene. Representative examples of colored nutrients (under visible light) include fat soluble vitamins such as Vitamin A (yellow); water soluble vitamins such as Vitamin B12 (pink-red) and folic acid (yellow-orange); carotenoids such as a-carotene (yellow-purple) and lycopene (red). Other examples of coloring agents include natural product (berry and fruit) extracts such as anthrocyanin (purple) and saffron extract (dark red). The coloring agent may be a fluorescent or phosphorescent compound such as a-tocopherolquinol (a Vitamin E
derivative) or L-tryptophan. Derivatives, analogues, and isomers of any of the above colored compound also may be used. The method for incorporating a colorant into an implant or therapeutic composition may be varied depending on the properties of and the desired location for the colorant. For example, a hydrophobic colorant may be selected for hydrophobic matrices. The colorant may be incorporated into a carrier matrix, such as micelles. Further, the pH
of the environment may be controlled to further control the color and intensity.
In one aspect, the composition of the present invention include one or more coloring agents, also referred to as dyestuffs, which will be present in an effective amount to impart observable coloration to the composition, e.g., the gel. Examples of coloring agents include dyes suitable for food such as those known as F.D. & C. dyes and natural coloring agents such as grape skin extract, beet red powder, beta carotene, annato, carmine, turmeric, paprika, and so forth. Derivatives, analogues, and isomers of any of the above colored compound also may be used. The method for incorporating a colorant into an implant or therapeutic composition may be varied depending on the properties of and the desired location for the colorant. For example, a hydrophobic colorant may be selected for hydrophobic matrices. The colorant may be incorporated into a carrier matrix, such as micelles. Further, the pH of the environment may be controlled to further control the color and intensity.
In one aspect, the compositions of the present invention include one or more preservatives or bacteriostatic agents, present in an effective amount to preserve the composition and/or inhibit bacterial growth in the composition, for example, bismuth tribromophenate, methyl hydroxybenzoate, bacitracin, ethyl hydroxybenzoate, propyl hydroxybenzoate, erythromycin, 5-fluorouracil, methotrexate, doxorubicin, mitoxantrone, rifamycin, chlorocresol, benzalkonium chlorides, and the like. Examples of the preservative include paraoxybenzoic acid esters, chlorobutanol, benzylalcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid, etc. In one aspect, the compositions of the present invention include one or more bactericidal (also known as bacteriacidal) agents.
In one aspect, the compositions of the present invention include one or more antioxidants, present in an effective amount. Examples of the antioxidant include sulfites, alpha-tocopherol and ascorbic acid.
Within certain aspects of the present invention, the therapeutic composition should be biocompatible, and release one or more fibrosis-inhibiting agents over a period of several hours, days, or, months. As described above, "release of an agent" refers to any statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the compositions and/or remains active on the surface of (or within) the composition. The compositions of the present invention may release the anti-scarring agent at one or more phases, the one or more phases having similar or different performance (e.g., release) profiles. The therapeutic agent may be made available to the tissue at amounts which may be sustainable, intermittent, or continuous; in one or more phases; and/or rates of delivery; effective to reduce or inhibit any one or more components of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue).
Thus, release rate may be programmed to impact fibrosis (or scarring) by releasing an anti-scarring agent at a time such that at least one of the components of fibrosis is inhibited or reduced. Moreover, the predetermined release rate may reduce agent loading and/or concentration as well as potentially providing minimal drug washout and thus, increases efficiency of drug effect. Any one of the at least one anti-scarring agents may perform one or more functions, including inhibiting the formation of new blood vessels (angiogenesis), inhibiting the migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), inhibiting the deposition of extracellular matrix (ECM), and inhibiting remodeling (maturation and organization of the fibrous tissue). In one embodiment, the rate of release may provide a sustainable level of the anti-scarring agent to the susceptible tissue site. In another embodiment, the rate of release is substantially constant.
The rate may decrease and/or increase over time, and it may optionally include a substantially non-release period. The release rate may comprise a plurality of rates. In an embodiment, the plurality of release rates may include rates selected from the group consisting of substantially constant, decreasing, increasing, substantially non-releasing.
The total amount of anti-scarring agent made available on, in or near the device may be in an amount ranging from about 0.01 pg (micrograms) to about 2500 mg (milligrams). Generally, the anti-scarring agent may be in the amount ranging from 0.01 pg to about 10 pg; or from 10 pg to about 1 mg; or from 1 mg to about 10 mg; or from 10 mg to about 100 mg; or from 100 mg to about 500 mg; or from 500 mg to about 2500 mg.
The total surface amount of anti-scarring agent on, in or near the device may be in an amount ranging from less than 0.01 pg to about 2500 pg per mm2 of device surface area. Generally, the anti-scarring agent may be in the amount ranging from less than 0.01 pg; or from 0.01 pg to about 10 pg; or from 10 pg to about 250 pg; or from 250 pg to about 2500 pg, The anti-scarring agent that is on, in or near the device may be released from the composition in a time period that may be measured from the time of implantation, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 7 days; from 7 days to about 14 days; from 14 days to about 28 days;
from 28 days to about 56 days; from 56 days to about 90 days; from 90 days to about 180 days.
The amount of anti-scarring agent released from the composition as a function of time may be determined based on the in vitro release characteristics of the agent from the composition. The in vitro release rate may be determined by placing the anti-scarring agent within the composition or device in an appropriate buffer such as 0.1 M phosphate buffer (pH 7.4)) at 37°C. Samples of the buffer solution are then periodically removed for analysis by HPLC, and the buffer is replaced to avoid any saturation effects.
Based on the in vitro release rates, the release of anti-scarring agent per day may range from an amount ranging from about 0.01 pg (micrograms) to about 2500 mg (milligrams). Generally, the anti-scarring agent that may be released in a day may be in the amount ranging from 0.01 pg to about 10 pg; or from 10 pg to about 1 mg; or from 1 mg to about 10 mg; or from 10 mg to about 100 mg; or from 100 mg to about 500 mg; or from 500 mg to about 2500 mg.
In one embodiment, the anti-scarring agent is made available to the susceptible tissue site in a programmed, sustained, and/or controlled manner which results in increased efficiency and/or efficacy. Further, the release rates may vary during either or both of the initial and subsequent release phases. There may also be additional phases) for release of the same substances) and/or different substance(s).
Further, therapeutic compositions and devices of the present invention should preferably be have a stable shelf-life for several months and capable of being produced and maintained under sterile conditions. Many pharmaceuticals are manufactured to be sterile and this criterion is defined by the USP XXII <1211 >. The term "USP" refers to U.S. Pharmacopeia (see www.usp.org, Rockville, MD). Sterilization may be accomplished by a number of means accepted in the industry and listed in the USP XXII <1211 >, including gas sterilization, ionizing radiation or, when appropriate, filtration.
Sterilization . may be maintained by what is termed asceptic processing, defined also in USP
XXII <1211 >. Acceptable gases used for gas sterilization include ethylene oxide. Acceptable radiation types used for ionizing radiation methods include gamma, for instance from a cobalt 60 source and electron beam. A typical dose of gamma radiation is 2.5 MRad. Filtration may be accomplished using a filter with suitable pore size, for example 0.22 ~.m and of a suitable material, for instance polytetrafluoroethylene (e.g., TEFLON from E.I. DuPont De Nemours and Company, Wilmington, DE).
In another aspect, the compositions and devices of the present invention are contained in a container that allows them to be used for their intended purpose, i.e., as a pharmaceutical composition. Properties of the container that are important are a volume of empty space to allow for the addition of a constitution medium, such as water or other aqueous medium, e.g., saline, acceptable light transmission characteristics in order to prevent light energy from damaging the composition in the container (refer to USP XXII
<661 >), an acceptable limit of extractables within the container material (refer to USP XXII), an acceptable barrier capacity for moisture (refer to USP XXII
<671 >) or oxygen. In the case of oxygen penetration, this may be controlled by including in the container, a positive pressure of an inert gas, such as high purity nitrogen, or a noble gas, such as argon.
Typical materials used to make containers for pharmaceuticals include USP Type I through III and Type NP glass (refer to USP X)CII <661 >), polyethylene, TEFLON, silicone, and gray-butyl rubber.
In one embodiment, the product containers can be thermoformed plastics. In another embodiment, a seconday package can be used for the product. In another embodiment, product can be in a sterile container that is placed in a box that is labeled to describe the contents of the box.
5) Coating of devices with fibrosis-inhibiting agents As described above, a range of polymeric and non-polymeric materials can be used to incorporate the fibrosis-inhibiting agent onto or into a device. The anti-fibrosing agent composition can be incorporated into or onto the device in a variety of ways. Coating of the device with the fibrosis-inhibiting agent containing composition or with the fibrosis-inhibiting agent only is one process that can be used to incorporate the fibrosis-inhibiting agent into or onto the device. The anti-fibrosing agent or anti-fibrosing composition may be coated onto the entire device or a portion of the device using a method, such as by dipping, spraying, painting or vacuum deposition, that is appropriate for the particular type of device.

a) Dip coating Dip coating is one coating process that can be used. In one embodiment, the fibrosis-inhibiting agent is dissolved in a solvent for the fibrosis agent and is then coated onto the device.
Fibrosis-inhibiting aetent with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time. The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to ~50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being coated on the surface of the device.
Fibrosis-inhibiting agent with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agentlsolvent solution for a specific period of time (seconds to days). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec).
The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting agent with a solvent In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to hours). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried.
The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device as well as being surface associated. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are no significant permanent dimensional changes to the device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer, surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In one embodiment, the fibrosis-inhibiting agent and a polymer are dissolved in a solvent, for both the polymer and the fibrosis -inhibiting agent, and are then coated onto the device.
In any one the above dip coating methods, the surface of the device can be treated with a plasma polymerization method prior to coating of the scarring agent or scarring agent containing composition, such that a thin polymeric layer is deposited onto the device surface. Examples of such methods include parylene coating of devices and the use of various monomers such hydrocyclosiloxane monomers. Parylene coating may be especially advantageous if the device, or portions of the device, is composed of materials (e.g., stainless steel, nitinol) that do not allow incorporation of the therapeutic agents) into the surface layer using one of the above methods. A parylene primer layer may be deposited onto the device using a parylene coater (e.g., PDS 2010 LABCOTER2 from Cookson Electronics) and a suitable reagent (e.g., di-p-xylylene or dichloro-di-p-xylylene) as the coating feed material.
Parylene compounds are commercially available, for example, from Specialty Coating Systems, Indianapolis, IN), including PARYLENE N (di-p-xylylene), PARYLENE C (a monchlorinated derivative of PARYLENE N, and PARYLENE
D, a dichlorinated derivative of PARYLENE N).
Fibrosis-inhibiting aaent/polymer with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agentlpolymer/solvent solution for a specific period of time. The rate of immersion into the fibrosis-inhibiting agent/polymer/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated on the surface of the device.
Fibrosis-inhibiting aaent/polymer with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/polymer/solvent solution for a specific period of time (seconds to days). The rate of immersion into the fibrosis-inhibiting agent/polymer/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried.
The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated onto the surface of the device as well as the potential for the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting aaentlpolymer with a solvent In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to hours). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried.
The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are not significant permanent dimensional changes to the device (other than those associated with the coating itself).
The fibrosis-inhibiting agent may also be present on the surface of the device.
The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In another embodiment, a suspension of the fibrosis-inhibiting agent in a polymer solution can be prepared. The suspension can be prepared by choosing a solvent that can dissolve the polymer but not the fibrosis-inhibiting agent or a solvent that can dissolve the polymer and in which the fibrosis-inhibiting agent is above its solubility limit. In similar processes described above, a device can be dipped into the suspension of the fibrosis-inhibiting and polymer solution such that the device is coated with a polymer that has a fibrosis-inhibiting agent suspended within it.
b) Spray coating Spray coating is another coating process that can be used. In the spray coating process, a solution or suspension of the fibrosis-inhibiting agent, with or without a polymeric or non-polymeric carrier, is nebulized and directed to the device to be coated by a stream of gas. One can use spray devices such as an air-brush (for example models 2020, 360, 175, 100, 200, 150, 350, 250, 400, 3000, 4000, 5000, 6000 from Badger Air-brush Company, Franklin Park, IL), spray painting equipment, TLC reagent sprayers (for example Part # 14545 and 14654, Alltech Associates, Inc. Deerfield, IL, and ultrasonic spray devices (for example those available from Sono-Tek, Milton, NY). One can also use powder sprayers and electrostatic sprayers.
In one embodiment, the fibrosis-inhibiting agent is dissolved in a solvent for the fibrosis agent and is then sprayed onto the device.
Fibrosis-inhibiting agent with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be held in place or the device can be mounted onto a mandrel or rod that has the ability to move in an X, Y or Z plane or a combination of these planes. Using one of the above described spray devices, the device can be spray coated such that the device is either partially or completely coated with the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agentlsolvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained.
The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being coated on the surface of the device.

Fibrosis-inhibiting agent with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL
per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting agent with a solvent In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution.
The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried.
The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device as well as being surface associated. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are not significant permanent dimensional changes to the device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.

In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In one embodiment, the fibrosis-inhibiting agent and a polymer are dissolved in a solvent, for both the polymer and the anti-fibrosing agent, and are then spray coated onto the device.
Fibrosis-inhibiting actent/polymer with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/polymer/solvent solution for a specific period of time. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated on the surface of the device.
Fibrosis-inhibiting agent/polymer with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/polymerlsolvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated onto the surface of the device as well as the potential for the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting ae~ent/polymer with a solvent In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution.
The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried.
The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are not significant permanent dimensional changes to the device (other than those associated with the coating itself). The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In another embodiment, a suspension of the fibrosis-inhibiting agent in a polymer solution can be prepared. The suspension can be prepared by choosing a solvent that can dissolve the polymer but not the fibrosis-inhibiting agent or a solvent that can dissolve the polymer and in which the fibrosis-inhibiting agent is above its solubility limit. In similar processes described above, the suspension of the fibrosis-inhibiting and polymer solution can be sprayed onto the device such that the device is coated with a polymer that has a fibrosis-inhibiting agent suspended within it.

D_ Methods for Utilizing Medical Implants There are numerous medical devices where the occurrence of a fibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used. Representative examples of implants or devices that can be coated with or otherwise constructed to contain and/or release the therapeutic agents provided herein include cardiovascular devices (e.g., implantable venous catheters, venous ports, tunneled venous catheters, chronic infusion lines or ports, including hepatic artery infusion catheters, pacemakers and pacemaker leads, implantable defibrillators; neurologic/neurosurgical devices (e.g., ventricular peritoneal shunts, ventricular atrial shunts, dural patches and implants to prevent epidural fibrosis post-laminectomy, devices for continuous subarachnoid infusions); gastrointestinal devices (e.g., chronic indwelling catheters, feeding tubes, portosystemic shunts, shunts for ascites, peritoneal implants for drug delivery, peritoneal dialysis catheters, and suspensions or solid implants to prevent surgical adhesions); genitourinary devices (e.g., uterine implants, including intrauterine devices (IUDs) and devices to prevent endometrial hyperplasia, fallopian tubal implants, including reversible sterilization devices, fallopian tubal stents, ureteric stents, chronic indwelling catheters, bladder augmentations, or wraps or splints for vasovasostomy, central venous catheters, urinary catheters; prosthetic heart valves, vascular grafts, ophthalmologic implants (e.g., multino implants and other implants for neovascular glaucoma, drug eluting contact lenses for pterygiums, splints for failed dacrocystalrhinostomy, drug eluting contact lenses for corneal neovascularity, implants for diabetic retinopathy, drug eluting contact lenses for high risk corneal transplants); otolaryngology devices (e.g., ossicular implants, Eustachian tube splints or stents for glue ear or chronic otitis as an alternative to transtempanic drains); catheter cuffs and orthopedic implants (e.g., cemented orthopedic prostheses).
Other examples of implants include drainage tubes, biliary T-tubes, clips, sutures, braids, meshes (e.g., hernia meshes, tissue support meshes), barriers (for the prevention of adhesions), anastomotic devices, anastomotic connectors, ventrical assist devices (e.g., LVAD's), artificial hearts, artificial joints, conduits, irrigation fluids, packing agents, stents, staples, inferior vena cava filters, pumps (e.g., for the delivery of therapeutics), hemostatic implants (e.g., sponges), tissue fillers, surgical adhesion barriers (e.g., INTERCEED, degradable polyester films (e.g., PLLA/PDLLA), CMC/PEO
association complexes (e.g., 07CIPLEX from Fziomed), hyaluronic acid/CMC
films (e.g., SEPRAFILM from Genzyme Corporation), bone grafts, skin grafts, tissue sealants, intrauterine devices (IUD), ligatures, titanium implants (particularly for use in dental applications), chest tubes, nasogastric tubes, percutaneous feeding tubes, colostomy devices, bone wax, and Penrose drains, hair plugs, ear rings, nose rings, and other piercing-associated implants, as well as anaesthetic solutions.
The coating of fibrosis-inhibiting agents) onto or incorporation of a fibrosis-inhibiting agents) into medical devices provides a solution to the clinical problems that can be encountered with these devices. Alternatively, or additional, compositions that comprise anti-scarring agents can be infiltrated in to the space or onto tissue surrounding the area where medical devices are implanted either before, during or after implantation of the devices.
Described below are examples of medical devices whose functioning can be improved by the use of a fibrosis-inhibiting agent as well as methods for incorporating fibrosis-inhibiting agents into or onto these devices and methods for using such devices.
Intravascular Devices The present invention provides for the combination of an anti-scarring agent and an intravascular device. "Intravascular devices" refers to devices that are implanted at least partially within the vasculature (e.g., blood vessels). Examples of intravascular devices that can be used to deliver anti-scarring agents to the desired location include, e.g., catheters, balloon catheters, balloons, stents, covered stents, stent grafts, anastomotic connectors, and guidewires.
In one aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti-scarring agent and an intravascular stent.
"Stent" refers to devices comprising a cylindrical tube (composed of a metal, textile, non-degradable or degradable polymer, and/or other suitable material (such as biological tissue) which maintains the flow of blood from one portion of a blood vessel to another. In one aspect, a stent is an endovascular scaffolding which maintains the lumen of a body passageway (e.g., an artery) and allows bloodflow. Representative examples of stents that can benefit from being coated with or having incorporated therein, a fibrosis-inhibiting agent include vascular stents, such as coronary stents, peripheral stents, and covered stents.
Stents that can be used in the present invention include metallic stents, polymeric stents, biodegradable stents and covered stents. Stents may be self-expandable or balloon-expandable, composed of a variety of metal compounds and/or polymeric materials, fabricated in innumerable designs, used in coronary or peripheral vessels, composed of degradable and/or nondegradable components, fully or partially covered with vascular graft materials (so called "covered stents") or "sleeves", and can be bare metal or drug-eluting.
Stents may be comprise a metal or metal alloy such as stainless steel, spring tempered stainless steel, stainless steel alloys, gold, platinum, super elastic alloys, cobalt-chromium alloys and other cobalt-containing alloys (including ELGILOY (Combined Metals of Chicago, Grove Village, IL), PHYNOX (Alloy Wire International, United Kingdom) and CONICHROME
(Carpenter Technology Corporation, Wyomissing, PA)), titanium-containing alloys, platinum-tungsten alloys, nickel-containing alloys, nickel-titanium alloys (including nitinol), malleable metals (including tantalum); a composite material or a clad composite material and/or other functionally equivalent materials;
and/or a polymeric (non-biodegradable or biodegradable) material.
Representative examples of polymers that may be included in the stent construction include polyethylene, polypropylene, polyurethanes, polyesters, such as polyethylene terephthalate (e.g., DACRON or MYLAR (E. I. DuPont De Nemours and Company, Wilmington, DE)), polyamides, polyaramids (e.g., KEVLAR from E.I. DuPont De Nemours and Company), polyfluorocarbons such as poly(tetrafluoroethylene with and without copolymerized hexafluoropropylene) (available, e.g., under the trade name TEFLON (E. I.
DuPont De Nemours and Company), silk, as well as the mixtures, blends and copolymers of these polymers. Stents also may be made with engineering plastics, such as thermotropic liquid crystal polymers (LCP), such as those formed from p,p'-dihydroxy-polynuclear-aromatics or dicarboxy-polynuclear-aromatics.
Further types of stents that can be used with the described therapeutic agents are described, e.g., in PCT Publication No. WO 01/01957 and U.S. Patent Nos. 6,165, 210; 6,099,561; 6,071,305; 6,063,101; 5,997,468;

5,980,551; 5,980,566; 5,972,027; 5,968,092; 5,951,586; 5,893,840; 5,891,108;
5,851,231; 5,843,172; 5,837,008; 5,766,237; 5,769,883; 5,735,811; 5,700,286;
5,683,448; 5,679,400; 5,665,115; 5,649,977; 5,637,113; 5,591,227; 5,551,954;
5,545,208; 5,500,013; 5,464,450; 5,419,760; 5,411,550; 5,342,348; 5,286,254;
and 5,163,952. Removable drug-eluting stents are described, e.g., in Lambert, T. (1993) J. Am. Col!. Cardiol.: 21: 483A. Moreover, the stent may be adapted to release the desired agent at only the distal ends, or along the entire body of the stent.
Balloon over stent devices, such as are described in Wilensky, R.L. (1993) J. Am. Col!. Cardiol.: 21: 185A, also are suitable for local delivery of a fibrosing agent to a treatment site.
In addition to using the more traditional stents, stents that are specifically designed for drug delivery can be used. Examples of these specialized drug delivery stents as well as traditional stents include those from Conor Medsystems (Palo Alto, CA) (e.g., U.S. Patent. Nos. 6,527,799;
6,293,967; 6,290,673; 6,241,762; U.S. Patent Application Publicatiori Nos.
200310199970 and 2003/0167085; and PCT Publication No. WO 03/015664).
Examples of intravascular stents, which may be combined with one or more therapeutic agents according to the present invention, include commercially available products. The stent may be self-expanding or balloon expandable (e.g., STRECKER stent by Medi-Tech/Boston Scientific Corporation), or implanted by a change in temperature (e.g., nitinol stent).
Self-expanding stents that can be used include the coronary WALLSTENT and the SCIMED RADIUS stent from Boston Scientific Corporation (Natick, MA) and the GIANTURCO stents from Cook Group, Inc. (Bloomington, IN). Examples of balloon expandable stents that can be used include the CROSSFLEX stent, BX-VELOCITY stent and the PALMAZ-SCHAT2 crown and spiral stents from Cordis Corporation (Miami Lakes, FL), the V-FLEX PLUS stent by Cook Group, Inc., the NIR, EXPRESS and LIBRERTE stents from Boston Scientific Corporation, the ACS MULTILINK, MULTILINK PENTA, SPIRIT, and CHAMPION stents from Guidant Corporation, and the Coronary Stent S670 and S7 by Medtronic, Inc. (Minneapolis, MN).
Other examples of stents that can be combined with a fibrosing agent in accordance with the invention include those from Boston Scientific Corporation, (e.g., the drug-eluting TAXUS EXPRESS Paclitaxel-Eluting Coronary Stent System; over the wire stent stents such as the Express Coronary Stent System and NIR Elite OTW Stent System; rapid exchange stents such as the EXPRESSZ Coronary Stent System and the NIR ELITE
MONORAIL Stent System; and self-expanding stents such as the MAGIC
WALLSTENT Stent System and RADIUS Self Expanding Stent); Medtronic, Inc.
(Minneapolis, MN) (e.g., DRIVER ABT578-eluting stent, DRIVER ZIPPER MX
Multi-Exchange Coronary Stent System and the DRIVER Over-the-Wire Coronary Stent System; the S7 ZIPPER MX Multi-Exchange Coronary Stent System; S7, S670, S660, and BESTENT2 with Discrete Technology Over-the-Wire Coronary Stent System); Guidant Corporation (e.g., cobalt chromium stents such as the MULTI-LINK VISION Coronary Stent System; MULTI-LINK
ZETA Coronary Stent System; MULTI-LINK PIXEL Coronary Stent System;
MULTI-LINK ULTRA Coronary Stent System; and the MULTI-LINK
FRONTIER); Johnson & Johnson/Cordis Corporation (e.g., CYPHER sirolimus-eluting Stent; PALMAZ-SCHATZ Balloon Expandable Stent; and S.M.A.R.T.
Stents); Abbott Vascular (Redwood City, California) (e.g., MATRIX LO Stent;
TRIMAXX Stent; and DEXAMET stent); Conor Medsystems (Menlo Park, California) (e.g., MEDSTENT and COSTAR stent); AMG GmbH (Germany) (e.g.,~ PICO Elite stent); Biosensors International (Singapore) (e.g., MATRIX
stent, CHAMPION Stent (formerly the S-STENT), and CHALLENGE Stent);
Biotronik (Switzerland) (e.g., MAGIC AMS stent); Clearstream Technologies (Ireland) (e.g., CLEARFLEX stent); Cook Inc. (Bloomington, Indiana) (e.g., V-FLEX PLUS stent, ZILVER PTX self-expanding vascular stent coating, LOGIX
PTX stent (in development); Devax (e.g., AXXESS stent) (Irvine, CA); DISA
Vascular (Pty) Ltd (South Africa) (e.g., CHROMOFLEX Stent, S-FLEX Stent, S-FLEX Micro Stent, and TAXOCHROME DES); Intek Technology (Baar, Switzerland) (e.g., APOLLO stent); Orbus Medical Technologies (Hoevelaken, The Netherlands) (e.g., GENOUS); Sorin Biomedica (Saluggia, Italy) (e.g., JANUS and CARBOSTENT); and stents from Bard/Angiomed GmbH
Medizintechnik KG (Murray Hill, NJ), and Blue Medical Supply & Equipment (Mariettta, GA), Aachen Resonance GmbH (Germany); Eucatech AG
(Germany), Eurocor GmbH (Bonn, Gemany), Prot, Goodman, Terumo (Japan), Translumina GmbH (Germany), MIV Therapeutics (Canada), Occam International B.V. (Eindhoven, The Netherlands), Sahajanand Medical Technologies PVT LTD. (India); AVI Biopharma/Medtronic/ Interventional Technologies (Portland, OR) (e.g., RESTEN NG-coated stent); and Jomed (e.g., FLEXMASTER drug-eluting stent) (Sweden).

Generally, stents are inserted in a similar fashion regardless of the site or the disease being treated. Briefly, a preinsertion examination, usually a diagnostic imaging procedure, endoscopy, or direct visualization at the time of surgery, is generally first performed in order to determine the appropriate positioning for stent insertion. A guidewire is then advanced through the lesion or proposed site of insertion, and over this is passed a delivery catheter which allows a stent in its collapsed form to be inserted.
Intravascular stents may be inserted into an artery such as the femoral artery in the groin and advanced through the circulation under radiological guidance until they reach the anatomical location of the plaque in the coronary or peripheral circulation. Typically, stents are capable of being compressed, so that they can be inserted through tiny cavities via small catheters, and then expanded to a larger diameter once they are at the desired location. The delivery catheter then is removed, leaving the stent standing on its own as a scaffold. Once expanded, the stent physically forces the walls of the passageway apart and holds them open. A post insertion examination, usually an x-ray, is often utilized to confirm appropriate positioning.
Stents are typically maneuvered into place' under, radiologic or direct visual control, taking particular care to place the stent precisely within the vessel being treated. In certain aspects, the stent can further include a radio-opaque, echogenic material, or MRI responsive material (e.g., MRI contrast agent) to aid in visualization of the device under ultrasound, fluoroscopy and/or magnetic resonance imaging. The radio-opaque or MRI visible material may be in the form of one or more markers (e.g., bands of material that are disposed on either end of the stent) that may be used to orient and guide the device during the implantation procedure.
In another aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti-scarring agent and an intravascular catheter.
"Intravascular Catheter" refers to any intravascular catheter containing one or more lumens suitable for the delivery of aqueous, microparticulate, fluid, or gel formulations into the bloodstream or into the vascular wall. These formulations may contain a biologically active agent (e.g., an anti-scarring agent). Numerous intravascular catheters have been described for direct, site-specific drug delivery (e.g., microinjector catheters, catheters placed within or immediately adjacent to the target tissue), regional drug delivery (i.e., catheters placed in an artery that supplies the target organ or tissue), or systemic drug delivery (i.e., intra-arterial and intravenous catheters placed in the peripheral circulation). . For example, catheters and balloon catheters can deliver anti-fibrosing agents from an end orifice, through one or more side ports, through a microporous outer structure, or through direct injection into the desired tissue or vascular location.
A variety of catheters are available for regional or localized arterial drug-delivery. Intravascular balloon and non-balloon catheters for delivering drugs are described, for example, in U.S. Patent Nos. 5,180,366; 5,171,217;
5,049,132; 5,021,044; 6,592,568; 5,304,121; 5,295,962; 5,286,254; 5,254,089;
5,112,305; PCT Publication Nos WO 93/08866, WO 92/11890, and WO
92/11895; and Riessen et al. (1994) JACC 23: 1234-1244, Kandarpa K. (2000) J. Vasc. Interv. Radio. 11 (suppl.): 419-423, and Yang, X. (2003) Imaging of Vascular Gene Therapy 228(1): 36-49.
Representative examples of drug delivery catheters include balloon catheters, such as the CHANNEL and TRANSPORT balloon catheters from Boston Scientific Corporation (Natick, MA) and Stack Perfusion Coronary Dilitation catheters from Advanced Cardiovascular Systems, Inc. (Santa Clara, CA). Other examples of drug delivery catheters include infusion catheters, such as the CRESCENDO coronary infusion catheter available from Cordis Corporation (Miami Lakes, FL), the Cragg-McNamara Valved Infusion Catheter available from Microtherapeutics, Inc. (San Clemente, CA), the DISPATCH
catheter from Boston Scientific Corporation, the GALILEO Centering Catheter from Guidant Corporation (Houston, TX), and infusion sleeve catheters, such as the INFUSASLEEVE catheter from LocaIMed, Inc. (Sunnyvale, CA). Infusion sleeve catheters are described in, e.g., U.S. Patent Nos. 5,318,531;
5,336,178;
5,279,565; 5,364,356; 5,772,629; 5,810,767; and 5,941,868. Catheters that mechanically or electrically enhance drug delivery include, for example, pressure driven catheters (e.g., needle injection catheters having injector ports, such as the INFILTRATOR catheter available from InterVentional Technologies, Inc. (San Diego, CA)) (see, e.g., U.S. Patent No. 5,354,279) and ultrasonically assisted (phonophoresis) and iontophoresis catheters (see, e.g., Singh, J., et al. (1989) Drug Des. Deliv.: 4: 1-12 and U.S. Patent Nos. 5,362,309;
5,318,014;
5,315,998; 5,304,120; 5,282,785; and 5,267,985).

In one aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti-scarring agent and a drug delivery balloon.
"Drug-Delivery Balloon" refers to an intra-arterial balloon (typically based upon percutaneous angioplasty balloons) suitable for insertion into a peripheral artery (typically the femoral artery) and manipulated via a catheter to the treatment site (either in the coronary or peripheral circulation).
Numerous drug delivery balloons have been developed for local delivery of therapeutic agents to the arterial wall such as "sweaty balloons," "channel balloons,"
"microinjector balloons," "double balloons," "spiral balloons" and other specialized drug-delivery balloons. Other examples of balloons include BHP
balloons and Transurethral and Radiofrequency Needle Ablation (TUNA or RFNA)) balloons for prostate applications.
In addition, numerous drug delivery balloons have been developed for local delivery of therapeutic agents to the arterial wall.
Representative examples of drug delivery balloons include porous (WOLINSKY) balloons, available from Advanced Polymers (Salem, NH), described in, e.g., U.S. Patent No. 5,087,244. Microporous and macroporous balloons (i.e., "sweaty balloons") for use in infusion catheters are described in, e.g., Lambert, C.R. et al. (1992) Circ. Res. 71: 27-33. Other types of specialized drug delivery balloons include hydrogel coated balloons (e.g., ULTRATHIN GLIDES from Boston Scientific Corporation) (see, e.g., Fram, D.B.
et al. (1992) Circulation: 86 Suppl. I: 1-380), "channel balloons" (see, e.g., U.S.
Patent Nos. 5,860,954; 5,843,033; and 5,254,089, and Hong, M.K., et al. (1992) Circulation: 86 Suppl. I: 1-380), "microinjector balloons" (see, e.g., U.S.
Patent Nos. 5,681,281 and 5,746,716), "double balloons," described in, e.g., U.S.
Patent No. 6,544,221, and double-layer channeled perfusion balloons (such as the REMEDY balloon from Boston Scientific Corportion), and "spiral balloons"
(see, e.g., U.S. Patent Nos. 6,527,739 and 6,605,056). Drug delivery catheters that include helical (i.e., spiral) balloons are described in, e.g., U.S.
Patent Nos.
6,190,356; 5,279,546; 5236424, 5,226,888; 5,181,911; 4,824,436; and 4,636,195.
The balloon catheter systems that can be used include systems in which the balloon can be inflated at the desired location the desired fibrosis-inducing agents can be delivered through holes that are located in the balloon wall. Other balloon catheters that can be used include systems that have a plurality of holes that are located between two balloons. The system can be guided into the desired location such that the inflatable balloon components are located on either side of the specific site that is to be treated. The balloons can then be inflated to isolate the treatment area. The compositions containing the fibrosing agent are then injected into the isolated area through the plurality of holes between the two balloons. Representative examples of these types of drug delivery balloons are described in U.S. Patent. Nos. 5,087,244, 6,623,452, 5,397,307, 4,636,195 and 4,994,033.
The compositions of the invention can be delivered using a catheter that has the ability to enhance uptake or efficacy of the compositions of the invention. The stimulus for enhanced uptake can include the use of heat, the use of cooling, the use of electrical fields or the use of radiation (e.g., ultraviolet light, visible light, infrared, microwaves, ultrasound or X-rays).
Further Representative examples of catheter systems that can be used are described in U.S. Patent. Nos. 5,362,309 and 6,623,444; U.S. Patent Application Publication Nos. 2002/0138036 and 2002/0068869; and PCT
Publication Nos. WO 01/15771; WO 94/05361; WO 96/04955 and WO
96/22111.
In another aspect of the invention, the compositions of the inventions can be delivered into the treatment site and/or into the tissue surrounding the treatment site by using catheter systems that have one or more injectors that can penetrate the surrounding tissue. Following insertion into the appropriate vessel, the catheter can be maneuvered into the desired position such that the injectors are aligned with or adjacent to the tissue. The injectors) are inserted into the desired location, for example by direct insertion into the tissue, by inflating the balloon or mechanical rotation of the injector, and the composition of the invention is injected into the desired location.
Representative examples of catheters that can be used for this application are described in and U.S. Patent Application Publication No. 2002/0082594 and U.S. Patent. Nos. 6,443,949; 6,488,659; 6,569,144; 5,609,151; 5,385,148;
5,551,427; 5,746,716; 5,681,281; and 5,713,863.
In another aspect of the invention, the catheter may be adapted to deliver a thermoreversible polymer composition. For the site-specific delivery of these materials, a catheter delivery system has the ability to either heat the composition to above body temperature or to cool the composition to below body temperature such that the composition remains in a fluent state within the catheter delivery system. The catheter delivery system can be guided to the desired location and the composition of the invention can be delivered to the surface of the surrounding tissue or can be injected directly into the surrounding tissue. A representative example of a catheter delivery system for direct injection of a thermoreversible material is described in U.S. Patent. No.
6,488,659. Representative examples of catheter delivery systems that can deliver the thermoreversible compositions to the surface of the vessel are described in U.S. Patent. Nos. 6,443,941; 6,290,729; 5,947;977; 5,800,538; .
and 5,749,922.
In another aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti-scarring agent and an anastomotic connector device.
"Anasomotic connector device" refers to any vascular device that mechanizes the creation of a vascular anastomosis (i.e., artery-to-artery, vein-to-artery, artery-to-vein, artery-to-synthetic graft, synthetic graft-to-artery, vein-to-synthetic graft or synthetic graft-to-vein anastomosis) without the manual suturing that is typically done in the creation of an anastomosis. The term also refers to anastomotic connector devices (described below), designed to produce a facilitated semiautomatic vascular anastomosis without the use of suture and reduce connection time substantially (often to several seconds), where there are numerous types and designs of such devices. The term also refers to devices which facilitate attachment of a vascular graft to an aperture or orifice (e.g., in the side or at the end of a vessel) in a target vessel.
Anastomotic connector devices may be anchored to the outside of a blood vessel, and/or into the wall of a blood vessel (e.g., into the adventitial, intramural, or intimal layer of the tissue), and/or a portion of the device may reside within the lumen of the vessel.
Anastomotic connector devices also may be used to create new flow from one structure to another through a channel or diversionary shunt.
Accordingly, such devices (also referred to herein as "bypass devices") typically include at least one tubular structure, wherein a tubular structure defines a lumen. Anastomotic connector devices may include one tubular structure or a plurality of tubular structures through which blood can flow. At least a portion of the tubular structure resides external to a blood vessel (i.e., extravascular) to provide a diversionary passageway. A portion of the device also may reside within the lumen and/or within the tissue of the blood vessel.

Examples of anastomotic connector devices are described in co-pending application entitled, "Anastomotic Connector Devices", filed May 23, 2003 (U.S. Ser. No. 60/473,135). Representative examples of anastomotic connector devices include, without limitation, vascular clips, vascular sutures, vascular staples, vascular clamps, suturing devices, anastomotic coupling devices (i.e., anastomotic couplers), including couplers that include tubular segments for carrying blood, anastomotic rings, and percutaneous in situ coronary artery bypass (PISCAB and PICVA) devices. Broadly, anastomotic connector devices may be classified into three categories: (1 ) automated and modified suturing methods and devices, (2) micromechanical devices, and (3) anastomotic coupling devices.
(1 ) Automated and Modified Suturing Methods and Devices Automated sutures and modified suturing methods generally facilitate the rapid deployment of multiple sutures, usually in a single step, and eliminate the need for knot tying or the use of aortic side-biting clamps.
Suturing devices include those devices that are adapted to be minimally invasive such that anastomoses are formed between vascular conduits and hollow organ structures by applying sutures or other surgical fasteners through device ports or other small. openings. .With these devices, sutures and other fasteners are applied in a relatively quick and automated manner within bodily areas that have limited access. By using minimally invasive means for establishing anastomoses, there is less blood loss and there is no need to temporarily stop the flow of blood distal to the operating site. For example, the suturing device may be composed of a shaft-supported vascular conduit that is adapted for anastomosis and a collar that is slideable on the shaft configured to hold a plurality of needles and sutures that passes through the vascular conduit. See, e.g., U.S. Patent No. 6,709,441. The suturing device may be composed of a carrier portion for inserting 'graft, arm portions that extend to support the graft into position, and a needle assembly adapted to retain and advance coil fasteners into engagement with the vessel wall and the graft flange to complete the anastomosis. See, e.g., U.S. Patent No. 6,709,442.
The suturing device may include two oblong interlinked members that include a split bush adapted for suturing (e.g., U.S. Patent No. 4,350,160).
One representative example of a suturing device is the HEARTFLOW device, made by Perclose-Abbott Labs, Redwood City, CA (see generally, U.S. Patent Nos. 6,358,258, 6,355,050, 6,190,396, and 6,036,699, and PCT Publication No. WO 01/19257) The nitinol U-CLIP suture clip device by Coalescent Surgical (Sunnyvale, CA) consists of a self-closing nitinol wire loop attached to a flexible member and a needle with a quick release mechanism. This device facilitates the construction of anastomosis by simplifying suture management and eliminating knot tying (see generally, U.S. Patent Nos. 6,074,401 and 6,149,658, and PCT Publication Nos. WO 99/62406, WO 99/62409, WO
00/59380, W O 01 /17441 ).
The ENCLOSE Anastomotic Assist Device (Novare Surgical Systems, Cupertino, CA) allows a surgeon to create a sutured anastomosis using standard suturing techniques but without the use of a partial occluding side-biting aortic clamp, avoiding aortic wall distortion (see U.S. Patent Nos.
6,312,445 and 6,165,186).
In one aspect, automated and modified suturing methods and devices can deliver a surgical fastener (e.g., a suture or suture clip) that comprises an anti-scarring agent. In another aspect, automated and modified suturing methods and devices can deliver a vascular graft that comprises an anti-scarring agent to complete an anastomosis.
(2) Micromechanical devices Micromechanical devices are used to create an anastomosis and/or secure a graft vessel to the site of an anastomosis. Representative examples of micromechanical devices include staples (either penetrating or non-penetrating) and clips.
Anastomotic staple and clip devices may take a variety of forms and may be made from different types of materials. For example, staples and clips may be formed of a metal or metal alloy, such as titanium, nickel-titanium alloy, or stainless steel, or a polymeric material, such as silicone, poly(urethane), rubber, or a thermoplastic elastomer.
The polymeric material may be an absorbable or biodegradable material designed to dissolve after completion of the anastomosis.
Biodegradable polymers include, for example, homopolymers and copolymers that comprise one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, s-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y-decanolactone, b-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-Zone.
A variety of devices for guiding staples and clips into position also have been described.
One manufacturer of non-penetrating staples for use in the creation of anastomosis is United States Surgical Corp. (Norwalk, CT). The VCS system (Autosuture) is an automatic stapling device that applies non-penetrating, titanium vascular clips which are usually used in an interrupted fashion to avert tissue edges with high compressive forces. (See, e.g., U.S.
Patent Nos. 6,440,146, 6,391,039, 6,024,748, 5,833,698, 5,799,857, 5,779,718, 5,725,538, 5,725,537, 5,720,756, 5,360,154, 5,193,731, and 5,005,749 for the description of anastomotic connector devices made by U.S. Surgical).
An anastomotic clip may be composed of a shape memory material, such as nitinol, which is self closing between an open U-shaped configuration and a closed configuration. See, e.g., U.S. Patent No.
6,641,593.
The anastomotic clip may be composed of a wire having a shape memory that defines a closed configuration which may be substantially spiral-shaped and having a needle that may be releasably attached to the clip. See, e.g., U.S.
Patent No. 6,551,332. Other anastomotic clips are described in, e.g., U.S.
Patent Nos. 6,461,365; and 6,514,265.
Automatic stapling devices are also made by Bypass/Ethicon, Inc.
(Somerville, NJ) and are described in, e.g., U.S. Patent Nos. 6,193,129;
5,632,433; 5,609,285; 5,533,661; 5,439,156; 5,350,104; 5,333,773; 5,312,024;
5,292,053; 5,285,945; 5,275,322; 5,271,544; 5,271,543 and 5,205,459 and WO
03/02016. Resorbable surgical staples that include a polymer blend that is rich in glycolide (i.e., 65 to 85 weight °l° polymerized glycolide) are described in, e.g., U.S. Patent No. 4,741,337 and 4,889,119. Surgical staples made from a blend of lactide/glycolide-copolymer and polyp-dioxanone) are described in U.S. Patent No. 4,646,741. Other types of stapling devices are described in, e.g., U.S. Patent Nos. 5,234,447; 5,904,697 and 6,565,582; and U.S.
Publication No. 2002/0185517A1.
In another aspect, the micromechanical device may be an anastomotic clip. For example, an anastomotic clip may be composed of a shape memory material, such as nitinol, which is self-closing between an open U-shaped configuration and a closed configuration. See, e.g., U.S. Patent No.
6,641,593. The anastomotic clip may be composed of a wire having a shape memory that defines a closed configuration which may be substantially spiral-shaped and having a needle that may be releasably attached to the clip. See, e.g., U.S. Patent No. 6,551,332. Other anastomotic clips are described in, e.g., U.S. Patent Nos. 6,461,365; 6,187,019; and 6,514,265.
In one aspect, the present invention provides for the combination of a micromechanical anastomotic device (e.g., a staple or a clip) and an anti-scarring agent.
(3) Anastomotic Coupling Devices Anastomotic coupling devices may be used to connect a first blood vessel to a second vessel, either with or without a graft vessel, for completion of an anastomosis. In one aspect, anastomotic coupling devices facilitate automated attachment of a graft or vessel to an aperture or orifice (e.g., in the side or at the end of a vessel) in a target vessel without the use of sutures or staples. In another aspect, the anastomotic coupling device comprises a tubular structure defining a lumen through which blood may flow (described below).
Anastomotic coupling devices that facilitate automated attachment of a graft or vessel to an aperture or orifice in a target vessel may tafce a variety of forms and may be made from a variety of materials. Typically, such devices are made of a biocompatible material, such as a polymer or a metal or metal alloy. For example, the device may be formed from a synthetic material, such as a fluoropolymer, such as expanded poly(tetrafluoroethylene) (ePTFE) (ePTFE) sold under the trade name GORE-TEX available from W.L. Gore &
Associates, Inc. or fluorinated ethylene propylene (FEP), a polyurethane, polyethylene, polyamide (nylon), silicone, polypropylene, polysulfone, or a polyester.
Anastomotic coupling devices may include an absorbable or biodegradable material designed to dissolve after completion of the anastomosis. Biodegradable polymers include, for example, homopolymers and copolymers that comprise one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, s-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y-decanolactone, ~-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-Zone.

The device may include a metal or metal alloy (e.g., nitinol, stainless steel, titanium, iron, nickel, nickel-titanium, cobalt, platinum, tungsten, tantalum, silver, gold, molybdenum, chromium, and chrome), or a combination of a metal and a polymer.
The device may be anchored to the outside of a vessel, within the tissue that surrounds the lumen of a blood vessel, and/or a portion of the device may reside within the lumen of the vessel.
In one aspect, the anastomotic coupler may be an artificially formed aperture connector that is placed in the side wall of the target vessel so that the tubular graft conduit may be extended from the target vessel. The connector may include a plurality of tissue-piercing members and retention fingers disposed in a concentric annular array which may be passed through the side wall of the tubular graft conduit for securing and retaining the graft to the connector in a fluid-tight configuration. See, e.g., U.S. Patent No.
6,702,829 and 6,699,256.
In another aspect, the anastomotic coupler may be in the form of a frame. For example, the frame may be configured to be deformable and scissor-shaped such that spreading members are moveable to secure a graft vessel upon insertion into a target vessel. See, e.g., U.S. Patent No.
6,179,849.
In another aspect, the anastomotic coupler may be a ring-like device that is used as an anastomotic interface between a lumen of a graft and an opening in a lumen of a target vessel. For example, the anastomotic ring may be composed of stainless steel alloy, titanium alloy, or cobalt alloy and have a flange with an expandable diameter. See, e.g., U.S. Patent No.
6,699,257. Anastomosis rings are also described in, e.g., U.S. Patent No.
6,248,117.
In another aspect, the anastomotic coupler is resorbable.
Resorbable anastomotic coupling devices may include, for example, a polymeric blend that is rich in glycolide (i.e., 65 to 85 weight % polymerized glycolide) (see, e.g., U.S. Patent No. 4,741,337 and 4,889,119) or a blend of lactide/glycolide-copolymer and polyp-dioxanone) (see, e.g., U.S. Patent No.
4,646,741 ).
In another aspect, the anastomotic coupler includes a bioabsorbable, elastomeric material. Representative examples of elastomeric materials for use in resorbable devices are described in, e.g., U.S. Patent No.
5,468,253.
In another aspect, the anastomotic coupler may be used to connect a first blood vessel to a second vessel, either with or without a graft vessel. For example, the anastomotic coupler may be a device that serves to interconnect two vessels in a side-to-side anastomosis, such as when grafting two juxtaposed cardiac vessels. The anastomotic coupler may be configured as two partially opened cylindrical segments that are interconnected along the periphery by a flow opening whereby the device may be inserted in a minimally-invasive manner which then conforms to provide pressure against the interior wall when in the original configuration such that leakage is prevented. See, e.g., U.S. Patent Nos. 6,464,709; 6,458,140 and 6,251,116 and U.S.
Application Publication No. 2003/0100920A1.
In another aspect, the anastomotic coupler may also be incorporated in the design of a vascular graft to eliminate the step of attaching the interface prior to deployment. For example, the anastomotic coupler may have a leading and rear petal for dilating the vessel opening during advancement, and a base which is configured for attachment to a graft while forming a seal with the opening of the vessel. See, e.g., U.S. Patent No.
6,702,828.
In another aspect, the anastomotic coupler may be in the form of a frame. For example, the anastomotic coupler may be composed of a deformable, scissor-shaped frame with spreading members that is inserted into a target vessel. See, e.g., U.S. Patent No. 6,179,849.
In another aspect, the anastomotic coupling device may include a graft that incorporates fixation mechanisms (e.g., a collet or a grommet) at its opposite ends and a heating element to create a thermal bond between the graft and a blood vessel (see, e.g., U.S. Patent Nos. 6,652,544 and 6,293,955).
In another aspect, the anastomotic coupling device includes a compressible, expandable fitting for securing the ends of a bypass graft to two vessels. The fitting may be incorporated in the bypass graft design to eliminate the step of attaching the graft to the fitting prior to deployment (see, e.g., U.S.
Patent No. 6,494,889).
In another aspect, the anastomotic coupling device includes a pair of coupling disc members for joining two vessels in an end-to-end or end-to-side fashion. One of the members includes hook members, while the other member has receptor cavities aligned with the hooks for locking everted tissue of the vessels together (see, e.g., U.S. Patent No. 4,523,592).
Representative examples of anastomotic connector devices of Bypass/Ethicon, Inc. are described in U.S. Application Publication Nos.
US2002/0082625A1 and 2003/0100910A1 and U.S. Patent Nos. 6,036,703, 6,036,700, 6,015,416, and 5,346,501.
Other anastomotic coupling devices are those described in e.g., U.S. Patent No. 6,036,702; 6,508,822; 6,599,303; 6,673,084, 5,695,504;
6,569,173; 4,931,057; 5,868,763; 4,624,257; 4,917,090; 4,917,091; 5,697,943;
5,562,690; 5,454,825; 5,447,514; 5,437,684; 5,376,098; 6,652,542; 6,551,334;
and 6,726,694 and U.S. Application Publication Nos. 2003/0120293A1 and 2004/0030348A1.
Anastomotic coupling devices may include proximal aortic connectors and distal coronary connectors. For example, aortic anastomotic connectors include devices such as the SYMMETRY Bypass Aortic Connector device made by St. Jude Medical, Inc. (Maple Grove, MN), which consists of an aortic cutter or hole punch assembly and a graft delivery system. The aortic hole punch is a cylindrical cutter with a barbed needle that provides an anchor and back pressure for the rotating cutter to core a round hole in the wall of the aorta. The graft delivery system is a radially expandable nitinol device that holds the vein graft with small hooks which pierce through vein graft wall.
The graft is fixed to the aorta through use of an inner and outer ring of struts or flanges. This and other anastomotic connector devices by St. Jude are described in U.S. Patent Nos. 6,309,416, 6,302,905, 6,152,937, and PCT
Publication Nos. WO 00/27312 and WO 00/27311.
The CORLINK Automated Anastomotic connector device, which is produced by the CardioVations division of Ethicon, Inc. (Johnson & Johnson, Somerville, NJ), uses a nitinol metal alloy fastener to connect the grafted vessel to the aorta. It consists of a central cylindrical body made of interconnected elliptical arches and two sets of several pins radiating from each end. The graft is loaded into a CORLINK insertion instrument and deployed to create an anastomosis in one step.
Further examples of anastomotic coupling devices include those made by Cardica (see, U.S. Patent Nos. 6,719,769; 6,419,681 and 6,537,287), Converge Medical (formerly Advanced Bypass Technologies), Onux Medical (see, e.g., PCT Publication No. WO 01/34037) and Ventrica, Menlo Park, CA

(VENTRICA Magnetic Vascular Positioner) (see, e.g., U.S. Patent Nos.
6,719,768; 6,517,558 and 6,352,543).
As described above, an anastomotic coupling device may comprise a tubular structure defining a lumen through which blood may flow.
These types of devices (also referred to herein as "bypass devices") can function as an artificial passageway or conduit for fluid communication between blood vessels and can be used to divert (i.e., shunt) blood from one part of a blood vessel (e.g., an artery) to another part of the same vessel, or to a second vessel (e.g., an artery or a vein) or to multiple vessels (e.g., a vein and an artery). In one aspect of the invention, the anastomotic device is a bypass device.
Bypass devices may be used in a variety of end-to-end and end-to-side anastomotic procedures. The bypass device may be placed into a patient where it is desired to create a pathway between two or more vascular structures, or between two different parts of the same vascular structure. For example, bypass devices may be used to create a passageway which allows blood to flow around a blood vessel, such as an artery (e.g., coronary artery, carotid artery, or artery supplying the lower limb), which has become damaged or completely or partially obstructed. Bypass devices may be used in coronary artery bypass surgery to shunt blood from an artery, such as the aorta, to a portion of a coronary artery downstream from an occlusion in the artery.
Certain types of anastomotic coupling devices are configured to join two abutting vessels. The device can further include a tubular segment to shunt blood to another vessel. These types of connectors are often used for end-to-end anastomosis if a vessel is severed or injured.
Bypass devices include at least one tubular structure having a first end and a second end, which defines a single lumen through which blood can flow, or may include more than one tubular structure, defining multiple lumens through which blood can flow. The tubular structure includes an extravascular portion and may, optionally, include an intravascular portion. The extravascular portion resides external to the adventitial tissue of a blood vessel, whereas the intravascular portion may reside within the vessel lumen or within the intimal, medial, and/or adventitial tissue.
The configuration of the tubular segment may take a variety of forms. For example, the tubular portion may be generally straight, bent or curved (e.g., L-shaped or helical), tapered, branched (e.g., bifurcated or trifurcated), or may include a network of conduits through which blood may flow.
Generally, straight or bent devices have a single lumen through which blood may flow, while branched conduits (e.g., generally T-shaped and Y-shaped devices) and conduit networks (described below) have two or more lumens through which blood may flow. A tubular structure may be in the form, for example, of a hollow cylinder and may or may not include a support structure, such as a mesh or porous framework. Depending on the procedure, the device may be biodegradable or non-biodegradable; expandable or rigid; metal and/or polymeric; and/or may include a shape-memory material (e.g., nitinol). In certain embodiments, the device may include a self-expanding stent structure.
Bypass devices typically are made of a biocompatible material.
Any of the materials described above for other types of connectors may be used to make a bypass device, such as a synthetic or naturally-derived polymer, or a metal or metal alloy. For example, the device may be formed from a synthetic material, such as a fluoropolymer, such as expanded poly(tetrafluoroethylene) (ePTFE) or fluorinated ethylene propylene (FEP), a polyurethane, polyethylene, polyamide (nylon), silicone, polypropylene, polysulfone, or a polyester and/or a naturally derived material, such as collagen or a polysaccharide. The device may include a metal or metal alloy (e.g., nitinol, stainless steel, titanium, nickel, nickel-titanium, cobalt, platinum, iron, tungsten, tantalum, silver, gold, molybdenum, chromium and chrome), or a combination of a metal and a polymer. Other types of devices include a natural graft material (e.g., autologous vessel, homologous vessel, or xenograft), or a combination of a synthetic and a natural graft material. In another aspect, the bypass device may be formed of an absorbable or biodegradable material designed to dissolve after completion of the anastomosis (e.g., polylactide, polyglycolide, and copolymers of lactide and glycolide). In yet another aspect, demineralized bone may be used to provide a pliable tubular conduit (see, e.g., U.S. Patent No. 6,290,718).
The tubular structures) include a proximal end that may be configured for attachment to a proximal blood vessel and a distal end configured for attachment to a distal blood vessel. As described above, an anastomosis may be described as being either "proximal" or "distal" depending on its location relative to the vascular obstruction. The "proximal"
anastomosis may be formed in a proximal blood vessel, and the "distal" anastomosis may be formed in a distal blood vessel, which may the same vessel or a different vessel than the proximal vessel. The terms "distal" and "proximal" may also be used to describe the direction that blood flows through a tubular structure from one vessel into another vessel. For example, blood may flow from a proximal vessel (e.g., the aorta) into a distal vessel, such as a coronary artery to bypass an obstruction in the coronary artery.
The tubular structure may be attached directly to a proximal or distal blood vessel. Alternatively, the bypass device may further include a graft vessel or be configured to receive a graft vessel, which can be connected to the same or a different blood vessel for completion of the anastomosis.
Representative examples of graft vessels include, for example, vascular grafts or grafts used in hemodialysis applications (e.g., AV graft, AV shunt, or AV
graft).
In one aspect, a tubular anastomotic coupler includes a proximal end that is attached to a proximal vessel and a distal end that is used to attach a bypass graft. The bypass graft can be secured to the distal vessel to complete the anastomosis. The direction of blood flow can be from the proximal blood vessel and into the proximal end of the tubular structure.
Blood can exit through the distal end of the tubular structure and into the graft vessel.
In another aspect, the tubular anastomotic coupler includes a proximal end that is attached to a graft vessel, which is secured to the proximal blood vessel, and a distal end that is configured for attachment to a distal blood vessel. The direction of blood flow can be from the proximal vessel into the graft vessel and into the proximal end of the tubular structure. Blood can exit through the distal end of the tubular structure and into the distal vessel.
Anastomotic bypass devices may be anchored to a blood vessel in a variety of ways and may be attached to a blood vessel for the formation of an anastomosis with or without the use of sutures. Bypass devices may be attached to the outside of a blood vessel, and/or a portion of the device may be implanted into a vessel. For example, a portion of the implanted device may reside within the lumen of the vessel (i.e., endoluminally), and/or a portion of the implanted device may reside intravascularly (i.e., within the intimal, intramural, and/or adventitial tissue of the blood vessel). In one aspect, at least one of the tubular structures, or a portion thereof, may be inserted into the end of a vessel or into the side of a blood vessel. The device may be secured directly to the vessel using, for example, a fastener, such as sutures, staples, or clips and/or an adhesive. Bypass devices may include an interface to secure the conduit to a target vessel without the use of sutures. The interface may include means, such as, for example, hooks, barbs, pins, clamps, or a flange or lip for coupling the device to the site of an anastomosis.
Representative examples of anastomotic coupling devices that include at least one tubular portion include, without limitation, devices used for end-to-end anastomosis procedures (e.g., anastomotic stents and anastomotic sleeves) and end-to-side anastomosis procedures (e.g., single-lumen and multi-lumen bypass devices).
In one aspect of the invention, the anastomotic coupling device comprises a single tubular portion that may by used as a shunt to divert blood from a source vessel to a graft vessel (e.g., in an end-to-side anastomosis procedure). In one aspect, an end of the tubular portion may be connected directly or indirectly to a target vessel, as described above. The opposite end of the tubular portion may be attached to a graft vessel, where the graft vessel may be secured to a target vessel to complete the anastomosis.
The tubular portions) may be straight or may have a curved or bent shape (e.g., L-shaped or helical) and may be oriented orthogonally or at an angle relative to the vessel to which it is connected. In one aspect, the conduit may be secured into the site by, for example, a fastener, such as staples, clamps, or hooks, or by adhesives, radiofrequency sealing, or by other methods known to those skilled in the art.
In one aspect, the anastomotic coupling device may be, for example, a tubular metal braided graft with suture rings welded at the distal end to provide a means for securing in place to the target vessel. See, e.g., U.S.
Patent No. 6,235,054. Other types of conduits that are secured into the site include, e.g., U.S. Patent Nos. 4,368,736 and 4,366,819.
In certain types of single-lumen coupling devices, the conduit terminates in a flange that resides within the lumen of the vessel. For example, the conduit may have a tubular body with a connector which has a plurality of extensions and is configured for disposition annularly within the inside of a tubular vessel. See, e.g., U.S. Patent No. 6,660,015. In other devices, the flange may be attached into or onto the surface of the adventitial tissue of the blood vessel.
Other types of single-lumen bypass devices are described, for example, in U.S. Patent Nos. 6,241,743; 6,428,550; 6,241,743; 6,428,550;
5,904,697; 5,290,298; 6,007,576; 6,361,559; 6,648,901, 4,931,057 and U.S.

Application Publication Nos. 2004/0015180A1, 2003/0065344A1, and 2002/0116018A1.
In one aspect of the invention, the anastomotic coupling device comprises more than one lumen through which blood may travel. Multi-lumen bypass devices may include two or more tubular portions configured to interconnect multiple (two or more) blood vessels. Multi-lumen coupling devices may be used in a variety of anastomosis procedures. For example, such devices may be used in coronary artery bypass graft (CABG) surgery to divert blood from an occluded proximal vessel (e.g., an artery) into one or more target (i.e., distal) vessels (e.g., an artery or vein).
In one aspect, at least one tubular portion may by used as a shunt for diverting blood between a source vessel and a target vessel. In another aspect, the device may be configured as an interFace for securing a graft vessel to a target vessel for completion of an anastomosis. Depending on the procedure, the tubular arms may be of equal length and diameter or of unequal length and diameter and may include a tubular portions) that is expandable and/or includes a shape-memory material (e.g., nitinol). Furthermore, the tubular portions may be made of the same material or a different material.
In one aspect, one or more ends of a tubular portion may be inserted into the end or into the side of one or more blood vessels. In other embodiments, one or more tubular portions of the device may reside within the lumen of a blood or graft vessel. The device, optionally, may be secured to the blood vessel using a fastener or an adhesive, or another approach known to those skilled in the art.
At least one arm of the multi-lumen connector may be attached to a graft vessel. The graft vessel may be a synthetic graft, such as an ePTFE or polyester graft, or natural graft material (e.g., autologous vessel, homologous vessel, or xenograft), or a combination of a synthetic and a natural graft material. In certain embodiments, a graft vessel may be attached to an end of a tubular portion of the device, and a second graft vessel may be attached to the opposite end of the same tubular portion or to the end of another tubular portion. The graft vessels) may be further attached to a target vessels) for the completion of the anastomosis.
In one aspect, the device may include three or more tubular arms that extend from a junction site. For example, the multi-lumen device may be generally T-shaped or Y-shaped (i.e., having two or three lumens, respectively).

For example, the multi-lumen device may be a T-shaped tubular graft connector having a longitudinal member that extends into the target vessel and a second section that is exterior to the vessel which provides a connection to an alternate tubular structure. See, e.g., U.S. Patent Nos. 6,152,945 and 5,972,017. Other multi-lumen devices are described in, (see, e.g., U.S. Patent Nos. 6,152,945;
6,451,033; 5,755,778; 5,922,022; 6,293,965; 6,517,558 and 6,626,914 and U.S.
Publication No. 2004/0015180A1 ).
In another aspect, the device may be a tube for bypassing blood flow directly from a portion of the heart (e.g., left ventricle} to a coronary artery.
For example, the device may be a hollow tube that may be partially closable by a one-way valve in response to movement of the cardiac tissue during diastole while permitting blood flow during systole (see, e.g., U.S. Patent No.
6,641,610). The device may be an elongated rigid shunt body composed of a diversion tube having two apertures in which one may be disposed within the cyocardium of the left ventricle and the other may be disposed within the coronary artery (see, e.g., WO 00/15146 and U.S. Application Publication No.
200310055371A1). The device may be a valued, tubular apparatus that is L- or T-shaped which is adapted for insertion into the wall of the heart to provide blood communication from the heart to a coronary vessel (see, e.g., U.S.
Patent No.6,123,682).
In another aspect, the device may include a network of interconnected tubular conduits. For example, the device- may include two tubular portions that may be oriented generally axially or orthogonally relative to each other. See U.S. Patent No. 6,241,761 and 6,241,764. Communication between the two tubular structures may be achieved through a flow channel which facilitates blood to flow between the bores of each tube.
In another aspect, the anastomotic coupling device is a resorbable device that may be configured with two or three termini which provide a vessel interface without the need for sutures and provides a fluid communication through an intersecting lumen, such as a bypass graft or alternate vessel. See, e.g., U.S. Application Publication Nos. 2002/0052572A1 and PCT Publication No. WO 02/24114A2. An anastomotic connector may also be formed of a resorbable tubular structure configured to include snap-connectors or other components for securing it to the tissue as well as hemostasis inducing sealing rings to prevent blood leakage. See, e.g., U.S.
Patent Nos. 6,056,762. The anastomotic connector may be designed with three legs whereby two legs are adapted to be inserted within the continuous blood vessel in a contracted state and then enlarged to form a tight fit and the third leg is adapted for connecting and sealing with a third conduit. See, e.g., U.S. Patent No. 6,019,788.
An example of a commercially available multi-lumen anastomotic coupling device is the SOLEM graft connector (made by Jomed, Sweden). This device, which is described in more detail in PCT Publication No. WO 01/13820, and U.S. Patent Nos. 6,179,848, D438618 and D429334, includes a T-shaped connector composed of nitinol and an ePTFE graft for completion of a distal anastomosis.
Another example of an anastomotic connector is the HOLLY
GRAFT System (in development) for use in bypass surgery from CABG
Medical, Inc. (Minneapolis, MN), which is described, e.g., in U.S. Patent Nos.
6,241,761 and 6,241,764.
In one aspect, the present invention provides for the combination of an anastomotic coupling device and an anti-scarring agent or a composition comprising an anti-scarring device. In one aspect, the anastomotic coupling device may be attached to a blood vessel for the formation of an anastomosis without the use of sutures or staples. In certain aspects, the anastomotic coupling device may comprise a tubular structure defining a lumen through which blood may flow, and an anti-scarring agent. The device may include one, two, three, or more lumens defined by one, two, three, or more tubular structures, depending on the number of vessels to be connected.
Introduction of an anastomotic connector into or onto an intramural, luminal, or adventitial portion of a blood vessel may irritate or damage the endothelial tissue of the blood vessel and/or may alter the natural hemodynamic flow through the vessel. This irritation or damage may stimulate a cascade of biological events resulting in a fibrotic response which can lead to the formation of scar tissue in the vessel. Incorporation of a therapeutic agent in accordance with the invention into or onto a portion of the device that is in direct contact with the blood vessel (e.g., a terminal portion or edge of the device) may inhibit one or more of the scarring processes described above (e.g., smooth muscle cell proliferation, cell migration, inflammation), making the vessel less prone to the formation of intimal hyperplasia and stenosis.
Thus, in one aspect, the therapeutic agent may be associated only with the portion of the device that is in contact with the blood or endothelial tissue. For example, the anti-scarring agent may be incorporated into only an intravascular portion (i.e., that portion that resides within the lumen of the vessel or in the vessel tissue) of the device. The anti-scarring agent may be incorporated onto all or a portion of the intravascular portion of the device.
In other embodiments, the coating may reside on all or a portion of an extravascular portion of the device.
The anti-scarring agent or a composition that includes an anti-scarring agent may be coated onto a portion of or onto the entire surface of the device or may be incorporated into a portion of, or into the entire the structure of, the device (e.g., either within voids, reservoirs, or divets in the device or within the material used to construct the device). In other aspects, the agent or a composition comprising the agent is impregnated into or affixed onto the device surface.
As described above, the device may include a tubular portion that is disposed within the lumen of a blood vessel. The entire tubular portion may, for example, be coated with an anti-scarring agent or a composition comprising an anti-scarring. Alternatively, only a portion of the tubular portion may include the anti-scarring agent. For example, only an external (abluminal) surface or only the interior (endoluminal) surface of the tubular portion may be coated.
In other embodiments, one or both termini of the tubular portion may be coated.
For example, the endoluminal and/or abluminal surface of the tubular section through which blood enters into the device (i.e., proximal end) may be coated with the anti-scarring agent or composition comprising the anti-scarring agent.
In another aspect, the endoluminal and/or abluminal surface of the tubular section through which blood exits (i.e., distal end) from the device may be coated with the anti-scarring agent or composition comprising the anti-scarring agent.
In another embodiment, the anti-scarring agent or composition comprising the anti-scarring agent is associated (e.g., coated onto or incorporated into) with an anchoring member (e.g., a fastener, such as a staple or clip) that secures the device to a blood vessel.
As described above, anastomotic connector devices can include a fibrosis-inhibiting agent as a means to improve the clinical efficacy of the device. In another approach, the fibrosis-inhibiting agent can be incorporated into or onto a film or mesh (described in further detail below) that is applied in a perivascular manner to an anastomotic site (e.g., at the junction of a graft vessel and the blood vessel). These films or wraps can be used with any of the anastomotic connector devices described above and, typically, are placed around the outside of the anastomosis at the time of surgery. In other embodiments, the agent may be delivered to the anastomotic site in the form of a spray, paste, gel, or the like. In yet another approach, the fibrosis-inhibiting agent can be incorporated into or onto the graft vessel that is secured to the blood vessel with the connector device.
In yet another aspect, other specialized intravascular devices, such as coronary drug infusion guidewires, such as those available from TherOx, Inc., grafts and balloon over stent devices, such as are described in Wilensky, R.L. (1993) J. Am. Coll. Cardiol.: 21: 185A can also be utilized for local delivery of an anti-fibrosing agent.
As described above, the present invention provides intravascular devices (e.g., anastomotic connectors, stents, drug-delivery balloons, intravascular catheters) that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use with intravascular devices have been described above.
Methods for incorporating coating fibrosis-inhibiting agents and compositions onto or into intravascular devices include: (a) directly affixing to the intravascular device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition), (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) by inserting the device into a sleeve or mesh which contains or is coated with a fibrosis-inhibiting composition, (f) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.

The intravascular device (e.g., a stent) may be adapted to release the desired agent at only the distal ends, or along the entire body of the device.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, intravascular devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As intravascular devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of agents for use in intravascular devices include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus); (E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082);
(I) antimycotic agents (e.g., sulconizole), (J) p38 MAP kinase inhibitors (e.g., SB202190), arid (K) angiogenesis inhibitors (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the intravascular device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines.

The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 ~g-10 ~,g, or 10 ~.g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 p,g/mm2 - 1 ~g/mm2, or 1 ~g/mm2 - 10 pg/mm2, or 10 ~g/mm~ - 250 p,g/mm2, 250 pg/mm2 - 1000 p.g/mm2, or 1000 ~g/mm2 - 2500 ~g/mm2.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with intravascular devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total dose not to exceed 25 mg (range of 0.1 ~.g to 25 mg); preferred 1 ~,g to 5 mg. The dose per unit area of 0.01 ~g - 100 ~g per mm2; preferred dose of 0.1 ~g/mm2 -10 pg/mm2. Minimum concentration of 10-8 - 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 p,g to 5 mg); preferred 0.1 ~,g to 1 mg. The dose per unit area of the device of 0.01 ~,g - 20 ~g per mm2;
preferred dose of 0.05 ~,g/mm2 - 3 ~,g/mm2. Minimum concentration of 10-8- 104 M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred ~,g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~g per mm2;
preferred dose of 0.25 ~g/mm2 - 5 ~.g/mm2. Minimum concentration of 10-8 - 10-4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 10 mg (range of 0.1 p.g to 10 mg); preferred 1 ~,g to 3 mg. The dose per unit area of the device of 0.1 p,g - 10 ~.g per mm2; preferred dose of 0.25 p.g/mm2 p.glmm2. Minimum concentration of 10-$ - 104 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~g - 100 p,g per mm2; preferred dose of 0.5 ~g/mm2 - 10 ~,g/mm2. Minimum concentration of 10-$ - 104 M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 ~,g - 100 wg per mm2 of surface area; preferred dose of 0.3 ~g/mm2 - 10 ~g/mm2. Minimum concentration of 10-$ - 10-4 M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof:
total dose not to exceed 20 mg (range of 0.1 p,g to 20 mg); preferred 1 p,g to mg. The dose per unit area of the device of 0.1 pg - 10 ~.g per mm2; preferred dose of 0.25 ~g/mm2 - 5 ~,glmm2. Minimum concentration of 10'8- 10'~ M of geldanamycin is to be maintained on the device surface. (F) HMGCoA
reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof:
total dose not to exceed 2000 mg (range of 10.0 pg to 2000 mg); preferred 10 ~.g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 pg per mm2; preferred dose of 2.5 p,g/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 - 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 p,g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~,g per mm2;
preferred dose of 2.5 ~g/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 -10'3 M of mycophenolic acid is to be maintained on the device surface. (H) NF
kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof:
total dose not to exceed 200 mg (range of 1.0 pg to 200 mg); preferred 1 p,g to 50 mg. The dose per unit area of the device of 1.0 ~,g - 100 ~g per mm2;
preferred dose of 2.5 ~,g/mm2 - 50 ~g/mm2. Minimum concentration of 10'8- 10' 4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 pg to 2000 mg); preferred 10 ~.g to 300 mg.
The dose per unit area of the device of 1.0 pg - 1000 p,g per mm2; preferred dose of 2.5 pg/mm2 - 500 pg/mm2. Minimum concentration of 10-8- 10'3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP Kinase Inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 p.g to 2000 mg); preferred 10 pg to 300 mg. The dose per unit area of the device of 1.0 pg - 1000 p,g per mm2;
preferred dose of 2.5 p.g/mm2 - 500 ~g/mm2. Minimum concentration of 10'$ -10-3 M of SB202190 is to be maintained on the device surface. (K) anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred pg to 3 mg. The dose per unit area of the device of 0.1 pg - 10 ~g per mm2;

preferred dose of 0.25 pg/mm2 - 5 pg/mmz. Minimum concentration of 10-$ - 10-~ M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with intravascular devices include the following: (A) Biolimus and derivatives and analogues thereof: Total dose should not exceed mg (range of 0.1 p,g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 pg - 100 p,g per mm2 of surface area; preferred dose of 0.3 pg/mm2 - 10 ~g/mm~. Minimum concentration of 10-8- 10-4 M of everolimus is to be 10 maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p.g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 p,g - 100 p,g per mm2 of surface area; preferred dose of 0.3 p.g/mm2 -10 p,g/mm2. Minimum concentration of 10-8- 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 pg to 1 mg.
The dose per unit area of 0.1 p,g - 100 ~g per mm2 of surface area; preferred dose of 0.3 pg/mm2 - 10 pg/mm2. Minimum concentration of 10-8- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 p,g - 100 p,g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 pg/mm2. Minimum concentration of 10-8- 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 p,g - 100 p,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 - 10 p,g/mm2. Minimum concentration of 10-8- 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 p.g - 100 p,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 -10 pg/mm2. Minimum concentration of 10-8- 10-4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 wg to 1 mg.
The dose per unit area of 0.1 p.g - 100 p,g per mm2 of surface area; preferred dose of 0.3 pg/mm2 - 10 ~,g/mm2. Minimum concentration of 10-$ - 10-4 M of ABT-578 is to be maintained on the device surface.
Gastrointestinal Stents The present invention provides for the combination of a drug and a gastrointestinal (GI) stent. The term GI stent refers to devices that are located in the gastrointestinal tract including the biliary duct, pancreatic duct, colon, and the esophagus. GI stents are or comprise scaffoldings that are used to treat endoluminal body passageways that have become blocked due to disease or damage, including malignancy or benign disease.
In one aspect, the GI stent may be an esophageal stent used to keep the esophagus open whereby food is able to travel from the mouth to the stomach. For example, the esophageal stent may be composed of a cylindrical supporting mesh inner layer, retaining mesh outer layer and a semi-permeable membrane sandwiched between. See, e.g., U.S. Patent No. 6,146,416. The esophageal stent may be a radially, self-expanding stent of open weave construction with an elastomeric film formed along the stent to prevent tissue ingrowth and distal cuffs that resist stent migration. See, e.g., U.S. Patent No.
5,876,448. The esophageal stent may be composed of a flexible wire configuration to form a cylindrical tube with a deformed end portion increased to a larger diameter for anchoring pressure. See, e.g., U.S. Patent No.
5,876,445.
The esophageal stent may be a flexible, self-expandable tubular wall incorporating at least one truncated conical segment along the longitudinal axis.
See, e.g., U.S. Patent No. 6,533,810.
In another aspect, the GI stent may be a biliary stent used to keep the biliary duct open whereby bile is able to drain into the small intestines.
For example, the biliary stent may be composed of shape memory alloy. See, e.g., U.S. Patent No. 5,466,242. The biliary stent may be a plurality of radially extending wings with grooves which project from a helical core. See, e.g., U.S.
Patent Nos. 5,776,160 and 5,486,191.
In another aspect, the GI stent may be a colonic stent. For example, the colonic stent may be a hollow tubular body that may expand radially and be secured to the inner wall of the organ in a release fitting.
See, e.g., European Patent Application No. EP1092400A2.
In another aspect, the GI stent may be a pancreatic stent used to keep the pancreatic duct open to facilitate secretion into the small intestines.

For example, the pancreatic stent may be composed of a soft biocompatible material which is resiliently compliant which conforms to the duct's curvature and contains perforations that facilitates drainage. See, e.g., U.S. Patent No.
6,132,471.
GI stents, which may be combined with one or more drugs according to the present invention, include commercially available products, such as the NIR Biliary Stent System and the WALLSTENT Endoprostheses from Boston Scientific Corporation.
In one aspect, the present invention provides GI stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in GI
stents have been described above.
Methods for incorporating fibrosis-inhibiting agents or fibrosis-inhibiting compositions onto or into the GI stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the, internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
In addition to coating the GI stent with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device. This can include the GI stent structure itself, the outer covering or sleeve, if applicable, or both the stent structure and the outer covering or sleeve.

According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, GI stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As GI stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design.
However, certain principles can be applied in the application of this art.
Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of scarring agents for use in GI stents include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus); (E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082);
(I) antimycotic agents (e.g., sulconizole), (J) p38 MAP kinase inhibitors (e.g., SB202190), and (K) anti-angiogenic agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the GI
stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 p,g-10 pg, or 10 p,g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~.g/mm2 - 1 ~g/mm~, or 1 ~g/mm2 - 10 ~.g/mm~, or 10 ~g/mm2 - 250 p.g/mm2, 250 ~g/mm2 - 1000 ~g/mm2, or 1000 ~g/mm2 - 2500 ~g/mm2.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with GI stent devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total dose not to exceed 25 mg (range of 0.1 ~.g to 25 mg); preferred 1 ~g to 5 mg. The dose per unit area of 0.01 ~.g - 100 ~,g per mm2; preferred dose of 0.1 pg/mm2 -10 ~g/mm2. Minimum concentration of 10-8- 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 ~g to 5 mg); preferred 0.1 ~g to 1 mg. The dose per unit area of the device of 0.01 ~,g - 20 p,g per mm2;
preferred dose of 0.05 ~,g/mm2 - 3 ~,g/mm2. Minimum concentration of 10-8 -M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred ~g to 3 mg. The dose per unit area of the device of 0.1 ~,g - 10 ~g per mm2;
preferred dose of 0.25 ~g/mm2 - 5 ~g/mmz. Minimum concentration of 10-8 - 10-4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 10 mg (range of 0.1 pg to 10 mg); preferred 1 ~,g to 3 mg. The dose per unit area of the device of 0.1 p,g - 10 ~g per mm2; preferred dose of 0.25 p,g/mm2 -~,g/mm2. Minimum concentration of 10-8 - 104 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~.g per mm2; preferred dose of 0.5 ~,g/mm2 -10 ~,glmm2. Minimum concentration of 10-8- 10'4 M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg.
The dose per unit area of 0.1 ~,g - 100 ~.g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 -10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof:

total dose not to exceed 20 mg (range of 0.1 ~,g to 20 mg); preferred 1 pg to mg. The dose per unit area of the device of 0.1 ~.g - 10 ~.g per mm2;
preferred dose of 0.25 ~,g/mm2 - 5 ~,g/mm2. Minimum concentration of 10-8 - 10-4 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA
reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof:
total dose not to exceed 2000 mg (range of 10.0 ~.g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 p.g - 1000 ~g per mm2; preferred dose of 2.5 ~g/mm2 - 500 ~.g/mm2. Minimum concentration of 10-$- 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 ~g per mm2;
preferred dose of 2.5 ~g/mm2 - 500 p,g/mm2. Minimum concentration of 10-8 -10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF
kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof:
total dose not to exceed 200 mg (range of 1.0 ~,g to 200 mg); preferred 1 ~.g to 50 mg. The dose per unit area of the device of 1.0 ~.g - 100 ~g per mm2;
preferred dose of 2.5 ~.g/mm2 - 50 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 ~,g to 300 mg.
The dose per unit area of the device of 1.0 pg - 1000 ~.g per mm2; preferred dose of 2.5 ~g/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 - 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 pg to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~g per mm2;
preferred dose of 2.5 ~,g/mm2 - 500 ~g/mm2. Minimum concentration of 10-8 -10-3 M of SB202190 is to be maintained on the device surface. K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred ~,g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~,g per mm2;
preferred dose of 0.25 ~,g/mm2 - 5 p.g/mm2. Minimum concentration of 10-8- 10-4 M of halofuginone bromide is to be maintained on the device surface.

In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with gastrointestinal stent devices include the following:
(A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~,g - 100 p.g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2- 10 ~g/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 ~g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 -10 pg/mm2. Minimum concentration of 10-$- 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~,g to 1 mg.
The dose per unit area of 0.1 pg - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 -10 ~g/mm2. Minimum concentration of 10-$ - 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~.g - 100 p.g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 - 10 ~g/mm~. Minimum concentration of 10-$ - 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 - 10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 ~,g - 100 p,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 -10 ~g/mm~. Minimum concentration of 10-$- 10'4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 ~.g - 100 pg per mm2 of surface area; preferred dose of 0.3 pg/mm2 -10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of ABT-578 is to be maintained on the device surface.

Tracheal and Bronchial Stents The present invention provides for the combination of an anti-scarring agent and a tracheal or bronchial stent device.
Representative examples of tracheal or bronchial stents that can benefit from being coated with or having incorporated therein, a fibrosis-inhibiting agent include tracheal stents or bronchial stents, including metallic and polymeric tracheal or bronchial stents and tracheal or bronchial stents that have an external covering (e.g., polyurethane, polyethylene terephthalate), PTFE, or silicone rubber).
Tracheal and bronchial stents may be, for example, composed of an elastic plastic shaft with metal clasps that expands to form a lumen along the axis for opening the diseased portion of the trachea and having three sections to emulate the natural shape of the trachea. See, e.g., U.S. Patent No.
5,480,431. The tracheal/bronchial stent may be a T-shaped tube having a tracheotomy tubular portion that projects outwardly through a tracheotomy orifice which is configured to close and form a fluid seal. See, e.g., U.S.
Patent Nos. 5,184,610 and 3,721,233. The tracheal/bronchial stent may be composed of a flexible, synthetic polymeric resin with a tracheotomy tube mounted on the wall with a bifurcated bronchial end that is configured in a T-Y shape with specific curves at the intersections to minimize tissue damage. See, e.g., U.S.
Patent No. 4,795,465. The tracheal/bronchial stent may be a scaffolding configured to be substantially cylindrical with a shape-memory frame having geometrical patterns and having a coating of sufficient thickness to prevent epithelialization. See, e.g., U.S. Patent Application Publication No.
2003/0024534A1.
Tracheal/bronchial stents, which may be combined with one or more agents according to the present invention, include commercially available products, such as the WALLSTENT Tracheobronchial Endoprostheses and ULTRAFLEX Tracheobronchial Stent Systems from Boston Scientific Corporation and the DUMON Tracheobronchial Silicone Stents from Bryan Corporation (Woburn, MA).
In one aspect, the present invention provides tracheal and bronchial stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in tracheal and bronchial stents have been described above. Methods for incorporating fibrosis-inhibiting agents or fibrosis-inhibiting compositions onto or into the tracheal or bronchial stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition), (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device. This can include the stent structure itself, the outer covering or sleeve, if applicable, or both the stent structure and the outer covering or sleeve.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, tracheal and bronchial stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As tracheal and bronchial stents are made in a variety of configurations and sizes, the exact dose 'administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined.
Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application: Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several fibrosis-inhibiting agents for use in tracheal and bronchial stents include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE
and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus); (E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082); (I) antimycotic agents (e.g., sulconizole), (J) p38 MAP
kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the tracheal or bronchial stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines.
The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 ~,g-10 fig, or 10 ~,g-10 mg, or 10 mg-250 mg, or 250 mg-, 1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~,g/mm2 - 1 ~.g/mm2, or 1 ~,g/mm2 - 10 ~.g/mm2, or 10 ~g/mm2 - 250 ~.g/mm2, 250 ~g/mm2 - 1000 ~g/mm2, or 1000 ~.g/mm~ - 2500 ~g/mm~.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with tracheal and bronchia!
stent devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof:
total dose not to exceed 25 mg (range of 0.1 ~,g to 25 mg); preferred 1 ~g to mg. The dose per unit area of 0.01 ~,g - 100 ~.g per mm2; preferred dose of 0.1 ~g/mm2 -10 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 ~.g to 5 mg);
preferred 0.1 ~g to 1 mg. The dose per unit area of the device of 0.01 ~,g -~g per mmz; preferred dose of 0.05 pg/mm2 - 3 ~g/mm2. Minimum concentration of 10-8 - 104 M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 pg to 10 mg); preferred 1 ~.g to 3 mg. The dose per unit area of the device of 0.1 pg - 10 ~g per mm2; preferred dose of 0.25 ~g/mm2 - 5 ~,g/mm2.
Minimum concentration of 10-8 - 104 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg);
preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 ~,g - 10 ~,g per mm2; preferred dose of 0.25 p,g/mm2 - 5 ~.g/mm2. Minimum concentration of 10-8- 10-4 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of the device 0.1 p,g -100 ~.g per mm2; preferred dose of 0.5 ~,g/mm2 - 10 ~.g/mm2. Minimum concentration of 10-8- 104 M is to be maintained on the device surface.
Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~,g per mmz of surface area; preferred dose of 0.3 p.g/mm2 - 10 ~g/mm2. Minimum concentration of 10-8- 104 M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof: total dose not to exceed 20 mg (range of 0.1 ~,g to 20 mg); preferred 1 ~g to 5 mg. The dose per unit area of the device of 0.1 p,g - 10 ~g per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~.g/mm2. Minimum concentration of 10-8- 10-4 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 p.g to 2000 mg); preferred 10 ~,g to 300 mg.
The dose per unit area of the device of 1.0 ~.g - 1000 pg per mma; preferred dose of 2.5 ~g/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 - 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~g to 300 mg.
The dose per unit area of the device of 1.0 ~,g - 1000 ~.g per mm2; preferred dose of 2.5 ~,g/mm2 - 500 ~g/mm2. Minimum concentration of 10-8- 10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF kappa B
inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 1.0 ~.g to 200 mg); preferred 1 p.g to 50 mg.
The dose per unit area of the device of 1.0 ~,g - 100 ~g per mm2; preferred dose of 2.5 p.g/mm2 - 50 ~,g/mm2. Minimum concentration of 10-8- 10-4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 pg - 1000 ~,g per mm2; preferred dose of 2.5 ~,g/mm2 - 500 ~g/mm2. Minimum concentration of 10-8- 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 pg - 1000 ~.g per mm2; preferred dose of 2.5 ~,g/mm2 - 500 ~.g/mm2. Minimum concentration of 10-8- 10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 1 pg to 3 mg. The dose per unit area of the device of 0.1 ~,g - 10 ~,g per mm2; preferred dose of 0.25 ~.g/mm2 - 5 ~,g/mm2. Minimum concentration of 10-$ - 10-4 M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with intravascular devices include the following: (A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 pg - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 - 10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 -10 p,g/mm2. Minimum concentration of 10-$ - 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 p,g to 1 mg.
The dose per unit area of 0.1 pg - 100 ~g per mm2 of surface area; preferred dose of 0.3 pg/mm2 -10 p.g/mm2. Minimum concentration of 10'$- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 p,g - 100 ~,g per mm~ of surface area; preferred dose of 0.3 pg/mm~ - 10 pg/mm2. Minimum concentration of 10-$ - 10-~ M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 p,g to 1 mg. The dose per unit area of 0.1 p.g - 100 pg per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 - 10 ~glmm~. Minimum concentration of 10'$- 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 p,g to 1 mg. The dose per unit area of 0.1 p,g - 100 p,g per mm2 of surface area; preferred dose of 0.3 p.glmm2 -10 p,g/mm2. Minimum concentration of 10-$- 10-4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 p,g - 100 pg per mm2 of surface area; preferred dose of 0.3 p.g/mm2 - 10 pg/mm2. Minimum concentration of 10-$- 10-4 M of ABT-578 is to be maintained on the device surface.
_Genital-Urinary Stents The present invention provides for the combination of an anti-scarring agent and genital-urinary (GU) stent device.
Representative examples genital-urinary (GU) stents that can benefit from being coated with or having incorporated therein, a fibrosis-inhibiting agent include ureteric and urethral stents, fallopian tube stents, prostate stents, including metallic and polymeric GU stents and GU stents that have an external covering (e.g., polyurethane, polyethylene terephthalate), PTFE or silicone rubber).
In one aspect, genital-urinary stents include ureteric and urethral stents. Ureteral stents are hollow tubes with holes along the sides and coils at either end to prevent migration. Ureteral stents are used to relieve obstructions (caused by stones or malignancy), to facilitate the passage of stones, or to allow healing of ureteral anastomoses or leaks following surgery or trauma.
They are placed endoscopically via the bladder or percutaneously via the kidney.
Urethral stents are used for the treatment of recurrent urethral strictures, detruso-external sphincter dyssynergia and bladder outlet obstruction due to benign prostatic hypertrophy. In addition, procedures that are conducted for the prostate, such as external radiation or brachytherapy, may lead to fibrosis due to tissue insult resulting from these procedures. The incidence of urethral stricture in prostate cancer patients treated with external beam radiation is about 2%. Development of urethral stricture may also occur in other conditions such as following urinary catheterization or surgery, which results in damage to the epithelium of the urethra. The clinical manifestation of urinary tract obstruction includes decreased force and caliber of the urinary stream, intermittency, postvoid dribbling, hesitance and nocturia. Complete closure of the urethra can result in numerous problems including eventual kidney failure.
To maintain patency in the urethra, urethral stents may be used. The stents are typically self-expanding and composed of metal superalloy, titanium, stainless steel or polyurethane.
For example, the ureteric/urethral stent may be composed of a main catheter body of flexible polymeric material having an enlarged entry end with a hydrophilic tip that dissolves when contacted with body fluids. See, e.g., U.S. Patent No. 5,401,257. The ureteric/urethral stent may be composed of a multi-sections including a closed section at that the bladder end which does not contain any fluid passageways such that it acts as an anti-reflux device to prevent reflux of urine back into the kidney. See, e.g., U.S. Patent No.
5,647,343. The ureteric/urethral stent may be composed of a central catheter tube made of shape memory material that forms a stent with a retention coil for anchoring to the ureter. See, e.g., U.S. Patent No. 5,631,274. The ureteric/urethral stent may be a composed of an elongated flexible tubular stent with preformed set curls at both ends and an elongated tubular rigid extension attached to the distal end which allows the combination function as an externalized ureteral catheter. See, e.g., U.S. Patent Nos. 5,221,253 and 5,116,309. The ureteric/urethral stent may be composed of an elongated member, a proximal retention structure, and a resilient portion connecting them together, whereby they are all in fluid communication with each other with a slideable portion providing a retracted and expanded position. See, e.g., U.S.
Patent No. 6,685,744. The ureteric/urethral stent may be a hollow cylindrical' tube that has a flexible connecting means and locating means that expands and selectively contracts. See, e.g., U.S. Patent No. 5,322,501. The ureteric/urethral stent may be composed of a stiff polymeric body that affords superior columnar and axial strength for advancement into the ureter, and a softer bladder coil portion for reducing the risk of irritation. See, e.g., U.S.
Patent No. 5,141,502. The ureteric/urethral stem may be composed of an elongated tubular segment that has a pliable wall at the proximal region and a plurality of members that prevent blockage of fluid drainage upon compression.
See, e.g., U.S. Patent No. 6,676,623. The ureteric/urethral stent may be a catheter composed of a conduit which is part of an assembly that allows for non-contaminated insertion into a urinary canal by providing a sealing member that surrounds the catheter during dismantling. See, e.g., U.S. Patent Application Publication No. 2003/0060807A1.
In another aspect, genital-urinary stents include prostatic stents.
For example, the prostatic stent may be composed of two polymeric rings constructed of tubing with a plurality of connecting arm members connecting the rings in ~ parallel manner. See, e.g., U.S. Patent No. 5,269,802. The prostatic stent may be composed of thermoplastic material and a circumferential reinforcing helical spring, which provides rigid mechanical support while being flexible to accommodate the natural anatomical bend of the prostatic urethra. See, e.g., U.S. Patent No. 5,069,169.
In another aspect, genital-urinary stents include fallopian stents and other female genital-urinary devices. For example, the genital-urinary device may be a~female urinary incontinence device composed of a vaginal-insertable supporting portion that is resilient and flexible, which is capable of self support by expansion against the vaginal wall and extending about the urethral orifice. See, e.g., U.S. Patent No. 3,661,155. The genital-urinary device may be a urinary evacuation device composed of a ovular bulbous concave wall having an opening to a body engaging perimetal edge integral with the wall and an attached tubular member with a pleated body. See, e.g., U.S. Patent No. 6,041,448.
Genital-urinary stents, which may be combined with one or more agents according to the present invention, include commercially available products, such as the UROLUME Endoprosthesis Stents from American Medical Systems, Inc. (Minnetonka, MN), the RELIEVE ProstaticlUrethral Endoscopic Device from InjecTx, Inc. (San Jose, CA), the PERCUFLEX
Ureteral Stents from Boston Scientific Corporation, and the TARKINGTON
Urethra) Stents and FIRLIT-KLUGE Urethra) Stents from Cook Group lnc (Bloomington, IN).
In one aspect, the present invention provides GU stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in GU
stents have been described above. Methods for incorporating fibrosing agents or fibrosis-inhibiting compositions onto or into the GU stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, GU stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As GU stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design.
However, certain principles can be applied in the application of this art.
Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of scarring agents for use in GU stents include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus); (E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7032);
(I) antimycotic agents (e.g., sulconizole), (J) p33 MAP kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents.(e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the GU
stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 p,g-10 pg, or 10 ~,g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~g/mm2 - 1 ~,g/mma, or 1 p,g/mm2 -10 p.g/mm2, or 10 ~g/mm2 - 250 p,g/mm2, 250 ~.g/mm2 - 1000 p.g/mm2, or 1000 ~g/mm2 - 2500 pg/mm2.

Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with GU stent devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total dose not to exceed 25 mg (range of 0.1 ~g to 25 mg); preferred 1 p.g to 5 mg. The dose per unit area of 0.01 pg - 100 p.g per mm2; preferred dose of 0.1 p.g/mm2 -pg/mm2. Minimum concentration of 10-8 - 10~ M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 p,g to 5 mg); preferred 0.1 10 p,g to 1 mg. The dose per unit area of the device of 0.01 wg - 20 pg per mm2;
preferred dose of 0.05 ~.g/mm2 - 3 ~,g/mm2. Minimum concentration of 10-8 -M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred ~g to 3 mg. The dose per unit area of the device of 0.1 p.g - 10 p,g per mm2;
preferred dose of 0.25 p,g/mm2 - 5 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 1 pg to 3 mg. The dose per unit area of the device of 0.1 p,g - 10 p.g per mm2; preferred dose of 0.25 pg/mm2 -pg/mmz. Minimum concentration of 10-8 - 104 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 p.g - 100 ~g per mm2; preferred dose of 0.5 p,g/mm2 -10 p.g/mm2. Minimum concentration of 10-8- 104 M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 p.g - 100 pg per mm2 of surface area; preferred dose of 0.3 pg/mm2 -10 p,g/mm2. Minimum concentration of 10-8- 104 M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof:
total dose not to exceed 20 mg (range of 0.1 p.g to 20 mg); preferred 1 p,g to mg. The dose per unit area of the device of 0.1 pg - 10 pg per mm2; preferred dose of 0.25 ~,g/mm2 - 5 p,g/mm2. Minimum concentration of 10-8 - 10-4 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA

reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof:
total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 p,g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 pg per mm2; preferred dose of 2.5 ~glmm2 - 500 pg/mm2. Minimum concentration of 10-8- 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 p,g to 2000 mg); preferred 10 p,g to 300 mg. The dose per unit area of the device of 1.0 p,g - 1000 ~g per mm2;
preferred dose of 2.5 ~glmm2 - 500 p,g/mm2. Minimum concentration of 10-8 -10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF
kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof:
total dose not to exceed 200 mg (range of 1.0 p,g to 200 mg); preferred 1 pg to 50 mg. The dose per unit area of the device of 1.0 p.g - 100 pg per mm2;
preferred dose of 2.5 ~,g/mm2 - 50 p,g/mm2. Minimum concentration of 10-$ - 10-4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 p,g to 2000 mg); preferred 10 ~g to 300 mg.
The dose per unit area of the device of 1.0 p,g - 1000 pg per mm2; preferred dose of 2.5 pg/mm2 - 500 p,g/mm2. Minimum concentration of 10-$ - 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 p.g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 pg per mm2;
preferred dose of 2.5 p.g/mm2 - 500 ~,g/mmz. Minimum concentration of 10-$ -10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 pg to 10 mg); preferred p.g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 p,g per mm2;
preferred dose of 0.25 ~glmm2- 5 p.g/mm2. Minimum concentration of 10-8- 10-4 M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with genital-urinary stent devices include the following:
(A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~.g - 100 p.g per mm2 of surface area; preferred dose of 0.3 ~g/mm~ -10 pg/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 ~g/mm~. Minimum concentration of 10-$- 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 ~,g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 -10 ~,g/mm2. Minimum concentration of 10'$- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 - 10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~g per mm2 of surface area; preferred dose of 0.3 pg/mm2 -10 ~,g/mm2. Minimum concentration of 10-$ - 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 pg - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 ~g/mm2. Minimum concentration of 10'$- 10-4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 ~,g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 -10 p.g/mm2. Minimum concentration of 10-$- 10-4 M of ABT-578 is to be maintained on the device surface.
Ear and Nose Stents The present invention provides for the combination of an anti-scarring agent and an ear-nose-throat (ENT) stent device (e.g., a lacrimal duct stent, Eustachian tube stent, nasal stent, or sinus stent).

The sinuses are four pairs of hollow regions contained in the bones of the skull named after the bones in which they are located (ethmoid, maxillary, frontal and sphenoid). All are lined by respiratory mucosa which is directly attached to the bone. Following an inflammatory insult such as an upper respiratory tract infection or allergic rhinitis, a purulent form of sinusitis can develop. Occasionally secretions can be retained in the sinus due to altered ciliary function or obstruction of the opening (ostea) that drains the sinus. Incomplete drainage makes the sinus prone to infection typically with Haemophilus influenza, Streptococcus pneumoniae, Moraxella catarrhalis, Veillonella, Peptococcus, Corynebacterium acnes and certain species of fungi.
When initial treatment such as antibiotics, intranasal steroid sprays and decongestants are ineffective, it may become necessary to perform surgical drainage of the infected sinus. Surgical therapy often involves debridement of the ostea to remove anatomic obstructions and removal of parts of the mucosa. Occasionally a stent (a cylindrical tube which physically holds the lumen of the ostea open) is left in the osta to ensure drainage is maintained even in the presence of postoperative swelling. ENT stents, typically made of stainless steel or plastic, remain in place for several days or several weeks before being removed.
Representative examples of ENT stents that can benefit from being coated with or having incorporated therein a fibrosis-inhibiting agent include lacrimal duct stents, Eustachian tube stents, nasal stents, and sinus stents.
In one aspect, the present invention provides for the combination of a lacrimal duct stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
In another aspect, the present invention provides for the combination of a Eustachian tube stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
In yet another aspect, the present invention provides for the combination of a sinus stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
In yet another aspect, the present invention provides for the combination of a nasal stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.

The ENT stent may be a choanal atresia stent composed of two long hollow tubes that are bridged by a flexible transverse tube. See, e.g., U.S.
Patent No. 6,606,995. The ENT stent may be an expandable nasal stmt for postoperative nasal packing composed of a highly porous, pliable and absorbent foam material capable of expanding outwardly, which has a nonadherent surface. See, e.g., U.S. Patent No. 5,336,163. The ENT stent may be a nasal stent composed of a deformable cylinder with a breathing passageway that has a smooth outer non-absorbent surface used for packing the nasal cavity following surgery. See, e.g., U.S. Patent No. 5,601,594. The ENT scent may be,a ventilation tube composed of a flexible, plastic, tubular vent with a rectangular flexible flange which is used for the nasal sinuses following endoscopic antrostomy. See, e.g., U.S. Patent No. 5,246,455. The ENT stent may be a ventilating ear tube composed of a shaft and an extended tab which is used for equalizing the pressure between the middle ear and outer ear. See, e.g., U.S. Patent No. 6,042,574. The ENT stent may be a middle ear vent tube composed of a non-compressible, tubular base and an eccentric flange. See, e.g., U.S. Patent No. 5,047,053.
ENT stents, which may be combined with one or more agents according to the present invention, include commercially available products such as Genzyme Corporation (Ridgefield, NJ) SEPRAGEL Sinus Stents and MEROGEL Nasal Dressing and Sinus Stents from Medtronic Xomed Surgical Products, Inc. (Jacksonville, FL).
In one aspect, the present invention provides ENT stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in ENT scents have been described above. Methods for incorporating fibrosis-inhibiting compositions onto or into the ENT stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the sfient structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of fihe specific stent, (b) coat the internal (luminal) surface of the stent, or (c) coat all or parts of both the internal and external surfaces of the device.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, ENT stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As ENT stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design.
However, certain principles can be applied in the application of this art.
Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in ENT
stents include the following: Cell Cycle Inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE
and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, fiacrolimus); (E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082); (I) antimycotic agents (e.g., sulconizole), (J) p38 MAP
kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the ENT
stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 pg-10 fig, or 10 g,g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 p.g/mm2 - 1 ~g/mm2, or 1 p,g/mm2 - 10 ~g/mm2, or 10 ~g/mm2 - 250 ~g/mm~, 250 ~,g/mm~ - 1000 ~g/mm2, or 1000 ~.g/mm2 - 2500 ~g/mm~.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with ENT stent devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total dose not to exceed 25 mg (range of 0.1 ~g to 25 mg); preferred 1 ~.g to 5 mg. The dose per unit area of 0.01 p,g - 100 p,g per mm2; preferred dose of 0.1 p,g/mm2 -10 pg/mm2. Minimum concentration of 10-8- 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 p,g to 5 mg); preferred 0.1 ~,g to 1 mg. The dose per unit area of the device of 0.01 ~.g - 20 ~g per mm2;
preferred dose of 0.05 ~g/mm2 - 3 ~g/mm2. Minimum concentration of 10-8 - 104 M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred g,g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~.g per mm2;
preferred dose of 0.25 ~,g/mm2 - 5 pg/mm2. Minimum concentration of 10-$ - 10-4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 10 mg (range of 0.1 p.g to 10 mg); preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 ~.g - 10 ~.g per mm2; preferred dose of 0.25 p,g/mm2 p,g/mm2. Minimum concentration of 10-8- 10~ M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 ~g - 100 ~.g per mm2; preferred dose of 0.5 ~.g/mm2 -10 ~g/mm2. Minimum concentration of 10-8 - 10~ M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~.g to 1 mg.
The dose per unit area of 0.1 ~g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 - 10 ~.g/mm2. Minimum concentration of 10-8 -~ 10~ M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof:
total dose not to exceed 20 mg (range of 0.1 ~g to 20 mg); preferred 1 ~,g to mg. The dose per unit area of the device of 0.1 p,g - 10 ~g per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA
reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof:
total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 pg per mm2; preferred dose of 2.5 p.g/mm2 - 500 ~.g/mm2. Minimum concentration of 10-8- 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 p.g to 300 mg. The dose per unit area of the device of 1.0 pg - 1000 pg per mm2;
preferred dose of 2.5 ~,g/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 -10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF
kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof:
total dose not to exceed 200 mg (range of 1.0 ~,g to 200 mg); preferred 1 ~,g to 50 mg. The dose per unit area of the device of 1.0 pg - 100 p,g per mm2;
preferred dose of 2.5 pg/mm2 - 50 pg/mm2. Minimum concentration of 10-8 - 10-4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~.g to 300 mg.
The dose per unit area of the device of 1.0 pg - 1000 p,g per mm2; preferred dose of 2.5 ~g/mmz - 500 p.g/mm2. Minimum concentration of 10-8- 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP Icinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 p,g - 1000 ~g per mm2;
preferred dose of 2.5 ~g/mm2 - 500 p,g/mm2. Minimum concentration of 10-8-10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred pg to 3 mg. The dose per unit area of the device of 0.1 pg - 10 pg per mm2;
preferred dose of 0.25 p,g/mm2 - 5 p.g/mm2. Minimum concentration of 10-8 - 10-4 M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with ENT stent devices include the following: (A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~g/mm~
- 10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 p.g/mm2 -10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~.g to 1 mg.
The dose per unit area of 0:1 p,g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 pg to 1 mg. The dose per unit area of 0.1 pg - 100 ~,g per mm2 of surface area; preferred dose of 0.3 pg/mm2 - 10 ~g/mm2. Minimum concentration of 10'$- 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~.g - 100 pg per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 p,g/mm2. Minimum concentration of 10'$- 10'4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 -10 ~g/mm~. Minimum concentration of 10'$- 10'4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 pg - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~g/mm~ -10 pg/mm~. Minimum concentration of 10'$- 10'4 M of ABT-578 is to be maintained on the device surface.
Ear Ventilation Tubes In another aspect, the present invention provides for the combination of an anti-scarring agent and an ear ventilation tube (also referred to as a tympanostomy tube).
Acute otitis media is the most common bacterial infection, the most frequent indication for surgical therapy, the leading cause of hearing loss and a common cause of impaired language development in children. The cost of treating this condition in children under the age of five is estimated at $5 billion annually in the United States alone. In fact, 85% of all children will have at least one episode of otitis media and 600,000 will require surgical therapy annually. The prevalence of otitis media is increasing and for severe cases surgical therapy is more cost effective than conservative management.
Acute otitis media (bacterial infection of the middle ear) is characterized by Eustachian tube dysfunction leading to failure of the middle ear clearance mechanism. The most common causes of otitis media are Streptococcus pneumoniae (30%), Haemophilus influenza (20%), Branhamella catarrhalis (12%), Streptococcus pyogenes (3%), and Staphylococcus aureus (1.5%). The end result is the accumulation of bacteria, white blood cells and fluid which, in the absence of an ability to drain through the Eustachian tube, results in increased pressure in the middle ear. For many cases antibiotic therapy is sufficient treatment and the condition resolves. However, for a significant number of patients the condition becomes frequently recurrent or does not resolve completely. In recurrent otitis media or chronic otitis media with effusion, there is a continuous build-up of fluid and bacteria that creates a pressure gradient across the tympanic membrane causing pain and impaired hearing. Fenestration of the tympanic membrane (typically with placement of a tympanostomy tube) relieves the pressure gradient and facilitates drainage of the middle ear (through the outer ear instead of through the Eustachian tube -a form of "Eustachian tube bypass").
Recurrent otitis media or otitis media with effusion may be treated with tympanostomy tubes or artificial Eustachian tubes/stents, such as described above. These ventilation tubes are indicated for chronic otitis media with effusion, recurrent acute otitis media, tympanic membrane atelectasis, and complications of acute otitis media in children. The excessive formation of granulation tissue around these devices can result in a decreased functioning of these devices. This can then result in a second procedure to either clear the obstruction or to insert a new device. The incorporation of a fibrosis-inhibiting agent into or onto the ventilation tubes may prevent the overgrowth of this granulation tissue.
Surgical placement of tympanostomy tubes is the most widely used treatment for chronic otitis media because, although not curative, it improves hearing (which in turn improves language development) and reduces the incidence of acute otitis media. Tympanostomy tube placement is one of the most common surgical procedures in the United States with 1.3 million surgical placements per year.
Representative examples of ear ventilation tubes that can benefit from being coated with or having incorporated therein a fibrosis-inhibiting agent include, without limitation, grommet-shaped tubes, T-tubes, tympanostomy tubes, drain tubes, tympanic tubes, otological tubes, myringotomy tubes, artificial Eustachian tubes, Eustachian tube prostheses, and Eustachian stents.
Ear ventilation tubes have been made out of, e.g., polytetrafluoroethylene (e.g., TEFLON), silicone, nylon, polyethylene and other polymers, stainless steel, titanium, and gold plated steel.
In one aspect, the ear ventilation tube may be a tympanostomy tube that is used to provide an alternative conduit for ventilation of the middle ear cavity via the external ear canal. Typically, ventilation of the middle ear is performed by conducting a myringotomy, in which a slit or opening in the tympanic membrane is surgically made to alleviate a buildup or reduction of pressure in the middle ear cavity and to drain accumulated fluids.
Tympanostomy tubes may be inserted into the surgical slit of the tympanic membrane to serve as a bypass for the normal Eustachian tube, which drains the middle ear cavity under normal conditions. For example, the tympanostomy tube may be an elongated uniform tubular member composed of pure titanium or titanium alloy that has a concavity inwardly spaced from one end that forms a flange. See, e.g., U.S. Patent No. 5,645,584. The tympanostomy tube may be composed of a micro-pitted titanium exterior flangeless surface used to ventilate the middle ear. See, e.g., U.S. Patent No. 4,971,076. The tympanostomy tube may be composed of a shaft with a tab that extends outwardly perpendicular from the bottom of the shaft. See, e.g., U.S. Patent No. 6,042,574. The tympanostomy tube may be a permanent ear ventilation device composed of an elongated tubular base having a flange eccentrically connected made of a non-compressible material. See, e.g., U.S. Patent No.
5,047,053. The tympanostomy tube may be composed of a cap-plug, central body and end cap, which together form a plurality of lumens within the tube.
See, e.g., U.S. Patent No. 5,851,199. The tympanostomy tube may be composed of a microporous resin cured to form a gas-permeable matrix containing a homogenous dispersion of silver particles capable of migrating to the surface of the tube sidewalls to provide antimicrobial activity. See, e.g., U.S. Patent No. 6,361,526. The tympanostomy tube may be composed of tubular body and a rib structure that projects outwardly to define a channel spiraling around the tubular body. See, e.g., U.S. Patent No. 5,775,336. The tympanostomy tube may be composed of an integral cutting tang extending from one of two flanges of a grommet for incising the tympanic membrane.
See, e.g., U.S. Patent Nos. 5,827,295 and 5,643,280. The tympanostomy tube may be composed of a tubular member having two opposed flanges in which the insertion of the tube is facilitated by a cutting edge on the flange which induces an incision of the tympanic membrane. See, e.g., U.S. Patent Nos.
5,489,286; 5,466,239; 5,254,120 and 5,207,685. Other tympanostomy tubes are described in, e.g., U.S. Patent Nos. 6,406,453; 5,178,623; 4,808,171 and 4,744,792.
In another aspect, the ear ventilation tube may be used to establish the normal function of the Eustachian tube and thus, attempt to resolve the stenosis that prevents its normal function. Fluid in the middle ear cavity normally secretes away from the tympanic membrane and thus, restoring the normal function of the Eustachian tube may provide optimal ventilation and drainage. For example, the ventilation tube may be an Eustachian stent composed of a hollow tubular body having a compressible core with two, connected parallel arms and a radially-oriented flange, which is placed in the Eustachian tube to maintain patency. See, e.g., U.S. Patent No. 6,589,286.
The ventilation tube may be an Eustachian tube prosthesis composed of a flexible tube having a flange that extends radially for positioning within the Eustachian tube passageway. See, e.g., U.S. Patent No. 4,015,607.
Tympanostomy tubes, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Medtronic Xomed, Inc. (Jackonsville, FL) sells a variety of ear ventilation tubes, including Long-Term Ventilation Tubes and Grommet Style Ventilation Tubes, including ARMSTRONG Grommets, GOODE T-Grommets, VENTURI Style Ventilation Tubes, SHEEHY Type Collar Buttons, REUTER Bobbins, COHEN T-Grommets, and SOILEAU TYTAN Titanium Tubes. Micromedics, Inc. (Eagan, MN) also sells a variety of ear ventilation tubes, including BAXTER Bevel Buttons, TINY TOUMA, SPOONER, TOUMA
T-Tubes, SHOEHORN Bobbins, SHAH, and SILVERSTEIN MICROWICK
Eustachian Tubes. Gyrus ENT LLC (Bartlett, TN) also sells a variety of ear ventilation tubes, including ULTRASIL Ventilation Tubes, RICHARDS COLLAR
Bobbins, BALDWIN BUTTERFLY Ventilation Tubes and PAPARELLA 2000 Tubes.
In one aspect, the present invention provides ear ventilation tube devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in ear ventilation tubes have been described above. These compositions can further include one or more fibrosis-inhibiting agents such that the overgrowth of granulation tissue is inhibited or reduced.
Numerous polymeric and non-polymeric delivery systems for use in ear ventilation tubes have been described above. Methods for incorporating the fibrosis-inhibiting agent or a composition comprising the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) a coating applied to the external surface of the ear ventilation tube; (b) a coating applied to the internal (luminal) surface of the ear ventilation tube;
or (c) a coating applied to all or parts of both surfaces.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin) and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, ear ventilation tubes may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As ear ventilation tube devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined.
Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in ear ventilation tubes include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus);
(E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA
reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082); (I) antimycotic agents (e.g., sulconizole), (J) p38 MAP kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the ear ventilation tube device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines.
The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 ~,g-10 ~,g, or 10 ~g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~g/mm2 - 1 ~glmm~, or 1 ~g/mm~ - 10 ~,g/mm~, or 10 ~,g/mm2 - 250 pglmm2, 250 ~.g/mm2 - 1000 ~glmm~, or 1000 ~,g/mm2 - 2500 ~.g/mm2.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with ear ventilation tube devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof:
total dose not to exceed 25 mg (range of 0.1 ~g to 25 mg); preferred 1 ~,g to mg. The dose per unit area of 0.01 ~g - 100 pg per mm~; preferred dose of 0.1 ~g/mm2 - 10 ~,g/mm2. Minimum concentration of 10-8 - 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 ~.g to 5 mg);
preferred 0.1 ~.g to 1 mg. The dose per unit area of the device of 0.01 ~,g -~g per mm2; preferred dose of 0.05 ~g/mm2 - 3 ~.g/mm2. Minimum concentration of 10-8 - 104 M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~,g per mm2; preferred dose of 0.25 ~g/mm2 - 5 ~g/mm2.
Minimum concentration of 10-8 - 10-4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg);
preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~,g per mmz; preferred dose of 0.25 ~.g/mm2 - 5 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~g per mm2; preferred dose of 0.5 ~g/mm2 -10 ~,g/mm2. Minimum concentration of 10-8- 10-4 M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 ~g/mm2. Minimum concentration of 10-8 - 10~' M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof: total dose not to exceed 20 mg (range of 0.1 ~g to 20 mg); preferred 1 ~,g to 5 mg. The dose per unit area of the device of 0.1 ~,g - 10 ~.g per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~.g/mm2.
Minimum concentration of 10-8 - 104 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~.g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~g per mm2; preferred dose of 2.5 ~g/mm2 - 500 ~.g/mm2. Minimum concentration of 10-8 - 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~g per mmz; preferred dose of 2.5 ~g/mm2 - 500 ~.g/mm2. Minimum concentration of 10-8 - 10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 1.0 ~,g to 200 mg); preferred 1 pg to 50 mg. The dose per unit area of the device of 1.0 ~.g - 100 ~,g per mm2; preferred dose of 2.5 ~,g/mmz - 50 ~,glmm2. Minimum concentration of 10-g- 10~ M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 pg - 1000 ~,g per mm2; preferred dose of 2.5 ~g/mm2 - 500 ~g/mm2. Minimum concentration of 10-8 - 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~.g to 2000 mg); preferred 10 p,g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~,g per mm2; preferred dose of 2.5 wg/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 - 10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 1 ~,g to 3 mg. The dose per unit area of the device of 0.1 ~,g - 10 ~.g per mm2; preferred dose of 0.25 ~g/mm2 - 5 ~.g/mm2. Minimum concentration of 10-8 - 10-4 M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with ear ventilation devices include the following: (A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 - 10 ~.g/mm~. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~.g to 1 mg.
The dose per unit area of 0.1 ~g - 100 ~.g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 - 10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p.g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~g per mm~ of surface area; preferred dose of 0.3 pg/mm2 -10 ~,g/mm2. Minimum concentration of 10-g- 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 p.g - 100 ~g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 - 10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 ~g - 100 pg per mm2 of surface area; preferred dose of 0.3 ~,g/mm~ - 10 p.g/mm2. Minimum concentration of 10-$- 10-4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg.
The dose per unit area of 0.1 ~g - 100 ~,g per mm~ of surface area; preferred dose of 0.3 ~,g/mm~ -10 ~g/mm2. Minimum concentration of 10-$- 10-4 M of ABT-578 is to be maintained on the device surface.
In addition to those described above (e.g., paclitaxel, TAXOTERE, and docetaxel), several other examples of anti-microtubule agents and appropriate dosages ranges for use with ear ventilation devices include vinca alkaloids such as vinblastine and vincristine sulfate and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg);
preferred 1 ~g to 3 mg. Dose per unit area of the device of 0.1 ~,g - 10 pg per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~g/mm2. Minimum concentration of 10-8- 10-4 M of drug is to be maintained on the device surface.
_Intraocular Implants In another aspect, the present invention provides for the combination of an anti-scarring agent and an intraocular implant.
In one embodiment, the intraocular implant is an intraocular lens device for the prevention of lens (e.g., anterior or posterior lens) opacification.
Eyesight deficiencies that may be treated with intraocular lenses include, without limitation, cataracts, myopia, hyperopia, astigmatism and other eye diseases. Intraocular lenses are most commonly used to replace the natural crystalline lens which is removed during cataract surgery. A cataract results from a change in the transparency of the normal crystalline lens in the eye.
When the lens becomes opaque from calcification (e.g., yellow and/or cloudy), the light cannot enter the eye properly and vision is impaired.
Implantation of intraocular lenses into the eye is a standard technique to restore useful vision in diseased or damaged eyes. The number of intraocular lenses implanted in the United States has grown exponentially over the last decade. Currently, over 1 million intraocular lenses are implanted annually, with the vast majority (90%) being placed in the posterior chamber of the eye. The intent of intraocular lenses is to replace the natural crystalline lens (i.e., aphakic eye) or to supplement and correct refractive errors (i.e., phakic eye, natural crystalline lens is not removed).
Implanted intraocular lenses may develop complications caused by mechanical trauma, inflammation, infection or optical problems. Mechanical and inflammatory injury may lead to reduced vision, chronic pain, secondary cataracts, corneal decompensation, cystoid macular edema, hyphema, uveitis or glaucoma. One common problem that occurs with cataract extraction is opacification which results from the tissue's reaction to the surgical procedure or to the artificial lens. Opacification leads to clouding of the intraocular lens, thus reducing the long-term benefits. Opacification typically results when proliferation and migration of epithelial cells occur along the posterior capsule behind the intraocular lens. Subsequent surgery may be required to correct this reaction; however, it involves a complex technical process and may lead to further serious, sight-threatening complications. Therefore, coating or incorporating the intraocular lens with a fibrosis-inhibiting agent may reduce these complications.
Representative examples of intraocular lenses that can benefit from being coated with or having incorporated therein a fibrosis-inhibiting agent include, without limitation, polymethylmethacrylate (PMMA) intraocular lenses, silicone intraocular lenses, achromatic lenses, pseudophakos, phakic lenses, aphakic lenses, multi-focal intraocular lenses, hydrophilic and hydrophobic acrylic intraocular lenses, intraocular implants, optic lenses and rigid gas permeable (RGP) lenses.
In one aspect, intraocular lenses may be foldable or rigid. The foldable lenses may be inserted in a small incision site using a tiny tube whereas the hard lenses are inserted through a larger incision site. Foldable lenses may be composed of silicone, acrylic or hydrogel whereas rigid lenses may be composed of hard polymeric compositions (PMMA).
In one aspect, the intraocular fens may be used as an implant for the treatment of cataracts, where the natural crystalline lens of the eye has been removed (i.e., aphakic lens). For example, the intraocular lens may be composed of two lenses having distinct refractive indices and distinct optical powers being joined together as an achromatic lens that may be connected within a posterior or anterior chamber of the eye. See, e.g., U.S. Patent No.
5,201,762. The intraocular lens may be secured in the posterior chamber by a system of posts that protrude through the iris attached to retaining rings.
See, e.g., U.S. Patent No. 4,053,953. The intraocular lens may be hard with a shape memory which is capable of deforming for insertion into the eye but will harden at normal body temperature. See, e.g., U.S. Patent No. 4,946,470. The intraocular lens may be coated with proteins, polypeptides, polyamino acids, polyamines or carbohydrates bound to the surface of the implant. See, e.g., U.S. Patent Nos. 6,454,802 and 6,106,554. Other examples of aphakic intraocular lenses are described in, e.g., U.S. Patent Nos. 6,599,317;
6,585,768; 6,558,419; 6,533,813; 6,210,438; 5,266,074; 4,753,654; 4,718,904 and 4,704,123.
In another aspect, the intraocular lens may be used as a corrective implant for vision impairment, where the natural crystalline lens of the eye has not been removed (i.e., phakic lens). For example, the intraocular lens may be a narrow profile, glare reducing, phakic anterior chamber lens that may be composed of an optic zone and a transition zone that has a curvature shaped to minimize direct glare. See, e.g., U.S. Patent No. 6,596,025. The intraocular lens may be a self-centering phakic lens inserted in the posterior chamber lens in which arms (i.e., haptic bodies) extend outwardly and protrude into the pupil such that the iris provides centering force to keep lens in place.
See, e.g., U.S. Patent No. 6,015,435. The intraocular lens may be composed of a circumferential edge and two haptics extending from the edge to a transverse member which is substantially straight or bowed inward toward the lens. See, e.g., U.S. Patent No. 6,241,777. Other examples of phakic intraocular lenses are described in, e.g., U.S. Patent Nos. 6,228,115;
5,480,428 and 5,222,981.

In another aspect, the intraocular lens may be a multi-focal lens capable of variable accommodation to enable the user to look through different portions of the lens to achieve different levels of focusing power. For example, the intraocular lens may be a variable focus lens composed of two lens portions with .an optical zone between the lenses which may contain a fluid reservoir and channel containing charged solution. See, e.g., U.S. Patent No. 5,443,506.
In another aspect, intraocular lenses may be deformable such that the lens may be folded for insertion through a tunnel incision. For example, the intraocular lens may be composed of a lens with fixation members for retaining the lens in the eye which may be configured for folding or rolling from a normal optical condition into an insertion condition to permit the lens to be passed through an incision into the eye. See, e.g., U.S. Patent No. 5,476,513. The intraocular lens may be composed of a resilient, deformable silicone based optic with a fixation means coupled to the optic for retaining the optic in the eye.
See, e.g., U.S. Patent No. 5,201,763. The intraocular lens may be composed of a copolymer of three constituents which may be deformable from its original shape. See, e.g., U.S. Patent No. 5,359,021. The intraocular lens may be composed of a transparent, flexible membrane with an interior sac and an attached bladder, in which optical fluid medium is shunted from the optical element to the bladder to aid in its deformity during insertion. See, e.g., U.S.
Patent No. 6,048,364. The intraocular lens may be a biocomposite composed of an optic portion made of high water content hydrogel capable of being folded and a haptic portion of low water content hydrogel having strength and rigidity.
See, e.g., U.S. Patent No. 5,211,662. Other deformable intraocular lenses are described in, e.g., U.S. Patent Nos. 6,267,784; 5,507,806 and U.S. Patent Application Publication No. 2003/0114928A1.
Other related devices and/or compositions (e.g., insertion devices) that may be used in conjunction with intraocular lenses are described in, e.g., U.S. Patent Nos. 6,629,979; 6,187,042; 6,113,633; 4,740,282 and U.S.
Patent Application Publication Nos. 2003/0212409A1 and 2003/0187455A1.
Intraocular lenses, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Alcon Laboratories, Inc. (Fort Worth, TX) sells the foldable ACRYSOF Intraocular Lens. Bausch & Lomb Surgical, Inc. (San Dimas, CA) sells the foldable SOFLEX SE Intraocular Lens. Advanced Medical Optics, Inc (Santa Ana, CA) sells the CLARIFLEX Foldable Intraocular Lens, SENSAR Acrylic lntraocular Lens, and PHACOFLEX II S140NB and S130NB.
The intraocular implant may comprise the fibrosis-inhibiting agent or a composition that includes the fibrosis-inhibiting agent directly.
Alternatively, or in addition, the agent may be coated, absorbed into, or bound onto the lens surface (e.g., to the haptics), or may be released from a hole (pore) or cavity outside the optical part of the lens surface.
The intraocular implants of the invention may be used in various surgical procedures. For example, the intraocular implant may be used in conjunction with a transplant for the cornea. Synthetic corneas can be used in patients loosing vision due to a degenarative cornea. Implanted synthetic corneas can restore patient vision, however, they often induce a fibrous foreign body response that limits their use. The intraocular implant of the present invention can prevent the foreign body response to the synthetic cornea and extend the cornea longevity. In another example, the synthetic cornea itself is coated with the agents of the invention, thus minimizing tissue reaction to corneal implantation.
In another aspect, the intraocular lens may be used in conjunction with treatment of secondary cataract after extracapsular cataract extraction.
As described above, the present invention provides intraocular lenses and other implants that include an anti-scarring agent or a composition that includes an anti-scarring agent. In one aspect, the anti-scarring agent is not paclitaxel or a derivative thereof.
Numerous polymeric and non-polymeric delivery systems for use in intraocular implants have been described above.
Methods for coating fibrosis-inhibiting compositions onto or into the implants include: (a) directly affixing to the implants a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the implant a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the implant with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) constructing the implant itself or a portion of the lens with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis-inhibiting agent directly to the lens surface or to a linker (small molecule or polymer) that is coated or attached to the implant surface. For these devices, the coating process can be performed in such a manner as to (a) coat the posterior surface of the specific implant, (b) coat the anterior surface of the implant or (c) coat all or parts of both the posterior and anterior surfaces of the device. The protruding arms of the implant can also be coated with the fibrosis-inhibiting agent.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
The process of coating these implants with a fibrosis-inhibiting agent or incorporating the fibrosis-inhibiting agent into the implant and the materials selected for these processes are such that they do not significantly alter the refractive index of the intraocular implant or the visible light transmission of the implant or lens.
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, intraocular implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As intraocular implants are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art.
Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surfiace concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in intraocular implants include the following: cell cycle inhibitor s including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus);
(E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA
reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7032); (I) antimycotic agents (e.g., sulconizole), (J) p3~ MAP kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the intraocular implant, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines.
The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 pg-10 ~,g, or 10 pg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~.g/mm~ - 1 pg/mm~, or 1 ~g/mm~ - 10 ~,g/mm2, or 10 pg/mm~ - 250 ~,g/mm2, 250 p,g/mm2 - 1000 ~.g/mm2, or 1000 ~,g/mm2 - 2500 ~,g/mm~.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with intraocular implants in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total dose not to exceed 25 mg (range of 0.1 p,g to 25 mg); preferred 1 p,g to 5 mg. The dose per unit area of 0.01 ~g - 100 ~g per mmz; preferred dose of 0.1 pg/mm2 -10 p,g/mm2. Minimum concentration of 10-8 - 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 5 mg (range of 0.01 ~g to 5 mg); preferred 0.1 pg to 1 mg. The dose per unit area of the device of 0.01 p,g - 20 ~,g per mm2;
preferred dose of 0.05 ~.g/mm2 - 3 p,g/mm2. Minimum concentration of 10-8 -M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 10 mg (range of 0.1 pg to 10 mg); preferred ~g to 3 mg. The dose per unit area of the device of 0.1 pg - 10 ~,g per mm2;
preferred dose of 0.25 p.g/mm2 - 5 p.g/mm2. Minimum concentration of 10-8 - 10 4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed mg (range of 0.1 ~g to 10 mg); preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 p,g - 10 ~.g per mm2; preferred dose of 0.25 ~,g/mm2 ~,g/mm2. Minimum concentration of 10'g - 10'4 M of etoposide is to be 5 maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 pg - 100 ~g per mm2; preferred dose of 0.5 ~,g/mm2 -10 ~,g/mm2. Minimum concentration of 10'8 - 10'4 M is to be maintained on the 10 device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.~1 ~.g to 10 mg); preferred 10 ~.g to 1 mg.
The dose per unit area of 0.1 ~.g - 100 ~.g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 -10 ~,g/mm2. Minimum concentration of 10-8 - 10'4 M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof:
total dose not to exceed 20 mg (range of 0.1 ~,g to 20 mg); preferred 1 ~g to mg. The dose per unit area of the device of 0.1 ~g - 10 p.g per mm2; preferred dose of 0.25 pg/mm2 - 5 ~,g/mm2. Minimum concentration of 10'8 - 104 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA
reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof:
total dose not to exceed 200 mg (range of 10.0 p,g to 200 mg); preferred 10 p.g to 100 mg. The dose per unit area of the device of 1.0 p.g - 1000 ~,g per mm2;
preferred dose of 2.5 ~.g/mmz - 500 ~.g/mm2. Minimum concentration of 10'$ -10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 10.0 ~,g to 200 mg); preferred 10 ~.g to 100 mg. The dose per unit area of the device of 1.0 ~.g - 1000 pg per mm2; preferred dose of 2.5 ~,g/mm2 - 500 ~,g/mm2. Minimum concentration of 10'8- 10'3 M of mycophenolic acid is to be maintained on the device surface. (H) NF kappa B
inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 1.0 ~.g to 200 mg); preferred 1 ~.g to 50 mg.
The dose per unit area of the device of 1.0 pg - 100 pg per mm2; preferred dose of 2.5 ~g/mm2 - 50 pglmm2. Minimum concentration of 10'8- 10'4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 10.0 ~,g to 200 mg); preferred 10 p,g to 100 mg. The dose per unit area of the device of 1.0 p.g - 1000 ~,g per mm2; preferred dose of 2.5 ~.g/mm2 - 500 pg/mm2. Minimum concentration of 10-8- 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 10.0 ~g to 200 mg); preferred 10 p,g to 100 mg. The dose per unit area of the device of 1.0 p,g -1000 ~.g per mm2; preferred dose of 2.5 wg/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 - 10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 1 ~.g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~,g per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~,g/mm2. Minimum concentration of 10-8 - 10-4 M of halofuginone bromide is to be maintained on the device surface.
Hypertrophic Scars and Keloids In another aspect, the present invention provides for the combination of an anti-scarring agent and a device for use in treating hypertrophic scars and keloids.
Hypertrophic scars and keloids are the result of an excessive fibroproliferative wound healing response. Briefly, healing of wounds and scar formation occurs in three phases: inflammation, proliferation, and maturation.
The first phase, inflammation, occurs in response to an injury which is severe enough to break the skin. During this phase, which lasts 3 to 4 days, blood and tissue fluid form an adhesive coagulum and fibrinous network which serves to bind the wound surfaces together. This is then followed by a proliferative phase in which there is ingrowth of capillaries and connective tissue from the wound edges, and closure of the skin defect. Finally, once capillary and fibroblastic proliferation has ceased, the maturation process begins wherein the scar contracts and becomes less cellular, less vascular, and appears flat and white.
This final phase may take between 6 and 12 months. If too much connective tissue is produced and the wound remains persistently cellular, the scar may become red and raised. If the scar remains within the boundaries of the original wound it is referred to as a hypertrophic scar, but if it extends beyond the original scar and into the surrounding tissue, the lesion is referred to as a keloid. Hypertrophic scars and keloids are produced during the second and third phases of scar formation. Several wounds are particularly prone to excessive endothelial and fibroblastic proliferation, including burns, open wounds, and infected wounds. With hypertrophic scars, some degree of maturation occurs and gradual improvement occurs. In the case of keloids however, an actual tumor is produced which can become quite large.
Spontaneous improvement in such cases rarely occurs.
A variety of devices for treating hypertrophic scars and keloids have been described. For example, the device may be an external tissue expansion device composed of two suture steel plates with adhesive attached foam cushions which apply constant continuous low grade force to skin and tissue to provide removal of hypertrophic scars and keloids. See, e.g., U.S.
Patent No. 6,254,624. The device may be a masking element which is pressed onto the scar tissue with an adjustable force by means of a pressure control unit and is connected with inflatable or suction members in the masking element. See, e.g., U.S. Patent No. 6,013,094. The treatment may be a device having locking elements and grasping structures such that the dermal and epidermal layers of a skin wound can be pushed together such that the tissue edges are abutting, such that a wound may be closed with minimal scarring.
See, e.g., U.S. Patent No. 5,591,206.
In another aspect, the hypertrophic scar or keloid may be treated by using a device in conjunction with a coating or sheet that may be used to deliver either anti-scarring agents alone, or anti-scarring compositions as described above. For example, the coating or sheet may be a copolymer composed of a hydrophilic polymer, such as polyethylene glycol, that is bound to a polymer that adsorbs readily to the surfaces of body tissues, such as phenylboronic acid. See, e.g., U.S. Patent No. 6,596,267. The coating or sheet may be a self-adhering silicone sheet which is impregnated with an antioxidant and/or antimicrobial. See, e.g., U.S. Patent No. 6,572,878. The coating or sheet may be a wound dressing garment composed of an outer pliable layer and a self-adhesive inner gel lining which serves as a dressing for contacting wounds. See, e.g., U.S. Patent No. 6,548,728. The coating or sheet may be a liquid composition composed of a film-forming carrier such as a collodion which contains one or more active ingredients such as a topical steroid, silicone gel and vitamin E. See, e.g., U.S. Patent No. 6,337,076. The coating or sheet may be a bandage with a scar treatment pad with a layer of silicone elastomer or silicone gel. See, e.g., U.S. Patent Nos. 6,284,941 and 5,891,076.
In another aspect, a medical device may be used in conjunction with an injectable composition that may be directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. The frequency of injections will depend upon the release kinetics of the polymer used (if present), and the clinical response. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., burns), and is preferably initiated after the proliferative phase has had time to progress (approximately 14 days after the initial injury), but before hypertrophic scar or keloid development. For example, an injectable treatment for hypertrophic scars and keloids may include the administration of an effective amount of angiogenesis inhibitor (e.g., fumagillol, thalidomide) as a systemic or local treatment to decrease excessive scarring. See, e.g., U.S. Patent No. 6,638,949. The injectable treatment may be a cryoprobe containing cryogen whereby it is positioned within the hypertrophic scar or keloid to freeze the tissue. See, e.g., U.S. Patent No. 6,503,246. The injectable treatment may be a method of locally administering an amount of botulinum toxin in or in close proximity to the skin wound, such that the healing is enhanced. See, e.g., U.S. Patent No.
6,447,787. The injectable treatment may be a method of administering an antifibrotic amount of fluoroquinolone to prevent or treat scar tissue formation.
See, e.g., U.S. Patent No. 6,060,474. The injectable treatment may be a composition of an effective amount of calcium antagonist and protein synthesis inhibitor sufficient to cause matrix degradation at a scar site so as to control scar formation. See, e.g., U.S. Patent No. 5,902,609. The injectable treatment may be a composition of non-biodegradable microspheres with a substantial surFace charge in a pharmaceutically acceptable carrier. See, e.g., U.S.
Patent No. 5,861,149. The injectable treatment may be a composition of endothelial cell growth factor and heparin which may be administered topically or by intralesional injection. See, e.g., U.S. Patent No. 5,500,409.
Treatments and devices used for hypertrophic scars and keloids, which may be combined with one or more agents according to the present invention, include commercially available products. Representative products include, for example, PROXIDERM External Tissue Expansion product for wound healing from Progressive Surgical Products (Westbury, NY), CICA-CARE Gel Sheet dressing product from Smith & Nephew Healthcare Ltd.
(India), and MEPIFORM Self-Adherent Silicone Dressing from Molnlycke Health Care (Eddystone, PA).
In one aspect, devices for the treatment of hypertrophic scars and keloids may be combined with a topical or injectable composition that includes an anti-scarring agent and a polymeric carrier suitable for application on or into hypertrophic scars or keloids. Incorporation of a fibrosis-inhibiting agent into a topical formulation or an injectable formulation is one approach to treat this condition. The topical formulation can be in the form of a solution, a suspension, an emulsion, a gel, an ointment, a cream, film or mesh. The injectable formulation can be in the form of a solution, a suspension, an emulsion or a gel. Polymeric and non-polymeric components that can be used to prepare these topical or injectable compositions are described above.
In another embodiment, the therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, devices for the treatment of hypertrophic scars and keloids may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including:
formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As devices for preventing hypertrophic scarring or keloids are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application.
Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use devices for treating hypertrophic scars and keloids include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus);
(E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA
reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082); (I) antimycotic agents (e.g., sulconizole), (J) p38 MAP kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 ~,g-~.g, or 10 ~.g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~g/mm~ - 1 ~,g/mm2, or 1 pg/mm2 - 10 p,g/mm2, or 10 ~,g/mm2 - 250 ~g/mm2, 250 ~glmm2 -1000 ~,g/mm~, or 1000 ~,g/mm~ - 2500 p.g/mm2.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with devices for treating hypertrophic scars and keloids in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total dose not to exceed 250 mg (range of 1.0 ~g to 250 mg); preferred 1 p,g to 100 mg. The dose per unit area of 0.01 ~,g - 500 ~g per mm2; preferred dose of 0.1 ~,g/mm2 -100 p.g/mm2. Minimum concentration of 10-8- 10~ M of doxorubicin is to be maintained on the device surface.
Mitoxantrone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 1.0 p,g to 200 mg); preferred 0.1 p,g to 75 mg. The dose per unit area of the device of 0.01 ~g - 300 ~g per mm2; preferred dose of 0.05 ~,g/mm2 - 75 ~g/mm2. Minimum concentration of 10-8 - 104 M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total dose not to exceed 250 mg (range of 1.0 ~g to 250 mg); preferred 1 ~.g to 100 mg.
The dose per unit area of the device of 0.1 ~g - 500 ~,g per mm2; preferred dose of 0.25 ~,g/mm2 -100 ~.g/mm2. Minimum concentration of 10-$ - 104 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total dose not to exceed 250 mg (range of 1.0 ~g to 250 mg); preferred 1 ~g to 100 mg. The dose per unit area of the device of 0.1 ~g - 500 O,g per mm2; preferred dose of 0.25 ~,glmm2 -~g/mm2. Minimum concentration of 10-8 - 104 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total dose not to exceed 250 mg (range of 1.0 ~,g to 250 mg); preferred 1 ~g to 100 mg. The dose per unit area of the device of 0.1 ~,g - 500 ~,g per mm2; preferred dose of 0.25 ~g/mm2 -100 ~,g/mm2. Minimum concentration of 10-8- 104 M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 250 mg (range of 1.0 ~.g to 250 mg);
preferred 1 ~,g to 100 mg. The dose per unit area of the device of 0.1 ~.g -~g per mm2; preferred dose of 0.25 ~.g/mm2 - 100 ~g/mm2. Minimum concentration of 10-8- 10-4 M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof: total dose not to exceed 250 mg (range of 1.0 ~,g to 250 mg); preferred 1 ~g to 100 mg. The dose per unit area of the device of 0.1 ~.g - 500 ~g per mm2; preferred dose of 0.25 ~,g/mm2 -100 ~.g/mm2. Minimum concentration of 10-8 - 104 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 ~,g per mm2; preferred dose of 2.5 ~,g/mm2 - 500 ~,g/mm2. Minimum concentration of 10-8 - 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~,g per mm2; preferred dose of 2.5 ~.g/mm2 - 500 ~g/mm2. Minimum concentration of 10-8 - 10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 1.0 ~g to 200 mg); preferred 1 ~,g to 50 mg. The dose per unit area of the device of 1.0 p,g - 100 ~,g per mm2; preferred dose of 2.5 ~,g/mm2 - 50 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~.g per mm2; preferred dose of 2.5 pg/mm2 - 500 ~g/mm2. Minimum concentration of 10-8 - 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total dose not to exceed 2000 mg (range of 10.0 p,g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~.g per mm2; preferred dose of 2.5 ~,g/mm2 - 500 ~g/mm2. Minimum concentration of 10-8 - 10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 p.g to 10 mg); preferred 1 ~,g to 3 mg. The dose per unit area of the device of 0.1 p,g - 10 ~,g per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of halofuginone bromide is to be maintained on the device surface.
Vascular Grafts In one aspect, the present invention provides for the combination of an anti-scarring agent and a vascular graft. Vascular graft devices that include a fibrosis-inhibiting agent are capable of inhibiting or reducing the overgrowth of granulation tissue, which can improve the clinical efficacy of these devices.
The vascular graft may be an extravascular graft or an intravascular (i.e., endoluminal) graft. The vascular graft may be, without limitation, in the form of a peripheral bypass application or a coronary bypass application. Vascular grafts may be used to replace or substitute damaged or diseased veins and arteries, including, without limitation, blood vessels damaged by aneurysms, intimal hyperplasia and thrombosis. Vascular grafts may also be used to provide access to blood vessels, for example, for hemodialysis access. Vascular grafts are implanted, for example, to provide an alternative conduit for blood flow through damaged or diseased areas in veins and arteries, including, without limitation, blood vessels damaged by aneurysms, intimal hyperplasia and thrombosis, however, the graft may lead to further complications, including, without limitation, infections, inflammation, thrombosis and intimal hyperplasia. The lack of long-term patency with vascular grafts may be due, for example, to surgical injury and abnormal hemodynamics and material mismatch at the suture line. Typically, further disease (e.g., restenosis) of the vessel occurs along the bed of the artery.
Some forms of improvements to vascular grafts have been made in an attempt to reduce the restenosis that occurs at the anastomosis site.
Improvements include: (a) using a Miller cuff, which is a small piece of natural vein to make a short cuff that is joined by stitching it to the artery opening and the prosthetic graft; (b) using a flanged graft whereby the graft has a terminal skirt or cuff that facilitates an end-to-side anastomosis; (c) using a graft with an enlarged chamber having a large diameter for suture at the anastomosis site;
and (d) using a graft that dispensing an agent that prevents thrombosis and/or intimal hyperplasia.
Representative examples of vascular grafts include, without limitation, synthetic bypass grafts (e.g., femoral-popliteal, femoral-femoral, axillary-femoral, and the like), vein grafts (e.g., peripheral and coronary), and internal mammary (e.g., coronary) grafts, bifurcated vascular grafts, intraluminal grafts, endovascular grafts and prosthetic grafts. Synthetic grafts can be made from a variety of polymeric materials, such as, for example, polytetrafluoroethylene (e.g., ePTFE), polyesters such as DACRON, polyurethanes, and combinations of polymeric materials.
Endoluminal vascular grafts may be used to treat aneurysms. For example, the vascular graft may be composed of a tubular graft with two tubular self-expanding stents that may be implanted for the treatment of aneurysms by means of minimally invasive procedures. See, e.g., U.S. Patent No. 6,168,620.
The vascular graft may be composed of a flexible tubular body and a compressible frame positioned against the tubular body for support which has pores on the surface to promote ingrowth. See, e.g., U.S. Patent No.
5,693,088. The vascular graft may be bifurcated endovascular graft having a tubular trunk and two tubular limbs. See, e.g., U.S. Patent No. 6,454,796. The vascular graft may be a kink-resistant endoluminal bifurcated graft having two separate lumens contacted by a single lumen section. See, e.g., U.S. Patent No. 6,551,350. The vascular graft may be an intraluminal tube composed of ePTFE that has a seamline formed by overlapping the edges such that the microstructure fibrils are oriented in perpendicular directions. See, e.g., U.S.
Patent No. 5,718,973.
In another aspect, the vascular graft may be used as a conduit to bypass vascular stenosis or other vascular abnormalities. For example, the vascular graft may be composed of a porous material having a layer of porous hollow fibers positioned along the inner surface which allows for tissue growth while inhibiting bleeding during the healing process. See, e.g., U.S. Patent No.
5,024,671. The vascular graft may be a flexible, monolithic, reinforced polymer tube having a microporous ePTFE tubular member and external ePTFE rib members projecting outwardly from the outer wall. See, e.g., U.S. Patent No.
5,609,624. The vascular graft may be composed of a tubular wall having longitudinally extending pleats that respond flexurally to changes in blood pressure while maintaining high compliance with reduced kinking. See, e.g., U.S. Patent No. 5,653,745. The vascular graft may be a radially supported ePTFE tube that is reinforced with greater density ring-shaped regions. See, e.g., U.S. Patent No. 5,747,128. The vascular graft may be porous PTFE
tubing composed of a microstructure of nodes interconnected by fibrils which has a coating of elastomer on the outer wall. See, e.g., U.S. Patent Nos.
5,152,782 and 4,955,899. The vascular graft may be a plurality of polymeric fibers knitted together composed of at least three different fibers in which two fibers are absorbable and one is non-absorbable. See, e.g., U.S. Patent Nos.
4,997,440; 4,871,365 and 4,652,264.
In another aspect, the vascular graft may be modified to reduce thrombus formation or intimal hyperplasia at the anastomotic site. For example, the vascular graft may have an enlarged chamber having a first diameter parallel to the axis of the tubular wall and a second diameter transverse to the axis of the tube. See, e.g., U.S. Patent No. 6,589,278. The vascular graft may have a flanged skirt or cuff section with facilitates an end-to-side anastomosis directly between the artery and the end of the flanged bypass graft. See, e.g., U.S. Patent No. 6,273,912. The vascular graft may be composed of a tubular wall having a non-thrombogenic agent within the luminal layer and a thrombogenic layer forming the exterior of the vascular graft. See, e.g., U.S.

Patent No. 6,440,166. The vascular graft may be composed of a smooth luminal surface made of ePTFE with a small pore size to reduce adherence of occlusive blood components. See, e.g., U.S. Patent No. 6,517,571. The vascular graft may be composed of hollow tubing that contains drug that is helically wrapped around the outer wall of a porous ePTFE graft whereby drug is dispensed by infusion through the porous interstices of the graft wall.
See, e.g., U.S. Patent No. 6,355,063.
In another aspect, the vascular graft may be a harvested blood vessel that is used for bypass grafting. For example, vascular grafts may be composed of harvested arterial vessels from a host, such as the internal mammary arteries or inferior epigastric arteries. See, e.g., U.S. Patent No.
5,797,946. Vascular grafts may also be composed of saphenous veins which may be harvested from the host and used for coronary bypass or peripheral bypass procedures. See, e.g., U.S. Patent No. 6,558,313.
Other examples of vascular grafts are described in U.S. Patent Nos. 3,096,560, 3,805,301, 3,945,052, 4,140,126, 4,323,525, 4,355,426, 4,475,972, 4,530,113, 4,550,447, 4,562,596, 4,601,718, 4,647,416, 4,878,908, 5,024,671, 5,104,399, 5,116,360, 5,151,105, 5,197,977, 5,282,824, 5,405,379, 5,609,624, 5,693,088, and 5,910,168.
Vascular grafts, which may be combined with one or more agents according to the present invention, include commercially available products.
GORE-TEX Vascular Grafts and GORE-TEX INTERING Vascular Grafts are sold by Gore Medical Division (W. L. Gore & Associates, Inc. Newark, DE).
C.R. Bard, Inc. (Murray Hill, NJ) sells the DISTAFLO Bypass Grafts and IMPRA
CARBOFLO Vascular Grafts.
In one aspect, the anti-scarring agent or a composition containing the anti-scarring agent is combined with a vascular graft.
Numerous polymeric and non-polymeric delivery systems for use in vascular grafts have been described above. Methods for incorporating fibrosis-inhibiting agents or fibrosis-inhibiting compositions onto or into the graft include: (a) affixing (directly or indirectly) to the graft a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) incorporating or impregnating into the graft a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the graft with a substance such as a hydrogel which will in turn absorb the fibrosis inhibiting composition, (d} constructing the graft itself or a portion of the graft with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis-inhibiting agent directly to the graft surface or to a linker (small molecule or polymer) that is coated or attached to the graft surface. For these grafts, the coating process can be performed in such a manner as to (a) coat the external surface of the graft, (b) coat the interior (luminal) surface of the graft, or (c) coat all or parts of both the external and internal surfaces of the graft, or (d) coat at least one end of the graft.
The fibrosis-inhibiting agent can be incorporated directly into the coating composition or into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In yet another embodiment, a gel, paste, thermogel or in situ forming gel that includes a fibrosis-inhibiting agent can be applied in a perivascular manner to the anastomosis produced during implantation of the graft device. Numerous polymeric and non-polymeric delivery systems for use in paste and gel formulations have been described above. The fibrosis-inhibiting agent can be incorporated directly into the gel or paste composition, or the therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above).
In another aspect, the fibrosis-inhibiting agent can be incorporated into or onto an implant (e.g., a film or mesh material), which can be used in conjunction with a vascular graft to inhibit scarring at an anastomotic site.
For example, a film or mesh material may be placed or wrapped in a perivascular (periadventitial) manner around the outside of the anastomosis at the time of surgery. Film and mesh implants may be used with a various types of vascular grafts, including synthetic bypass grafts (femoral-popliteal, femoral-femoral, axillary-femoral etc.), vein grafts (peripheral and coronary), internal mammary (coronary) grafts or hemodialysis grafts (AV fistulas, AV access grafts).
Representative examples of films and meshes are described in further detail below.

In addition to the fibrosis-inhibiting agent, the vascular graft devices compositions for use with vascular graft devices can also further contain an anti-inflammatory agent (e.g., dexamethazone or aspirin) and/or an anti-thrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, dipyridamole, or aspirin). The combination of agents may be coated onto the entire or portions of the vascular graft such that the thrombogenicity and/or fibrosis is reduced or inhibited. In certain embodiments, these agents may be coated onto the vascular graft using biodegradable polymers. For example, polymeric material that forms a gel in the pores and/or on the surface of the graft may be used, such as alginates, chitosan and chitosan sulfate, hyaluronic acid, dextran sulfate, PLURONIC polymers, chain extended PLURONIC polymers, polyester-polyether block copolymers of the various configurations (e.g., MePEG-PLA, PLA-PEG-PLA, and the like).
In one aspect, synthetic vascular grafts are provided that comprise, in addition to the anti-fibrosing agent, a composition in the form of a biodegradable gel. The gel composition can have anti-thrombogenic properties or include an agent having anti-thrombogenic properties, which may or may not be released from the gel composition. Gel coated grafts may reduce or prevent early thrombotic events commonly associated with implantation of synthetic grafts.
Polymeric biodegradable gels may comprise, for example, a chain extended PLURONIC polymer. Chain extended polymers may include a PLURONIC polymer (e.g., F127, Fi37, or the like) that has been reacted with a difunctional molecule such as succinyl chloride to increase the molecular weight of the polymer and thereby increase the viscosity of the PLURONIC polymer.
Chain extended polymers can be dissolved in a solvent and then coated onto the synthetic vascular graft.
Gel compositions may be formed from a combination of small and/or polymeric molecules having two or more electrophilic groups and two or more nucleophilic groups. For example, the formulations may include a combination of a multi-armed PEG molecule in which the terminal hydroxyl groups are activated with succinimidyl moieties and a multi-armed PEG
molecule having terminal amino and/or sulfhydryl groups. The multi-armed PEG reagents may be dissolved separately in an appropriate solvent (e.g., aqueous buffer, IPA, dichloromethane, or a combination of solvents) and then sprayed sequentially or simultaneously onto the desired surface of the graft, such that the two components react to produce a crosslinked gel. The solvent may then be removed by air or vacuum drying.
In another embodiment, the composition may be formed from a polymer having two or more succinimidyl groups and a small molecule having two or more amino or sulfhydryl groups (e.g., dilysine). Alternatively, the polymer components can include two or more sulfhydryl groups or amino groups, and the small molecule contains two or more succinimidyl groups.
In yet another embodiment, gel coatings may be produced from polyester-polyether block copolymers of various configurations (e.g., X-Y, X-Y-X or Y-X-Y, R-(Y-X)", R-(X-Y)" where X is a polyalkylene oxide and Y is a polyester (e.g., polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y-decanolactone, ~-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-Zone.), R is a multifunctional initiator and copolymers as well as blends and copolymers thereof.) may be used to form the gel coating.
In one embodiment, the synthetic vascular graft is formed of a porous synthetic material such as expanded PTFE (ePTFE). A coating comprising a gel composition, such as described above, may be applied onto the entire graft or a portion of the graft surface (e.g., the interior surface of the graft or the ends of the graft). Further, the pores of the graft may be either partially or fully filled with the coating composition. The extent to which the coating occupies the pores of the device can be altered by changing the solvent Z5 used to dissolve the polymer. For example, a surface coating may be achieved by using a hydrophilic solvent such as water which will not wet the hydrophobic surface of an ePTFE graft. Coating from a solvent such as dichloromethane, which wets an ePTFE surface, can be used to coat the polymer composition onto the inner pore structure of the graft.
The gel formulations may have anti-thrombogenic properties due to the hydrophilicity. Hydrophilic coatings may be physically removed from the surface of the graft over time which may reduce the adhesion of platelets to the graft surface. Additionally, an anti-thrombogenic agent (e.g., heparin, fragments of heparin, organic soluble salts of heparin, sulfonated carbohydrates, warfarin, coumadin, coumarin, heparinoid, danaparoid, ' argatroban chitosan sulfate, or chondroitin sulfate) may be included into the formulation. In one embodiment, the anti-thrombotic agents) may be incorporated into microspheres. Other additives which may be added into gel compositions for use with vascular grafts include buffers, osmolality modifiers, viscosity modifiers, and hydrating agents (e.g., PEG, MePEG, and various sugars).
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, vascular grafts may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As vascular grafts are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art.
Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use with vascular grafts include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus);
(E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA
reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082); (I) antimycotic agents (e.g., sulconizole), (J) p38 MAP kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
Regardless of the method of application of the drug to the vascular graft, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines.
The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 ~.g-10 ~.g, or 10 ~g-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~,g/mm2 - 1 pg/mm2, or 1 ~,g/mm2 - 10 pg/mm2, or 10 ~,g/mm~ - 250 g.g/mm2, 250 ~g/mm~ - 1000 pg/mm2, or 1000 ~glmm2 - 2500 pg/mm2.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with vascular graft devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof: total amount of drug on the device not to exceed 25 mg (range of 0.1 ~,g to 25 mg);
preferred 1 ~g to 5 mg. The dose per unit area of 0.01 pg - 100 ~g per mm2;
preferred dose of 0.1 p,g/mm2'-10 ~g/mm2. Minimum concentration of 10-8- 10-4 M of doxorubicin is to be maintained on the device surface. Mitoxantrone and analogues and derivatives thereof: total amount of drug on the device not to exceed 5 mg (range of 0.01 ~.g to 5 mg); preferred 0.1 ~.g to 1 mg. The dose per unit area of the device of 0.01 pg - 20 p,g per mm2; preferred dose of 0.05 pg/mm2 - 3 ~g/mm2. Minimum concentration of 10-8 - 104 M of mitoxantrone is to be maintained on the device surface. B) Cell cycle inhibitors including Paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total amount of drug on the device not to exceed 10 mg (range of 0.1 pg to 10 mg);
preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 pg - 10 p.g per mm2;
preferred dose of 0.25 pg/mm2 - 5 p.g/mm2. Minimum concentration of 10-$- 10-4 M of paclitaxel is to be maintained on the device surface. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total amount of drug on the device not to exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 1 p,g to mg. The dose per unit area of the device of 0.1 ~,g - 10 p,g per mm2;
preferred dose of 0.25 g.g/mm2 - 5 pg/mm2. Minimum concentration of 10-$ - 10-4 M of etoposide is to be maintained on the device surface. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE):
Total amount of drug on the device not to exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~,g per mm2; preferred dose of 0.5 pg/mm2 -10 ~g/mm2. Minimum concentration of 10-8 - 10~ M is to be maintained on the device surface. Everolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~g - 100 pg per mm2 of surface area; preferred dose of 0.3 pg/mm2 -10 ~.g/mm2. Minimum concentration of 10-8 - 10~ M of everolimus is to be maintained on the device surface. (E) Heat shock protein 90 antagonists (e.g., geldanamycin} and analogues and derivatives thereof: total amount of drug on the device not to exceed 20 mg (range of 0.1 ~g to 20 mg); preferred 1 ~g to 5 mg. The dose per unit area of the device of 0.1 ~g - 10 p.g per mm2; preferred dose of 0.25 ~,g/mm2 - 5 ~g/mm2. Minimum concentration of 10-8- 10-4 M of geldanamycin is to be maintained on the device surface. (F) HMGCoA reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 ~.g to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 pg per mm2; preferred dose of 2.5 pg/mm2 - 500 ~g/mmz. Minimum concentration of 10-$ - 10-3 M of simvastatin is to be maintained on the device surface. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 pg to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 pg - 1000 ~,g per mm2; preferred dose of 2.5 ~g/mm2 - 500 p.glmm2. Minimum concentration of 10-8 - 10-3 M of mycophenolic acid is to be maintained on the device surface. (H) NF kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof: total amount of drug on the device not to exceed 200 mg (range of 1.0 ~g to 200 mg); preferred 1 ~g to 50 mg. The dose per unit area of the device of 1.0 ~g - 100 ~,g per mm2; preferred dose of 2.5 pg/mm2 -50 ~,g/mm2. Minimum concentration of 10-8- 10-4 M of Bay 11-7082 is to be maintained on the device surface. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~,g to 300 mg.
The dose per unit area of the device of 1.0 ~,g - 1000 ~g per mm2; preferred dose of 2.5 ~g/mm2 - 500 ~g/mm2. Minimum concentration of 10-g - 10-3 M of sulconizole is to be maintained on the device surface. (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 ~.g per mmz; preferred dose of 2.5 ~g/mm2 - 500 ~.g/mm2. Minimum concentration of 10-8 - 10-3 M of SB202190 is to be maintained on the device surface. (K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 1 ~.g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~g per mm2; preferred dose of 0.25 ~g/mm2 - 5 ~.g/mm2. Minimum concentration of 10-8- 10~ M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with vascular graft devices include the following: (A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~.g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 pg/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 - 10 ~g/mm2. Minimum concentration of 10'$- 10'4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 ~,g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~,g/mm2 -10 ~,g/mm~. Minimum concentration of 10-$- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 - 10 ~,g/mm2. Minimum concentration of 10-$ - 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~g/mm~ -10 ~,g/mm~. Minimum concentration of 10-$ - 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 -10 p,g/mm2. Minimum concentration of 10'$- 10-4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg.
The dose per unit area of 0.1 ~g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 -10 pg/mm2. Minimum concentration of 10-$- 10-4 M of ABT-578 is to be maintained on the device surface.
In addition to those described above (e.g., paclitaxel, TAXOTERE, and docetaxel), several other examples of anti-microtubule agents and appropriate dosages ranges for use with vascular graft devices include vinca alkaloids such as vinblastine and vincristine sulfate and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~.g to 10 mg);
preferred 1 p,g to 3 mg. Dose per unit area of the device of 0.1 ~g - 10 p.g per mm2; preferred dose of 0.25 pg/mm2 - 5 pg/mm2. Minimum concentration of 10-$- 10-4 M of drug is to be maintained on the device surface.
Hemodialysis Access Devices In one aspect, the present invention provides for the combination of an anti-scarring agent and a hemodialysis access device. Hemodialysis dialysis access devices that include a fibrosis-inhibiting agent are capable of inhibiting or reducing the overgrowth of granulation tissue, which can improve the clinical efficacy of these devices.
Hemodialysis access devices may be used when blood needs to be removed, cleansed and then returned to the body. Hemodialysis regulates the body's fluid and chemical balances as well as removes waste from the blood stream that cannot be cleansed by a normally functioning kidney due to disease or injury. For hemodialysis to occur, the blood may be obtained through a hemodialysis access or vascular access, in which minor surgery is performed to provide access through an AV fistula or AV access graft. These hemodialysis access devices may develop complications, including infections, inflammation, thrombosis and intimal hyperplasia of the associated blood vessels. The lack of long-term patency with hemodialysis access may be due to surgical injury, abnormal hemodynamics and material mismatch at the suture line. Typically, further disease (e.g., restenosis) of the vessel occurs along the bed of the artery and/or at the site of anastomosis.
In addition to the AV fistulas and AV access grafts described above, implantable subcutaneous hemodialysis access systems such as the commercially available catheters, ports, and shunts, may also be used for hemodialysis patients. These access systems may consist of a small metallic or polymeric device or devices implanted underneath the skin. These devices may be connected to flexible tubes, which are inserted into a vessel to allow for blood access.
Representative examples of hemodialysis access devices include, without limitation, AV access grafts, venous catheters, vascular grafts, implantable ports, and AV shunts. Synthetic hemodialysis access devices can , be made from metals or polymers, such as polytetrafluoroethylene (e.g., ePTFE), polyesters such as DACRON, polyurethanes, or combinations of these materials.
In one aspect, the hemodialysis access device may be an AV
access graft. For example, the AV access graft may be composed of an implantable self-expanding flexible percutaneous stent-graft of open weave construction with ends being compressible and having an elastic layer arranged along a portion of its length. See, e.g., U.S. Patent Nos. 5,755,775 and 5,591,226. The AV access graft may be composed of a tubular section with a generally constant diameter which tapers towards the venous end. See, e.g., U.S. Patent No. 6,585,762. The AV access graft may be composed of a two microporous ePTFE tubes that are circumferentially disposed over each other with a polymeric layer interposed between such that the graft is self-sealing and exhibits superior radial tensile strength and suture hole elongation resistance.
See, e.g., U.S. Patent No. 6,428,571. The AV access graft may be composed of a coaxial double lumen tube with an inner and outer tube having a self-sealing, nonbiodegradable, polymeric adhesive between the tubes. See, e.g., U.S. Patent No. 4,619,641. The AV access graft may be composed of a synthetic fabric having a high external velour profile which is woven or knitted to form a tubular prosthesis which has elastic fibers that allows self-sealing following a punctured state. See, e.g., U.S. Patent No. 6,547,820. The AV
access graft may be of tubular form having a base tube with the ablumenal surface covered with a deflectable material, such as a porous film, which is arranged adjacently to allow movement. See, e.g., U.S. Patent No. 5,910,168.

In another aspect, the hemodialysis access device may be a catheter system. For example, the catheter system may be composed of a suction and return line that are adapted for disposition in the vascular system of the body and are connected to a subcutaneous connector port. See, e.g., U.S.
Patent Nos. 6,620,118 and 5,989,206. The catheter system may be an apparatus that is used to arterialize a vein by creating an AV fistula by inserting a catheter into a vein and a catheter into an adjacent artery. See, e.g., U.S.
Patent No. 6,464,665. The catheter system may be composed of a hollow sheath that provides percutaneous introduction of fistula-generating vascular catheters through a perforation in a vessel wall, such that the catheters generate an intervascular fistula on-demand between adjacent vessels. See, e.g., U.S. Patent Nos. 6,099,542 and 5,830,224.
In another aspect, the hemodialysis access device may be used for an AV fistula. For example, the hemodialysis access device may be an AV
fistula assembly composed of a synthetic coiled stent graft with helically extending turns with gaps used to enhance the function of an AV fistula. See, e.g., U.S. Patent No. 6,585,760.
In another aspect, the hemodialysis access device may be an implantable access port, shunt or valve. These devices may be implanted subcutaneously with communication to the blood supply and accessed using a percutaneous puncture. For example, the hemodialysis access device may be composed of housing having an entry port and an exit port to a passageway which has an elastomeric sealing valve that provides access into the exit port for a needle. See, e.g., U.S. Patent No. 5,741,228. The hemodialysis access device may be a shunt composed of a slideable valve and flexible lid that has a fluid communication tube between the arterial and venous ends. See, e.g., U.S. Patent No. 5,879,320. The hemodialysis access device may be a shunt in the form of a junction that has a connector with two legs that are inserted into the native blood vessel and one leg that is adapted for sealing to another blood vessel without punctures. See, e.g., U.S. Patent No. 6,019,788. The hemodialysis access device may be a surface access double hemostatic valve that may be mounted on the wall of an AV graft for hemodialysis access. See, e.g., U.S. Patent Nos. 6,004,301 and 6,090,067.
Hemodialysis access devices, which may be combined with one or more agents according to the present invention, include commercially available products. For example, hemodialysis access devices include products, such as the LIFESITE (Vasca Inc., Tewksbury, MA) and the DIALOCK catheters from Biolink Corp. (Middleboro, MA), VECTRA Vascular Access Grafts and VENAFLO Vascular Grafts from C.R. Bard, Inc. (Murray Hill, NJ), and GORE-TEX Vascular Grafts and Stretch Vascular Grafts from Gore Medical Division (W. L. Gore & Associates, Inc. Newark, DE).
In one aspect, the anti-scarring agent or a composition containing the anti-scarring agent is combined with a hemodialysis access device.
Numerous polymeric and non-polymeric delivery systems for use in hemodialysis access devices have been described above. Methods for incorporating fibrosis-inhibiting agents or compositions comprising fibrosis-inhibiting agents onto or into the hemodialysis access device include: (a) directly affixing to the hemodialysis access device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the hemodialysis access device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the hemodialysis access device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) constructing the hemodialysis access device itself or a portion of the graft with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis-inhibiting agent directly to the hemodialysis access device surface or to a linker (small molecule or polymer) that is coated or attached to the hemodialysis access device surface. For devices that are coated, the coating process can be performed in such a manner as to (a) coat only the external surface of the device; (b) coat the internal (luminal) surface of the device; or (c) coat all or parts of both the external and internal surfaces.
In another aspect, the fibrosis-inhibiting agent or a composition containing the fibrosis-inhibiting agent can be incorporated into an implant, such as a film or mesh, which can be used in conjunction with a hemodialysis access device to inhibit scarring at the site of an anastomosis or fistula. These films or meshes may be placed or wrapped in a perivascular (periadventitial) manner around the outside of the fistula or anastomosis at the time of surgery.
Representative examples of implants (i.e., meshes and films) for use with hemodialysis access devices are described below.
In yet another aspect, a composition in the form of, for example, a gel, paste, thermogel, or in situ forming gel, which includes a fibrosis-inhibiting agent can be applied in a perivascular manner to the fistula or anastomosis produced during implantation of the hemodialysis access device.
The fibrosis-inhibiting agent can be incorporated directly into the gel or paste composition, or the therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above) that is then incorporated into the composition that is to be delivered. Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly (hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In addition to the fibrosis-inhibiting agent, hemodialysis access devices and compositions for use with hemodialysis access devices can also further contain an anti-inflammatory agent (e.g., dexamethazone or aspirin) and/or an anti-thrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, dipyridamole, or aspirin).
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, hemodialysis access devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Several examples of fibrosis-inhibiting agents for use with hemodialysis access devices include the following: cell cycle inhibitors including (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) taxanes (e.g., paclitaxel, TAXOTERE and docetaxel), and (C) podophyllotoxins (e.g., etoposide); (D) immunomodulators (e.g., sirolimus, everolimus, tacrolimus);
(E) heat shock protein 90 antagonists (e.g., geldanamycin); (F) HMGCoA
reductase inhibitors (e.g., simvastatin); (G) inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3); (H) NF kappa B inhibitors (e.g., Bay 11-7082); (I) antimycotic agents (e.g., sulconizole), (J) p38 MAP kinase inhibitors (e.g., SB202190), and (K) and anti-angiogenesis agents (e.g., halofuginone bromide), as well as analogues and derivatives of the aforementioned.
As hemodialysis access devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), and total amount of drug on the device can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 p.g-pg, or 10 pg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 ~g/mm~ - 1 ~g/mm2, or 1 ~g/mm2 - 10 ~g/mm2, or 10 ~,g/mm2 - 250 ~,g/mm~, 250 p,g/mm2 -1000 ~,g/mm2, or 1000 ~g/mm2 - 2500 ~.g/mm2.
Provided below are exemplary dosage ranges for various anti-scarring agents that can be used in conjunction with hemodialysis access devices in accordance with the invention. A) Cell cycle inhibitors including doxorubicin and mitoxantrone. Doxorubicin analogues and derivatives thereof:
total amount of drug on the device not to exceed 25 mg (range of 0.1 ~g to 25 mg); preferred 1 ~,g to 5 mg. The dose per unit area of 0.01 p,g - 100 ~.g per mm2; preferred dose of 0.1 ~,g/mm2 - 10 wg/mm2. Minimum concentration of 10-8 - 10-4 M of doxorubicin is to be maintained on the device surface.
Mitoxantrone and analogues and derivatives thereof: total amount of drug on the device not to exceed 5 mg (range of 0.01 ~g to 5 mg); preferred 0.1 ~,g to mg. The dose per unit area of the device of 0.01 p.g - 20 ~,g per mm2;
preferred dose of 0.05 ~.g/mm2 - 3 ~.g/mm2. Minimum concentration of 10-$ - 104 M of mitoxantrone is to be maintained on the device surface for up to 90 days. B) Cell cycle inhibitors including paclitaxel and analogues and derivatives (e.g., docetaxel) thereof: total amount of drug on the device not to exceed 10 mg (range of 0.1 pg to 10 mg); preferred 1 ~,g to 3 mg. The dose per unit area of the device of 0.1 p.g - 10 p,g per mm2; preferred dose of 0.25 p,g/mm2 - 5 ~.g/mmz. Minimum concentration of 10-8- 10~ M of paclitaxel is to be maintained on the device surface for up to 90 days. (C) Cell cycle inhibitors such as podophyllotoxins (e.g., etoposide): total amount of drug on the device not to exceed 10 mg (range of 0.1 p.g to 10 mg); preferred 1 p.g to 3 mg. The dose per unit area of the device of 0.1 pg - 10 ~g per mm2; preferred dose of 0.25 pg/mm2 - 5 pg/mm2. Minimum concentration of 10-8 - 10~ M of etoposide is to be maintained on the device surface for up to 90 days. (D) Immunomodulators including sirolimus and everolimus. Sirolimus (i.e., rapamycin, RAPAMUNE): Total amount of drug on the device not to exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 pg - 100 p,g per mm2; preferred dose of 0.5 p.g/mm2 -10 pg/mm2.
Minimum concentration of 10-8 - 10-4 M is to be maintained on the device surface for up to 90 days. Everolimus and derivatives and analogues thereof:
Total dose should not exceed 10 mg (range of 0.1 pg to 10 mg); preferred 10 ~,g to 1 mg. The dose per unit area of 0.1 p.g - 100 pg per mm2 of surface area;
preferred dose of 0.3 ~,g/mm2 -10 ~g/mm2. Minimum concentration of 10-8 - 10-4 M of everolimus is to be maintained on the device surface for up to 90 days.
(E) Heat shock protein 90 antagonists (e.g., geldanamycin) and analogues and derivatives thereof: total dose not to exceed 20 mg (range of 0.1 p.g to 20 mg);
preferred 1 pg to 5 mg. The dose per unit area of the device of 0.1 pg - 10 pg per mm2; preferred dose of 0.25 p,g/mm2 - 5 pg/mmz. Minimum concentration of 10-8- 10-4 M of paclitaxel is to be maintained on the device surface for up to 90 days. (F) HMGCoA reductase inhibitors (e.g., simvastatin) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~g to 2000 mg); preferred 10 pg to 300 mg. The dose per unit area of the device of 1.0 ~g - 1000 ~g per mm2; preferred dose of 2.5 ~,g/mm2 - 500 p,g/mm2. Minimum concentration of 10-8 - 10-3 M of simvastatin is to be maintained on the device surface for up to 90 days. (G) Inosine monophosphate dehydrogenase inhibitors (e.g., mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg);
preferred 10 p,g to 300 mg. The dose per unit area of the device of 1.0 ~g -1000 pg per mm2; preferred dose of 2.5 ~,g/mm2 - 500 p,g/mm2. Minimum concentration of 10-8 - 10'3 M of mycophenolic acid is to be maintained on the device surface for up to 90 days. (H) NF kappa B inhibitors (e.g., Bay 11-7082) and analogues and derivatives thereof: total amount of drug on the device not to exceed 200 mg (range of 1.0 p,g to 200 mg); preferred 1 ~,g to 50 mg. The dose per unit area of the device of 1.0 ~.g - 100 ~g per mm2; preferred dose of 2.5 p.g/mm2 - 50 ~,g/mm2. Minimum concentration of 10'8- 10~ M of Bay 11-7082 is to be maintained on the device surface for up to 90 days. (I) Antimycotic agents (e.g., sulconizole) and analogues and derivatives thereof:
total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~.g to 2000 mg); preferred 10 ~g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 ~g per mm2; preferred dose of 2.5 ~.g/mm2 - 500 p,g/mm2.
Minimum concentration of 10'8- 10-3 M of sulconizole is to be maintained on the device surface for up to 90 days and (J) p38 MAP kinase inhibitors (e.g., SB202190) and analogues and derivatives thereof: total amount of drug on the device not to exceed 2000 mg (range of 10.0 ~,g to 2000 mg); preferred 10 ~,g to 300 mg. The dose per unit area of the device of 1.0 ~,g - 1000 pg per mm2;
preferred dose of 2.5 ~,g/mm2 - 500 ~g/mm2. Minimum concentration of 10'8 -10'3 M of SB202190 is to be maintained on the device surface for up to 90 days.
(K) Anti-angiogenic agents (e.g., halofuginone bromide) and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg);
preferred 1 ~g to 3 mg. The dose per unit area of the device of 0.1 ~g - 10 ~.g per mm2; preferred dose of 0.25 ~g/mm2 - 5 ~g/mm2. Minimum concentration of 10'$ - 10'4 M of halofuginone bromide is to be maintained on the device surface.
In addition to those described above (e.g., sirolimus, everolimus, and tacrolimus), several other examples of immunomodulators and appropriate dosages ranges for use with hemodialysis access devices include the following:
(A) Biolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~.g to 1 mg. The dose per unit area of 0.1 ~,g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 - 10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of everolimus is to be maintained on the device surface. (B) Tresperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 -10 ~,g/mm2. Minimum concentration of 10-$- 10-4 M of tresperimus is to be maintained on the device surface. (C) Auranofin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~.g to 10 mg); preferred 10 ~,g to 1 mg.
The dose per unit area of 0.1 p.g - 100 p.g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 p,g/mm2. Minimum concentration of 10'$- 10-4 M of auranofin is to be maintained on the device surface. (D) 27-0-Demethylrapamycin and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 ~g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 p.g/mm2 - 10 ~g/mm~. Minimum concentration of 10-$- 10-4 M of 27-0-Demethylrapamycin is to be maintained on the device surface. (E) Gusperimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 p.g to 1 mg. The dose per unit area of 0.1 ~.g - 100 ~.g per mm2 of surface area; preferred dose of 0.3 ~g/mm2 -10 p,g/mm2. Minimum concentration of 10-$ - 10-4 M of gusperimus is to be maintained on the device surface. (F) Pimecrolimus and derivatives and analogues thereof: Total dose should not exceed 10 mg (range of 0.1 p,g to 10 mg); preferred 10 ~g to 1 mg. The dose per unit area of 0.1 pg - 100 p.g per mm2 of surface area; preferred dose of 0.3 p,g/mm2 -10 ~.g/mm2. Minimum concentration of 10-$- 10-4 M of pimecrolimus is to be maintained on the device surface and (G) ABT-578 and analogues and derivatives thereof: Total dose should not exceed 10 mg (range of 0.1 ~g to 10 mg); preferred 10 ~,g to 1 mg.
The dose per unit area of 0.1 p,g - 100 ~,g per mm2 of surface area; preferred dose of 0.3 ~.g/mm2 -10 ~g/mm2. Minimum concentration of 10-$- 10'4 M of ABT-578 is to be maintained on the device surface.
In addition to those described above (e.g., paclitaxel, TAXOTERE, and docetaxel), several other examples of anti-microtubule agents and appropriate dosages ranges for use with hemodialysis access devices include vinca alkaloids such as vinblastine and vincristine sulfate and analogues and derivatives thereof: total dose not to exceed 10 mg (range of 0.1 ~,g to 10 mg);
preferred 1 pg to 3 mg. Dose per unit area of the device of 0.1 pg - 10 pg per mm2; preferred dose of 0.25 pg/mm2 - 5 p,g/mma. Minimum concentration of 10-$ - 10-4 M of drug is to be maintained on the device surface.
Films and Meshes In one aspect, the present invention provides for the combination of an anti-scarring agent and a film or mesh. incorporation of a fibrosis-inhibiting agent into or onto a film or mesh can minimize fibrosis (or scarring) in the vicinity of the implant and may reduce or prevent the formation of adhesions between the implant and.the surrounding tissue. In certain aspects, the film or mesh may be used as a drug-delivery vehicle (e.g., as a perivascular delivery device for the prevention of neointimal hyperplasia at an anastomotic site).
Films or meshes may take a variety of forms including, but not limited to, surgical barriers, surgical adhesion barriers, membranes (e.g., barrier membranes), surgical sheets, surgical patches (e.g., dural patches), surgical wraps (e.g., vascular, perivascular, adventitial, periadventitital wraps, and adventitial sheets), meshes (e.g., perivascular meshes), bandages, liquid bandages, surgical dressings, gauze, fabrics, tapes, surgical membranes, polymer matrices, shells, envelopes, tissue coverings, and other types of surgical matrices, scaffolds, and coatings.
In one aspect, the device comprises or may be in the form of a film. The film may be formed into one of many geometric shapes. Depending on the application, the film may be formed into the shape of a tube or may be a thin, elastic sheet of polymer. Generally, films are less than 5, 4, 3, 2, or 1 mm thick, more preferably less than 0.75 mm, 0.5 mm, 0.25 mm, or, 0.10 mm thick.
Films can also be generated of thicknesses less than 50 Vim, 25 ~.m or 10 Vim.
Films generally are flexible with a good tensile strength (e.g., greater than 50, preferably greater than 100, and more preferably greater than 150 or 200 Nicm2), good adhesive properties (i.e., adheres to moist or wet surfaces), and have controlled permeability. Polymeric films (which may be porous or non-porous) are particularly useful for application to the surface of a device or implant as well as to the surface of tissue, cavity or an organ.
Films may be made by several processes, including for example, by casting, and by spraying, or may be formed at the treatment site in situ.
For example, a sprayable formulation may be applied onto the treatment site which then forms into a solid film.
In another aspect, the device may comprise or be in the form of a polymer, wherein at least some of the polymer is in the form of a mesh. A
mesh, as used herein, is a material composed of a plurality of fibers or filaments (i.e., a fibrous material), where the fibers or filaments are arranged in such a manner (e.g., interwoven, knotted, braided, overlapping, looped, knitted, interlaced, intertwined, webbed, felted, and the like) so as to form a porous structure. Typically, a mesh is a pliable material, such that it has sufficient flexibility to be wrapped around the external surface of a body passageway or cavity, or a portion thereof. The mesh may be capable of providing support to the structure (e.g., the vessel or cavity wall) and may be adapted to release an amount of the therapeutic agent.
Mesh materials may take a variety of forms. For example, the mesh may be in a woven, knit, or non-woven form and may include fibers or filaments that are randomly oriented relative to each other or that are arranged in an ordered array or pattern. In one embodiment, for example, a mesh may be in the form of a fabric, such as, for example, a knitted, braided, crocheted, woven, non-woven (e.g., a melt-blown or wet-laid) or webbed fabric. In one embodiment, a mesh may include a natural or synthetic biodegradable polymer that may be formed into a knit mesh, a weave mesh, a sprayed mesh, a web mesh, a braided mesh, a looped mesh, and the like. Preferably, a mesh or wrap has intertwined threads that form a porous structure, which may be, for example, knitted, woven, or webbed.
The structure and properties of the mesh used in a device depend on the application and the desired mechanical (i.e., flexibility, tensile strength, and elasticity), degradation properties, and the desired loading and release characteristics for the selected therapeutic.agent(s). The mesh should have mechanical properties, such that the device will remain sufficiently strong until the surrounding tissue has healed. Factors that affect the flexibility and mechanical strength of the mesh include, for example, the porosity, fabric thickness, fiber diameter, polymer composition (e.g., type of monomers and initiators), process conditions, and the additives that are used to prepare the material.
Typically, the mesh possesses sufficient porosity to permit the flow of fluids through the pores of the fiber network and to facilitate tissue ingrowth. Generally, the interstices of the mesh should be sufficiently wide apart to allow light visible by eye, or fluids, to pass through the pores.
However, materials having a more compact structure also may be used. The flow of fluid through the interstices of the mesh depends on a variety of factors, including, for example, the stitch count or thread density. The porosity of the mesh may be further tailored by, for example, filling the interstices of the mesh with another material (e.g., particles or polymer) or by processing the mesh (e.g., by heating) in order to reduce the pore size and to create non-fibrous areas. Fluid flow through the mesh of the invention will vary depending on the properties of the fluid, such as viscosity, hydrophilicity/hydrophobicity, ionic concentration, temperature, elasticity, pseudoplasticity, particulate content, and the like. Preferably, the interstices do not prevent the release of impregnated or coated therapeutic agents) from the mesh, and the interstices preferably do not prevent the exchange of tissue fluid at the application site.
Mesh materials should be sufficiently flexible so as to be capable of being wrapped around all or a portion of the external surface of a body passageway or cavity. Flexible mesh materials are typically in the form of flexible woven or knitted sheets having a thickness ranging from about 25 microns to about 3000 microns; preferably from about 50 to about 1000 microns. Mesh material suitable for wrapping around arteries and veins typically ranges from about 100 to 400 microns in thickness.
The diameter and length of the fibers or filaments may range in size depending on the form of the material (e.g., knit, woven, or non-woven), and the desired elasticity, porosity, surface area, flexibility, and tensile strength.
The fibers may be of any length, ranging from short filaments to long threads (i.e., several microns to hundreds of meters in length). Depending on the application, the fibers may have a monofilament or a multifilament construction.
The mesh may include fibers that are of same dimension or of different dimensions, and the fibers may be formed from the same or different types of biodegradable polymers. Woven materials, for example, may include a regular or irregular array of warp and weft strands and may include one type of polymer in the weft direction and another type (having the same or a different degradation profile from the first polymer) in the warp direction. The degradation profile of the weft polymer may be different than or the same as the degradation profile of the warp polymer. Similarly, knit materials may include one or more types (e.g., monofilament, multi-filament) and sizes of fibers and may include fibers made from the same or from different types of biodegradable polymers.
The structure of the mesh (e.g., fiber density and porosity) may impact the amount of therapeutic agent that may be loaded into or onto the device. For example, a fabric having a loose weave characterized by a low DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter 1e Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS
THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

Claims (5416)

1. A device, comprising an intravascular implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2. The device of claim 1 wherein the agent inhibits cell regeneration.
3. The device of claim 1 wherein the agent inhibits angiogenesis.
4. The device of claim 1 wherein the agent inhibits fibroblast migration.
5. The device of claim 1 wherein the agent inhibits fibroblast proliferation.
6. The device of claim 1 wherein the agent inhibits deposition of extracellular matrix.
7. The device of claim 1 wherein the agent inhibits tissue remodeling.
8. The device of claim 1 wherein the agent is an angiogenesis inhibitor.
9. The device of claim 1 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
10. The device of claim 1 wherein the agent is a chemokine receptor antagonist.
11. The device of claim 1 wherein the agent is a cell cycle inhibitor.
12. The device of claim 1 wherein the agent is a taxane.
13. The device of claim 1 wherein the agent is an anti-microtubule agent.
14. The device of claim 1 wherein the agent is paclitaxel.
15. The device of claim 1 wherein the agent is not paclitaxel.
16. The device of claim 1 wherein the agent is an analogue or derivative of paclitaxel.
17. The device of claim 1 wherein the agent is a vinca alkaloid.
18. The device of claim 1 wherein the agent is camptothecin or an analogue or derivative thereof.
19. The device of claim 1 wherein the agent is a podophyllotoxin.
20. The device of claim 1 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
21. The device of claim 1 wherein the agent is an anthracycline.
22. The device of claim 1 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
23. The device of claim 1 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
24. The device of claim 1 wherein the agent is a platinum compound.
25. The device of claim 1 wherein the agent is a nitrosourea.
26. The device of claim 1 wherein the agent is a nitroimidazole.
27. The device of claim 1 wherein the agent is a folic acid antagonist.
28. The device of claim 1 wherein the agent is a cytidine analogue.
29. The device of claim 1 wherein the agent is a pyrimidine analogue.
30. The device of claim 1 wherein the agent is a fluoropyrimidine analogue.
31. The device of claim 1 wherein the agent is a purine analogue.
32. The device of claim 1 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
33. The device of claim 1 wherein the agent is a hydroxyurea.
34. The device of claim 1 wherein the agent is a mytomicin or an analogue or derivative thereof.
35. The device of claim 1 wherein the agent is an alkyl sulfonate.
36. The device of claim 1 wherein the agent is a benzamide or an analogue or derivative thereof.
37. The device of claim 1 wherein the agent is a nicotinamide or an analogue or derivative thereof.
38. The device of claim 1 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
39. The device of claim 1 wherein the agent is a DNA
alkylating agent.
40. The device of claim 1 wherein the agent is an anti-microtubule agent.
41. The device of claim 1 wherein the agent is a topoisomerase inhibitor.
42. The device of claim 1 wherein the agent is a DNA cleaving agent.
43. The device of claim 1 wherein the agent is an antimetabolite.
44. The device of claim 1 wherein the agent inhibits adenosine deaminase.
45. The device of claim 1 wherein the agent inhibits purine ring synthesis.
46. The device of claim 1 wherein the agent is a nucleotide interconversion inhibitor.
47. The device of claim 1 wherein the agent inhibits dihydrofolate reduction.
48. The device of claim 1 wherein the agent blocks thymidine monophosphate.
49. The device of claim 1 wherein the agent causes DNA
damage.
50. The device of claim 1 wherein the agent is a DNA
intercalation agent.
51. The device of claim 1 wherein the agent is a RNA
synthesis inhibitor.
52. The device of claim 1 wherein the agent is a pyrimidine synthesis inhibitor.
53. The device of claim 1 wherein the agent inhibits ribonucleotide synthesis or function.
54. The device of claim 1 wherein the agent inhibits thymidine monophosphate synthesis or function.
55. The device of claim 1 wherein the agent inhibits DNA
synthesis.
56. The device of claim 1 wherein the agent causes DNA
adduct formation.
57. The device of claim 1 wherein the agent inhibits protein synthesis.
58. The device of claim 1 wherein the agent inhibits microtubule function.
59. The device of claim 1 wherein the agent is a cyclin dependent protein kinase inhibitor.
60. The device of claim 1 wherein the agent is an epidermal growth factor kinase inhibitor.
61. The device of claim 1 wherein the agent is an elastase inhibitor.
62. The device of claim 1 wherein the agent is a factor Xa inhibitor.
63. The device of claim 1 wherein the agent is a farnesyltransferase inhibitor.
64. The device of claim 1 wherein the agent is a fibrinogen antagonist.
65. The device of claim 1 wherein the agent is a guanylate cyclase stimulant.
66. The device of claim 1 wherein the agent is a heat shock protein 90 antagonist.
67. The device of claim 1 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
68. The device of claim 1 wherein the agent is a guanylate cyclase stimulant.
69. The device of claim 1 wherein the agent is a HMGCoA
reductase inhibitor.
70. The device of claim 1 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
71. The device of claim 1 wherein the agent is a hydroorotate dehydrogenase inhibitor.
72. The device of claim 1 wherein the agent is an IKK2 inhibitor.
73. The device of claim 1 wherein the agent is an IL-1 antagonist.
74. The device of claim 1 wherein the agent is an ICE
antagonist.
75. The device of claim 1 wherein the agent is an IRAK
antagonist.
76. The device of claim 1 wherein the agent is an IL-4 agonist.
77. The device of claim 1 wherein the agent is an immunomodulatory agent.
78. The device of claim 1 wherein the agent is sirolimus or an analogue or derivative thereof.
79. The device of claim 1 wherein the agent is not sirolimus.
80. The device of claim 1 wherein the agent is everolimus or an analogue or derivative thereof.
81. The device of claim 1 wherein the agent is tacrolimus or an analogue or derivative thereof.
82. The device of claim 1 wherein the agent is not tacrolimus.
83. The device of claim 1 wherein the agent is biolmus or an analogue or derivative thereof.
84. The device of claim 1 wherein the agent is tresperimus or an analogue or derivative thereof.
85. The device of claim 1 wherein the agent is auranofin or an analogue or derivative thereof.
86. The device of claim 1 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
87. The device of claim 1 wherein the agent is gusperimus or an analogue or derivative thereof.
88. The device of claim 1 wherein the agent is pimecrolimus or an analogue or derivative thereof.
89. The device of claim 1 wherein the agent is ABT-578 or an analogue or derivative thereof.
90. The device of claim 1 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
91. The device of claim 1 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
92. The device of claim 1 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
93. The device of claim 1 wherein the agent is a leukotriene inhibitor.
94. The device of claim 1 wherein the agent is a MCP-1 antagonist.
95. The device of claim 1 wherein the agent is a MMP inhibitor.
96. The device of claim 1 wherein the agent is an NF kappa B
inhibitor.
97. The device of claim 1 wherein the agent is an NF kappa B
inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
98. The device of claim 1 wherein the agent is an NO agonist.
99. The device of claim 1 wherein the agent is a p38 MAP
kinase inhibitor.
100. The device of claim 1 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
101. The device of claim 1 wherein the agent is a phosphodiesterase inhibitor.
102. The device of claim 1 wherein the agent is a TGF beta inhibitor.
103. The device of claim 1 wherein the agent is a thromboxane A2 antagonist.
104. The device of claim 1 wherein the agent is a TNFa antagonist.
105. The device of claim 1 wherein the agent is a TACE
inhibitor.
106. The device of claim 1 wherein the agent is a tyrosine kinase inhibitor.
107. The device of claim 1 wherein the agent is a vitronectin inhibitor.
108. The device of claim 1 wherein the agent is a fibroblast growth factor inhibitor.
109. The device of claim 1 wherein the agent is a protein kinase inhibitor.
110. The device of claim 1 wherein the agent is a PDGF
receptor kinase inhibitor.
111. The device of claim 1 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
112. The device of claim 1 wherein the agent is a retinoic acid receptor antagonist.
113. The device of claim 1 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
114. The device of claim 1 wherein the agent is a fibronogin antagonist.
115. The device of claim 1 wherein the agent is an antimycotic agent.
116. The device of claim 1 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
117. The device of claim 1 wherein the agent is a bisphosphonate.
118. The device of claim 1 wherein the agent is a phospholipase A1 inhibitor.
119. The device of claim 1 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
120. The device of claim 1 wherein the agent is a macrolide antibiotic.
121. The device of claim 1 wherein the agent is a GPIIb/IIIa receptor antagonist.
122. The device of claim 1 wherein the agent is an endothelin receptor antagonist.
123. The device of claim 1 wherein the agent is a peroxisome proliferator-activated receptor agonist.
124. The device of claim 1 wherein the agent is an estrogen receptor agent.
125. The device of claim 1 wherein the agent is a somastostatin analogue.
126. The device of claim 1 wherein the agent is a neurokinin 1 antagonist.
127. The device of claim 1 wherein the agent is a neurokinin 3 antagonist.
128. The device of claim 1 wherein the agent is a VLA-4 antagonist.
129. The device of claim 1 wherein the agent is an osteoclast inhibitor.
130. The device of claim 1 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
131. The device of claim 1 wherein the agent is an angiotensin I
converting enzyme inhibitor.
132. The device of claim 1 wherein the agent is an angiotensin II
antagonist.
133. The device of claim 1 wherein the agent is an enkephalinase inhibitor.
134. The device of claim 1 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
135. The device of claim 1 wherein the agent is a protein kinase C inhibitor.
136. The device of claim 1 wherein the agent is a ROCK (rho-associated kinase) inhibitor.
137. The device of claim 1 wherein the agent is a CXCR3 inhibitor.
138. The device of claim 1 wherein the agent is an Itk inhibitor.
139. The device of claim 1 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
140. The device of claim 1 wherein the agent is a PPAR agonist.
141. The device of claim 1 wherein the agent is an immunosuppressant.
142. The device of claim 1 wherein the agent is an Erb inhibitor.
143. The device of claim 1 wherein the agent is an apoptosis agonist.
144. The device of claim 1 wherein the agent is a lipocortin agonist.
145. The device of claim 1 wherein the agent is a VCAM-1 antagonist.
146. The device of claim 1 wherein the agent is a collagen antagonist.
147. The device of claim 1 wherein the agent is an alpha 2 integrin antagonist.
148. The device of claim 1 wherein the agent is a TNF alpha inhibitor.
149. The device of claim 1 wherein the agent is a nitric oxide inhibitor
150. The device of claim 1 wherein the agent is a cathepsin inhibitor.
151. The device of claim 1 wherein the agent is not an anti-inflammatory agent.
152. The device of claim 1 wherein the agent is not a steroid.
153. The device of claim 1 wherein the agent is not a glucocorticosteroid.
154. The device of claim 1 wherein the agent is not dexamethasone.
155. The device of claim 1 wherein the agent is not an anti-infective agent.
156. The device of claim 1 wherein the agent is not an antibiotic.
157. The device of claim 1 wherein the agent is not an anti-fungal agent.
158. The device of claim 1, further comprising a polymer.
159. The device of claim 1, further comprising a polymeric carrier.
160. The device of claim 1 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
161. The device of claim 1 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
162. The device of claim 1, further comprising a coating, wherein the coating comprises the anti-scarring agent.
163. The device of claim 1, further comprising a coating, wherein the coating is disposed on a surface of the device.
164. The device of claim 1, further comprising a coating, wherein the coating directly contacts the device.
165. The device of claim 1, further comprising a coating, wherein the coating indirectly contacts the device.
166. The device of claim 1, further comprising a coating, wherein the coating partially covers the device.
167. The device of claim 1, further comprising a coating, wherein the coating completely covers the device.
168. The device of claim 1, further comprising a coating, wherein the coating is a uniform coating.
169. The device of claim 1, further comprising a coating, wherein the coating is a non-uniform coating.
170. The device of claim 1, further comprising a coating, wherein the coating is a discontinuous coating.
171. The device of claim 1, further comprising a coating, wherein the coating is a patterned coating.
172. The device of claim 1, further comprising a coating, wherein the coating has a thickness of 100 µm or less
173. The device of claim 1, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
174. The device of claim 1, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
175. The device of claim 1, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
176. The device of claim 1, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
177. The device of claim 1, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
178. The device of claim 1, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
179. The device of claim 1, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
180. The device of claim 1, further comprising a coating, wherein the coating further comprises a polymer.
181. The device of claim 1, further comprising a first coating having a first composition and the second coating having a second composition.
182. The device of claim 1, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
183. The device of claim 1, further comprising a polymer.
184. The device of claim 1, further comprising a polymeric carrier.
185. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
186. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
187. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
188. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
189. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
190. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
191. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
192. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
193. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
194. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
195. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
196. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
197. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
198. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
199. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
200. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
201. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
202. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
203. The device of claim 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
204. The device of claim 1, further comprising a lubricious coating.
205. The device of claim 1 wherein the anti-scarring agent is located within pores or holes of the device.
206. The device of claim 1 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
207. The device of claim 1, further comprising a second pharmaceutically active agent.
208. The device of claim 1, further comprising an anti-inflammatory agent.
209. The device of claim 1, further comprising an agent that inhibits infection.
210. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
211. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
212. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
213. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
214. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
215. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
216. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
217. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
218. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is etoposide.
219. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
220. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.~
221. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
222. The device of claim 1, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
223. The device of claim 1, further comprising an anti-thrombotic agent.
224. The device of claim 1, further comprising a visualization agent.
225. The device of claim 1, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
226. The device of claim 1, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
227. The device of claim 1, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
228. The device of claim 1, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
229. The device of claim 1, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
230. The device of claim 1, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
231. The device of claim 1, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
232. The device of claim 1, further comprising an echogenic material.
233. The device of claim 1, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
234. The device of claim 1 wherein the device is sterile.
235. The device of claim 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
236. The device of claim 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
237. The device of claim 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
238. The device of claim 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
239. The device of claim 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
240. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
241. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
242. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
243. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
244. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
245. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
246. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
247. The device of claim 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
248. The device of claim 1 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
249. The device of claim 1 wherein the device comprises about µg to about 10 mg of the anti-scarring agent.
250. The device of claim 1 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
251. The device of claim 1 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
252. The device of claim 1 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
253. The device of claim 1 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
254. The device of claim 1 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
255. The device of claim 1 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
256. The device of claim 1 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
257. The device of claim 1 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
258. The device of claim 1 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
259. The device of any one of claims 1-258 wherein the implant is a stent.
260. The device of any one of claims 1-258 wherein the implant is a coronary stent.
261. The device of any one of claims 1-258 wherein the implant is a peripheral stent.
262. The device of any one of claims 1-258 wherein the implant is a covered stent.
263. The device of any one of claims 1-258 wherein the implant is an intravascular catheter.
264. The device of any one of claims 1-258 wherein the implant is a microinjector catheter.
265. The device of any one of claims 1-258 wherein the implant is a drug delivery balloon.
266. The device of any one of claims 1-258 wherein the implant is a sweaty balloon.
267. The device of any one of claims 1-258 wherein the implant is a channel balloon.
268. The device of any one of claims 1-258 wherein the implant is a microinjector balloon.
269. The device of any one of claims 1-258 wherein the implant is a double balloon.
270. The device of any one of claims 1-258 wherein the implant is a spiral balloon.
271. The device of any one of claims 1-258 wherein the implant is a BHP balloon.
272. The device of any one of claims 1-258 wherein the implant is a transurethral needle ablation (TUNA) balloon.
273. The device of any one of claims 1-258 wherein the implant is a radio frequency needle ablation (RFNA) balloon.
274. The device of any one of claims 1-258 wherein the implant is a coronary drug infection guidewire.
275. A device, comprising a vascular graft or wrap implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
276. The device of claim 275 wherein the agent inhibits cell regeneration.
277. The device of claim 275 wherein the agent inhibits angiogenesis.
278. The device of claim 275 wherein the agent inhibits fibroblast migration.
279. The device of claim 275 wherein the agent inhibits fibroblast proliferation.
280. The device of claim 275 wherein the agent inhibits deposition of extracellular matrix.
281. The device of claim 275 wherein the agent inhibits tissue remodeling.
282. The device of claim 275 wherein the agent is an angiogenesis inhibitor.
283. The device of claim 275 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
284. The device of claim 275 wherein the agent is a chemokine receptor antagonist.
285. The device of claim 275 wherein the agent is a cell cycle inhibitor.
286. The device of claim 275 wherein the agent is a taxane.
287. The device of claim 275 wherein the agent is an anti-microtubule agent.
288. The device of claim 275 wherein the agent is paclitaxel.
289. The device of claim 275 wherein the agent is not paclitaxel.
290. The device of claim 275 wherein the agent is an analogue or derivative of paclitaxel.
291. The device of claim 275 wherein the agent is a vinca alkaloid.
292. The device of claim 275 wherein the agent is camptothecin or an analogue or derivative thereof.
293. The device of claim 275 wherein the agent is a podophyllotoxin.
294. The device of claim 275 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
295. The device of claim 275 wherein the agent is an anthracycline.
296. The device of claim 275 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
297. The device of claim 275 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
298. The device of claim 275 wherein the agent is a platinum compound.
299. The device of claim 275 wherein the agent is a nitrosourea.
300. The device of claim 275 wherein the agent is a nitroimidazole.
301. The device of claim 275 wherein the agent is a folic acid antagonist.
302. The device of claim 275 wherein the agent is a cytidine analogue.
303. The device of claim 275 wherein the agent is a pyrimidine analogue.
304. The device of claim 275 wherein the agent is a fluoropyrimidine analogue.
305. The device of claim 275 wherein the agent is a purine analogue.
306. The device of claim 275 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
307. The device of claim 275 wherein the agent is a hydroxyurea.
308. The device of claim 275 wherein the agent is a mytomicin or an analogue or derivative thereof.
309. The device of claim 275 wherein the agent is an alkyl sulfonate.
310. The device of claim 275 wherein the agent is a benzamide or an analogue or derivative thereof.
311. The device of claim 275 wherein the agent is a nicotinamide or an analogue or derivative thereof.
312. The device of claim 275 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
313. The device of claim 275 wherein the agent is a DNA
alkylating agent.
314. The device of claim 275 wherein the agent is an anti-microtubule agent.
315. The device of claim 275 wherein the agent is a topoisomerase inhibitor.
316. The device of claim 275 wherein the agent is a DNA
cleaving agent.
317. The device of claim 275 wherein the agent is an antimetabolite.
318. The device of claim 275 wherein the agent inhibits adenosine deaminase.
319. The device of claim 275 wherein the agent inhibits purine ring synthesis.
320. The device of claim 275 wherein the agent is a nucleotide interconversion inhibitor.
321. The device of claim 275 wherein the agent inhibits dihydrofolate reduction.
322. The device of claim 275 wherein the agent blocks thymidine monophosphate.
323. The device of claim 275 wherein the agent causes DNA
damage.
324. The device of claim 275 wherein the agent is a DNA
intercalation agent.
325. The device of claim 275 wherein the agent is a RNA
synthesis inhibitor.
326. The device of claim 275 wherein the agent is a pyrimidine synthesis inhibitor.
327. The device of claim 275 wherein the agent inhibits ribonucleotide synthesis or function.
328. The device of claim 275 wherein the agent inhibits thymidine monophosphate synthesis or function.
329. The device of claim 275 wherein the agent inhibits DNA
synthesis.
330. The device of claim 275 wherein the agent causes DNA
adduct formation.
331. The device of claim 275 wherein the agent inhibits protein synthesis.
332. The device of claim 275 wherein the agent inhibits microtubule function.
333. The device of claim 275 wherein the agent is a cyclin dependent protein kinase inhibitor.
334. The device of claim 275 wherein the agent is an epidermal growth factor kinase inhibitor.
335. The device of claim 275 wherein the agent is an elastase inhibitor.
336. The device of claim 275 wherein the agent is a factor Xa inhibitor.
337. The device of claim 275 wherein the agent is a farnesyltransferase inhibitor.
1774 333. The device of claim 275 wherein the agent is a fibrinogen antagonist.
339. The device of claim 275 wherein the agent is a guanylate cyclase stimulant.
340. The device of claim 275 wherein the agent is a heat shock protein 90 antagonist.
341. The device of claim 275 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
342. The device of claim 275 wherein the agent is a guanylate cyclase stimulant.
343. The device of claim 275 wherein the agent is a HMGCoA
reductase inhibitor.
344. The device of claim 275 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
345. The device of claim 275 wherein the agent is a hydroorotate dehydrogenase inhibitor.
346. The device of claim 275 wherein the agent is an IKK2 inhibitor.
347. The device of claim 275 wherein the agent is an IL-1 antagonist.
343. The device of claim 275 wherein the agent is an ICE
antagonist.
349. The device of claim 275 wherein the agent is an IRAK
antagonist.
350. The device of claim 275 wherein the agent is an IL-4 agonist.
351. The device of claim 275 wherein the agent is an immunomodulatory agent.
352. The device of claim 275 wherein the agent is sirolimus or an analogue or derivative thereof.
353. The device of claim 275 wherein the agent is not sirolimus.
354. The device of claim 275 wherein the agent is everolimus or an analogue or derivative thereof.
355. The device of claim 275 wherein the agent is tacrolimus or an analogue or derivative thereof.
356. The device of claim 275 wherein the agent is not tacrolimus.
357. The device of claim 275 wherein the agent is biolmus or an analogue or derivative thereof.
358. The device of claim 275 wherein the agent is tresperimus or an analogue or derivative thereof.
359. The device of claim 275 wherein the agent is auranofin or an analogue or derivative thereof.
360. The device of claim 275 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
361. The device of claim 275 wherein the agent is gusperimus or an analogue or derivative thereof.
362. The device of claim 275 wherein the agent is pimecrolimus or an analogue or derivative thereof.
363. The device of claim 275 wherein the agent is ABT-578 or an analogue or derivative thereof.
364. The device of claim 275 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
365. The device of claim 275 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
366. The device of claim 275 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
367. The device of claim 275 wherein the agent is a leukotriene inhibitor.
368. The device of claim 275 wherein the agent is a MCP-1 antagonist.
369. The device of claim 275 wherein the agent is a MMP
inhibitor.
370. The device of claim 275 wherein the agent is an NF kappa B inhibitor.
371. The device of claim 275 wherein the agent is an NF kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
372. The device of claim 275 wherein the agent is an NO
agonist.
373. The device of claim 275 wherein the agent is a p38 MAP
kinase inhibitor.
374. The device of claim 275 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
375. The device of claim 275 wherein the agent is a phosphodiesterase inhibitor.
376. The device of claim 275 wherein the agent is a TGF beta inhibitor.
377. The device of claim 275 wherein the agent is a thromboxane A2 antagonist.
378. The device of claim 275 wherein the agent is a TNFa antagonist.
379. The device of claim 275 wherein the agent is a TACE
inhibitor.
380. The device of claim 275 wherein the agent is a tyrosine kinase inhibitor.
381. The device of claim 275 wherein the agent is a vitronectin inhibitor.
382. The device of claim 275 wherein the agent is a fibroblast growth factor inhibitor.
383. The device of claim 275 wherein the agent is a protein kinase inhibitor.
384. The device of claim 275 wherein the agent is a PDGF
receptor kinase inhibitor.
385. The device of claim 275 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
386. The device of claim 275 wherein the agent is a retinoic acid receptor antagonist.
387. The device of claim 275 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
388. The device of claim 275 wherein the agent is a fibronogin antagonist.
389. The device of claim 275 wherein the agent is an antimycotic agent.
390. The device of claim 275 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
391. The device of claim 275 wherein the agent is a bisphosphonate.
392. The device of claim 275 wherein the agent is a phospholipase A1 inhibitor.
393. The device of claim 275 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
394. The device of claim 275 wherein the agent is a macrolide antibiotic.
395. The device of claim 275 wherein the agent is a GPIIb/IIIa receptor antagonist.
396. The device of claim 275 wherein the agent is an endothelin receptor antagonist.
397. The device of claim 275 wherein the agent is a peroxisome proliferator-activated receptor agonist.
398. The device of claim 275 wherein the agent is an estrogen receptor agent.
399. The device of claim 275 wherein the agent is a somastostatin analogue.
400. The device of claim 275 wherein the agent is a neurokinin 1 antagonist.
401. The device of claim 275 wherein the agent is a neurokinin 3 antagonist.
402. The device of claim 275 wherein the agent is a VLA-4 antagonist.
403. The device of claim 275 wherein the agent is an osteoclast inhibitor.
404. The device of claim 275 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
405. The device of claim 275 wherein the agent is an angiotensin I converting enzyme inhibitor.
406. The device of claim 275 wherein the agent is an angiotensin II antagonist.
407. The device of claim 275 wherein the agent is an enkephalinase inhibitor.
408. The device of claim 275 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
409. The device of claim 275 wherein the agent is a protein kinase C inhibitor.
410. The device of claim 275 wherein the agent is a ROCK (rho-associated kinase) inhibitor.
411. The device of claim 275 wherein the agent is a CXCR3 inhibitor.
412. The device of claim 275 wherein the agent is an Itk inhibitor.
413. The device of claim 275 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
414. The device of claim 275 wherein the agent is a PPAR
agonist.
415. The device of claim 275 wherein the agent is an immunosuppressant.
416. The device of claim 275 wherein the agent is an Erb inhibitor.
417. The device of claim 275 wherein the agent is an apoptosis agonist.
418. The device of claim 275 wherein the agent is a lipocortin agonist.
419. The device of claim 275 wherein the agent is a VCAM-1 antagonist.
420. The device of claim 275 wherein the agent is a collagen antagonist.
421. The device of claim 275 wherein the agent is an alpha 2 integrin antagonist.
422. The device of claim 275 wherein the agent is a TNF alpha inhibitor.
423. The device of claim 275 wherein the agent is a nitric oxide inhibitor
424. The device of claim 275 wherein the agent is a cathepsin inhibitor.
425. The device of claim 275 wherein the agent is not an anti-inflammatory agent.
426. The device of claim 275 wherein the agent is not a steroid.
427. The device of claim 275 wherein the agent is not a glucocorticosteroid.
428. The device of claim 275 wherein the agent is not dexamethasone.
429. The device of claim 275 wherein the agent is not an anti-infective agent.
430. The device of claim 275 wherein the agent is not an antibiotic.
431. The device of claim 275 wherein the agent is not an anti-fungal agent.
432. The device of claim 275, further comprising a polymer.
433. The device of claim 275, further comprising a polymeric carrier.
434. The device of claim 275 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
435. The device of claim 275 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
436. The device of claim 275, further comprising a coating, wherein the coating comprises the anti-scarring agent.
437. The device of claim 275, further comprising a coating, wherein the coating is disposed on a surface of the device.
438. The device of claim 275, further comprising a coating, wherein the coating directly contacts the device.
439. The device of claim 275, further comprising a coating, wherein the coating indirectly contacts the device.
440. The device of claim 275, further comprising a coating, wherein the coating partially covers the device.
441. The device of claim 275, further comprising a coating, wherein the coating completely covers the device.
442. The device of claim 275, further comprising a coating, wherein the coating is a uniform coating.
443. The device of claim 275, further comprising a coating, wherein the coating is a non-uniform coating.
444. The device of claim 275, further comprising a coating, wherein the coating is a discontinuous coating.
445. The device of claim 275, further comprising a coating, wherein the coating is a patterned coating.
446. The device of claim 275, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
447. The device of claim 275, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
448. The device of claim 275, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
449. The device of claim 275, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
450. The device of claim 275, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
451. The device of claim 275, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
452. The device of claim 275, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
453. The device of claim 275, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
454. The device of claim 275, further comprising a coating, wherein the coating further comprises a polymer.
455. The device of claim 275, further comprising a first coating having a first composition and the second coating having a second composition.
456. The device of claim 275, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
457. The device of claim 275, further comprising a polymer.
458. The device of claim 275, further comprising a polymeric carrier.
459. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
460. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
461. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
462. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
463. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
464. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
465. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
466. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
467. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
468. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
469. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
470. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
471. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
472. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
473. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
474. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
475. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
476. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
477. The device of claim 275, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
478. The device of claim 275, further comprising a lubricious coating.
479. The device of claim 275 wherein the anti-scarring agent is located within pores or holes of the device.
480. The device of claim 275 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
481. The device of claim 275, further comprising a second pharmaceutically active agent.
482. The device of claim 275, further comprising an anti-inflammatory agent.
483. The device of claim 275, further comprising an agent that inhibits infection.
484. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
485. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
486. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
487. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
488. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
489. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
490. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
491. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
492. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is etoposide.
493. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
494. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
495. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
496. The device of claim 275, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
497. The device of claim 275, further comprising an anti-thrombotic agent.
498. The device of claim 275, further comprising a visualization agent.
499. The device of claim 275, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
500. The device of claim 275, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
501. The device of claim 275, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
502. The device of claim 275, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
503. The device of claim 275, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
504. The device of claim 275, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
505. The device of claim 275, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
506. The device of claim 275, further comprising an echogenic material.
507. The device of claim 275, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
508. The device of claim 275 wherein the device is sterile.
509. The device of claim 275 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
510. The device of claim 275 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
511. The device of claim 275 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
512. The device of claim 275 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
513. The device of claim 275 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
514. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
515. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
516. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
517. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
518. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
519. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
520. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
521. The device of claim 275 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
522. The device of claim 275 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
523. The device of claim 275 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
524. The device of claim 275 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
525. The device of claim 275 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
526. The device of claim 275 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
527. The device of claim 275 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
528. The device of claim 275 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
529. The device of claim 275 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
530. The device of claim 275 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
531. The device of claim 275 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
532. The device of claim 275 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
533. The device of any one of claims 275-532 wherein the implant is a synthetic bypass graft.
534. The device of any one of claims 275-532 wherein the implant is a femoral-popliteal synthetic bypass graft.
535. The device of any one of claims 275-532 wherein the implant is a femoral-femoral synthetic bypass graft.
536. The device of any one of claims 275-532 wherein the implant is an axillary-femoral synthetic bypass graft.
537. The device of any one of claims 275-532 wherein the implant is a vein graft.
538. The device of any one of claims 275-532 wherein the implant is a peripheral vein graft.
539. The device of any one of claims 275-532 wherein the implant is a coronary vein graft.
540. The device of any one of claims 275-532 wherein the implant is an internal mammary graft.
541. The device of any one of claims 275-532 wherein the implant is an internal mammary coronary graft.
542. The device of any one of claims 275-532 wherein the implant is a bifurcated vascular graft.
543. The device of any one of claims 275-532 wherein the implant is a vascular wrap.
544. A device, comprising an implant for hemodialysis access (i.e., a hemodialysis access device) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
545. The device of claim 544 wherein the agent inhibits cell regeneration.
546. The device of claim 544 wherein the agent inhibits angiogenesis.
547. The device of claim 544 wherein the agent inhibits fibroblast migration.
548. The device of claim 544 wherein the agent inhibits fibroblast proliferation.
549. The device of claim 544 wherein the agent inhibits deposition of extracellular matrix.
550. The device of claim 544 wherein the agent inhibits tissue remodeling.
551. The device of claim 544 wherein the agent is an angiogenesis inhibitor.
552. The device of claim 544 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
553. The device of claim 544 wherein the agent is a chemokine receptor antagonist.
554. The device of claim 544 wherein the agent is a cell cycle inhibitor.
555. The device of claim 544 wherein the agent is a taxane.
556. The device of claim 544 wherein the agent is an anti-microtubule agent.
557. The device of claim 544 wherein the agent is paclitaxel.
559. The device of claim 544 wherein the agent is not paclitaxel.
559. The device of claim 544 wherein the agent is an analogue or derivative of paclitaxel.
560. The device of claim 544 wherein the agent is a vinca alkaloid.
561. The device of claim 544 wherein the agent is camptothecin or an analogue or derivative thereof.
562. The device of claim 544 wherein the agent is a podophyllotoxin.
563. The device of claim 544 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
564. The device of claim 544 wherein the agent is an anthracycline.
565. The device of claim 544 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
566. The device of claim 544 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
567. The device of claim 544 wherein the agent is a platinum compound.
568. The device of claim 544 wherein the agent is a nitrosourea.
569. The device of claim 544 wherein the agent is a nitroimidazole.
570. The device of claim 544 wherein the agent is a folic acid antagonist.
571. The device of claim 544 wherein the agent is a cytidine analogue.
572. The device of claim 544 wherein the agent is a pyrimidine analogue.
573. The device of claim 544 wherein the agent is a fluoropyrimidine analogue.
574. The device of claim 544 wherein the agent is a purine analogue.
575. The device of claim 544 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
576. The device of claim 544 wherein the agent is a hydroxyurea.
577. The device of claim 544 wherein the agent is a mytomicin or an analogue or derivative thereof.
578. The device of claim 544 wherein the agent is an alkyl sulfonate.
579. The device of claim 544 wherein the agent is a benzamide or an analogue or derivative thereof.
580. The device of claim 544 wherein the agent is a nicotinamide or an analogue or derivative thereof.
581. The device of claim 544 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
582. The device of claim 544 wherein the agent is a DNA
alkylating agent.
583. The device of claim 544 wherein the agent is an anti-microtubule agent.
584. The device of claim 544 wherein the agent is a topoisomerase inhibitor.
585. The device of claim 544 wherein the agent is a DNA
cleaving agent.
586. The device of claim 544 wherein the agent is an antimetabolite.
587. The device of claim 544 wherein the agent inhibits adenosine deaminase.
588. The device of claim 544 wherein the agent inhibits purine ring synthesis.
589. The device of claim 544 wherein the agent is a nucleotide interconversion inhibitor.
590. The device of claim 544 wherein the agent inhibits dihydrofolate reduction.
591. The device of claim 544 wherein the agent blocks thymidine monophosphate.
592. The device of claim 544 wherein the agent causes DNA
damage.
593. The device of claim 544 wherein the agent is a DNA
intercalation agent.
594. The device of claim 544 wherein the agent is a RNA
synthesis inhibitor.
595. The device of claim 544 wherein the agent is a pyrimidine synthesis inhibitor.
596. The device of claim 544 wherein the agent inhibits ribonucleotide synthesis or function.
597. The device of claim 544 wherein the agent inhibits thymidine monophosphate synthesis or function.
598. The device of claim 544 wherein the agent inhibits DNA
synthesis.
599. The device of claim 544 wherein the agent causes DNA
adduct formation.
600. The device of claim 544 wherein the agent inhibits protein synthesis.
601. The device of claim 544 wherein the agent inhibits microtubule function.
602. The device of claim 544 wherein the agent is a cyclin dependent protein kinase inhibitor.
603. The device of claim 544 wherein the agent is an epidermal growth factor kinase inhibitor.
604. The device of claim 544 wherein the agent is an elastase inhibitor.
605. The device of claim 544 wherein the agent is a factor Xa inhibitor.
606. The device of claim 544 wherein the agent is a farnesyltransferase inhibitor.
607. The device of claim 544 wherein the agent is a fibrinogen antagonist.
608. The device of claim 544 wherein the agent is a guanylate cyclase stimulant.
609. The device of claim 544 wherein the agent is a heat shock protein 90 antagonist.
610. The device of claim 544 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
611. The device of claim 544 wherein the agent is a guanylate cyclase stimulant.
612. The device of claim 544 wherein the agent is a HMGCoA
reductase inhibitor.
613. The device of claim 544 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
614. The device of claim 544 wherein the agent is a hydroorotate dehydrogenase inhibitor.
615. The device of claim 544 wherein the agent is an IKK2 inhibitor.
616. The device of claim 544 wherein the agent is an IL-1 antagonist.
617. The device of claim 544 wherein the agent is an ICE
antagonist.
618. The device of claim 544 wherein the agent is an IRAK
antagonist.
619. The device of claim 544 wherein the agent is an IL-4 agonist.
620. The device of claim 544 wherein the agent is an immunomodulatory agent.
621. The device of claim 544 wherein the agent is sirolimus or an analogue or derivative thereof.
622. The device of claim 544 wherein the agent is not sirolimus.
623. The device of claim 544 wherein the agent is everolimus or an analogue or derivative thereof.
624. The device of claim 544 wherein the agent is tacrolimus or an analogue or derivative thereof.
625. The device of claim 544 wherein the agent is not tacrolimus.
626. The device of claim 544 wherein the agent is biolmus or an analogue or derivative thereof.
627. The device of claim 544 wherein the agent is tresperimus or an analogue or derivative thereof.
628. The device of claim 544 wherein the agent is auranofin or an analogue or derivative thereof.
629. The device of claim 544 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
630. The device of claim 544 wherein the agent is gusperimus or an analogue or derivative thereof.
631. The device of claim 544 wherein the agent is pimecrolimus or an analogue or derivative thereof.
632. The device of claim 544 wherein the agent is ABT-578 or an analogue or derivative thereof.
633. The device of claim 544 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
634. The device of claim 544 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
635. The device of claim 544 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
636. The device of claim 544 wherein the agent is a leukotriene inhibitor.
637. The device of claim 544 wherein the agent is a MCP-1 antagonist.
638. The device of claim 544 wherein the agent is a MMP
inhibitor.
639. The device of claim 544 wherein the agent is an NF kappa B inhibitor.
640. The device of claim 544 wherein the agent is an NF kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
641. The device of claim 544 wherein the agent is an NO
agonist.
642. The device of claim 544 wherein the agent is a p38 MAP
kinase inhibitor.
643. The device of claim 544 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
644. The device of claim 544 wherein the agent is a phosphodiesterase inhibitor.
645. The device of claim 544 wherein the agent is a TGF beta inhibitor.
646. The device of claim 544 wherein the agent is a thromboxane A2 antagonist.
647. The device of claim 544 wherein the agent is a TNFa antagonist.
648. The device of claim 544 wherein the agent is a TALE
inhibitor.
649. The device of claim 544 wherein the agent is a tyrosine kinase inhibitor.
650. The device of claim 544 wherein the agent is a vitronectin inhibitor.
651. The device of claim 544 wherein the agent is a fibroblast growth factor inhibitor.
652. The device of claim 544 wherein the agent is a protein kinase inhibitor.
653. The device of claim 544 wherein the agent is a PDGF
receptor kinase inhibitor.
654. The device of claim 544 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
655. The device of claim 544 wherein the agent is a retinoic acid receptor antagonist.
656. The device of claim 544 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
657. The device of claim 544 wherein the agent is a fibronogin antagonist.
658. The device of claim 544 wherein the agent is an antimycotic agent.
659. The device of claim 544 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
660. The device of claim 544 wherein the agent is a bisphosphonate.
661. The device of claim 544 wherein the agent is a phospholipase A1 inhibitor.
662. The device of claim 544 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
663. The device of claim 544 wherein the agent is a macrolide antibiotic.
664. The device of claim 544 wherein the agent is a GPIIb/IIIa receptor antagonist.
665. The device of claim 544 wherein the agent is an endothelin receptor antagonist.
666. The device of claim 544 wherein the agent is a peroxisome proliferator-activated receptor agonist.
667. The device of claim 544 wherein the agent is an estrogen receptor agent.
668. The device of claim 544 wherein the agent is a somastostatin analogue.
669. The device of claim 544 wherein the agent is a neurokinin 1 antagonist.
670. The device of claim 544 wherein the agent is a neurokinin 3 antagonist.
671. The device of claim 544 wherein the agent is a VLA-4 antagonist.
672. The device of claim 544 wherein the agent is an osteoclast inhibitor.
673. The device of claim 544 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
674. The device of claim 544 wherein the agent is an angiotensin I converting enzyme inhibitor.
675. The device of claim 544 wherein the agent is an angiotensin II antagonist.
676. The device of claim 544 wherein the agent is an enkephalinase inhibitor.
677. The device of claim 544 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
678. The device of claim 544 wherein the agent is a protein kinase C inhibitor.
679. The device of claim 544 wherein the agent is a ROCK (rho-associated kinase) inhibitor.
680. The device of claim 544 wherein the agent is a CXCR3 inhibitor.
681. The device of claim 544 wherein the agent is an Itk inhibitor.
682. The device of claim 544 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
683. The device of claim 544 wherein the agent is a PPAR
agonist.
684. The device of claim 544 wherein the agent is an immunosuppressant.
685. The device of claim 544 wherein the agent is an Erb inhibitor.
686. The device of claim 544 wherein the agent is an apoptosis agonist.
687. The device of claim 544 wherein the agent is a lipocortin agonist.
688. The device of claim 544 wherein the agent is a VCAM-1 antagonist.
689. The device of claim 544 wherein the agent is a collagen antagonist.
690. The device of claim 544 wherein the agent is an alpha 2 integrin antagonist.
691. The device of claim 544 wherein the agent is a TNF alpha inhibitor.
692. The device of claim 544 wherein the agent is a nitric oxide inhibitor
693. The device of claim 544 wherein the agent is a cathepsin inhibitor.
694. The device of claim 544 wherein the agent is not an anti-inflammatory agent.
695. The device of claim 544 wherein the agent is not a steroid.
696. The device of claim 544 wherein the agent is not a glucocorticosteroid.
697. The device of claim 544 wherein the agent is not dexamethasone.
698. The device of claim 544 wherein the agent is not an anti-infective agent.
699. The device of claim 544 wherein the agent is not an antibiotic.
700. The device of claim 544 wherein the agent is not an anti-fungal agent.
701. The device of claim 544, further comprising a polymer.
702. The device of claim 544, further comprising a polymeric carrier.
703. The device of claim 544 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
704. The device of claim 544 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
705. The device of claim 544, further comprising a coating, wherein the coating comprises the anti-scarring agent.
706. The device of claim 544, further comprising a coating, wherein the coating is disposed on a surface of the device.
707. The device of claim 544, further comprising a coating, wherein the coating directly contacts the device.
708. The device of claim 544, further comprising a coating, wherein the coating indirectly contacts the device.
709. The device of claim 544, further comprising a coating, wherein the coating partially covers the device.
710. The device of claim 544, further comprising a coating, wherein the coating completely covers the device.
711. The device of claim 544, further comprising a coating, wherein the coating is a uniform coating.
712. The device of claim 544, further comprising a coating, wherein the coating is a non-uniform coating.
713. The device of claim 544, further comprising a coating, wherein the coating is a discontinuous coating.
714. The device of claim 544, further comprising a coating, wherein the coating is a patterned coating.
715. The device of claim 544, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
716. The device of claim 544, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
717. The device of claim 544, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
718. The device of claim 544, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
719. The device of claim 544, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
720. The device of claim 544, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
721. The device of claim 544, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
722. The device of claim 544, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
723. The device of claim 544, further comprising a coating, wherein the coating further comprises a polymer.
724. The device of claim 544, further comprising a first coating having a first composition and the second coating having a second composition.
725. The device of claim 544, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
726. The device of claim 544, further comprising a polymer.
727. The device of claim 544, further comprising a polymeric carrier.
728. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
729. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
730. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
731. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
732. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
733. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
734. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
735. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
736. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
737. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
738. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
739. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
740. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
741. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
742. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
743. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
744. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
745. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
746. The device of claim 544, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
747. The device of claim 544, further comprising a lubricious coating.
748. The device of claim 544 wherein the anti-scarring agent is located within pores or holes of the device.
749. The device of claim 544 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
750. The device of claim 544, further comprising a second pharmaceutically active agent.
751. The device of claim 544, further comprising an anti-inflammatory agent.
752. The device of claim 544, further comprising an agent that inhibits infection.
753. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
754. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
755. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
756. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
757. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
758. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
759. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
760. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
761. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is etoposide.
762. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
763. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
764. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
765. The device of claim 544, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
766. The device of claim 544, further comprising an anti-thrombotic agent.
767. The device of claim 544, further comprising a visualization agent.
768. The device of claim 544, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
769. The device of claim 544, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
770. The device of claim 544, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.~
771. The device of claim 544, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
772. The device of claim 544, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
773. The device of claim 544, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
774. The device of claim 544, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.

1812~
775. The device of claim 544, further comprising an echogenic material.
776. The device of claim 544, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
777. The device of claim 544 wherein the device is sterile.
778. The device of claim 544 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
779. The device of claim 544 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
780. The device of claim 544 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
781. The device of claim 544 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
782. The device of claim 544 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
783. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
784. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
785. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
786. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
787. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
788. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
789. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
790. The device of claim 544 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
791. The device of claim 544 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
792. The device of claim 544 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
793. The device of claim 544 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
794. The device of claim 544 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
795. The device of claim 544 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
796. The device of claim 544 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
797. The device of claim 544 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
798. The device of claim 544 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
799. The device of claim 544 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
800. The device of claim 544 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
801. The device of claim 544 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
802. The device of any one of claims 544-801 wherein the implant is an AV fistula.
803. The device of any one of claims 544-801 wherein the implant is an AV access graft.
804. The device of any one of claims 544-801 wherein the implant is a venous catheter.
805. The device of any one of claims 544-801 wherein the implant is an implantable port.
806. The device of any one of claims 544-801 wherein the implant is an AV shunt.
807. A device, comprising an implant that provides an anastomotic connection (i.e., an anastomotic connector device) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
808. The device of claim 807 wherein the agent inhibits cell regeneration.
809. The device of claim 807 wherein the agent inhibits angiogenesis.
810. The device of claim 807 wherein the agent inhibits fibroblast migration.
811. The device of claim 807 wherein the agent inhibits fibroblast proliferation.
812. The device of claim 807 wherein the agent inhibits deposition of extracellular matrix.
813. The device of claim 807 wherein the agent inhibits tissue remodeling.
814. The device of claim 807 wherein the agent is an angiogenesis inhibitor.
815. The device of claim 807 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
816. The device of claim 807 wherein the agent is a chemokine receptor antagonist.
817. The device of claim 807 wherein the agent is a cell cycle inhibitor.
818. The device of claim 807 wherein the agent is a taxane.
819. The device of claim 807 wherein the agent is an anti-microtubule agent.
820. The device of claim 807 wherein the agent is paclitaxel.
821. The device of claim 807 wherein the agent is not paclitaxel.
822. The device of claim 807 wherein the agent is an analogue or derivative of paclitaxel.
823. The device of claim 807 wherein the agent is a vinca alkaloid.
824. The device of claim 807 wherein the agent is camptothecin or an analogue or derivative thereof.
825. The device of claim 807 wherein the agent is a podophyllotoxin.
826. The device of claim 807 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
827. The device of claim 807 wherein the agent is an anthracycline.~
828. The device of claim 807 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
829. The device of claim 807 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
830. The device of claim 807 wherein the agent is a platinum compound.
831. The device of claim 807 wherein the agent is a nitrosourea.
832. The device of claim 807 wherein the agent is a nitroimidazole.
833. The device of claim 807 wherein the agent is a folic acid antagonist.
834. The device of claim 807 wherein the agent is a cytidine analogue.
835. The device of claim 807 wherein the agent is a pyrimidine analogue.
836. The device of claim 807 wherein the agent is a fluoropyrimidine analogue.
837. The device of claim 807 wherein the agent is a purine analogue.
838. The device of claim 807 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
839. The device of claim 807 wherein the agent is a hydroxyurea.
840. The device of claim 807 wherein the agent is a mytomicin or an analogue or derivative thereof.
841. The device of claim 807 wherein the agent is an alkyl sulfonate.
842. The device of claim 807 wherein the agent is a benzamide or an analogue or derivative thereof.
843. The device of claim 807 wherein the agent is a nicotinamide or an analogue or derivative thereof.
844. The device of claim 807 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
845. The device of claim 807 wherein the agent is a DNA
alkylating agent.
846. The device of claim 807 wherein the agent is an anti-microtubule agent.
847. The device of claim 807 wherein the agent is a topoisomerase inhibitor.
848. The device of claim 807 wherein the agent is a DNA
cleaving agent.
849. The device of claim 807 wherein the agent is an antimetabolite.
850. The device of claim 807 wherein the agent inhibits adenosine deaminase.
851. The device of claim 807 wherein the agent inhibits purine ring synthesis.
852. The device of claim 807 wherein the agent is a nucleotide interconversion inhibitor.
853. The device of claim 807 wherein the agent inhibits dihydrofolate reduction.
854. The device of claim 807 wherein the agent blocks thymidine monophosphate.
855. The device of claim 807 wherein the agent causes DNA
damage.
856. The device of claim 807 wherein the agent is a DNA
intercalation agent.
857. The device of claim 807 wherein the agent is a RNA
synthesis inhibitor.
858. The device of claim 807 wherein the agent is a pyrimidine synthesis inhibitor.
859. The device of claim 807 wherein the agent inhibits ribonucleotide synthesis or function.
860. The device of claim 807 wherein the agent inhibits thymidine monophosphate synthesis or function.
861. The device of claim 807 wherein the agent inhibits DNA
synthesis.
862. The device of claim 807 wherein the agent causes DNA
adduct formation.
863. The device of claim 807 wherein the agent inhibits protein synthesis.
864. The device of claim 807 wherein the agent inhibits microtubule function.
865. The device of claim 807 wherein the agent is a cyclin dependent protein kinase inhibitor.
866. The device of claim 807 wherein the agent is an epidermal growth factor kinase inhibitor.
867. The device of claim 807 wherein the agent is an elastase inhibitor.
868. The device of claim 807 wherein the agent is a factor Xa inhibitor.
869. The device of claim 807 wherein the agent is a farnesyltransferase inhibitor.
870. The device of claim 807 wherein the agent is a fibrinogen antagonist.
871. The device of claim 807 wherein the agent is a guanylate cyclase stimulant.
872. The device of claim 807 wherein the agent is a heat shock protein 90 antagonist.
873. The device of claim 807 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
874. The device of claim 807 wherein the agent is a guanylate cyclase stimulant.
875. The device of claim 807 wherein the agent is a HMGCoA
reductase inhibitor.
876. The device of claim 807 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
877. The device of claim 807 wherein the agent is a hydroorotate dehydrogenase inhibitor.
878. The device of claim 807 wherein the agent is an IKK2 inhibitor.
879. The device of claim 807 wherein the agent is an IL-1 antagonist.
880. The device of claim 807 wherein the agent is an ICE
antagonist.
881. The device of claim 807 wherein the agent is an IRAK
antagonist.
882. The device of claim 807 wherein the agent is an IL-4 agonist.
883. The device of claim 807 wherein the agent is an immunomodulatory agent.
884. The device of claim 807 wherein the agent is sirolimus or an analogue or derivative thereof.
885. The device of claim 807 wherein the agent is not sirolimus.
886. The device of claim 807 wherein the agent is everolimus or an analogue or derivative thereof.
887. The device of claim 807 wherein the agent is tacrolimus or an analogue or derivative thereof.
888. The device of claim 807 wherein the agent is not tacrolimus.
889. The device of claim 807 wherein the agent is biolmus or an analogue or derivative thereof.
890. The device of claim 807 wherein the agent is tresperimus or an analogue or derivative thereof.
891. The device of claim 807 wherein the agent is auranofin or an analogue or derivative thereof.
892. The device of claim 807 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
893. The device of claim 807 wherein the agent is gusperimus or an analogue or derivative thereof.
894. The device of claim 807 wherein the agent is pimecrolimus or an analogue or derivative thereof.
895. The device of claim 807 wherein the agent is ABT-578 or an analogue or derivative thereof.
896. The device of claim 807 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
897. The device of claim 807 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
898. The device of claim 807 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
899. The device of claim 807 wherein the agent is a leukotriene inhibitor.
900. The device of claim 807 wherein the agent is a MCP-1 antagonist.
901. The device of claim 807 wherein the agent is a MMP
inhibitor.
902. The device of claim 807 wherein the agent is an NF kappa B inhibitor.
903. The device of claim 807 wherein the agent is an NF kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
904. The device of claim 807 wherein the agent is an NO
agonist.
905. The device of claim 807 wherein the agent is a p38 MAP
kinase inhibitor.
906. The device of claim 807 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
907. The device of claim 807 wherein the agent is a phosphodiesterase inhibitor.
908. The device of claim 807 wherein the agent is a TGF beta inhibitor.
909. The device of claim 807 wherein the agent is a thromboxane A2 antagonist.
910. The device of claim 807 wherein the agent is a TNFa antagonist.
911. The device of claim 807 wherein the agent is a TACE
inhibitor.
912. The device of claim 807 wherein the agent is a tyrosine kinase inhibitor.
913. The device of claim 807 wherein the agent is a vitronectin inhibitor.
914. The device of claim 807 wherein the agent is a fibroblast growth factor inhibitor.
915. The device of claim 807 wherein the agent is a protein kinase inhibitor.
916. The device of claim 807 wherein the agent is a PDGF
receptor kinase inhibitor.
917. The device of claim 807 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
918. The device of claim 807 wherein the agent is a retinoic acid receptor antagonist.
919. The device of claim 807 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
920. The device of claim 807 wherein the agent is a fibronogin antagonist.
921. The device of claim 807 wherein the agent is an antimycotic agent.
922. The device of claim 807 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
923. The device of claim 807 wherein the agent is a bisphosphonate.
924. The device of claim 807 wherein the agent is a phospholipase A1 inhibitor.
925. The device of claim 807 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
926. The device of claim 807 wherein the agent is a macrolide antibiotic.
927. The device of claim 807 wherein the agent is a GPIIb/IIIa receptor antagonist.
928. The device of claim 807 wherein the agent is an endothelin receptor antagonist.
929. The device of claim 807 wherein the agent is a peroxisome proliferator-activated receptor agonist.
930. The device of claim 807 wherein the agent is an estrogen receptor agent.
931. The device of claim 807 wherein the agent is a somastostatin analogue.
932. The device of claim 807 wherein the agent is a neurokinin 1 antagonist.
933. The device of claim 807 wherein the agent is a neurokinin 3 antagonist.
934. The device of claim 807 wherein the agent is a VLA-4 antagonist.
935. The device of claim 807 wherein the agent is an osteoclast inhibitor.
936. The device of claim 807 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
937. The device of claim 807 wherein the agent is an angiotensin I converting enzyme inhibitor.
938. The device of claim 807 wherein the agent is an angiotensin II antagonist.
939. The device of claim 807 wherein the agent is an enkephalinase inhibitor.
940. The device of claim 807 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
941. The device of claim 807 wherein the agent is a protein kinase C inhibitor.
942. The device of claim 807 wherein the agent is a ROCK (rho-associated kinase) inhibitor.
943. The device of claim 807 wherein the agent is a CXCR3 inhibitor.
944. The device of claim 807 wherein the agent is an Itk inhibitor.
945. The device of claim 807 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
946. The device of claim 807 wherein the agent is a PPAR
agonist.
947. The device of claim 807 wherein the agent is an immunosuppressant.
948. The device of claim 807 wherein the agent is an Erb inhibitor.
949. The device of claim 807 wherein the agent is an apoptosis agonist.
950. The device of claim 807 wherein the agent is a lipocortin agonist.
951. The device of claim 807 wherein the agent is a VCAM-1 antagonist.
952. The device of claim 807 wherein the agent is a collagen antagonist.
953. The device of claim 807 wherein the agent is an alpha 2 integrin antagonist.
954. The device of claim 807 wherein the agent is a TNF alpha inhibitor.
955. The device of claim 807 wherein the agent is a nitric oxide inhibitor
956. The device of claim 807 wherein the agent is a cathepsin inhibitor.
957. The device of claim 807 wherein the agent is not an anti-inflammatory agent.
958. The device of claim 807 wherein the agent is not a steroid.
959. The device of claim 807 wherein the agent is not a glucocorticosteroid.
960. The device of claim 807 wherein the agent is not dexamethasone.
961. The device of claim 807 wherein the agent is not an anti-infective agent.
962. The device of claim 807 wherein the agent is not an antibiotic.
963. The device of claim 807 wherein the agent is not an anti-fungal agent.
964. The device of claim 807, further comprising a polymer.
965. The device of claim 807, further comprising a polymeric carrier.
966. The device of claim 807 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
967. The device of claim 807 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
968. The device of claim 807, further comprising a coating, wherein the coating comprises the anti-scarring agent.
969. The device of claim 807, further comprising a coating, wherein the coating is disposed on a surface of the device.
970. The device of claim 807, further comprising a coating, wherein the coating directly contacts the device.
971. The device of claim 807, further comprising a coating, wherein the coating indirectly contacts the device.
972. The device of claim 807, further comprising a coating, wherein the coating partially covers the device.
973. The device of claim 807, further comprising a coating, wherein the coating completely covers the device.
974. The device of claim 807, further comprising a coating, wherein the coating is a uniform coating.
975. The device of claim 807, further comprising a coating, wherein the coating is a non-uniform coating.
976. The device of claim 807, further comprising a coating, wherein the coating is a discontinuous coating.
977. The device of claim 807, further comprising a coating, wherein the coating is a patterned coating.
978. The device of claim 807, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
979. The device of claim 807, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
980. The device of claim 807, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
981. The device of claim 807, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
982. The device of claim 807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
983. The device of claim 807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
984. The device of claim 807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
985. The device of claim 807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
986. The device of claim 807, further comprising a coating wherein the coating further comprises a polymer.
987. The device of claim 807, further comprising a first coating having a first composition and the second coating having a second composition.
988. The device of claim 807, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
989. The device of claim 807, further comprising a polymer.
990. The device of claim 807, further comprising a polymeric carrier.
991. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
992. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
993. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
994. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
995. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
996. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
997. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
998. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
999. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1000. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1001. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1002. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1003. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1004. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1005. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1006. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1007. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1008. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1009. The device of claim 807, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1010. The device of claim 807, further comprising a lubricious coating.
1011. The device of claim 807 wherein the anti-scarring agent is located within pores or holes of the device.
1012. The device of claim 807 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1013. The device of claim 807, further comprising a second pharmaceutically active agent.
1014. The device of claim 807, further comprising an anti-inflammatory agent.
1015. The device of claim 807, further comprising an agent that inhibits infection.
1016. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1017. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1018. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1019. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1020. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1021. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1022. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1023. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1024. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1025. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1026. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1027. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1028. The device of claim 807, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1029. The device of claim 807, further comprising an anti-thrombotic agent.
1030. The device of claim 807, further comprising a visualization agent.
1031. The device of claim 807, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1032. The device of claim 807, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1033. The device of claim 807, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1034. The device of claim 807, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1035. The device of claim 807, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1036. The device of claim 807, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1037. The device of claim 807, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1038. The device of claim 807, further comprising an echogenic material.
1039. The device of claim 807, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1040. The device of claim 807 wherein the device is sterile.
1041. The device of claim 807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1042. The device of claim 807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1043. The device of claim 807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1044. The device of claim 807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1045. The device of claim 807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1046. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1047. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1048. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1049. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1050. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1051. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1052. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1053. The device of claim 807 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1054. The device of claim 807 wherein the device comprises about. 0.01 µg to about 10 µg of the anti-scarring agent.
1055. The device of claim 807 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
1056. The device of claim 807 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1057. The device of claim 807 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1058. The device of claim 807 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1059. The device of claim 807 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1060. The device of claim 807 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1061. The device of claim 807 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1062. The device of claim 807 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1063. The device of claim 807 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1064. The device of claim 807 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1065. A device, comprising a ventricular assist implant (i.e., a ventricular assist device) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1066. The device of claim 1065 wherein the agent inhibits cell regeneration.
1067. The device of claim 1065 wherein the agent inhibits angiogenesis.
1068. The device of claim 1065 wherein the agent inhibits fibroblast migration.
1069. The device of claim 1065 wherein the agent inhibits fibroblast proliferation.
1070. The device of claim 1065 wherein the agent inhibits deposition of extracellular matrix.
1071. The device of claim 1065 wherein the agent inhibits tissue remodeling.
1072. The device of claim 1065 wherein the agent is an angiogenesis inhibitor.
1073. The device of claim 1065 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
1074. The device of claim 1065 wherein the agent is a chemoleine receptor antagonist.
1075. The device of claim 1065 wherein the agent is a cell cycle inhibitor.
1076. The device of claim 1065 wherein the agent is a taxane.
1077. The device of claim 1065 wherein the agent is an anti-microtubule agent.
1078. The device of claim 1065 wherein the agent is paclitaxel.
1079. The device of claim 1065 wherein the agent is not paclitaxel.
1080. The device of claim 1065 wherein the agent is an analogue or derivative of paclitaxel.
1081. The device of claim 1065 wherein the agent is a vinca alkaloid.
1082. The device of claim 1065 wherein the agent is camptothecin or an analogue or derivative thereof.
1083. The device of claim 1065 wherein the agent is a podophyllotoxin.
1084. The device of claim 1065 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
1085. The device of claim 1065 wherein the agent is an anthracycline.
1086. The device of claim 1065 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
1087. The device of claim 1065 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
1088. The device of claim 1065 wherein the agent is a platinum compound.
1089. The device of claim 1065 wherein the agent is a nitrosourea.
1090. The device of claim 1065 wherein the agent is a nitroimidazole.
1091. The device of claim 1065 wherein the agent is a folic acid antagonist.
1092. The device of claim 1065 wherein the agent is a cytidine analogue.
1093. The device of claim 1065 wherein the agent is a pyrimidine analogue.
1094. The device of claim 1065 wherein the agent is a fluoropyrimidine analogue.
1095. The device of claim 1065 wherein the agent is a purine analogue.
1096. The device of claim 1065 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
1097. The device of claim 1065 wherein the agent is a hydroxyurea.
1098. The device of claim 1065 wherein the agent is a mytomicin or an analogue or derivative thereof.
1099. The device of claim 1065 wherein the agent is an alkyl sulfonate.
1100. The device of claim 1065 wherein the agent is a benzamide or an analogue or derivative thereof.
1101. The device of claim 1065 wherein the agent is a nicotinamide or an analogue or derivative thereof.
1102. The device of claim 1065 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
1103. The device of claim 1065 wherein the agent is a DNA
alkylating agent.
1104. The device of claim 1065 wherein the agent is an anti-microtubule agent.
1105. The device of claim 1065 wherein the agent is a topoisomerase inhibitor.
1106. The device of claim 1065 wherein the agent is a DNA
cleaving agent.
1107. The device of claim 1065 wherein the agent is an antimetabolite.
1108. The device of claim 1065 wherein the agent inhibits adenosine deaminase.
1109. The device of claim 1065 wherein the agent inhibits purine ring synthesis.
1110. The device of claim 1065 wherein the agent is a nucleotide interconversion inhibitor.
1111. The device of claim 1065 wherein the agent inhibits dihydrofolate reduction.
1112. The device of claim 1065 wherein the agent blocks thymidine monophosphate.
1113. The device of claim 1065 wherein the agent causes DNA
damage.
1114. The device of claim 1065 wherein the agent is a DNA
intercalation agent.
1115. The device of claim 1065 wherein the agent is a RNA
synthesis inhibitor.
1116. The device of claim 1065 wherein the agent is a pyrimidine synthesis inhibitor.
1117. The device of claim 1065 wherein the agent inhibits , ribonucleotide synthesis or function.
1118. The device of claim 1065 wherein the agent inhibits thymidine monophosphate synthesis or function.
1119. The device of claim 1065 wherein the agent inhibits DNA
synthesis.
1120. The device of claim 1065 wherein the agent causes DNA
adduct formation.
1121. The device of claim 1065 wherein the agent inhibits protein synthesis.
1122. The device of claim 1065 wherein the agent inhibits microtubule function.
1123. The device of claim 1065 wherein the agent is a cyclin dependent protein kinase inhibitor.
1124. The device of claim 1065 wherein the agent is an epidermal growth factor kinase inhibitor.
1125. The device of claim 1065 wherein the agent is an elastase inhibitor.
1126. The device of claim 1065 wherein the agent is a factor Xa inhibitor.
1127. The device of claim 1065 wherein the agent is a farnesyltransferase inhibitor.
1128. The device of claim 1065 wherein the agent is a fibrinogen antagonist.
1129. The device of claim 1065 wherein the agent is a guanylate cyclase stimulant.
1130. The device of claim 1065 wherein the agent is a heat shock protein 90 antagonist.
1131. The device of claim 1065 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
1132. The device of claim 1065 wherein the agent is a guanylate cyclase stimulant.
1133. The device of claim 1065 wherein the agent is a HMGCoA
reductase inhibitor.
1134. The device of claim 1065 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
1135. The device of claim 1065 wherein the agent is a hydroorotate dehydrogenase inhibitor.
1136. The device of claim 1065 wherein the agent is an IKK2 inhibitor.
1137. The device of claim 1065 wherein the agent is an IL-1 antagonist.
1138. The device of claim 1065 wherein the agent is an ICE
antagonist.
1139. The device of claim 1065 wherein the agent is an IRAK
antagonist.
1140. The device of claim 1065 wherein the agent is an IL-4 agonist.
1141. The device of claim 1065 wherein the agent is an immunomodulatory agent.
1142. The device of claim 1065 wherein the agent is sirolimus or an analogue or derivative thereof.
1143. The device of claim 1065 wherein the agent is not sirolimus.
1144. The device of claim 1065 wherein the agent is everolimus or an analogue or derivative thereof.
1145. The device of claim 1065 wherein the agent is tacrolimus or an analogue or derivative thereof.
1146. The device of claim 1065 wherein the agent is not tacrolimus.
1147. The device of claim 1065 wherein the agent is biolmus or an analogue or derivative thereof.
1148. The device of claim 1065 wherein the agent is tresperimus or an analogue or derivative thereof.
1149. The device of claim 1065 wherein the agent is auranofin or an analogue or derivative thereof.
1150. The device of claim 1065 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
1151. The device of claim 1065 wherein the agent is gusperimus or an analogue or derivative thereof.
1152. The device of claim 1065 wherein the agent is pimecrolimus or an analogue or derivative thereof.
1153. The device of claim 1065 wherein the agent is ACT-578 or an analogue or derivative thereof.
1154. The device of claim 1065 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
1155. The device of claim 1065 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
1156. The device of claim 1065 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
1157. The device of claim 1065 wherein the agent is a leukotriene inhibitor.
1158. The device of claim 1065 wherein the agent is a MCP-1 antagonist.
1159. The device of claim 1065 wherein the agent is a MMP
inhibitor.
1160. The device of claim 1065 wherein the agent is an NF
kappa B inhibitor.
1161. The device of claim 1065 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
1162. The device of claim 1065 wherein the agent is an NO
agonist.
1163. The device of claim 1065 wherein the agent is a p38 MAP
kinase inhibitor.
1164. The device of claim 1065 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
1165. The device of claim 1065 wherein the agent is a phosphodiesterase inhibitor.
1166. The device of claim 1065 wherein the agent is a TGF beta inhibitor.
1167. The device of claim 1065 wherein the agent is a thromboxane A2 antagonist.
1168. The device of claim 1065 wherein the agent is a TNFa antagonist.
1169. The device of claim 1065 wherein the agent is a TALE
inhibitor.
1170. The device of claim 1065 wherein the agent is a tyrosine kinase inhibitor.
1171. The device of claim 1065 wherein the agent is a vitronectin inhibitor.
1172. The device of claim 1065 wherein the agent is a fibroblast growth factor inhibitor.
1173. The device of claim 1065 wherein the agent is a protein kinase inhibitor.
1174. The device of claim 1065 wherein the agent is a PDGF
receptor kinase inhibitor.
1175. The device of claim 1065 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
1176. The device of claim 1065 wherein the agent is a retinoic acid receptor antagonist.
1177. The device of claim 1065 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
1178. The device of claim 1065 wherein the agent is a fibronogin antagonist.
1179. The device of claim 1065 wherein the agent is an antimycotic agent.
1180. The device of claim 1065 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
1181. The device of claim 1065 wherein the agent is a bisphosphonate.
1182. The device of claim 1065 wherein the agent is a phospholipase A1 inhibitor.
1183. The device of claim 1065 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
1184. The device of claim 1065 wherein the agent is a macrolide antibiotic.
1185. The device of claim 1065 wherein the agent is a GPIIb/IIIa receptor antagonist.
1186. The device of claim 1065 wherein the agent is an endothelin receptor antagonist.
1187. The device of claim 1065 wherein the agent is a peroxisome proliferator-activated receptor agonist.
1188. The device of claim 1065 wherein the agent is an estrogen receptor agent.
1189. The device of claim 1065 wherein the agent is a somastostatin analogue.
1190. The device of claim 1065 wherein the agent is a neurokinin 1 antagonist.
1191. The device of claim 1065 wherein the agent is a neurokinin 3 antagonist.
1192. The device of claim 1065 wherein the agent is a VLA-4 antagonist.
1193. The device of claim 1065 wherein the agent is an osteoclast inhibitor.
1194. The device of claim 1065 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
1195. The device of claim 1065 wherein the agent is an angiotensin I converting enzyme inhibitor.
1196. The device of claim 1065 wherein the agent is an angiotensin II antagonist.
1197. The device of claim 1065 wherein the agent is an enkephalinase inhibitor.
1198. The device of claim 1065 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
1199. The device of claim 1065 wherein the agent is a protein kinase C inhibitor.
1200. The device of claim 1065 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
1201. The device of claim 1065 wherein the agent is a CXCR3 inhibitor.
1202. The device of claim 1065 wherein the agent is an Itk inhibitor.
1203. The device of claim 1065 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
1204. The device of claim 1065 wherein the agent is a PPAR
agonist.
1205. The device of claim 1065 wherein the agent is an immunosuppressant.
1206. The device of claim 1065 wherein the agent is an Erb inhibitor.
1207. The device of claim 1065 wherein the agent is an apoptosis agonist.
1208. The device of claim 1065 wherein the agent is a lipocortin agonist.
1209. The device of claim 1065 wherein the agent is a VCAM-1 antagonist.
1210. The device of claim 1065 wherein the agent is a collagen antagonist.
1211. The device of claim 1065 wherein the agent is an alpha 2 integrin antagonist.
1212. The device of claim 1065 wherein the agent is a TNF alpha inhibitor.
1213. The device of claim 1065 wherein the agent is a nitric oxide inhibitor
1214. The device of claim 1065 wherein the agent is a cathepsin inhibitor.
1215. The device of claim 1065 wherein the agent is not an anti-inflammatory agent.
1216. The device of claim 1065 wherein the agent is not a steroid.
1217. The device of claim 1065 wherein the agent is not a glucocorticosteroid.
1218. The device of claim 1065 wherein the agent is not dexamethasone.
1219. The device of claim 1065 wherein the agent is not an anti-infective agent.
1220. The device of claim 1065 wherein the agent is not an antibiotic.
1221. The device of claim 1065 wherein the agent is not an anti-fungal agent.
1222. The device of claim 1065, further comprising a polymer.
1223. The device of claim 1065, further comprising a polymeric carrier.
1224. The device of claim 1065 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1225. The device of claim 1065 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1226. The device of claim 1065, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1227. The device of claim 1065, further comprising a coating, wherein the coating is disposed on a surface of the device.
1228. The device of claim 1065, further comprising a coating, wherein the coating directly contacts the device.
1229. The device of claim 1065, further comprising a coating, wherein the coating indirectly contacts the device.
1230. The device of claim 1065, further comprising a coating, wherein the coating partially covers the device.
1231. The device of claim 1065, further comprising a coating, wherein the coating completely covers the device.
1232. The device of claim 1065, further comprising a coating, wherein the coating is a uniform coating.
1233. The device of claim 1065, further comprising a coating, wherein the coating is a non-uniform coating.
1234. The device of claim 1065, further comprising a coating, wherein the coating is a discontinuous coating.
1235. The device of claim 1065, further comprising a coating, wherein the coating is a patterned coating.
1236. The device of claim 1065, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
1237. The device of claim 1065, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
1238. The device of claim 1065, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1239. The device of claim 1065, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1240. The device of claim 1065, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1241. The device of claim 1065, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1242. The device of claim 1065, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1243. The device of claim 1065, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1244. The device of claim 1065, further comprising a coating, wherein the coating further comprises a polymer.
1245. The device of claim 1065, further comprising a first coating having a first composition and the second coating having a second composition.
1246. The device of claim 1065, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1247. The device of claim 1065, further comprising a polymer.
1248. The device of claim 1065, further comprising a polymeric carrier.
1249. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1250. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1251. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1252. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1253. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1254. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1255. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1256. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1257. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1258. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1259. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1260. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1261. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1262. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1263. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1264. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1265. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1266. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1267. The device of claim 1065, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1268. The device of claim 1065, further comprising a lubricious coating.
1269. The device of claim 1065 wherein the anti-scarring agent is located within pores or holes of the device.
1270. The device of claim 1065 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1271. The device of claim 1065, further comprising a second pharmaceutically active agent.
1272. The device of claim 1065, further comprising an anti-inflammatory agent.
1273. The device of claim 1065, further comprising an agent that inhibits infection.
1274. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1275. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1276. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1277. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1278. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1279. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1280. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1281. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1282. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1283. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1284. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1285. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1286. The device of claim 1065, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1287. The device of claim 1065, further comprising an anti-thrombotic agent.
1288. The device of claim 1065, further comprising a visualization agent.
1289. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1290. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1291. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1292. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1293. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1294. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1295. The device of claim 1065, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1296. The device of claim 1065, further comprising an echogenic material.
1297. The device of claim 1065, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1298. The device of claim 1065 wherein the device is sterile.
1299. The device of claim 1065 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1300. The device of claim 1065 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1301. The device of claim 1065 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1302. The device of claim 1065 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1303. The device of claim 1065 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1304. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1305. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1306. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1307. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1308. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1309. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1310. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1311. The device of claim 1065 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1312. The device of claim 1065 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
1313. The device of claim 1065 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
1314. The device of claim 1065 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1315. The device of claim 1065 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1316. The device of claim 1065 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1317. The device of claim 1065 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1318. The device of claim 1065 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1319. The device of claim 1065 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1320. The device of claim 1065 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1321. The device of claim 1065 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1322. The device of claim 1065 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1323. The device of any one of claims 1065-1322 wherein the implant is a left ventricular assist device.
1324. The device of any one of claims 1065-1322 wherein the implant is a right ventricular assist device.
1325. The device of any one of claims 1065-1322 wherein the implant is a biventricular assist device.
1326. The device of any one of claims 1065-1322 wherein the implant is a cardiac assist device.
1327. A device, comprising a prosthetic heart valve implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1328. The device of claim 1327 wherein the agent inhibits cell regeneration.
1329. The device of claim 1327 wherein the agent inhibits angiogenesis.
1330. The device of claim 1327 wherein the agent inhibits fibroblast migration.
1331. The device of claim 1327 wherein the agent inhibits fibroblast proliferation.
1332. The device of claim 1327 wherein the agent inhibits deposition of extracellular matrix.
1333. The device of claim 1327 wherein the agent inhibits tissue remodeling.
1334. The device of claim 1327 wherein the agent is an angiogenesis inhibitor.
1335. The device of claim 1327 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
1336. The device of claim 1327 wherein the agent is a chemokine receptor antagonist.
1337. The device of claim 1327 wherein the agent is a cell cycle inhibitor.
1338. The device of claim 1327 wherein the agent is a taxane.
1339. The device of claim 1327 wherein the agent is an anti-microtubule agent.
1340. The device of claim 1327 wherein the agent is paclitaxel.
1341. The device of claim 1327 wherein the agent is not paclitaxel.
1342. The device of claim 1327 wherein the agent is an analogue or derivative of paclitaxel.
1343. The device of claim 1327 wherein the agent is a vinca alkaloid.
1344. The device of claim 1327 wherein the agent is camptothecin or an analogue or derivative thereof.
1345. The device of claim 1327 wherein the agent is a podophyllotoxin.
1346. The device of claim 1327 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
1347. The device of claim 1327 wherein the agent is an anthracycline.
1348. The device of claim 1327 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
1349. The device of claim 1327 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
1350. The device of claim 1327 wherein the agent is a platinum compound.
1351. The device of claim 1327 wherein the agent is a nitrosourea.
1352. The device of claim 1327 wherein the agent is a nitroimidazole.
1353. The device of claim 1327 wherein the agent is a folic acid antagonist.
1354. The device of claim 1327 wherein the agent is a cytidine analogue.
1355. The device of claim 1327 wherein the agent is a pyrimidine analogue.
1356. The device of claim 1327 wherein the agent is a fluoropyrimidine analogue.
1357. The device of claim 1327 wherein the agent is a purine analogue.
1358. The device of claim 1327 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
1359. The device of claim 1327 wherein the agent is a hydroxyurea.
1360. The device of claim 1327 wherein the agent is a mytomicin or an analogue or derivative thereof.
1361. The device of claim 1327 wherein the agent is an alkyl sulfonate.
1362. The device of claim 1327 wherein the agent is a benzamide or an analogue or derivative thereof.
1363. The device of claim 1327 wherein the agent is a nicotinamide or an analogue or derivative thereof.
1364. The device of claim 1327 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
1365. The device of claim 1327 wherein the agent is a DNA
alkylating agent.
1366. The device of claim 1327 wherein the agent is an anti-microtubule agent.
1367. The device of claim 1327 wherein the agent is a topoisomerase inhibitor.
1368. The device of claim 1327 wherein the agent is a DNA
cleaving agent.
1369. The device of claim 1327 wherein the agent is an antimetabolite.
1370. The device of claim 1327 wherein the agent inhibits adenosine deaminase.
1371. The device of claim 1327 wherein the agent inhibits purine ring synthesis.
1372. The device of claim 1327 wherein the agent is a nucleotide interconversion inhibitor.
1373. The device of claim 1327 wherein the agent inhibits dihydrofolate reduction.
1374. The device of claim 1327 wherein the agent blocks thymidine monophosphate.
1375. The device of claim 1327 wherein the agent causes DNA
damage.
1376. The device of claim 1327 wherein the agent is a DNA
intercalation agent.
1377. The device of claim 1327 wherein the agent is a RNA
synthesis inhibitor.
1378. The device of claim 1327 wherein the agent is a pyrimidine synthesis inhibitor.
1379. The device of claim 1327 wherein the agent inhibits ribonucleotide synthesis or function.
1380. The device of claim 1327 wherein the agent inhibits thymidine monophosphate synthesis or function.
1381. The device of claim 1327 wherein the agent inhibits DNA
synthesis.
1382. The device of claim 1327 wherein the agent causes DNA
adduct formation.
1383. The device of claim 1327 wherein the agent inhibits protein synthesis.
1384. The device of claim 1327 wherein the agent inhibits microtubule function.
1385. The device of claim 1327 wherein the agent is a cyclin dependent protein kinase inhibitor.
1386. The device of claim 1327 wherein the agent is an epidermal growth factor kinase inhibitor.
1387. The device of claim 1327 wherein the agent is an elastase inhibitor.
1388. The device of claim 1327 wherein the agent is a factor Xa inhibitor.
1389. The device of claim 1327 wherein the agent is a farnesyltransferase inhibitor.
1390. The device of claim 1327 wherein the agent is a fibrinogen antagonist.
1391. The device of claim 1327 wherein the agent is a guanylate cyclase stimulant.
1392. The device of claim 1327 wherein the agent is a heat shock protein 90 antagonist.
1393. The device of claim 1327 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
1394. The device of claim 1327 wherein the agent is a guanylate cyclase stimulant.
1395. The device of claim 1327 wherein the agent is a HMGCoA
reductase inhibitor.
1396. The device of claim 1327 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
1397. The device of claim 1327 wherein the agent is a hydroorotate dehydrogenase inhibitor.
1398. The device of claim 1327 wherein the agent is an IKK2 inhibitor.
1399. The device of claim 1327 wherein the agent is an IL-1 antagonist.
1400. The device of claim 1327 wherein the agent is an ICE
antagonist.
1401. The device of claim 1327 wherein the agent is an IRAK
antagonist.
1402. The device of claim 1327 wherein the agent is an IL-4 agonist.
1403. The device of claim 1327 wherein the agent is an immunomodulatory agent.
1404. The device of claim 1327 wherein the agent is sirolimus or an analogue or derivative thereof.
1405. The device of claim 1327 wherein the agent is not sirolimus.
1406. The device of claim 1327 wherein the agent is everolimus or an analogue or derivative thereof.
1407. The device of claim 1327 wherein the agent is tacrolimus or an analogue or derivative thereof.
1408. The device of claim 1327 wherein the agent is not tacrolimus.
1409. The device of claim 1327 wherein the agent is biolmus or an analogue or derivative thereof.
1410. The device of claim 1327 wherein the agent is tresperimus or an analogue or derivative thereof.
1411. The device of claim 1327 wherein the agent is auranofin or an analogue or derivative thereof.
1412. The device of claim 1327 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
1413. The device of claim 1327 wherein the agent is gusperimus or an analogue or derivative thereof.
1414. The device of claim 1327 wherein the agent is pimecrolimus or an analogue or derivative thereof.
1415. The device of claim 1327 wherein the agent is ABT-578 or an analogue or derivative thereof.
1416. The device of claim 1327 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
1417. The device of claim 1327 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
1418. The device of claim 1327 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
1419. The device of claim 1327 wherein the agent is a leukotriene inhibitor.
1420. The device of claim 1327 wherein the agent is a MCP-1 antagonist.
1421. The device of claim 1327 wherein the agent is a MMP
inhibitor.
1422. The device of claim 1327 wherein the agent is an NF
kappa B inhibitor.
1423. The device of claim 1327 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
1424. The device of claim 1327 wherein the agent is an NO
agonist.
1425. The device of claim 1327 wherein the agent is a p38 MAP
kinase inhibitor.
1426. The device of claim 1327 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
1427. The device of claim 1327 wherein the agent is a phosphodiesterase inhibitor.
1428. The device of claim 1327 wherein the agent is a TGF beta inhibitor.
1429. The device of claim 1327 wherein the agent is a thromboxane A2 antagonist.
1430. The device of claim 1327 wherein the agent is a TNFa antagonist.
1431. The device of claim 1327 wherein the agent is a TACE
inhibitor.
1432. The device of claim 1327 wherein the agent is a tyrosine kinase inhibitor.
1433. The device of claim 1327 wherein the agent is a vitronectin inhibitor.
1434. The device of claim 1327 wherein the agent is a fibroblast growth factor inhibitor.
1435. The device of claim 1327 wherein the agent is a protein kinase inhibitor.
1436. The device of claim 1327 wherein the agent is a PDGF
receptor kinase inhibitor.
1437. The device of claim 1327 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
1438. The device of claim 1327 wherein the agent is a retinoic acid receptor antagonist.
1439. The device of claim 1327 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
1440. The device of claim 1327 wherein the agent is a fibronogin antagonist.
1441. The device of claim 1327 wherein the agent is an antimycotic agent.
1442. The device of claim 1327 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
1443. The device of claim 1327 wherein the agent is a bisphosphonate.
1444. The device of claim 1327 wherein the agent is a phospholipase A1 inhibitor.
1445. The device of claim 1327 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
1446. The device of claim 1327 wherein the agent is a macrolide antibiotic.
1447. The device of claim 1327 wherein the agent is a GPIIb/IIIa receptor antagonist.
1448. The device of claim 1327 wherein the agent is an endothelin receptor antagonist.
1449. The device of claim 1327 wherein the agent is a peroxisome proliferator-activated receptor agonist.
1450. The device of claim 1327 wherein the agent is an estrogen receptor agent.
1451. The device of claim 1327 wherein the agent is a somastostatin analogue.
1452. The device of claim 1327 wherein the agent is a neurokinin 1 antagonist.
1453. The device of claim 1327 wherein the agent is a neurokinin 3 antagonist.
1454. The device of claim 1327 wherein the agent is a VLA-4 antagonist.
1455. The device of claim 1327 wherein the agent is an osteoclast inhibitor.
1456. The device of claim 1327 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
1457. The device of claim 1327 wherein the agent is an angiotensin I converting enzyme inhibitor.
1458. The device of claim 1327 wherein the agent is an angiotensin II antagonist.
1459. The device of claim 1327 wherein the agent is an enkephalinase inhibitor.
1460. The device of claim 1327 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
1461. The device of claim 1327 wherein the agent is a protein kinase C inhibitor.
1462. The device of claim 1327 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
1463. The device of claim 1327 wherein the agent is a CXCR3 inhibitor.
1464. The device of claim 1327 wherein the agent is an Itk inhibitor.
1465. The device of claim 1327 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
1466. The device of claim 1327 wherein the agent is a PPAR
agonist.
1467. The device of claim 1327 wherein the agent is an immunosuppressant.
1468. The device of claim 1327 wherein the agent is an Erb inhibitor.
1469. The device of claim 1327 wherein the agent is an apoptosis agonist.
1470. The device of claim 1327 wherein the agent is a lipocortin agonist.
1471. The device of claim 1327 wherein the agent is a VCAM-1 antagonist.
1472. The device of claim 1327 wherein the agent is a collagen antagonist.
1473. The device of claim 1327 wherein the agent is an alpha 2 integrin antagonist.
1474. The device of claim 1327 wherein the agent is a TNF alpha inhibitor.
1475. The device of claim 1327 wherein the agent is a nitric oxide inhibitor
1476. The device of claim 1327 wherein the agent is a cathepsin inhibitor.
1477. The device of claim 1327 wherein the agent is not an anti-inflammatory agent.
1478. The device of claim 1327 wherein the agent is not a steroid.
1479. The device of claim 1327 wherein the agent is not a glucocorticosteroid.
1480. The device of claim 1327 wherein the agent is not dexamethasone.
1481. The device of claim 1327 wherein the agent is not an anti-infective agent.
1482. The device of claim 1327 wherein the agent is not an antibiotic.
1483. The device of claim 1327 wherein the agent is not an anti-fungal agent.
1484. The device of claim 1327, further comprising a polymer.
1485. The device of claim 1327, further comprising a polymeric carrier.
1486. The device of claim 1327 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1487. The device of claim 1327 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1488. The device of claim 1327, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1489. The device of claim 1327, further comprising a coating, wherein the coating is disposed on a surface of the device.
1490. The device of claim 1327, further comprising a coating, wherein the coating directly contacts the device.
1491. The device of claim 1327, further comprising a coating, wherein the coating indirectly contacts the device.
1492. The device of claim 1327, further comprising a coating, wherein the coating partially covers the device.
1493. The device of claim 1327, further comprising a coating, wherein the coating completely covers the device.
1494. The device of claim 1327, further comprising a coating, wherein the coating is a uniform coating.
1495. The device of claim 1327, further comprising a coating, wherein the coating is a non-uniform coating.
1496. The device of claim 1327, further comprising a coating, wherein the coating is a discontinuous coating.
1497. The device of claim 1327, further comprising a coating, wherein the coating is a patterned coating.
1498. The device of claim 1327, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
1499. The device of claim 1327, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
1500. The device of claim 1327, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1501. The device of claim 1327, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1502. The device of claim 1327, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1503. The device of claim 1327, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1504. The device of claim 1327, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1505. The device of claim 1327, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1506. The device of claim 1327, further comprising a coating, wherein the coating further comprises a polymer.
1507. The device of claim 1327, further comprising a first coating having a first composition and the second coating having a second composition.
1508. The device of claim 1327, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1509. The device of claim 1327, further comprising a polymer.
1510. The device of claim 1327, further comprising a polymeric carrier.
1511. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1512. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1513. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1514. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1515. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1516. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1517. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1518. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1519. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1520. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1521. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1522. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1523. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1524. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1525. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1526. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1527. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1528. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1529. The device of claim 1327, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1530. The device of claim 1327, further comprising a lubricious coating.
1531. The device of claim 1327 wherein the anti-scarring agent is located within pores or holes of the device.
1532. The device of claim 1327 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1533. The device of claim 1327, further comprising a second pharmaceutically active agent.
1534. The device of claim 1327, further comprising an anti-inflammatory agent.
1535. The device of claim 1327, further comprising an agent that inhibits infection.
1536. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1537. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1538. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1539. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1540. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1541. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1542. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1543. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1544. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1545. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1546. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1547. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1548. The device of claim 1327, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1549. The device of claim 1327, further comprising an anti-thrombotic agent.
1550. The device of claim 1327, further comprising a visualization agent.
1551. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1552. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1553. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1554. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1555. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1556. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1557. The device of claim 1327, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1558. The device of claim 1327, further comprising an echogenic material.
1559. The device of claim 1327, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1560. The device of claim 1327 wherein the device is sterile.
1561. The device of claim 1327 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1562. The device of claim 1327 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1563. The device of claim 1327 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1564. The device of claim 1327 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1565. The device of claim 1327 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1566. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1567. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1568. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1569. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1570. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1571. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1572. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1573. The device of claim 1327 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1574. The device of claim 1327 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
1575. The device of claim 1327 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
1576. The device of claim 1327 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1577. The device of claim 1327 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1578. The device of claim 1327 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1579. The device of claim 1327 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1580. The device of claim 1327 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1581. The device of claim 1327 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1582. The device of claim 1327 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1583. The device of claim 1327 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1584. The device of claim 1327 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1585. The device of any one of claims 1327-1584 wherein the implant is a mechanical prosthesis.
1586. The device of any one of claims 1327-1584 wherein the implant is a bioprosthetic heart valve.
1587. The device of any one of claims 1327-1584 wherein the implant is a bioprosthetic heart valve formed, at least in part, from porcine valve.
1588. The device of any one of claims 1327-1584 wherein the implant is a bioprosthetic heart valve formed, at least in part, from bovine pericardial valve.
1589. A device, comprising an inferior vena cava filter implant an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1590. The device of claim 1589 wherein the agent inhibits cell regeneration.
1591. The device of claim 1589 wherein the agent inhibits angiogenesis.
1592. The device of claim 1589 wherein the agent inhibits fibroblast migration.
1593. The device of claim 1589 wherein the agent inhibits fibroblast proliferation.
1594. The device of claim 1589 wherein the agent inhibits deposition of extracellular matrix.
1595. The device of claim 1589 wherein the agent inhibits tissue remodeling.
1596. The device of claim 1589 wherein the agent is an angiogenesis inhibitor.
1597. The device of claim 1589 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
1598. The device of claim 1589 wherein the agent is a chemokine receptor antagonist.
1599. The device of claim 1589 wherein the agent is a cell cycle inhibitor.
1600. The device of claim 1589 wherein the agent is a taxane.
1601. The device of claim 1589 wherein the agent is an anti-microtubule agent.
1602. The device of claim 1589 wherein the agent is paclitaxel.
1603. The device of claim 1589 wherein the agent is not paclitaxel.
1604. The device of claim 1589 wherein the agent is an analogue or derivative of paclitaxel.
1605. The device of claim 1589 wherein the agent is a vinca alkaloid.
1606. The device of claim 1589 wherein the agent is camptothecin or an analogue or derivative thereof.
1607. The device of claim 1589 wherein the agent is a podophyllotoxin.
1608. The device of claim 1589 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
1609. The device of claim 1589 wherein the agent is an anthracycline.
1610. The device of claim 1589 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
1611. The device of claim 1589 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
1612. The device of claim 1589 wherein the agent is a platinum compound.
1613. The device of claim 1589 wherein the agent is a nitrosourea.
1614. The device of claim 1589 wherein the agent is a nitroimidazole.
1615. The device of claim 1589 wherein the agent is a folic acid antagonist.
1616. The device of claim 1589 wherein the agent is a cytidine analogue.
1617. The device of claim 1589 wherein the agent is a pyrimidine analogue.
1618. The device of claim 1589 wherein the agent is a fluoropyrimidine analogue.
1619. The device of claim 1589 wherein the agent is a purine analogue.
1620. The device of claim 1589 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
1621. The device of claim 1589 wherein the agent is a hydroxyurea.
1622. The device of claim 1589 wherein the agent is a mytomicin or an analogue or derivative thereof.
1623. The device of claim 1589 wherein the agent is an alkyl sulfonate.
1624. The device of claim 1589 wherein the agent is a benzamide or an analogue or derivative thereof.
1625. The device of claim 1589 wherein the agent is a nicotinamide or an analogue or derivative thereof.
1626. The device of claim 1589 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
1627. The device of claim 1589 wherein the agent is a DNA
alkylating agent.
1628. The device of claim 1589 wherein the agent is an anti-microtubule agent.
1629. The device of claim 1589 wherein the agent is a topoisomerase inhibitor.
1630. The device of claim 1589 wherein the agent is a DNA
cleaving agent.
1631. The device of claim 1589 wherein the agent is an antimetabolite.
1632. The device of claim 1589 wherein the agent inhibits adenosine deaminase.
1633. The device of claim 1589 wherein the agent inhibits purine ring synthesis.
1634. The device of claim 1589 wherein the agent is a nucleotide interconversion inhibitor.
1635. The device of claim 1589 wherein the agent inhibits dihydrofolate reduction.
1636. The device of claim 1589 wherein the agent blocks thymidine monophosphate.
1637. The device of claim 1589 wherein the agent causes DNA
damage.
1638. The device of claim 1589 wherein the agent is a DNA
intercalation agent.
1639. The device of claim 1589 wherein the agent is a RNA
synthesis inhibitor.
1640. The device of claim 1589 wherein the agent is a pyrimidine synthesis inhibitor.
1641. The device of claim 1589 wherein the agent inhibits ribonucleotide synthesis or function.
1642. The device of claim 1589 wherein the agent inhibits thymidine monophosphate synthesis or function.
1643. The device of claim 1589 wherein the agent inhibits DNA
synthesis.
1644. The device of claim 1589 wherein the agent causes DNA
adduct formation.
1645. The device of claim 1589 wherein the agent inhibits protein synthesis.
1646. The device of claim 1589 wherein the agent inhibits microtubule function.
1647. The device of claim 1589 wherein the agent is a cyclin dependent protein kinase inhibitor.
1648. The device of claim 1589 wherein the agent is an epidermal growth factor kinase inhibitor.
1649. The device of claim 1589 wherein the agent is an elastase inhibitor.
1650. The device of claim 1589 wherein the agent is a factor Xa inhibitor.
1651. The device of claim 1589 wherein the agent is a farnesyltransferase inhibitor.
1652. The device of claim 1589 wherein the agent is a fibrinogen antagonist.
1653. The device of claim 1589 wherein the agent is a guanylate cyclase stimulant.
1654. The device of claim 1589 wherein the agent is a heat shock protein 90 antagonist.
1655. The device of claim 1589 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
1656. The device of claim 1589 wherein the agent is a guanylate cyclase stimulant.
1657. The device of claim 1589 wherein the agent is a HMGCoA
reductase inhibitor.
1658. The device of claim 1589 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
1659. The device of claim 1589 wherein the agent is a hydroorotate dehydrogenase inhibitor.
1660. The device of claim 1589 wherein the agent is an IKK2 inhibitor.
1661. The device of claim 1589 wherein the agent is an IL-1 antagonist.
1662. The device of claim 1589 wherein the agent is an ICE
antagonist.
1663. The device of claim 1589 wherein the agent is an IRAK
antagonist.
1664. The device of claim 1589 wherein the agent is an IL-4 agonist.
1665. The device of claim 1589 wherein the agent is an immunomodulatory agent.
1666. The device of claim 1589 wherein the agent is sirolimus or an analogue or derivative thereof.
1667. The device of claim 1589 wherein the agent is not sirolimus.
1668. The device of claim 1589 wherein the agent is everolimus or an analogue or derivative thereof.
1669. The device of claim 1589 wherein the agent is tacrolimus or an analogue or derivative thereof.
1670. The device of claim 1589 wherein the agent is not tacrolimus.
1671. The device of claim 1589 wherein the agent is biolmus or an analogue or derivative thereof.
1672. The device of claim 1589 wherein the agent is tresperimus or an analogue or derivative thereof.
1673. The device of claim 1589 wherein the agent is auranofin or an analogue or derivative thereof.
1674. The device of claim 1589 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
1675. The device of claim 1589 wherein the agent is gusperimus or an analogue or derivative thereof.
1676. The device of claim 1589 wherein the agent is pimecrolimus or an analogue or derivative thereof.
1677. The device of claim 1589 wherein the agent is ABT-578 or an analogue or derivative thereof.
1678. The device of claim 1589 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
1679. The device of claim 1589 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
1680. The device of claim 1589 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
1681. The device of claim 1589 wherein the agent is a leukotriene inhibitor.
1682. The device of claim 1589 wherein the agent is a MCP-1 antagonist.
1683. The device of claim 1589 wherein the agent is a MMP
inhibitor.
1684. The device of claim 1589 wherein the agent is an NF
kappa B inhibitor.
1685. The device of claim 1589 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
1686. The device of claim 1589 wherein the agent is an NO
agonist.
1687. The device of claim 1589 wherein the agent is a p38 MAP
kinase inhibitor.
1688. The device of claim 1589 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
1689. The device of claim 1589 wherein the agent is a phosphodiesterase inhibitor.
1690. The device of claim 1589 wherein the agent is a TGF beta inhibitor.
1691. The device of claim 1589 wherein the agent is a thromboxane A2 antagonist.
1692. The device of claim 1589 wherein the agent is a TNFa antagonist.
1693. The device of claim 1589 wherein the agent is a TACE
inhibitor.
1694. The device of claim 1589 wherein the agent is a tyrosine kinase inhibitor.
1695. The device of claim 1589 wherein the agent is a vitronectin inhibitor.
1696. The device of claim 1589 wherein the agent is a fibroblast growth factor inhibitor.
1697. The device of claim 1589 wherein the agent is a protein kinase inhibitor.
1698. The device of claim 1589 wherein the agent is a PDGF
receptor kinase inhibitor.
1699. The device of claim 1589 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
1700. The device of claim 1589 wherein the agent is a retinoic acid receptor antagonist.
1701. The device of claim 1589 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
1702. The device of claim 1589 wherein the agent is a fibronogin antagonist.
1703. The device of claim 1589 wherein the agent is an antimycotic agent.
1704. The device of claim 1589 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
1705. The device of claim 1589 wherein the agent is a bisphosphonate.
1706. The device of claim 1589 wherein the agent is a phospholipase A1 inhibitor.
1707. The device of claim 1589 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
1708. The device of claim 1589 wherein the agent is a macrolide antibiotic.
1709. The device of claim 1589 wherein the agent is a GPIIb/IIIa receptor antagonist.
1710. The device of claim 1589 wherein the agent is an endothelin receptor antagonist.
1711. The device of claim 1589 wherein the agent is a peroxisome proliferator-activated receptor agonist.
1712. The device of claim 1589 wherein the agent is an estrogen receptor agent.
1713. The device of claim 1589 wherein the agent is a somastostatin analogue.
1714. The device of claim 1589 wherein the agent is a neurokinin 1 antagonist.
1715. The device of claim 1589 wherein the agent is a neurokinin 3 antagonist.
1716. The device of claim 1589 wherein the agent is a VLA-4 antagonist.
1717. The device of claim 1589 wherein the agent is an osteoclast inhibitor.
1718. The device of claim 1589 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
1719. The device of claim 1589 wherein the agent is an angiotensin I converting enzyme inhibitor.
1720. The device of claim 1589 wherein the agent is an angiotensin II antagonist.
1721. The device of claim 1589 wherein the agent is an enkephalinase inhibitor.
1722. The device of claim 1589 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
1723. The device of claim 1589 wherein the agent is a protein kinase C inhibitor.
1724. The device of claim 1589 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
1725. The device of claim 1589 wherein the agent is a CXCR3 inhibitor.
1726. The device of claim 1589 wherein the agent is an Itk inhibitor.
1727. The device of claim 1589 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
1728. The device of claim 1589 wherein the agent is a PPAR
agonist.
1729. The device of claim 1589 wherein the agent is an immunosuppressant.
1730. The device of claim 1589 wherein the agent is an Erb inhibitor.
1731. The device of claim 1589 wherein the agent is an apoptosis agonist.
1732. The device of claim 1589 wherein the agent is a lipocortin agonist.
1733. The device of claim 1589 wherein the agent is a VCAM-1 antagonist.
1734. The device of claim 1589 wherein the agent is a collagen antagonist.
1735. The device of claim 1589 wherein the agent is an alpha 2 integrin antagonist.
1736. The device of claim 1589 wherein the agent is a TNF alpha inhibitor.
1737. The device of claim 1589 wherein the agent is a nitric oxide inhibitor
1738. The device of claim 1589 wherein the agent is a cathepsin inhibitor.
1739. The device of claim 1589 wherein the agent is not an anti-inflammatory agent.
1740. The device of claim 1589 wherein the agent is not a steroid.
1741. The device of claim 1589 wherein the agent is not a glucocorticosteroid.
1742. The device of claim 1589 wherein the agent is not dexamethasone.
1743. The device of claim 1589 wherein the agent is not an anti-infective agent.
1744. The device of claim 1589 wherein the agent is not an antibiotic.
1745. The device of claim 1589 wherein the agent is not an anti-fungal agent.
1746. The device of claim 1589, further comprising a polymer.
1747. The device of claim 1589, further comprising a polymeric carrier.
1748. The device of claim 1589 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1749. The device of claim 1589 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1750. The device of claim 1589, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1751. The device of claim 1589, further comprising a coating, wherein the coating is disposed on a surface of the device.
1752. The device of claim 1589, further comprising a coating, wherein the coating directly contacts the device.
1753. The device of claim 1589, further comprising a coating, wherein the coating indirectly contacts the device.
1754. The device of claim 1589, further comprising a coating, wherein the coating partially covers the device.
1755. The device of claim 1589, further comprising a coating, wherein the coating completely covers the device.
1756. The device of claim 1589, further comprising a coating, wherein the coating is a uniform coating.
1757. The device of claim 1589, further comprising a coating, wherein the coating is a non-uniform coating.
1758. The device of claim 1589, further comprising a coating, wherein the coating is a discontinuous coating.
1759. The device of claim 1589, further comprising a coating, wherein the coating is a patterned coating.
1760. The device of claim 1589, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
1761. The device of claim 1589, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
1762. The device of claim 1589, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1763. The device of claim 1589, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1764. The device of claim 1589, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1765. The device of claim 1589, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1766. The device of claim 1589, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1767. The device of claim 1589, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1768. The device of claim 1589, further comprising a coating, wherein the coating further comprises a polymer.
1769. The device of claim 1589, further comprising a first coating having a first composition and the second coating having a second composition.
1770. The device of claim 1589, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1771. The device of claim 1589, further comprising a polymer.
1772. The device of claim 1589, further comprising a polymeric carrier.
1773. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1774. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1775. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1776. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1777. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1778. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1779. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1780. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1781. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1782. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1783. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1784. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1785. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1786. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1787. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1788. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1789. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1790. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1791. The device of claim 1589, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1792. The device of claim 1589, further comprising a lubricious coating.
1793. The device of claim 1589 wherein the anti-scarring agent is located within pores or holes of the device.
1794. The device of claim 1589 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1795. The device of claim 1589, further comprising a second pharmaceutically active agent.
1796. The device of claim 1589, further comprising an anti-inflammatory agent.
1797. The device of claim 1589, further comprising an agent that inhibits infection.
1798. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1799. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1800. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1801. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1802. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1803. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1804. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1805. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1806. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1807. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1808. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1809. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1810. The device of claim 1589, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1811. The device of claim 1589, further comprising an anti-thrombotic agent.
1812. The device of claim 1589, further comprising a visualization agent.
1813. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1814. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1815. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1816. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1817. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1818. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1819. The device of claim 1589, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1820. The device of claim 1589, further comprising an echogenic material.
1821. The device of claim 1589, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1822. The device of claim 1589 wherein the device is sterile.
1823. The device of claim 1589 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1824. The device of claim 1589 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1825. The device of claim 1589 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1826. The device of claim 1589 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1827. The device of claim 1589 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1828. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1829. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1830. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1831. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1832. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1833. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1834. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1835. The device of claim 1589 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1836. The device of claim 1589 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
1837. The device of claim 1589 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
1838. The device of claim 1589 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1839. The device of claim 1589 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1840. The device of claim 1589 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1841. The device of claim 1589 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1842. The device of claim 1589 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1843. The device of claim 1589 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1844. The device of claim 1589 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1845. The device of claim 1589 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1846. The device of claim 1589 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1847. A device, comprising a peritoneal dialysis catheter implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1848. The device of claim 1847 wherein the agent inhibits cell regeneration.
1849. The device of claim 1847 wherein the agent inhibits angiogenesis.
1850. The device of claim 1847 wherein the agent inhibits fibroblast migration.
1851. The device of claim 1847 wherein the agent inhibits fibroblast proliferation.
1852. The device of claim 1847 wherein the agent inhibits deposition of extracellular matrix.
1853. The device of claim 1847 wherein the agent inhibits tissue remodeling.
1854. The device of claim 1847 wherein the agent is an angiogenesis inhibitor.
1855. The device of claim 1847 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
1856. The device of claim 1847 wherein the agent is a chemokine receptor antagonist.
1857. The device of claim 1847 wherein the agent is a cell cycle inhibitor.
1858. The device of claim 1847 wherein the agent is a taxane.
1859. The device of claim 1847 wherein the agent is an anti-microtubule agent.
1860. The device of claim 1847 wherein the agent is paclitaxel.
1861. The device of claim 1847 wherein the agent is not paclitaxel.
1862. The device of claim 1847 wherein the agent is an analogue or derivative of paclitaxel.
1863. The device of claim 1847 wherein the agent is a vinca alkaloid.
1864. The device of claim 1847 wherein the agent is camptothecin or an analogue or derivative thereof.
1865. The device of claim 1847 wherein the agent is a podophyllotoxin.
1866. The device of claim 1847 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
1867. The device of claim 1847 wherein the agent is an anthracycline.
1868. The device of claim 1847 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
1869. The device of claim 1847 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
1870. The device of claim 1847 wherein the agent is a platinum compound.
1871. The device of claim 1847 wherein the agent is a nitrosourea.
1872. The device of claim 1847 wherein the agent is a nitroimidazole.
1873. The device of claim 1847 wherein the agent is a folic acid antagonist.
1874. The device of claim 1847 wherein the agent is a cytidine analogue.
1875. The device of claim 1847 wherein the agent is a pyrimidine analogue.
1876. The device of claim 1847 wherein the agent is a fluoropyrimidine analogue.
1877. The device of claim 1847 wherein the agent is a purine analogue.
1878. The device of claim 1847 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
1879. The device of claim 1847 wherein the agent is a hydroxyurea.
1880. The device of claim 1847 wherein the agent is a mytomicin or an analogue or derivative thereof.
1881. The device of claim 1847 wherein the agent is an alkyl sulfonate.
1882. The device of claim 1847 wherein the agent is a benzamide or an analogue or derivative thereof.
1883. The device of claim 1847 wherein the agent is a nicotinamide or an analogue or derivative thereof.
1884. The device of claim 1847 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
1885. The device of claim 1847 wherein the agent is a DNA
alkylating agent.
1886. The device of claim 1847 wherein the agent is an anti-microtubule agent.
1887. The device of claim 1847 wherein the agent is a topoisomerase inhibitor.
1888. The device of claim 1847 wherein the agent is a DNA
cleaving agent.
1889. The device of claim 1847 wherein the agent is an antimetabolite.
1890. The device of claim 1847 wherein the agent inhibits adenosine deaminase.
1891. The device of claim 1847 wherein the agent inhibits purine ring synthesis.
1892. The device of claim 1847 wherein the agent is a nucleotide interconversion inhibitor.
1893. The device of claim 1847 wherein the agent inhibits dihydrofolate reduction.
1894. The device of claim 1847 wherein the agent blocks thymidine monophosphate.
1895. The device of claim 1847 wherein the agent causes DNA
damage.
1896. The device of claim 1847 wherein the agent is a DNA
intercalation agent.
1897. The device of claim 1847 wherein the agent is a RNA
synthesis inhibitor.
1898. The device of claim 1847 wherein the agent is a pyrimidine synthesis inhibitor.
1899. The device of claim 1847 wherein the agent inhibits ribonucleotide synthesis or function.
1900. The device of claim 1847 wherein the agent inhibits thymidine monophosphate synthesis or function.
1901. The device of claim 1847 wherein the agent inhibits DNA
synthesis.
1902. The device of claim 1847 wherein the agent causes DNA
adduct formation.
1903. The device of claim 1847 wherein the agent inhibits protein synthesis.
1904. The device of claim 1847 wherein the agent inhibits microtubule function.
1905. The device of claim 1847 wherein the agent is a cyclin dependent protein kinase inhibitor.
1906. The device of claim 1847 wherein the agent is an epidermal growth factor kinase inhibitor.
1907. The device of claim 1847 wherein the agent is an elastase inhibitor.
1908. The device of claim 1847 wherein the agent is a factor Xa inhibitor.
1909. The device of claim 1847 wherein the agent is a farnesyltransferase inhibitor.
1910. The device of claim 1847 wherein the agent is a fibrinogen antagonist.
1911. The device of claim 1847 wherein the agent is a guanylate cyclase stimulant.
1912. The device of claim 1847 wherein the agent is a heat shock protein 90 antagonist.
1913. The device of claim 1847 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
1914. The device of claim 1847 wherein the agent is a guanylate cyclase stimulant.
1915. The device of claim 1847 wherein the agent is a HMGCoA
reductase inhibitor.
1916. The device of claim 1847 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
1917. The device of claim 1847 wherein the agent is a hydroorotate dehydrogenase inhibitor.
1918. The device of claim 1847 wherein the agent is an IKK2 inhibitor.
1919. The device of claim 1847 wherein the agent is an IL-1 antagonist.
1920. The device of claim 1847 wherein the agent is an ICE
antagonist.
1921. The device of claim 1847 wherein the agent is an IRAK
antagonist.
1922. The device of claim 1847 wherein the agent is an IL-4 agonist.
1923. The device of claim 1847 wherein the agent is an immunomodulatory agent.
1924. The device of claim 1847 wherein the agent is sirolimus or an analogue or derivative thereof.
1925. The device of claim 1847 wherein the agent is not sirolimus.
1926. The device of claim 1847 wherein the agent is everolimus or an analogue or derivative thereof.
1927. The device of claim 1847 wherein the agent is tacrolimus or an analogue or derivative thereof.
1928. The device of claim 1847 wherein the agent is not tacrolimus.
1929. The device of claim 1847 wherein the agent is biolmus or an analogue or derivative thereof.
1930. The device of claim 1847 wherein the agent is tresperimus or an analogue or derivative, thereof.
1931. The device of claim 1847 wherein the agent is auranofin or an analogue or derivative thereof.
1932. The device of claim 1847 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
1933. The device of claim 1847 wherein the agent is gusperimus or an analogue or derivative thereof.
1934. The device of claim 1847 wherein the agent is pimecrolimus or an analogue or derivative thereof.
1935. The device of claim 1847 wherein the agent is ABT-578 or an analogue or derivative thereof.
1936. The device of claim 1847 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
1937. The device of claim 1847 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
1938. The device of claim 1847 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
1939. The device of claim 1847 wherein the agent is a leukotriene inhibitor.
1940. The device of claim 1847 wherein the agent is a MCP-1 antagonist.
1941. The device of claim 1847 wherein the agent is a MMP
inhibitor.~
1942. The device of claim 1847 wherein the agent is an NF
kappa B inhibitor.
1943. The device of claim 1847 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
1944. The device of claim 1847 wherein the agent is an NO
agonist.
1945. The device of claim 1847 wherein the agent is a p38 MAP
kinase inhibitor.
1946. The device of claim 1847 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
1947. The device of claim 1847 wherein the agent is a phosphodiesterase inhibitor.
1948. The device of claim 1847 wherein the agent is a TGF beta inhibitor.
1949. The device of claim 1847 wherein the agent is a thromboxane A2 antagonist.
1950. The device of claim 1847 wherein the agent is a TNFa antagonist.
1951. The device of claim 1847 wherein the agent is a TACE
inhibitor.
1952. The device of claim 1847 wherein the agent is a tyrosine kinase inhibitor.
1953. The device of claim 1847 wherein the agent is a vitronectin inhibitor.
1954. The device of claim 1847 wherein the agent is a fibroblast growth factor inhibitor.
1955. The device of claim 1847 wherein the agent is a protein kinase inhibitor.
1956. The device of claim 1847 wherein the agent is a PDGF
receptor kinase inhibitor.
1957. The device of claim 1847 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
1958. The device of claim 1847 wherein the agent is a retinoic acid receptor antagonist.
1959. The device of claim 1847 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
1960. The device of claim 1847 wherein the agent is a fibronogin antagonist.
1961. The device of claim 1847 wherein the agent is an antimycotic agent.
1962. The device of claim 1847 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
1963. The device of claim 1847 wherein the agent is a bisphosphonate.
1964. The device of claim 1847 wherein the agent is a phospholipase A1 inhibitor.
1965. The device of claim 1847 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
1966. The device of claim 1847 wherein the agent is a macrolide antibiotic.
1967. The device of claim 1847 wherein the agent is a GPIIb/IIIa receptor antagonist.
1968. The device of claim 1847 wherein the agent is an endothelia receptor antagonist.
1969. The device of claim 1847 wherein the agent is a peroxisome proliferator-activated receptor agonist.
1970. The device of claim 1847 wherein the agent is an estrogen receptor agent.
1971. The device of claim 1847 wherein the agent is a somastostatin analogue.
1972. The device of claim 1847 wherein the agent is a neurokinin 1 antagonist.
1973. The device of claim 1847 wherein the agent is a neurokinin 3 antagonist.
1974. The device of claim 1847 wherein the agent is a VLA-4 antagonist.
1975. The device of claim 1847 wherein the agent is an osteoclast inhibitor.
1976. The device of claim 1847 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
1977. The device of claim 1847 wherein the agent is an angiotensin I converting enzyme inhibitor.
1978. The device of claim 1847 wherein the agent is an angiotensin II antagonist.
1979. The device of claim 1847 wherein the agent is an enkephalinase inhibitor.
1980. The device of claim 1847 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
1981. The device of claim 1847 wherein the agent is a protein kinase C inhibitor.
1982. The device of claim 1847 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
1983. The device of claim 1847 wherein the agent is a CXCR3 inhibitor.
1984. The device of claim 1847 wherein the agent is an Itk inhibitor.
1985. The device of claim 1847 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
1986. The device of claim 1847 wherein the agent is a PPAR
agonist.
1987. The device of claim 1847 wherein the agent is an immunosuppressant.
1988. The device of claim 1847 wherein the agent is an Erb inhibitor.
1989. The device of claim 1847 wherein the agent is an apoptosis agonist.
1990. The device of claim 1847 wherein the agent is a lipocortin agonist.
1991. The device of claim 1847 wherein the agent is a VCAM-1 antagonist.
1992. The device of claim 1847 wherein the agent is a collagen antagonist.
1993. The device of claim 1847 wherein the agent is an alpha 2 integrin antagonist.
1994. The device of claim 1847 wherein the agent is a TNF alpha inhibitor.
1995. The device of claim 1847 wherein the agent is a nitric oxide inhibitor
1996. The device of claim 1847 wherein the agent is a cathepsin inhibitor.
1997. The device of claim 1847 wherein the agent is not an anti-inflammatory agent.
1998. The device of claim 1847 wherein the agent is not a steroid.
1999. The device of claim 1847 wherein the agent is not a glucocorticosteroid.
2000. The device of claim 1847 wherein the agent is not dexamethasone.
2001. The device of claim 1847 wherein the agent is not an anti-infective agent.
2002. The device of claim 1847 wherein the agent is not an antibiotic.
2003. The device of claim 1847 wherein the agent is not an anti-fungal agent.
2004. The device of claim 1847, further comprising a polymer.
2005. The device of claim 1847, further comprising a polymeric carrier.
2006. The device of claim 1847 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2007. The device of claim 1847 wherein the device delivers the anti-scarring agent focally to tissue proximate to the device.
2008. The device of claim 1847, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2009. The device of claim 1847, further comprising a coating, wherein the coating is disposed on a surface of the device.
2010. The device of claim 1847, further comprising a coating, wherein the coating directly contacts the device.
2011. The device of claim 1847, further comprising a coating, wherein the coating indirectly contacts the device.
2012. The device of claim 1847, further comprising a coating, wherein the coating partially covers the device.
2013. The device of claim 1847, further comprising a coating, wherein the coating completely covers the device.
2014. The device of claim 1847, further comprising a coating, wherein the coating is a uniform coating.
2015. The device of claim 1847, further comprising a coating, wherein the coating is a non-uniform coating.
2016. The device of claim 1847, further comprising a coating, wherein the coating is a discontinuous coating.
2017. The device of claim 1847, further comprising a coating, wherein the coating is a patterned coating.
2018. The device of claim 1847, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
2019. The device of claim 1847, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
2020. The device of claim 1847, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2021. The device of claim 1847, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2022. The device of claim 1847, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2023. The device of claim 1847, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2024. The device of claim 1847, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2025. The device of claim 1847, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2026. The device of claim 1847, further comprising a coating, wherein the coating further comprises a polymer.~
2027. The device of claim 1847, further comprising a first coating having a first composition and the second coating having a second composition.
2028. The device of claim 1847, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2029. The device of claim 1847, further comprising a polymer.
2030. The device of claim 1847, further comprising a polymeric carrier.
2031. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2032. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2033. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2034. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2035. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2036. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2037. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2038. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2039. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2040. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2041. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2042. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2043. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2044. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2045. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2046. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2047. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2048. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2049. The device of claim 1847, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2050. The device of claim 1847, further comprising a lubricious coating.
2051. The device of claim 1847 wherein the anti-scarring agent is located within pores or holes of the device.
2052. The device of claim 1847 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2053. The device of claim 1847, further comprising a second pharmaceutically active agent.
2054. The device of claim 1847, further comprising an anti-inflammatory agent.
2055. The device of claim 1847, further comprising an agent that inhibits infection.
2056. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2057. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2058. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2059. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2060. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2061. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2062. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2063. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2064. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2065. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2066. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2067. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2068. The device of claim 1847, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2069. The device of claim 1847, further comprising an anti-thrombotic agent.
2070. The device of claim 1847, further comprising a visualization agent.
2071. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2072. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2073. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2074. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2075. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2076. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2077. The device of claim 1847, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2078. The device of claim 1847, further comprising an echogenic material.
2079. The device of claim 1847, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2080. The device of claim 1847 wherein the device is sterile.
2081. The device of claim 1847 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2082. The device of claim 1847 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2083. The device of claim 1847 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2084. The device of claim 1847 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2085. The device of claim 1847 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2086. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2087. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2088. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2089. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2090. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2091. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2092. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2093. The device of claim 1847 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2094. The device of claim 1847 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
2095. The device of claim 1847 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
2096. The device of claim 1847 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2097. The device of claim 1847 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2098. The device of claim 1847 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2099. The device of claim 1847 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2100. The device of claim 1847 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2101. The device of claim 1847 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2102. The device of claim 1847 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2103. The device of claim 1847 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2104. The device of claim 1847 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2105. A device, comprising a central nervous system shunt (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2106. The device of claim 2105 wherein the agent inhibits cell regeneration.
2107. The device of claim 2105 wherein the agent inhibits angiogenesis.
2108. The device of claim 2105 wherein the agent inhibits fibroblast migration.
2109. The device of claim 2105 wherein the agent inhibits fibroblast proliferation.
2110. The device of claim 2105 wherein the agent inhibits deposition of extracellular matrix.
2111. The device of claim 2105 wherein the agent inhibits tissue remodeling.
2112. The device of claim 2105 wherein the agent is an angiogenesis inhibitor.
2113. The device of claim 2105 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
2114. The device of claim 2105 wherein the agent is a chemokine receptor antagonist.
2115. The device of claim 2105 wherein the agent is a cell cycle inhibitor.
2116. The device of claim 2105 wherein the agent is a taxane.
2117. The device of claim 2105 wherein the agent is an anti-microtubule agent.
2118. The device of claim 2105 wherein the agent is paclitaxel.
2119. The device of claim 2105 wherein the agent is not paclitaxel.
2120. The device of claim 2105 wherein the agent is an analogue or derivative of paclitaxel.
2121. The device of claim 2105 wherein the agent is a vinca alkaloid.
2122. The device of claim 2105 wherein the agent is camptothecin or an analogue or derivative thereof.
2123. The device of claim 2105 wherein the agent is a podophyllotoxin.
2124. The device of claim 2105 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
2125. The device of claim 2105 wherein the agent is an anthracycline.
2126. The device of claim 2105 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
2127. The device of claim 2105 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
2128. The device of claim 2105 wherein the agent is a platinum compound.
2129. The device of claim 2105 wherein the agent is a nitrosourea.
2130. The device of claim 2105 wherein the agent is a nitroimidazole.
2131. The device of claim 2105 wherein the agent is a folic acid antagonist.
2132. The device of claim 2105 wherein the agent is a cytidine analogue.
2133. The device of claim 2105 wherein the agent is a pyrimidine analogue.
2134. The device of claim 2105 wherein the agent is a fluoropyrimidine analogue.
2135. The device of claim 2105 wherein the agent is a purine analogue.
2136. The device of claim 2105 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
2137. The device of claim 2105 wherein the agent is a hydroxyurea.
2138. The device of claim 2105 wherein the agent is a mytomicin or an analogue or derivative thereof.
2139. The device of claim 2105 wherein the agent is an alkyl sulfonate.
2140. The device of claim 2105 wherein the agent is a benzamide or an analogue or derivative thereof.
2141. The device of claim 2105 wherein the agent is a nicotinamide or an analogue or derivative thereof.
2142. The device of claim 2105 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
2143. The device of claim 2105 wherein the agent is a DNA
alkylating agent.
2144. The device of claim 2105 wherein the agent is an anti-microtubule agent.
2145. The device of claim 2105 wherein the agent is a topoisomerase inhibitor.
2146. The device of claim 2105 wherein the agent is a DNA
cleaving agent.
2147. The device of claim 2105 wherein the agent is an antimetabolite.
2143. The device of claim 2105 wherein the agent inhibits adenosine deaminase.
2149. The device of claim 2105 wherein the agent inhibits purine ring synthesis.
2150. The device of claim 2105 wherein the agent is a nucleotide interconversion inhibitor.
2151. The device of claim 2105 wherein the agent inhibits dihydrofolate reduction.
2152. The device of claim 2105 wherein the agent blocks thymidine monophosphate.
2153. The device of claim 2105 wherein the agent causes DNA
damage.
2154. The device of claim 2105 wherein the agent is a DNA
intercalation agent.
2155. The device of claim 2105 wherein the agent is a RNA
synthesis inhibitor.
2156. The device of claim 2105 wherein the agent is a pyrimidine synthesis inhibitor.
2157. The device of claim 2105 wherein the agent inhibits ribonucleotide synthesis or function.
2158. The device of claim 2105 wherein the agent inhibits thymidine monophosphate synthesis or function.
2159. The device of claim 2105 wherein the agent inhibits DNA
synthesis.
2160. The device of claim 2105 wherein the agent causes DNA
adduct formation.
2161. The device of claim 2105 wherein the agent inhibits protein synthesis.
2162. The device of claim 2105 wherein the agent inhibits microtubule function.
2163. The device of claim 2105 wherein the agent is a cyclin dependent protein kinase inhibitor.
2164. The device of claim 2105 wherein the agent is an epidermal growth factor kinase inhibitor.
2165. The device of claim 2105 wherein the agent is an elastase inhibitor.
2166. The device of claim 2105 wherein the agent is a factor Xa inhibitor.
2167. The device of claim 2105 wherein the agent is a farnesyltransferase inhibitor.
2168. The device of claim 2105 wherein the agent is a fibrinogen antagonist.
2169. The device of claim 2105 wherein the agent is a guanylate cyclase stimulant.
2170. The device of claim 2105 wherein the agent is a heat shock protein 90 antagonist.
2171. The device of claim 2105 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
2172. The device of claim 2105 wherein the agent is a guanylate cyclase stimulant.
2173. The device of claim 2105 wherein the agent is a HMGCoA
reductase inhibitor.
2174. The device of claim 2105 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
2175. The device of claim 2105 wherein the agent is a hydroorotate dehydrogenase inhibitor.
2176. The device of claim 2105 wherein the agent is an IKK2 inhibitor.
2177. The device of claim 2105 wherein the agent is an IL-1 antagonist.
2178. The device of claim 2105 wherein the agent is an ICE
antagonist.
2179. The device of claim 2105 wherein the agent is an IRAK
antagonist.
2180. The device of claim 2105 wherein the agent is an IL-4 agonist.
2181. The device of claim 2105 wherein the agent is an immunomodulatory agent.
2182. The device of claim 2105 wherein the agent is sirolimus or an analogue or derivative thereof.
2183. The device of claim 2105 wherein the agent is not sirolimus.
2184. The device of claim 2105 wherein the agent is everolimus or an analogue or derivative thereof.
2185. The device of claim 2105 wherein the agent is tacrolimus or an analogue or derivative thereof.
2186. The device of claim 2105 wherein the agent is not tacrolimus.
2187. The device of claim 2105 wherein the agent is biolmus or an analogue or derivative thereof.
2188. The device of claim 2105 wherein the agent is tresperimus or an analogue or derivative thereof.
2189. The device of claim 2105 wherein the agent is auranofin or an analogue or derivative thereof.
2190. The device of claim 2105 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
2191. The device of claim 2105 wherein the agent is gusperimus or an analogue or derivative thereof.
2192. The device of claim 2105 wherein the agent is pimecrolimus or an analogue or derivative thereof.
2193. The device of claim 2105 wherein the agent is ABT-578 or an analogue or derivative thereof.
2194. The device of claim 2105 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
2195. The device of claim 2105 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
2196. The device of claim 2105 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
2197. The device of claim 2105 wherein the agent is a leukotriene inhibitor.
2198. The device of claim 2105 wherein the agent is a MCP-1 antagonist.
2199. The device of claim 2105 wherein the agent is a MMP
inhibitor.
2200. The device of claim 2105 wherein the agent is an NF
kappa B inhibitor.
2201. The device of claim 2105 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
2202. The device of claim 2105 wherein the agent is an NO
agonist.
2203. The device of claim 2105 wherein the agent is a p38 MAP
kinase inhibitor.
2204. The device of claim 2105 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
2205. The device of claim 2105 wherein the agent is a phosphodiesterase inhibitor.
2206. The device of claim 2105 wherein the agent is a TGF beta inhibitor.
2207. The device of claim 2105 wherein the agent is a thromboxane A2 antagonist.
2208. The device of claim 2105 wherein the agent is a TNFa antagonist.
2209. The device of claim 2105 wherein the agent is a TACE
inhibitor.
2210. The device of claim 2105 wherein the agent is a tyrosine kinase inhibitor.
2211. The device of claim 2105 wherein the agent is a vitronectin inhibitor.
2212. The device of claim 2105 wherein the agent is a fibroblast growth factor inhibitor.
2213. The device of claim 2105 wherein the agent is a protein kinase inhibitor.
2214. The device of claim 2105 wherein the agent is a PDGF
receptor kinase inhibitor.
2215. The device of claim 2105 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
2216. The device of claim 2105 wherein the agent is a retinoic acid receptor antagonist.
2217. The device of claim 2105 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
2218. The device of claim 2105 wherein the agent is a fibronogin antagonist.
2219. The device of claim 2105 wherein the agent is an antimycotic agent.
2220. The device of claim 2105 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
2221. The device of claim 2105 wherein the agent is a bisphosphonate.
2222. The device of claim 2105 wherein the agent is a phospholipase A1 inhibitor.
2223. The device of claim 2105 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
2224. The device of claim 2105 wherein the agent is a macrolide antibiotic.
2225. The device of claim 2105 wherein the agent is a GPIIb/IIIa receptor antagonist.
2226. The device of claim 2105 wherein the agent is an endothelin receptor antagonist.
2227. The device of claim 2105 wherein the agent is a peroxisome proliferator-activated receptor agonist.
2228. The device of claim 2105 wherein the agent is an estrogen receptor agent.
2229. The device of claim 2105 wherein the agent is a somastostatin analogue.
2230. The device of claim 2105 wherein the agent is a neurokinin 1 antagonist.
2231. The device of claim 2105 wherein the agent is a neurokinin 3 antagonist.
2232. The device of claim 2105 wherein the agent is a VLA-4 antagonist.
2233. The device of claim 2105 wherein the agent is an osteoclast inhibitor.
2234. The device of claim 2105 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
2235. The device of claim 2105 wherein the agent is an angiotensin I converting enzyme inhibitor.
2236. The device of claim 2105 wherein the agent is an angiotensin II antagonist.
2237. The device of claim 2105 wherein the agent is an enkephalinase inhibitor.
2233. The device of claim 2105 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
2239. The device of claim 2105 wherein the agent is a protein kinase C inhibitor.
2240. The device of claim 2105 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
2241. The device of claim 2105 wherein the agent is a CXCR3 inhibitor.
2242. The device of claim 2105 wherein the agent is an Itk inhibitor.
2243. The device of claim 2105 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
2244. The device of claim 2105 wherein the agent is a PPAR
agonist.
2245. The device of claim 2105 wherein the agent is an immunosuppressant.
2246. The device of claim 2105 wherein the agent is an Erb inhibitor.
2247. The device of claim 2105 wherein the agent is an apoptosis agonist.
2248. The device of claim 2105 wherein the agent is a lipocortin agonist.
2249. The device of claim 2105 wherein the agent is a VCAM-1 antagonist.
2250. The device of claim 2105 wherein the agent is a collagen antagonist.
2251. The device of claim 2105 wherein the agent is an alpha 2 integrin antagonist.
2252. The device of claim 2105 wherein the agent is a TNF alpha inhibitor.
2253. The device of claim 2105 wherein the, agent is a nitric oxide inhibitor
2254. The device of claim 2105 wherein the agent is a cathepsin inhibitor.
2255. The device of claim 2105 wherein the agent is not an anti-inflammatory agent.
2256. The device of claim 2105 wherein the agent is not a steroid.
2257. The device of claim 2105 wherein the agent is not a glucocorticosteroid.
2258. The device of claim 2105 wherein the agent is not dexamethasone.
2259. The device of claim 2105 wherein the agent is not an anti-infective agent.
2260. The device of claim 2105 wherein the agent is not an antibiotic.
2261. The device of claim 2105 wherein the agent is not an anti-fungal agent.
2262. The device of claim 2105, further comprising a polymer.
2263. The device of claim 2105, further comprising a polymeric carrier.
2264. The device of claim 2105 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2265. The device of claim 2105 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2266. The device of claim 2105, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2267. The device of claim 2105, further comprising a coating, wherein the coating is disposed on a surface of the device.
2268. The device of claim 2105, further comprising a coating, wherein the coating directly contacts the device.
2269. The device of claim 2105, further comprising a coating, wherein the coating indirectly contacts the device.
2270. The device of claim 2105, further comprising a coating, wherein the coating partially covers the device.
2271. The device of claim 2105, further comprising a coating, wherein the coating completely covers the device.
2272. The device of claim 2105, further comprising a coating, wherein the coating is a uniform coating.
2273. The device of claim 2105, further comprising a coating, wherein the coating is a non-uniform coating.
2274. The device of claim 2105, further comprising a coating, wherein the coating is a discontinuous coating.
2275. The device of claim 2105, further comprising a coating, wherein the coating is a patterned coating.
2276. The device of claim 2105, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
2277. The device of claim 2105, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
2278. The device of claim 2105, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2279. The device of claim 2105, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2280. The device of claim 2105, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2281. The device of claim 2105, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2282. The device of claim 2105, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2283. The device of claim 2105, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2284. The device of claim 2105, further comprising a coating, wherein the coating further comprises a polymer.
2285. The device of claim 2105, further comprising a first coating having a first composition and the second coating having a second composition.
2286. The device of claim 2105, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2287. The device of claim 2105, further comprising a polymer.
2288. The device of claim 2105, further comprising a polymeric carrier.
2289. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2290. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2291. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2292. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2293. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2294. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2295. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2296. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2297. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2298. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2299. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2300. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2301. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2302. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2303. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2304. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2305. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2306. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2307. The device of claim 2105, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2303. The device of claim 2105, further comprising a lubricious coating.
2309. The device of claim 2105 wherein the anti-scarring agent is located within pores or holes of the device.
2310. The device of claim 2105 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2311. The device of claim 2105, further comprising a second pharmaceutically active agent.
2312. The device of claim 2105, further comprising an anti-inflammatory agent.
2313. The device of claim 2105, further comprising an agent that inhibits infection.
2314. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2315. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2316. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2317. The device of claim 2105, further comprising an agent that inhibits,infection, wherein the agent is a fluoropyrimidine.
2318. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2319. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2320. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2321. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2322. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2323. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2324. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2325. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2326. The device of claim 2105, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2327. The device of claim 2105, further comprising an anti-thrombotic agent.
2328. The device of claim 2105, further comprising a visualization agent.
2329. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2330. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2331. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2332. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2333. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2334. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2335. The device of claim 2105, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2336. The device of claim 2105, further comprising an echogenic material.
2337. The device of claim 2105, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2338. The device of claim 2105 wherein the device is sterile.
2339. The device of claim 2105 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2340. The device of claim 2105 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2341. The device of claim 2105 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2342. The device of claim 2105 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2343. The device of claim 2105 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2344. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2345. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2346. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2347. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2348. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2349. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2350. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2351. The device of claim 2105 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2352. The device of claim 2105 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
2353. The device of claim 2105 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
2354. The device of claim 2105 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2355. The device of claim 2105 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2356. The device of claim 2105 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2357. The device of claim 2105 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2358. The device of claim 2105 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2359. The device of claim 2105 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2360. The device of claim 2105 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2361. The device of claim 2105 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2362. The device of claim 2105 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2363. The device of any one of claims 2105-2362 wherein the implant is a ventriculopleural shunt.
2364. The device of any one of claims 2105-2362 wherein the implant is a jugular vein shunt.
2365. The device of any one of claims 2105-2362 wherein the implant is a vena cava (VA) shunt.
2366. The device of any one of claims 2105-2362 wherein the implant is a ventriculoperitoneal shunt (VP shunt).
2367. The device of any one of claims 2105-2362 wherein the implant is a gallbladder shunt.
2368. The device of any one of claims 2105-2362 wherein the implant is a peritoneum shunt.
2369. The device of any one of claims 2105-2362 wherein the implant is an external ventricular drainage (EVD) device.
2370. The device of any one of claims 2105-2362 wherein the implant is an intracranial pressure (ICP) monitoring device.
2371. The device of any one of claims 2105-2362 wherein the implant is a dural patch to prevent epidural fibrosis post-laminectomy.
2372. The device of any one of claims 2105-2362 wherein the implant is a device for continuous subarachnoid infusions.
2373. A device, comprising an intraocular lens (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2374. The device of claim 2373 wherein the agent inhibits cell regeneration.
2375. The device of claim 2373 wherein the agent inhibits angiogenesis.
2376. The device of claim 2373 wherein the agent inhibits fibroblast migration.
2377. The device of claim 2373 wherein the agent inhibits fibroblast proliferation.
2378. The device of claim 2373 wherein the agent inhibits deposition of extracellular matrix.
2379. The device of claim 2373 wherein the agent inhibits tissue remodeling.
2380. The device of claim 2373 wherein the agent is an angiogenesis inhibitor.
2381. The device of claim 2373 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
2382. The device of claim 2373 wherein the agent is a chemokine receptor antagonist.
2383. The device of claim 2373 wherein the agent is a cell cycle inhibitor.
2384. The device of claim 2373 wherein the agent is a taxane.
2385. The device of claim 2373 wherein the agent is an anti-microtubule agent.
2386. The device of claim 2373 wherein the agent is paclitaxel.
2387. The device of claim 2373 wherein the agent is not paclitaxel.
2388. The device of claim 2373 wherein the agent is an analogue or derivative of paclitaxel.
2389. The device of claim 2373 wherein the agent is a vinca alkaloid.
2390. The device of claim 2373 wherein the agent is camptothecin or an analogue or derivative thereof.
2391. The device of claim 2373 wherein the agent is a podophyllotoxin.
2392. The device of claim 2373 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
2393. The device of claim 2373 wherein the agent is an anthracycline.
2394. The device of claim 2373 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
2395. The device of claim 2373 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
2396. The device of claim 2373 wherein the agent is a platinum compound.
2397. The device of claim 2373 wherein the agent is a nitrosourea.
2398. The device of claim 2373 wherein the agent is a nitroimidazole.
2399. The device of claim 2373 wherein the agent is a folic acid antagonist.
2400. The device of claim 2373 wherein the agent is a cytidine analogue.
2401. The device of claim 2373 wherein the agent is a pyrimidine analogue.
2402. The device of claim 2373 wherein the agent is a fluoropyrimidine analogue.
2403. The device of claim 2373 wherein the agent is a purine analogue.
2404. The device of claim 2373 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
2405. The device of claim 2373 wherein the agent is a hydroxyurea.
2406. The device of claim 2373 wherein the agent is a mytomicin or an analogue or derivative thereof.
2407. The device of claim 2373 wherein the agent is an alkyl sulfonate.
2408. The device of claim 2373 wherein the agent is a benzamide or an analogue or derivative thereof.
2409. The device of claim 2373 wherein the agent is a nicotinamide or an analogue or derivative thereof.
2410. The device of claim 2373 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
2411. The device of claim 2373 wherein the agent is a DNA
alkylating agent.
2412. The device of claim 2373 wherein the agent is an anti-microtubule agent.
2413. The device of claim 2373 wherein the agent is a topoisomerase inhibitor.
2414. The device of claim 2373 wherein the agent is a DNA
cleaving agent.
2415. The device of claim 2373 wherein the agent is an antimetabolite.
2416. The device of claim 2373 wherein the agent inhibits adenosine deaminase.
2417. The device of claim 2373 wherein the agent inhibits purine ring synthesis.
2418. The device of claim 2373 wherein the agent is a nucleotide interconversion inhibitor.
2419. The device of claim 2373 wherein the agent inhibits dihydrofolate reduction.
2420. The device of claim 2373 wherein the agent blocks thymidine monophosphate.
2421. The device of claim 2373 wherein the agent causes DNA
damage.
2422. The device of claim 2373 wherein the agent is a DNA
intercalation agent.
2423. The device of claim 2373 wherein the agent is a RNA
synthesis inhibitor.
2424. The device of claim 2373 wherein the agent is a pyrimidine synthesis inhibitor.
2425. The device of claim 2373 wherein the agent inhibits ribonucleotide synthesis or function.
2426. The device of claim 2373 wherein the agent inhibits thymidine monophosphate synthesis or function.
2427. The device of claim 2373 wherein the agent inhibits DNA
synthesis.
2428. The device of claim 2373 wherein the agent causes DNA
adduct formation.
2429. The device of claim 2373 wherein the agent inhibits protein synthesis.
2430. The device of claim 2373 wherein the agent inhibits microtubule function.
2431. The device of claim 2373 wherein the agent is a cyclin dependent protein kinase inhibitor.
2432. The device of claim 2373 wherein the agent is an epidermal growth factor kinase inhibitor.
2433. The device of claim 2373 wherein the agent is an elastase inhibitor.
2434. The device of claim 2373 wherein the agent is a factor Xa inhibitor.
2435. The device of claim 2373 wherein the agent is a farnesyltransferase inhibitor.
2436. The device of claim 2373 wherein the agent is a fibrinogen antagonist.
2437. The device of claim 2373 wherein the agent is a guanylate cyclase stimulant.
2438. The device of claim 2373 wherein the agent is a heat shock protein 90 antagonist.
2439. The device of claim 2373 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
2440. The device of claim 2373 wherein the agent is a guanylate cyclase stimulant.
2441. The device of claim 2373 wherein the agent is a HMGCoA
reductase inhibitor.
2442. The device of claim 2373 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
2443. The device of claim 2373 wherein the agent is a hydroorotate dehydrogenase inhibitor.
2444. The device of claim 2373 wherein the agent is an IKK2 inhibitor.
2445. The device of claim 2373 wherein the agent is an IL-1 antagonist.
2446. The device of claim 2373 wherein the agent is an ICE
antagonist.
2447. The device of claim 2373 wherein the agent is an IRAK
antagonist.
2448. The device of claim 2373 wherein the agent is an IL-4 agonist.
2449. The device of claim 2373 wherein the agent is an immunomodulatory agent.
2450. The device of claim 2373 wherein the agent is sirolimus or an analogue or derivative thereof.
2451. The device of claim 2373 wherein the agent is not sirolimus.
2452. The device of claim 2373 wherein the agent is everolimus or an analogue or derivative thereof.
2453. The device of claim 2373 wherein the agent is tacrolimus or an analogue or derivative thereof.
2454. The device of claim 2373 wherein the agent is not tacrolimus.
2455. The device of claim 2373 wherein the agent is biolmus or an analogue or derivative thereof.
2456. The device of claim 2373 wherein the agent is tresperimus or an analogue or derivative thereof.
2457. The device of claim 2373 wherein the agent is auranofin or an analogue or derivative thereof.
2458. The device of claim 2373 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
2459. The device of claim 2373 wherein the agent is gusperimus or an analogue or derivative thereof.
2460. The device of claim 2373 wherein the agent is pimecrolimus or an analogue or derivative thereof.
2461. The device of claim 2373 wherein the agent is ABT-578 or an analogue or derivative thereof.
2462. The device of claim 2373 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
2463. The device of claim 2373 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
2464. The device of claim 2373 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
2465. The device of claim 2373 wherein the agent is a leukotriene inhibitor.
2466. The device of claim 2373 wherein the agent is a MCP-1 antagonist.
2467. The device of claim 2373 wherein the agent is a MMP
inhibitor.
2468. The device of claim 2373 wherein the agent is an NF
kappa B inhibitor.
2469. The device of claim 2373 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
2470. The device of claim 2373 wherein the agent is an NO
agonist.
2471. The device of claim 2373 wherein the agent is a p38 MAP
kinase inhibitor.
2472. The device of claim 2373 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
2473. The device of claim 2373 wherein the agent is a phosphodiesterase inhibitor.
2474. The device of claim 2373 wherein the agent is a TGF beta inhibitor.
2475. The device of claim 2373 wherein the agent is a thromboxane A2 antagonist.
2476. The device of claim 2373 wherein the agent is a TNFa antagonist.
2477. The device of claim 2373 wherein the agent is a TACE
inhibitor.
2478. The device of claim 2373 wherein the agent is a tyrosine kinase inhibitor.
2479. The device of claim 2373 wherein the agent is a vitronectin inhibitor.
2480. The device of claim 2373 wherein the agent is a fibroblast growth factor inhibitor.
2481. The device of claim 2373 wherein the agent is a protein kinase inhibitor.
2482. The device of claim 2373 wherein the agent is a PDGF
receptor kinase inhibitor.
2483. The device of claim 2373 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
2484. The device of claim 2373 wherein the agent is a retinoic acid receptor antagonist.
2485. The device of claim 2373 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
2486. The device of claim 2373 wherein the agent is a fibronogin antagonist.
2487. The device of claim 2373 wherein the agent is an antimycotic agent.
2488. The device of claim 2373 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
2489. The device of claim 2373 wherein the agent is a bisphosphonate.
2490. The device of claim 2373 wherein the agent is a phospholipase A1 inhibitor.
2491. The device of claim 2373 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
2492. The device of claim 2373 wherein the agent is a macrolide antibiotic.
2493. The device of claim 2373 wherein the agent is a GPIIb/IIIa receptor antagonist.
2494. The device of claim 2373 wherein the agent is an endothelin receptor antagonist.
2495. The device of claim 2373 wherein the agent is a peroxisome proliferator-activated receptor agonist.
2496. The device of claim 2373 wherein the agent is an estrogen receptor agent.
2497. The device of claim 2373 wherein the agent is a somastostatin analogue.
2498. The device of claim 2373 wherein the agent is a neurokinin 1 antagonist.
2499. The device of claim 2373 wherein the agent is a neurokinin 3 antagonist.
2500. The device of claim 2373 wherein the agent is a VLA-4 antagonist.
2501. The device of claim 2373 wherein the agent is an osteoclast inhibitor.
2502. The device of claim 2373 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
2503. The device of claim 2373 wherein the agent is an angiotensin I converting enzyme inhibitor.
2504. The device of claim 2373 wherein the agent is an angiotensin II antagonist.
2505. The device of claim 2373 wherein the agent is an enkephalinase inhibitor.
2506. The device of claim 2373 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
2507. The device of claim 2373 wherein the agent is a protein kinase C inhibitor.
2508. The device of claim 2373 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
2509. The device of claim 2373 wherein the agent is a CXCR3 inhibitor.
2510. The device of claim 2373 wherein the agent is an Itk inhibitor.
2511. The device of claim 2373 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
2512. The device of claim 2373 wherein the agent is a PPAR
agonist.
2513. The device of claim 2373 wherein the agent is an immunosuppressant.
2514. The device of claim 2373 wherein the agent is an Erb inhibitor.
2515. The device of claim 2373 wherein the agent is an apoptosis agonist.
2516. The device of claim 2373 wherein the agent is a lipocortin agonist.
2517. The device of claim 2373 wherein the agent is a VCAM-1 antagonist.
2518. The device of claim 2373 wherein the agent is a collagen antagonist.
2519. The device of claim 2373 wherein the agent is an alpha 2 integrin antagonist.
2520. The device of claim 2373 wherein the agent is a TNF alpha inhibitor.
2521. The device of claim 2373 wherein the agent is a nitric oxide inhibitor
2522. The device of claim 2373 wherein the agent is a cathepsin inhibitor.
2523. The device of claim 2373 wherein the agent is not an anti-inflammatory agent.
2524. The device of claim 2373 wherein the agent is not a steroid.
2525. The device of claim 2373 wherein the agent is not a glucocorticosteroid.
2526. The device of claim 2373 wherein the agent is not dexamethasone.
2527. The device of claim 2373 wherein the agent is not an anti-infective agent.
2528. The device of claim 2373 wherein the agent is not an antibiotic.
2529. The device of claim 2373 wherein the agent is not an anti-fungal agent.
2530. The device of claim 2373, further comprising a polymer.
2531. The device of claim 2373, further comprising a polymeric carrier.
2532. The device of claim 2373 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2533. The device of claim 2373 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2534. The device of claim 2373, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2535. The device of claim 2373, further comprising a coating, wherein the coating is disposed on a surface of the device.
2536. The device of claim 2373, further comprising a coating, wherein the coating directly contacts the device.
2537. The device of claim 2373, further comprising a coating, wherein the coating indirectly contacts the device.
2538. The device of claim 2373, further comprising a coating, wherein the coating partially covers the device.
2539. The device of claim 2373, further comprising a coating, wherein the coating completely covers the device.
2540. The device of claim 2373, further comprising a coating, wherein the coating is a uniform coating.
2541. The device of claim 2373, further comprising a coating, wherein the coating is a non-uniform coating.
2542. The device of claim 2373, further comprising a coating, wherein the coating is a discontinuous coating.
2543. The device of claim 2373, further comprising a coating, wherein the coating is a patterned coating.
2544. The device of claim 2373, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
2545. The device of claim 2373, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
2546. The device of claim 2373, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2547. The device of claim 2373, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2548. The device of claim 2373, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
2549. The device of claim 2373, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2550. The device of claim 2373, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2551. The device of claim 2373, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2552. The device of claim 2373, further comprising a coating, wherein the coating further comprises a polymer.
2553. The device of claim 2373, further comprising a first coating having a first composition and the second coating having a second composition.
2554. The device of claim 2373, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2555. The device of claim 2373, further comprising a polymer.
2556. The device of claim 2373, further comprising a polymeric carrier.
2557. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2558. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2559. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2560. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2561. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2562. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2563. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2564. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2565. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2566. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2567. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2568. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2569. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2570. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2571. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2572. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2573. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2574. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2575. The device of claim 2373, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2576. The device of claim 2373, further comprising a lubricious coating.
2577. The device of claim 2373 wherein the anti-scarring agent is located within pores or holes of the device.
2578. The device of claim 2373 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2579. The device of claim 2373, further comprising a second pharmaceutically active agent.
2580. The device of claim 2373, further comprising an anti-inflammatory agent.
2581. The device of claim 2373, further comprising an agent that inhibits infection.
2582. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2583. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2584. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2585. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2586. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2587. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2588. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2589. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2590. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2591. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2592. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2593. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2594. The device of claim 2373, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2595. The device of claim 2373, further comprising an anti-thrombotic agent.
2596. The device of claim 2373, further comprising a visualization agent.
2597. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2598. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2599. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2600. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2601. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2602. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2603. The device of claim 2373, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2604. The device of claim 2373, further comprising an echogenic material.
2605. The device of claim 2373, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2606. The device of claim 2373 wherein the device is sterile.
2607. The device of claim 2373 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2608. The device of claim 2373 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2609. The device of claim 2373 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2610. The device of claim 2373 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2611. The device of claim 2373 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2612. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2613. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2614. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2615. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2616. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2617. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2618. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2619. The device of claim 2373 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2620. The device of claim 2373 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
2621. The device of claim 2373 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
2622. The device of claim 2373 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2623. The device of claim 2373 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2624. The device of claim 2373 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2625. The device of claim 2373 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2626. The device of claim 2373 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2627. The device of claim 2373 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2628. The device of claim 2373 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2629. The device of claim 2373 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2630. The device of claim 2373 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2631. The device of any one of claims 2373-2630 wherein the implant is an aphakic lens.
2632. The device of any one of claims 2373-2630 wherein the implant is a phakic lens.
2633. The device of any one of claims 2373-2630 wherein the implant is a multi-focal lens.
2634. A device, comprising a glaucoma drainage device (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2635. The device of claim 2634 wherein the agent inhibits cell regeneration.
2636. The device of claim 2634 wherein the agent inhibits angiogenesis.
2637. The device of claim 2634 wherein the agent inhibits fibroblast migration.
2638. The device of claim 2634 wherein the agent inhibits fibroblast proliferation.
2639. The device of claim 2634 wherein the agent inhibits deposition of extracellular matrix.
2640. The device of claim 2634 wherein the agent inhibits tissue remodeling.
2641. The device of claim 2634 wherein the agent is an angiogenesis inhibitor.
2642. The device of claim 2634 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
2643. The device of claim 2634 wherein the agent is a chemokine receptor antagonist.
2644. The device of claim 2634 wherein the agent is a cell cycle inhibitor.
2645. The device of claim 2634 wherein the agent is a taxane.
2646. The device of claim 2634 wherein the agent is an anti-microtubule agent.
2647. The device of claim 2634 wherein the agent is paclitaxel.
2648. The device of claim 2634 wherein the agent is not paclitaxel.
2649. The device of claim 2634 wherein the agent is an analogue or derivative of paclitaxel.
2650. The device of claim 2634 wherein the agent is a vinca alkaloid.
2651. The device of claim 2634 wherein the agent is camptothecin or an analogue or derivative thereof.
2652. The device of claim 2634 wherein the agent is a podophyllotoxin.
2653. The device of claim 2634 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
2654. The device of claim 2634 wherein the agent is an anthracycline.
2655. The device of claim 2634 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
2656. The device of claim 2634 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
2657. The device of claim 2634 wherein the agent is a platinum compound.
2658. The device of claim 2634 wherein the agent is a nitrosourea.
2659. The device of claim 2634 wherein the agent is a nitroimidazole.
2660. The device of claim 2634 wherein the agent is a folic acid antagonist.
2661. The device of claim 2634 wherein the agent is a cytidine analogue.
2662. The device of claim 2634 wherein the agent is a pyrimidine analogue.
2663. The device of claim 2634 wherein the agent is a fluoropyrimidine analogue.
2664. The device of claim 2634 wherein the agent is a purine analogue.
2665. The device of claim 2634 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
2666. The device of claim 2634 wherein the agent is a hydroxyurea.
2667. The device of claim 2634 wherein the agent is a mytomicin or an analogue or derivative thereof.
2668. The device of claim 2634 wherein the agent is an alkyl sulfonate.
2669. The device of claim 2634 wherein the agent is a benzamide or an analogue or derivative thereof.
2670. The device of claim 2634 wherein the agent is a nicotinamide or an analogue or derivative thereof.
2671. The device of claim 2634 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
2672. The device of claim 2634 wherein the agent is a DNA
alkylating agent.
2673. The device of claim 2634 wherein the agent is an anti-microtubule agent.
2674. The device of claim 2634 wherein the agent is a topoisomerase inhibitor.
2675. The device of claim 2634 wherein the agent is a DNA
cleaving agent.
2676. The device of claim 2634 wherein the agent is an antimetabolite.
2677. The device of claim 2634 wherein the agent inhibits adenosine deaminase.
2678. The device of claim 2634 wherein the agent inhibits purine ring synthesis.
2679. The device of claim 2634 wherein the agent is a nucleotide interconversion inhibitor.
2680. The device of claim 2634 wherein the agent inhibits dihydrofolate reduction.
2681. The device of claim 2634 wherein the agent blocks thymidine monophosphate.
2682. The device of claim 2634 wherein the agent causes DNA
damage.
2683. The device of claim 2634 wherein the agent is a DNA
intercalation agent.
2684. The device of claim 2634 wherein the agent is a RNA
synthesis inhibitor.
2685. The device of claim 2634 wherein the agent is a pyrimidine synthesis inhibitor.
2686. The device of claim 2634 wherein the agent inhibits ribonucleotide synthesis or function.
2687. The device of claim 2634 wherein the agent inhibits thymidine monophosphate synthesis or function.
2688. The device of claim 2634 wherein the agent inhibits DNA
synthesis.
2689. The device of claim 2634 wherein the agent causes DNA
adduct formation.
2690. The device of claim 2634 wherein the agent inhibits protein synthesis.
2691. The device of claim 2634 wherein the agent inhibits microtubule function.
2692. The device of claim 2634 wherein the agent is a cyclin dependent protein kinase inhibitor.
2693. The device of claim 2634 wherein the agent is an epidermal growth factor kinase inhibitor.
2694. The device of claim 2634 wherein the agent is an elastase inhibitor.
2695. The device of claim 2634 wherein the agent is a factor Xa inhibitor.
2696. The device of claim 2634 wherein the agent is a farnesyltransferase inhibitor.
2697. The device of claim 2634 wherein the agent is a fibrinogen antagonist.
2698. The device of claim 2634 wherein the agent is a guanylate cyclase stimulant.
2699. The device of claim 2634 wherein the agent is a heat shock protein 90 antagonist.
2700. The device of claim 2634 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
2701. The device of claim 2634 wherein the agent is a guanylate cyclase stimulant.
2702. The device of claim 2634 wherein the agent is a HMGCoA
reductase inhibitor.
2703. The device of claim 2634 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
2704. The device of claim 2634 wherein the agent is a hydroorotate dehydrogenase inhibitor.
2705. The device of claim 2634 wherein the agent is an IKK2 inhibitor.
2706. The device of claim 2634 wherein the agent is an IL-1 antagonist.
2707. The device of claim 2634 wherein the agent is an ICE
antagonist.
2708. The device of claim 2634 wherein the agent is an IRAK
antagonist.
2709. The device of claim 2634 wherein the agent is an IL-4 agonist.
2710. The device of claim 2634 wherein the agent is an immunomodulatory agent.
2711. The device of claim 2634 wherein the agent is sirolimus or an analogue or derivative thereof.
2712. The device of claim 2634 wherein the agent is not sirolimus.
2713. The device of claim 2634 wherein the agent is everolimus or an analogue or derivative thereof.
2714. The device of claim 2634 wherein the agent is tacrolimus or an analogue or derivative thereof.
2715. The device of claim 2634 wherein the agent is not tacrolimus.
2716. The device of claim 2634 wherein the agent is biolmus or an analogue or derivative thereof.
2717. The device of claim 2634 wherein the agent is tresperimus or an analogue or derivative thereof.
2718. The device of claim 2634 wherein the agent is auranofin or an analogue or derivative thereof.
2719. The device of claim 2634 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
2720. The device of claim 2634 wherein the agent is gusperimus or an analogue or derivative thereof.
2721. The device of claim 2634 wherein the agent is pimecrolimus or an analogue or derivative thereof.
2722. The device of claim 2634 wherein the agent is ABT-578 or an analogue or derivative thereof.
2723. The device of claim 2634 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
2724. The device of claim 2634 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
2725. The device of claim 2634 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
2726. The device of claim 2634 wherein the agent is a leukotriene inhibitor.
2727. The device of claim 2634 wherein the agent is a MCP-1 antagonist.
2728. The device of claim 2634 wherein the agent is a MMP
inhibitor.
2729. The device of claim 2634 wherein the agent is an NF
kappa B inhibitor.
2730. The device of claim 2634 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
2731. The device of claim 2634 wherein the agent is an NO
agonist.
2732. The device of claim 2634 wherein the agent is a p38 MAP
kinase inhibitor.
2733. The device of claim 2634 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
2734. The device of claim 2634 wherein the agent is a phosphodiesterase inhibitor.
2735. The device of claim 2634 wherein the agent is a TGF beta inhibitor.
2736. The device of claim 2634 wherein the agent is a thromboxane A2 antagonist.
2737. The device of claim 2634 wherein the agent is a TNFa antagonist.
2738. The device of claim 2634 wherein the agent is a TACE
inhibitor.
2739. The device of claim 2634 wherein the agent is a tyrosine kinase inhibitor.
2740. The device of claim 2634 wherein the agent is a vitronectin inhibitor.
2741. The device of claim 2634 wherein the agent is a fibroblast growth factor inhibitor.
2742. The device of claim 2634 wherein the agent is a protein kinase inhibitor.
2743. The device of claim 2634 wherein the agent is a PDGF
receptor kinase inhibitor.
2744. The device of claim 2634 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
2745. The device of claim 2634 wherein the agent is a retinoic acid receptor antagonist.
2746. The device of claim 2634 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
2747. The device of claim 2634 wherein the agent is a fibronogin antagonist.
2748. The device of claim 2634 wherein the agent is an antimycotic agent.
2749. The device of claim 2634 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
2750. The device of claim 2634 wherein the agent is a bisphosphonate.
2751. The device of claim 2634 wherein the agent is a phospholipase A1 inhibitor.
2752. The device of claim 2634 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
2753. The device of claim 2634 wherein the agent is a macrolide antibiotic.
2754. The device of claim 2634 wherein the agent is a GPIIb/IIIa receptor antagonist.
2755. The device of claim 2634 wherein the agent is an endothelin receptor antagonist.
2756. The device of claim 2634 wherein the agent is a peroxisome proliferator-activated receptor agonist.
2757. The device of claim 2634 wherein the agent is an estrogen receptor agent.
2758. The device of claim 2634 wherein the agent is a somastostatin analogue.
2759. The device of claim 2634 wherein the agent is a neurokinin 1 antagonist.
2760. The device of claim 2634 wherein the agent is a neurokinin 3 antagonist.
2761. The device of claim 2634 wherein the agent is a VLA-4 antagonist.
2762. The device of claim 2634 wherein the agent is an osteoclast inhibitor.
2763. The device of claim 2634 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
2764. The device of claim 2634 wherein the agent is an angiotensin I converting enzyme inhibitor.
2765. The device of claim 2634 wherein the agent is an angiotensin II antagonist.
2766. The device of claim 2634 wherein the agent is an enkephalinase inhibitor.
2767. The device of claim 2634 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
2768. The device of claim 2634 wherein the agent is a protein kinase C inhibitor.
2769. The device of claim 2634 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
2770. The device of claim 2634 wherein the agent is a CXCR3 inhibitor.
2771. The device of claim 2634 wherein the agent is an Itk inhibitor.
2772. The device of claim 2634 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
2773. The device of claim 2634 wherein the agent is a PPAR
agonist.
2774. The device of claim 2634 wherein the agent is an immunosuppressant.
2775. The device of claim 2634 wherein the agent is an Erb inhibitor.
2776. The device of claim 2634 wherein the agent is an apoptosis agonist.
2777. The device of claim 2634 wherein the agent is a lipocortin agonist.
2778. The device of claim 2634 wherein the agent is a VCAM-1 antagonist.
2779. The device of claim 2634 wherein the agent is a collagen antagonist.
2780. The device of claim 2634 wherein the agent is an alpha 2 integrin antagonist.
2781. The device of claim 2634 wherein the agent is a TNF alpha inhibitor.
2782. The device of claim 2634 wherein the agent is a nitric oxide inhibitor
2783. The device of claim 2634 wherein the agent is a cathepsin inhibitor.
2784. The device of claim 2634 wherein the agent is not an anti-inflammatory agent.
2785. The device of claim 2634 wherein the agent is not a steroid.
2786. The device of claim 2634 wherein the agent is not a glucocorticosteroid.
2787. The device of claim 2634 wherein the agent is not dexamethasone.
2788. The device of claim 2634 wherein the agent is not an anti-infective agent.
2789. The device of claim 2634 wherein the agent is not an antibiotic.
2790. The device of claim 2634 wherein the agent is not an anti-fungal agent.
2791. The device of claim 2634, further comprising a polymer.
2792. The device of claim 2634, further comprising a polymeric carver.
2793. The device of claim 2634 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2794. The device of claim 2634 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2795. The device of claim 2634, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2796. The device of claim 2634, further comprising a coating, wherein the coating is disposed on a surface of the device.
2797. The device of claim 2634, further comprising a coating, wherein the coating directly contacts the device.
2798. The device of claim 2634, further comprising a coating, wherein the coating indirectly contacts the device.
2799. The device of claim 2634, further comprising a coating, wherein the coating partially covers the device.
2800. The device of claim 2634, further comprising a coating, wherein the coating completely covers the device.
2801. The device of claim 2634, further comprising a coating, wherein the coating is a uniform coating.
2802. The device of claim 2634, further comprising a coating, wherein the coating is a non-uniform coating.
2803. The device of claim 2634, further comprising a coating, wherein the coating is a discontinuous coating.
2804. The device of claim 2634, further comprising a coating, wherein the coating is a patterned coating.
2805. The device of claim 2634, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
2806. The device of claim 2634, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
2807. The device of claim 2634, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2808. The device of claim 2634, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2809. The device of claim 2634, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
2810. The device of claim 2634, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
2811. The device of claim 2634, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2812. The device of claim 2634, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2813. The device of claim 2634, further comprising a coating, wherein the coating further comprises a polymer.
2814. The device of claim 2634, further comprising a first coating having a first composition and the second coating having a second composition.
2815. The device of claim 2634, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2816. The device of claim 2634, further comprising a polymer.
2817. The device of claim 2634, further comprising a polymeric carrier.
2818. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2819. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2820. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2821. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2822. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2823. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2824. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2825. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2826. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2827. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2828. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2829. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
283. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2831. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2832. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2833. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2834. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2835. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2836. The device of claim 2634, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2837. The device of claim 2634, further comprising a lubricious coating.
2838. The device of claim 2634 wherein the anti-scarring agent is located within pores or holes of the device.
2839. The device of claim 2634 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2840. The device of claim 2634, further comprising a second pharmaceutically active agent.
2841. The device of claim 2634, further comprising an anti-inflammatory agent.
2842. The device of claim 2634, further comprising an agent that inhibits infection.
2843. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2844. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2845. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2846. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2847. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2848. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2849. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2850. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2851. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2852. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2853. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2854. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2855. The device of claim 2634, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2856. The device of claim 2634, further comprising an anti-thrombotic agent.
2857. The device of claim 2634, further comprising a visualization agent.
2858. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2859. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2860. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2861. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2862. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2863. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2864. The device of claim 2634, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2865. The device of claim 2634, further comprising an echogenic material.
2866. The device of claim 2634, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2867. The device of claim 2634 wherein the device is sterile.
2868. The device of claim 2634 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2869. The device of claim 2634 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2870. The device of claim 2634 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2871. The device of claim 2634 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2872. The device of claim 2634 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2873. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2874. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2875. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2876. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2877. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2878. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2879. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2880. The device of claim 2634 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2881. The device of claim 2634 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
2882. The device of claim 2634 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
2883. The device of claim 2634 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2884. The device of claim 2634 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2885. The device of claim 2634 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2886. The device of claim 2634 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2887. The device of claim 2634 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2888. The device of claim 2634 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2889. The device of claim 2634 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2890. The device of claim 2634 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2891. The device of claim 2634 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2892. The device of any one of claims 2634-2891 wherein the implant is an episcleral drainage plate or tube.
2893. A device, comprising a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2894. The device of claim 2893 wherein the agent inhibits cell regeneration.
2895. The device of claim 2893 wherein the agent inhibits angiogenesis.
2896. The device of claim 2893 wherein the agent inhibits fibroblast migration.
2897. The device of claim 2893 wherein the agent inhibits fibroblast proliferation.
2898. The device of claim 2893 wherein the agent inhibits deposition of extracellular matrix.
2899. The device of claim 2893 wherein the agent inhibits tissue remodeling.
2900. The device of claim 2893 wherein the agent is an angiogenesis inhibitor.
2901. The device of claim 2893 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
2902. The device of claim 2893 wherein the agent is a chemokine receptor antagonist.
2903. The device of claim 2893 wherein the agent is a cell cycle inhibitor.
2904. The device of claim 2893 wherein the agent is a taxane.
2905. The device of claim 2893 wherein the agent is an anti-microtubule agent.
2906. The device of claim 2893 wherein the agent is paclitaxel.
2907. The device of claim 2893 wherein the agent is not paclitaxel.
2908. The device of claim 2893 wherein the agent is an analogue or derivative of paclitaxel.
2909. The device of claim 2893 wherein the agent is a vinca alkaloid.
2910. The device of claim 2893 wherein the agent is camptothecin or an analogue or derivative thereof.
2911. The device of claim 2893 wherein the agent is a podophyllotoxin.
2912. The device of claim 2893 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
2913. The device of claim 2893 wherein the agent is an anthracycline.
2914. The device of claim 2893 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
2915. The device of claim 2893 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
2916. The device of claim 2893 wherein the agent is a platinum compound.
2917. The device of claim 2893 wherein the agent is a nitrosourea.
2918. The device of claim 2893 wherein the agent is a nitroimidazole.
2919. The device of claim 2893 wherein the agent is a folic acid antagonist.
2920. The device of claim 2893 wherein the agent is a cytidine analogue.
2921. The device of claim 2893 wherein the agent is a pyrimidine analogue.
2922. The device of claim 2893 wherein the agent is a fluoropyrimidine analogue.
2923. The device of claim 2893 wherein the agent is a purine analogue.
2924. The device of claim 2893 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
2925. The device of claim 2893 wherein the agent is a hydroxyurea.
2926. The device of claim 2893 wherein the agent is a mytomicin or an analogue or derivative thereof.
2927. The device of claim 2893 wherein the agent is an alkyl sulfonate.
2928. The device of claim 2893 wherein the agent is a benzamide or an analogue or derivative thereof.
2929. The device of claim 2893 wherein the agent is a nicotinamide or an analogue or derivative thereof.
2930. The device of claim 2893 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
2931. The device of claim 2893 wherein the agent is a DNA
alkylating agent.
2932. The device of claim 2893 wherein the agent is an anti-microtubule agent.
2933. The device of claim 2893 wherein the agent is a topoisomerase inhibitor.
2934. The device of claim 2893 wherein the agent is a DNA
cleaving agent.
2935. The device of claim 2893 wherein the agent is an antimetabolite.
2936. The device of claim 2893 wherein the agent inhibits adenosine deaminase.
2937. The device of claim 2893 wherein the agent inhibits purine ring synthesis.
2938. The device of claim 2893 wherein the agent is a nucleotide interconversion inhibitor.
2939. The device of claim 2893 wherein the agent inhibits dihydrofolate reduction.
2940. The device of claim 2893 wherein the agent blocks thymidine monophosphate.
2941. The device of claim 2893 wherein the agent causes DNA
damage.
2942. The device of claim 2893 wherein the agent is a DNA
intercalation agent.
2943. The device of claim 2893 wherein the agent is a RNA
synthesis inhibitor.
2944. The device of claim 2893 wherein the agent is a pyrimidine synthesis inhibitor.
2945. The device of claim 2893 wherein the agent inhibits ribonucleotide synthesis or function.
2946. The device of claim 2893 wherein the agent inhibits thymidine monophosphate synthesis or function.
2947. The device of claim 2893 wherein the agent inhibits DNA
synthesis.
2948. The device of claim 2893 wherein the agent causes DNA
adduct formation.
2949. The device of claim 2893 wherein the agent inhibits protein synthesis.
2950. The device of claim 2893 wherein the agent inhibits microtubule function.
2951. The device of claim 2893 wherein the agent is a cyclin dependent protein kinase inhibitor.
2952. The device of claim 2893 wherein the agent is an epidermal growth factor kinase inhibitor.
2953. The device of claim 2893 wherein the agent is an elastase inhibitor.
2954. The device of claim 2893 wherein the agent is a factor Xa inhibitor.
2955. The device of claim 2893 wherein the agent is a farnesyltransferase inhibitor.
2956. The device of claim 2893 wherein the agent is a fibrinogen antagonist.
2957. The device of claim 2893 wherein the agent is a guanylate cyclase stimulant.
2958. The device of claim 2893 wherein the agent is a heat shock protein 90 antagonist.
2959. The device of claim 2893 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
2960. The device of claim 2893 wherein the agent is a guanylate cyclase stimulant.
2961. The device of claim 2893 wherein the agent is a HMGCoA
reductase inhibitor.
2962. The device of claim 2893 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
2963. The device of claim 2893 wherein the agent is a hydroorotate dehydrogenase inhibitor.
2964. The device of claim 2893 wherein the agent is an IKK2 inhibitor.
2965. The device of claim 2893 wherein the agent is an IL-1 antagonist.
2966. The device of claim 2893 wherein the agent is an ICE
antagonist.
2967. The device of claim 2893 wherein the agent is an IRAK
antagonist.
2968. The device of claim 2893 wherein the agent is an IL-4 agonist.
2969. The device of claim 2893 wherein the agent is an immunomodulatory agent.
2970. The device of claim 2893 wherein the agent is sirolimus or an analogue or derivative thereof.
2971. The device of claim 2893 wherein the agent is not sirolimus.
2972. The device of claim 2893 wherein the agent is everolimus or an analogue or derivative thereof.
2973. The device of claim 2893 wherein the agent is tacrolimus or an analogue or derivative thereof.
2974. The device of claim 2893 wherein the agent is not tacrolimus.
2975. The device of claim 2893 wherein the agent is biolmus or an analogue or derivative thereof.
2976. The device of claim 2893 wherein the agent is tresperimus or an analogue or derivative thereof.
2977. The device of claim 2893 wherein the agent is auranofin or an analogue or derivative thereof.
2978. The device of claim 2893 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
2979. The device of claim 2893 wherein the agent is gusperimus or an analogue or derivative thereof.
2980. The device of claim 2893 wherein the agent is pimecrolimus or an analogue or derivative thereof.
2981. The device of claim 2893 wherein the agent is ABT-578 or an analogue or derivative thereof.
2982. The device of claim 2893 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
2983. The device of claim 2893 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
2984. The device of claim 2893 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
2985. The device of claim 2893 wherein the agent is a leukotriene inhibitor.
2986. The device of claim 2893 wherein the agent is a MCP-1 antagonist.
2987. The device of claim 2893 wherein the agent is a MMP
inhibitor.
2988. The device of claim 2893 wherein the agent is an NF
kappa B inhibitor.
2989. The device of claim 2893 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
2990. The device of claim 2893 wherein the agent is an NO
agonist.
2991. The device of claim 2893 wherein the agent is a p38 MAP
kinase inhibitor.
2992. The device of claim 2893 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
2993. The device of claim 2893 wherein the agent is a phosphodiesterase inhibitor.
2994. The device of claim 2893 wherein the agent is a TGF beta inhibitor.
2995. The device of claim 2893 wherein the agent is a thromboxane A2 antagonist.
2996. The device of claim 2893 wherein the agent is a TNFa antagonist.
2997. The device of claim 2893 wherein the agent is a TACE
inhibitor.
2998. The device of claim 2893 wherein the agent is a tyrosine kinase inhibitor.
2999. The device of claim 2893 wherein the agent is a vitronectin inhibitor.
3000. The device of claim 2893 wherein the agent is a fibroblast growth factor inhibitor.
3001. The device of claim 2893 wherein the agent is a protein kinase inhibitor.
3002. The device of claim 2893 wherein the agent is a PDGF
receptor kinase inhibitor.
3003. The device of claim 2893 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
3004. The device of claim 2893 wherein the agent is a retinoic acid receptor antagonist.
3005. The device of claim 2893 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
3006. The device of claim 2893 wherein the agent is a fibronogin antagonist.
3007. The device of claim 2893 wherein the agent is an antimycotic agent.
3008. The device of claim 2893 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
3009. The device of claim 2893 wherein the agent is a bisphosphonate.
3010. The device of claim 2893 wherein the agent is a phospholipase A1 inhibitor.
3011. The device of claim 2893 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
3012. The device of claim 2893 wherein the agent is a macrolide antibiotic.
3013. The device of claim 2893 wherein the agent is a GPIIb/IIIa receptor antagonist.
3014. The device of claim 2893 wherein the agent is an endothelin receptor antagonist.
3015. The device of claim 2893 wherein the agent is a peroxisome proliferator-activated receptor agonist.
3016. The device of claim 2893 wherein the agent is an estrogen receptor agent.
3017. The device of claim 2893 wherein the agent is a somastostatin analogue.
3018. The device of claim 2893 wherein the agent is a neurokinin 1 antagonist.
3019. The device of claim 2893 wherein the agent is a neurokinin 3 antagonist.
3020. The device of claim 2893 wherein the agent is a VLA-4 antagonist.
3021. The device of claim 2893 wherein the agent is an osteoclast inhibitor.
3022. The device of claim 2893 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
3023. The device of claim 2893 wherein the agent is an angiotensin I converting enzyme inhibitor.
3024. The device of claim 2893 wherein the agent is an angiotensin II antagonist.
3025. The device of claim 2893 wherein the agent is an enkephalinase inhibitor.
3026. The device of claim 2893 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
3027. The device of claim 2893 wherein the agent is a protein kinase C inhibitor.
3028. The device of claim 2893 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
3029. The device of claim 2893 wherein the agent is a CXCR3 inhibitor.
3030. The device of claim 2893 wherein the agent is an Itk inhibitor.
3031. The device of claim 2893 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
3032. The device of claim 2893 wherein the agent is a PPAR
agonist.
3033. The device of claim 2893 wherein the agent is an immunosuppressant.
3034. The device of claim 2893 wherein the agent is an Erb inhibitor.
3035. The device of claim 2893 wherein the agent is an apoptosis agonist.
3036. The device of claim 2893 wherein the agent is a lipocortin agonist.
3037. The device of claim 2893 wherein the agent is a VCAM-1 antagonist.
3038. The device of claim 2893 wherein the agent is a collagen antagonist.
3039. The device of claim 2893 wherein the agent is an alpha 2 integrin antagonist.
3040. The device of claim 2893 wherein the agent is a TNF alpha inhibitor.
3041. The device of claim 2893 wherein the agent is a nitric oxide inhibitor
3042. The device of claim 2893 wherein the agent is a cathepsin inhibitor.
3043. The device of claim 2893 wherein the agent is not an anti-inflammatory agent.
3044. The device of claim 2893 wherein the agent is not a steroid.
3045. The device of claim 2893 wherein the agent is not a glucocorticosteroid.
3046. The device of claim 2893 wherein the agent is not dexamethasone.
3047. The device of claim 2893 wherein the agent is not an anti-infective agent.
3048. The device of claim 2893 wherein the agent is not an antibiotic.
3049. The device of claim 2893 wherein the agent is not an anti-fungal agent.
3050. The device of claim 2893, further comprising a polymer.
3051. The device of claim 2893, further comprising a polymeric carrier.
3052. The device of claim 2893 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3053. The device of claim 2893 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3054. The device of claim 2893, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3055. The device of claim 2893, further comprising a coating, wherein the coating is disposed on a surface of the device.
3056. The device of claim 2893, further comprising a coating, wherein the coating directly contacts the device.
3057. The device of claim 2893, further comprising a coating, wherein the coating indirectly contacts the device.
3058. The device of claim 2893, further comprising a coating, wherein the coating partially covers the device.
3059. The device of claim 2893, further comprising a coating, wherein the coating completely covers the device.
3060. The device of claim 2893, further comprising a coating, wherein the coating is a uniform coating.
3061. The device of claim 2893, further comprising a coating, wherein the coating is a non-uniform coating.
3062. The device of claim 2893, further comprising a coating, wherein the coating is a discontinuous coating.
3063. The device of claim 2893, further comprising a coating, wherein the coating is a patterned coating.
3064. The device of claim 2893, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
3065. The device of claim 2893, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
3066. The device of claim 2893, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3067. The device of claim 2893, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3068. The device of claim 2893, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3069. The device of claim 2893, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3070. The device of claim 2893, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3071. The device of claim 2893, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3072. The device of claim 2893, further comprising a coating, wherein the coating further comprises a polymer.
3073. The device of claim 2893, further comprising a first coating having a first composition and the second coating having a second composition.
3074. The device of claim 2893, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3075. The device of claim 2893, further comprising a polymer.
3076. The device of claim 2893, further comprising a polymeric carrier.
3077. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3078. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3079. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3080. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3081. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3082. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
3083. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3084. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3085. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3086. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3087. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3088. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3089. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3090. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3091. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3092. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
3093. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3094. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
3095. The device of claim 2893, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3096. The device of claim 2893, further comprising a lubricious coating.
3097. The device of claim 2893 wherein the anti-scarring agent is located within pores or holes of the device.
3098. The device of claim 2893 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3099. The device of claim 2893, further comprising a second pharmaceutically active agent.
3100. The device of claim 2893, further comprising an anti-inflammatory agent.
3101. The device of claim 2893, further comprising an agent that inhibits infection.
3102. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3103. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3104. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3105. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3106. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3107. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3108. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3109. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3110. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3111. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3112. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3113. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3114. The device of claim 2893, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3115. The device of claim 2893, further comprising an anti-thrombotic agent.
3116. The device of claim 2893, further comprising a visualization agent.
3117. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3118. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3119. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3120. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3121. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3122. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3123. The device of claim 2893, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3124. The device of claim 2893, further comprising an echogenic material.
3125. The device of claim 2893, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3126. The device of claim 2893 wherein the device is sterile.
3127. The device of claim 2893 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3128. The device of claim 2893 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
3129. The device of claim 2893 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3130. The device of claim 2893 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3131. The device of claim 2893 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3132. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3133. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3134. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3135. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3136. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
3137. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3138. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3139. The device of claim 2893 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3140. The device of claim 2893 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
3141. The device of claim 2893 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
3142. The device of claim 2893 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3143. The device of claim 2893 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3144. The device of claim 2893 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3145. The device of claim 2893 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3146. The device of claim 2893 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3147. The device of claim 2893 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3148. The device of claim 2893 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3149. The device of claim 2893 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3150. The device of claim 2893 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3151. The device of any one of claims 2893-3150 wherein the implant is a flexible rod or coil.
3152. The device of any one of claims 2893-3150 wherein the implant comprises an inflatable tube and a pump.
3153. The device of any one of claims 2893-3150 wherein the implant comprises a pressure chamber.
3154. A device, comprising an endotracheal tube (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3155. The device of claim 3154 wherein the agent inhibits cell regeneration.
3156. The device of claim 3154 wherein the agent inhibits angiogenesis.
3157. The device of claim 3154 wherein the agent inhibits fibroblast migration.
3158. The device of claim 3154 wherein the agent inhibits fibroblast proliferation.
3159. The device of claim 3154 wherein the agent inhibits deposition of extracellular matrix.
3160. The device of claim 3154 wherein the agent inhibits tissue remodeling.
3161. The device of claim 3154 wherein the agent is an angiogenesis inhibitor.
3162. The device of claim 3154 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
3163. The device of claim 3154 wherein the agent is a chemokine receptor antagonist.
3164. The device of claim 3154 wherein the agent is a cell cycle inhibitor.
3165. The device of claim 3154 wherein the agent is a taxane.
3166. The device of claim 3154 wherein the agent is an anti-microtubule agent.
3167. The device of claim 3154 wherein the agent is paclitaxel.
3168. The device of claim 3154 wherein the agent is not paclitaxel.
3169. The device of claim 3154 wherein the agent is an analogue or derivative of paclitaxel.
3170. The device of claim 3154 wherein the agent is a vinca alkaloid.
3171. The device of claim 3154 wherein the agent is camptothecin or an analogue or derivative thereof.
3172. The device of claim 3154 wherein the agent is a podophyllotoxin.
3173. The device of claim 3154 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
3174. The device of claim 3154 wherein the agent is an anthracycline.
3175. The device of claim 3154 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
3176. The device of claim 3154 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
3177. The device of claim 3154 wherein the agent is a platinum compound.
3178. The device of claim 3154 wherein the agent is a nitrosourea.
3179. The device of claim 3154 wherein the agent is a nitroimidazole.
3180. The device of claim 3154 wherein the agent is a folic acid antagonist.
3181. The device of claim 3154 wherein the agent is a cytidine analogue.
3182. The device of claim 3154 wherein the agent is a pyrimidine analogue.
3183. The device of claim 3154 wherein the agent is a fluoropyrimidine analogue.
3184. The device of claim 3154 wherein the agent is a purine analogue.
3185. The device of claim 3154 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
3186. The device of claim 3154 wherein the agent is a hydroxyurea.
3187. The device of claim 3154 wherein the agent is a mytomicin or an analogue or derivative thereof.
3188. The device of claim 3154 wherein the agent is an alkyl sulfonate.
3189. The device of claim 3154 wherein the agent is a benzamide or an analogue or derivative thereof.
3190. The device of claim 3154 wherein the agent is a nicotinamide or an analogue or derivative thereof.
3191. The device of claim 3154 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
3192. The device of claim 3154 wherein the agent is a DNA
alkylating agent.
3193. The device of claim 3154 wherein the agent is an anti-microtubule agent.
3194. The device of claim 3154 wherein the agent is a topoisomerase inhibitor.
3195. The device of claim 3154 wherein the agent is a DNA
cleaving agent.
3196. The device of claim 3154 wherein the agent is an antimetabolite.
3197. The device of claim 3154 wherein the agent inhibits adenosine deaminase.
3198. The device of claim 3154 wherein the agent inhibits purine ring synthesis.
3199. The device of claim 3154 wherein the agent is a nucleotide interconversion inhibitor.
3200. The device of claim 3154 wherein the agent inhibits dihydrofolate reduction.
3201. The device of claim 3154 wherein the agent blocks thymidine monophosphate.
3202. The device of claim 3154 wherein the agent causes DNA
damage.
3203. The device of claim 3154 wherein the agent is a DNA
intercalation agent.
3204. The device of claim 3154 wherein the agent is a RNA
synthesis inhibitor.
3205. The device of claim 3154 wherein the agent is a pyrimidine synthesis inhibitor.
3206. The device of claim 3154 wherein the agent inhibits ribonucleotide synthesis or function.
3207. The device of claim 3154 wherein the agent inhibits thymidine monophosphate synthesis or function.
3208. The device of claim 3154 wherein the agent inhibits DNA
synthesis.
3209. The device of claim 3154 wherein the agent causes DNA
adduct formation.
3210. The device of claim 3154 wherein the agent inhibits protein synthesis.
3211. The device of claim 3154 wherein the agent inhibits microtubule function.
3212. The device of claim 3154 wherein the agent is a cyclin dependent protein kinase inhibitor.
3213. The device of claim 3154 wherein the agent is an epidermal growth factor kinase inhibitor.
3214. The device of claim 3154 wherein the agent is an elastase inhibitor.
3215. The device of claim 3154 wherein the agent is a factor Xa inhibitor.
3216. The device of claim 3154 wherein the agent is a farnesyltransferase inhibitor.
3217. The device of claim 3154 wherein the agent is a fibrinogen antagonist.
3218. The device of claim 3154 wherein the agent is a guanylate cyclase stimulant.
3219. The device of claim 3154 wherein the agent is a heat shock protein 90 antagonist.
3220. The device of claim 3154 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
3221. The device of claim 3154 wherein the agent is a guanylate cyclase stimulant.
3222. The device of claim 3154 wherein the agent is a HMGCoA
reductase inhibitor.
3223. The device of claim 3154 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
3224. The device of claim 3154 wherein the agent is a hydroorotate dehydrogenase inhibitor.
3225. The device of claim 3154 wherein the agent is an IKK2 inhibitor.
3226. The device of claim 3154 wherein the agent is an IL-1 antagonist.
3227. The device of claim 3154 wherein the agent is an ICE
antagonist.
3228. The device of claim 3154 wherein the agent is an IRAK
antagonist.
3229. The device of claim 3154 wherein the agent is an IL-4 agonist.
3230. The device of claim 3154 wherein the agent is an immunomodulatory agent.
3231. The device of claim 3154 wherein the agent is sirolimus or an analogue or derivative thereof.
3232. The device of claim 3154 wherein the agent is not sirolimus.
3233. The device of claim 3154 wherein the agent is everolimus or an analogue or derivative thereof.
3234. The device of claim 3154 wherein the agent is tacrolimus or an analogue or derivative thereof.
3235. The device of claim 3154 wherein the agent is not tacrolimus.
3236. The device of claim 3154 wherein the agent is biolmus or an analogue or derivative thereof.
3237. The device of claim 3154 wherein the agent is tresperimus or an analogue or derivative thereof.
3238. The device of claim 3154 wherein the agent is auranofin or an analogue or derivative thereof.
3239. The device of claim 3154 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
3240. The device of claim 3154 wherein the agent is gusperimus or an analogue or derivative thereof.
3241. The device of claim 3154 wherein the agent is pimecrolimus or an analogue or derivative thereof.
3242. The device of claim 3154 wherein the agent is ABT-578 or an analogue or derivative thereof.
3243. The device of claim 3154 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
3244. The device of claim 3154 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
3245. The device of claim 3154 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
3246. The device of claim 3154 wherein the agent is a leukotriene inhibitor.
3247. The device of claim 3154 wherein the agent is a MCP-1 antagonist.
3248. The device of claim 3154 wherein the agent is a MMP
inhibitor.
3249. The device of claim 3154 wherein the agent is an NF
kappa B inhibitor.
3250. The device of claim 3154 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
3251. The device of claim 3154 wherein the agent is an NO
agonist.
3252. The device of claim 3154 wherein the agent is a p38 MAP
kinase inhibitor.
3253. The device of claim 3154 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
3254. The device of claim 3154 wherein the agent is a phosphodiesterase inhibitor.
3255. The device of claim 3154 wherein the agent is a TGF beta inhibitor.
3256. The device of claim 3154 wherein the agent is a thromboxane A2 antagonist.
3257. The device of claim 3154 wherein the agent is a TNFa antagonist.
3258. The device of claim 3154 wherein the agent is a TACE
inhibitor.
3259. The device of claim 3154 wherein the agent is a tyrosine kinase inhibitor.
3260. The device of claim 3154 wherein the agent is a vitronectin inhibitor.
3261. The device of claim 3154 wherein the agent is a fibroblast growth factor inhibitor.
3262. The device of claim 3154 wherein the agent is a protein kinase inhibitor.
3263. The device of claim 3154 wherein the agent is a PDGF
receptor kinase inhibitor.
3264. The device of claim 3154 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
3265. The device of claim 3154 wherein the agent is a retinoic acid receptor antagonist.
3266. The device of claim 3154 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
3267. The device of claim 3154 wherein the agent is a fibronogin antagonist.
3268. The device of claim 3154 wherein the agent is an antimycotic agent.
3269. The device of claim 3154 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
3270. The device of claim 3154 wherein the agent is a bisphosphonate.
3271. The device of claim 3154 wherein the agent is a phospholipase A1 inhibitor.
3272. The device of claim 3154 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
3273. The device of claim 3154 wherein the agent is a macrolide antibiotic.
3274. The device of claim 3154 wherein the agent is a GPIIb/IIIa receptor antagonist.
3275. The device of claim 3154 wherein the agent is an endothelin receptor antagonist.
3276. The device of claim 3154 wherein the agent is a peroxisome proliferator-activated receptor agonist.
3277. The device of claim 3154 wherein the agent is an estrogen receptor agent.
3278. The device of claim 3154 wherein the agent is a somastostatin analogue.
3279. The device of claim 3154 wherein the agent is a neurokinin 1 antagonist.
3280. The device of claim 3154 wherein the agent is a neurokinin 3 antagonist.
3281. The device of claim 3154 wherein the agent is a VLA-4 antagonist.
3282. The device of claim 3154 wherein the agent is an osteoclast inhibitor.
3283. The device of claim 3154 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
3284. The device of claim 3154 wherein the agent is an angiotensin I converting enzyme inhibitor.
3285. The device of claim 3154 wherein the agent is an angiotensin II antagonist.
3286. The device of claim 3154 wherein the agent is an enkephalinase inhibitor.
3287. The device of claim 3154 wherein the agent is a peroxisome profiferator-activated receptor gamma agonist insulin sensitizer.
3288. The device of claim 3154 wherein the agent is a protein kinase C inhibitor.
3289. The device of claim 3154 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
3290. The device of claim 3154 wherein the agent is a CXCR3 inhibitor.
3291. The device of claim 3154 wherein the agent is an Itk inhibitor.
3292. The device of claim 3154 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
3293. The device of claim 3154 wherein the agent is a PPAR
agonist.
3294. The device of claim 3154 wherein the agent is an immunosuppressant.
3295. The device of claim 3154 wherein the agent is an Erb inhibitor.
3296. The device of claim 3154 wherein the agent is an apoptosis agonist.
3297. The device of claim 3154 wherein the agent is a lipocortin agonist.
3298. The device of claim 3154 wherein the agent is a VCAM-1 antagonist.
3299. The device of claim 3154 wherein the agent is a collagen antagonist.
3300. The device of claim 3154 wherein the agent is an alpha 2 integrin antagonist.
3301. The device of claim 3154 wherein the agent is a TNF alpha inhibitor.
3302. The device of claim 3154 wherein the agent is a nitric oxide inhibitor
3303. The device of claim 3154 wherein the agent is a cathepsin inhibitor.
3304. The device of claim 3154 wherein the agent is not an anti-inflammatory agent.
3305. The device of claim 3154 wherein the agent is not a steroid.
3306. The device of claim 3154 wherein the agent is not a glucocorticosteroid.
3307. The device of claim 3154 wherein the agent is not dexamethasone.
3308. The device of claim 3154 wherein the agent is not an anti-infective agent.
3309. The device of claim 3154 wherein the agent is not an antibiotic.
3310. The device of claim 3154 wherein the agent is not an anti-fungal agent.
3311. The device of claim 3154, further comprising a polymer.
3312. The device of claim 3154, further comprising a polymeric carrier.
3313. The device of claim 3154 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3314. The device of claim 3154 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3315. The device of claim 3154, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3316. The device of claim 3154, further comprising a coating, wherein the coating is disposed on a surface of the device.
3317. The device of claim 3154, further comprising a coating, wherein the coating directly contacts the device.
3318. The device of claim 3154, further comprising a coating, wherein the coating indirectly contacts the device.
3319. The device of claim 3154, further comprising a coating, wherein the coating partially covers the device.
3320. The device of claim 3154, further comprising a coating, wherein the coating completely covers the device.
3321. The device of claim 3154, further comprising a coating, wherein the coating is a uniform coating.
3322. The device of claim 3154, further comprising a coating, wherein the coating is a non-uniform coating.
3323. The device of claim 3154, further comprising a coating, wherein the coating is a discontinuous coating.
3324. The device of claim 3154, further comprising a coating, wherein the coating is a patterned coating.
3325. The device of claim 3154, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
3326. The device of claim 3154, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
3327. The device of claim 3154, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3328. The device of claim 3154, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3329. The device of claim 3154, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3330. The device of claim 3154, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3331. The device of claim 3154, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3332. The device of claim 3154, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3333. The device of claim 3154, further comprising a coating, wherein the coating further comprises a polymer.
3334. The device of claim 3154, further comprising a first coating having a first composition and the second coating having a second composition.
3335. The device of claim 3154, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3336. The device of claim 3154, further comprising a polymer.
3337. The device of claim 3154, further comprising a polymeric carrier.
3338. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3339. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3340. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3341. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3342. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3343. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
3344. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3345. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3346. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3347. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3348. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3349. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3350. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3351. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3352. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3353. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
3354. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3355. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polyethylene glycol) polymer.
3356. The device of claim 3154, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3357. The device of claim 3154, further comprising a lubricious coating.
3358. The device of claim 3154 wherein the anti-scarring agent is located within pores or holes of the device.
3359. The device of claim 3154 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3360. The device of claim 3154, further comprising a second pharmaceutically active agent.
3361. The device of claim 3154, further comprising an anti-inflammatory agent.
3362. The device of claim 3154, further comprising an agent that inhibits infection.
3363. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3364. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3365. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3366. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3367. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3368. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3369. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3370. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3371. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3372. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3373. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3374. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3375. The device of claim 3154, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3376. The device of claim 3154, further comprising an anti-thrombotic agent.
3377. The device of claim 3154, further comprising a visualization agent.
3378. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3379. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3380. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3381. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3382. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3383. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3384. The device of claim 3154, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3385. The device of claim 3154, further comprising an echogenic material.
3386. The device of claim 3154, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3387. The device of claim 3154 wherein the device is sterile.
3388. The device of claim 3154 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3389. The device of claim 3154 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
3390. The device of claim 3154 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3391. The device of claim 3154 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3392. The device of claim 3154 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3393. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3394. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3395. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3396. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3397. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
3398. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3399. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3400. The device of claim 3154 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3401. The device of claim 3154 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
3402. The device of claim 3154 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
3403. The device of claim 3154 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3404. The device of claim 3154 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3405. The device of claim 3154 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3406. The device of claim 3154 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3407. The device of claim 3154 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3408. The device of claim 3154 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3409. The device of claim 3154 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3410. The device of claim 3154 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3411. The device of claim 3154 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3412. A device, comprising a tracheostomy tube (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3413. The device of claim 3412 wherein the agent inhibits cell regeneration.
3414. The device of claim 3412 wherein the agent inhibits angiogenesis.
3415. The device of claim 3412 wherein the agent inhibits fibroblast migration.
3416. The device of claim 3412 wherein the agent inhibits fibroblast proliferation.
3417. The device of claim 3412 wherein the agent inhibits deposition of extracellular matrix.
3413. The device of claim 3412 wherein the agent inhibits tissue remodeling.
3419. The device of claim 3412 wherein the agent is an angiogenesis inhibitor.
3420. The device of claim 3412 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
3421. The device of claim 3412 wherein the agent is a chemokine receptor antagonist.
3422. The device of claim 3412 wherein the agent is a cell cycle inhibitor.
3423. The device of claim 3412 wherein the agent is a taxane.
3424. The device of claim 3412 wherein the agent is an anti-microtubule agent.
3425. The device of claim 3412 wherein the agent is paclitaxel.
3426. The device of claim 3412 wherein the agent is not paclitaxel.
3427. The device of claim 3412 wherein the agent is an analogue or derivative of paclitaxel.
3428. The device of claim 3412 wherein the agent is a vinca alkaloid.
3429. The device of claim 3412 wherein the agent is camptothecin or an analogue or derivative thereof.
3430. The device of claim 3412 wherein the agent is a podophyllotoxin.
3431. The device of claim 3412 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
3432. The device of claim 3412 wherein the agent is an anthracycline.
3433. The device of claim 3412 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
3434. The device of claim 3412 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
3435. The device of claim 3412 wherein the agent is a platinum compound.
3436. The device of claim 3412 wherein the agent is a nitrosourea.
3437. The device of claim 3412 wherein the agent is a nitroimidazole.
3438. The device of claim 3412 wherein the agent is a folic acid antagonist.
3439. The device of claim 3412 wherein the agent is a cytidine analogue.
3440. The device of claim 3412 wherein the agent is a pyrimidine analogue.
3441. The device of claim 3412 wherein the agent is a fluoropyrimidine analogue.
3442. The device of claim 3412 wherein the agent is a purine analogue.
3443. The device of claim 3412 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
3444. The device of claim 3412 wherein the agent is a hydroxyurea.
3445. The device of claim 3412 wherein the agent is a mytomicin or an analogue or derivative thereof.
3446. The device of claim 3412 wherein the agent is an alkyl sulfonate.
3447. The device of claim 3412 wherein the agent is a benzamide or an analogue or derivative thereof.
3448. The device of claim 3412 wherein the agent is a nicotinamide or an analogue or derivative thereof.
3449. The device of claim 3412 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
3450. The device of claim 3412 wherein the agent is a DNA
alkylating agent.
3451. The device of claim 3412 wherein the agent is an anti-microtubule agent.
3452. The device of claim 3412 wherein the agent is a topoisomerase inhibitor.
3453. The device of claim 3412 wherein the agent is a DNA
cleaving agent.
3454. The device of claim 3412 wherein the agent is an antimetabolite.
3455. The device of claim 3412 wherein the agent inhibits adenosine deaminase.
3456. The device of claim 3412 wherein the agent inhibits purine ring synthesis.
3457. The device of claim 3412 wherein the agent is a nucleotide interconversion inhibitor.
3458. The device of claim 3412 wherein the agent inhibits dihydrofolate reduction.
3459. The device of claim 3412 wherein the agent blocks thymidine monophosphate.
3460. The device of claim 3412 wherein the agent causes DNA
damage.
3461. The device of claim 3412 wherein the agent is a DNA
intercalation agent.
3462. The device of claim 3412 wherein the agent is a RNA
synthesis inhibitor.
3463. The device of claim 3412 wherein the agent is a pyrimidine synthesis inhibitor.
3464. The device of claim 3412 wherein the agent inhibits ribonucleotide synthesis or function.
3465. The device of claim 3412 wherein the agent inhibits thymidine monophosphate synthesis or function.
3466. The device of claim 3412 wherein the agent inhibits DNA
synthesis.
3467. The device of claim 3412 wherein the agent causes DNA
adduct formation.
3468. The device of claim 3412 wherein the agent inhibits protein synthesis.
3469. The device of claim 3412 wherein the agent inhibits microtubule function.
3470. The device of claim 3412 wherein the agent is a cyclin dependent protein kinase inhibitor.
3471. The device of claim 3412 wherein the agent is an epidermal growth factor kinase inhibitor.
3472. The device of claim 3412 wherein the agent is an elastase inhibitor.
3473. The device of claim 3412 wherein the agent is a factor Xa inhibitor.
3474. The device of claim 3412 wherein the agent is a farnesyltransferase inhibitor.
3475. The device of claim 3412 wherein the agent is a fibrinogen antagonist.
3476. The device of claim 3412 wherein the agent is a guanylate cyclase stimulant.
3477. The device of claim 3412 wherein the agent is a heat shock protein 90 antagonist.
3478. The device of claim 3412 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
3479. The device of claim 3412 wherein the agent is a guanylate cyclase stimulant.
3480. The device of claim 3412 wherein the agent is a HMGCoA
reductase inhibitor.
3481. The device of claim 3412 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
3482. The device of claim 3412 wherein the agent is a hydroorotate dehydrogenase inhibitor.
3483. The device of claim 3412 wherein the agent is an IKK2 inhibitor.
3484. The device of claim 3412 wherein the agent is an IL-1 antagonist.
3485. The device of claim 3412 wherein the agent is an ICE
antagonist.
3486. The device of claim 3412 wherein the agent is an IRAK
antagonist.
3487. The device of claim 3412 wherein the agent is an IL-4 agonist.
3488. The device of claim 3412 wherein the agent is an immunomodulatory agent.
3489. The device of claim 3412 wherein the agent is sirolimus or an analogue or derivative thereof.
3490. The device of claim 3412 wherein the agent is not sirolimus.
3491. The device of claim 3412 wherein the agent is everolimus or an analogue or derivative thereof.
3492. The device of claim 3412 wherein the agent is tacrolimus or an analogue or derivative thereof.
3493. The device of claim 3412 wherein the agent is not tacrolimus.
3494. The device of claim 3412 wherein the agent is biolmus or an analogue or derivative thereof.
3495. The device of claim 3412 wherein the agent is tresperimus or an analogue or derivative thereof.
3496. The device of claim 3412 wherein the agent is auranofin or an analogue or derivative thereof.
3497. The device of claim 3412 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
3498. The device of claim 3412 wherein the agent is gusperimus or an analogue or derivative thereof.
3499. The device of claim 3412 wherein the agent is pimecrolimus or an analogue or derivative thereof.
3500. The device of claim 3412 wherein the agent is ABT-578 or an analogue or derivative thereof.
3501. The device of claim 3412 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
3502. The device of claim 3412 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
3503. The device of claim 3412 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
3504. The device of claim 3412 wherein the agent is a leukotriene inhibitor.
3505. The device of claim 3412 wherein the agent is a MCP-1 antagonist.
3506. The device of claim 3412 wherein the agent is a MMP
inhibitor.
3507. The device of claim 3412 wherein the agent is an NF
kappa B inhibitor.
3508. The device of claim 3412 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
3509. The device of claim 3412 wherein the agent is an NO
agonist.
3510. The device of claim 3412 wherein the agent is a p38 MAP
kinase inhibitor.
3511. The device of claim 3412 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
3512. The device of claim 3412 wherein the agent is a phosphodiesterase inhibitor.
3513. The device of claim 3412 wherein the agent is a TGF beta inhibitor.
3514. The device of claim 3412 wherein the agent is a thromboxane A2 antagonist.
3515. The device of claim 3412 wherein the agent is a TNFa antagonist.
3516. The device of claim 3412 wherein the agent is a TACE
inhibitor.
3517. The device of claim 3412 wherein the agent is a tyrosine kinase inhibitor.
3518. The device of claim 3412 wherein the agent is a vitronectin inhibitor.
3519. The device of claim 3412 wherein the agent is a fibroblast growth factor inhibitor.
3520. The device of claim 3412 wherein the agent is a protein kinase inhibitor.
3521. The device of claim 3412 wherein the agent is a PDGF
receptor kinase inhibitor.
3522. The device of claim 3412 wherein the agent is an endothelial growth factor receptor kinase inhibitor,
3523. The device of claim 3412 wherein the agent is a retinoic acid receptor antagonist.
3524. The device of claim 3412 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
3525. The device of claim 3412 wherein the agent is a fibronogin antagonist.
3526. The device of claim 3412 wherein the agent is an antimycotic agent.
3527. The device of claim 3412 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
3528. The device of claim 3412 wherein the agent is a bisphosphonate.
3529. The device of claim 3412 wherein the agent is a phospholipase A1 inhibitor.
3530. The device of claim 3412 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
3531. The device of claim 3412 wherein the agent is a macrolide antibiotic.
3532. The device of claim 3412 wherein the agent is a GPIIb/IIIa receptor antagonist.
3533. The device of claim 3412 wherein the agent is an endothelin receptor antagonist.
3534. The device of claim 3412 wherein the agent is a peroxisome proliferator-activated receptor agonist.
3535. The device of claim 3412 wherein the agent is an estrogen receptor agent.
3536. The device of claim 3412 wherein the agent is a somastostatin analogue.
3537. The device of claim 3412 wherein the agent is a neurokinin 1 antagonist.
3538. The device of claim 3412 wherein the agent is a neurokinin 3 antagonist.
3539. The device of claim 3412 wherein the agent is a VLA-4 antagonist.
3540. The device of claim 3412 wherein the agent is an osteoclast inhibitor.
3541. The device of claim 3412 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
3542. The device of claim 3412 wherein the agent is an angiotensin I converting enzyme inhibitor.
3543. The device of claim 3412 wherein the agent is an angiotensin 1l antagonist.
3544. The device of claim 3412 wherein the agent is an enkephalinase inhibitor.
3545. The device of claim 3412 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
3546. The device of claim 3412 wherein the agent is a protein kinase C inhibitor.
3547. The device of claim 3412 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
3548. The device of claim 3412 wherein the agent is a CXCR3 inhibitor.
3549. The device of claim 3412 wherein the agent is an Itk inhibitor.
3550. The device of claim 3412 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
3551. The device of claim 3412 wherein the agent is a PPAR
agonist.
3552. The device of claim 3412 wherein the agent is an immunosuppressant.
3553. The device of claim 3412 wherein the agent is an Erb inhibitor.
3554. The device of claim 3412 wherein the agent is an apoptosis agonist.
3555. The device of claim 3412 wherein the agent is a lipocortin agonist.
3556. The device of claim 3412 wherein the agent is a VCAM-1 antagonist.
3557. The device of claim 3412 wherein the agent is a collagen antagonist.
3558. The device of claim 3412 wherein the agent is an alpha 2 integrin antagonist.
3559. The device of claim 3412 wherein the agent is a TNF alpha inhibitor.
3560. The device of claim 3412 wherein the agent is a nitric oxide inhibitor
3561. The device of claim 3412 wherein the agent is a cathepsin inhibitor.
3562. The device of claim 3412 wherein the agent is not an anti-inflammatory agent.
3563. The device of claim 3412 wherein the agent is not a steroid.
3564. The device of claim 3412 wherein the agent is not a glucocorticosteroid.
3565. The device of claim 3412 wherein the agent is not dexamethasone.
3566. The device of claim 3412 wherein the agent is not an anti-infective agent.
3567. The device of claim 3412 wherein the agent is not an antibiotic.
3568. The device of claim 3412 wherein the agent is not an anti-fungal agent.
3569. The device of claim 3412, further comprising a polymer.
3570. The device of claim 3412, further comprising a polymeric carrier.
3571. .The device of claim 3412 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3572. The device of claim 3412 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3573. The device of claim 3412, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3574. The device of claim 3412, further comprising a coating, wherein the coating is disposed on a surface of the device.
3575. The device of claim 3412, further comprising a coating, wherein the coating directly contacts the device.
3576. The device of claim 3412, further comprising a coating, wherein the coating indirectly contacts the device.
3577. The device of claim 3412, further comprising a coating, wherein the coating partially covers the device.
3578. The device of claim 3412, further comprising a coating, wherein the coating completely covers the device.
3579. The device of claim 3412, further comprising a coating, wherein the coating is a uniform coating.
3580. The device of claim 3412, further comprising a coating, wherein the coating is a non-uniform coating.
3581. The device of claim 3412, further comprising a coating, wherein the coating is a discontinuous coating.
3582. The device of claim 3412, further comprising a coating, wherein the coating is a patterned coating.
3583. The device of claim 3412, further comprising a coating, wherein the coating has a thickness of 100 ~,m or less.
3584. The device of claim 3412, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
3585. The device of claim 3412, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3586. The device of claim 3412, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3587. The device of claim 3412, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3588. The device of claim 3412, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3589. The device of claim 3412, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3590. The device of claim 3412, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3591. The device of claim 3412, further comprising a coating, wherein the coating further comprises a polymer.
3592. The device of claim 3412, further comprising a first coating having a first composition and the second coating having a second composition.
3593. The device of claim 3412, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3594. The device of claim 3412, further comprising a polymer.
3595. The device of claim 3412, further comprising a polymeric carrier.
3596. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3597. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3598. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3599. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3600. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3601. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
3602. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3603. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3604. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3605. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3606. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3607. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3608. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3609. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3610. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3611. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
3612. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3613. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polyethylene glycol) polymer.
3614. The device of claim 3412, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3615. The device of claim 3412, further comprising a lubricious coating.
3616. The device of claim 3412 wherein the anti-scarring agent is located within pores or holes of the device.
3617. The device of claim 3412 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3618. The device of claim 3412, further comprising a second pharmaceutically active agent.
3619. The device of claim 3412, further comprising an anti-inflammatory agent.
3620. The device of claim 3412, further comprising an agent that inhibits infection.
3621. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3622. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3623. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3624. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3625. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3626. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3627. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3628. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3629. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3630. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3631. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3632. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3633. The device of claim 3412, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3634. The device of claim 3412, further comprising an anti-thrombotic agent.
3635. The device of claim 3412, further comprising a visualization agent.
3636. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3637. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3638. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3639. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3640. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3641. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3642. The device of claim 3412, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3643. The device of claim 3412, further comprising an echogenic material.
3644. The device of claim 3412, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3645. The device of claim 3412 wherein the device is sterile.
3646. The device of claim 3412 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3647. The device of claim 3412 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
3648. The device of claim 3412 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3649. The device of claim 3412 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3650. The device of claim 3412 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3651. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3652. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3653. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3654. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3655. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
3656. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3657. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3658. The device of claim 3412 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3659. The device of claim 3412 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
3660. The device of claim 3412 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
3661. The device of claim 3412 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3662. The device of claim 3412 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3663. The device of claim 3412 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3664. The device of claim 3412 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3665. The device of claim 3412 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3666. The device of claim 3412 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3667. The device of claim 3412 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3668. The device of claim 3412 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3669. The device of claim 3412 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3670. A device, comprising a gastrointestinal device (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3671. The device of claim 3670 wherein the agent inhibits cell regeneration.
3672. The device of claim 3670 wherein the agent inhibits angiogenesis.
3673. The device of claim 3670 wherein the agent inhibits fibroblast migration.
3674. The device of claim 3670 wherein the agent inhibits fibroblast proliferation.
3675. The device of claim 3670 wherein the agent inhibits deposition of extracellular matrix.
3676. The device of claim 3670 wherein the agent inhibits tissue remodeling.
3677. The device of claim 3670 wherein the agent is an angiogenesis inhibitor.
3678. The device of claim 3670 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
3679. The device of claim 3670 wherein the agent is a chemokine receptor antagonist.
3680. The device of claim 3670 wherein the agent is a cell cycle inhibitor.
3681. The device of claim 3670 wherein the agent is a taxane.
3682. The device of claim 3670 wherein the agent is an anti-microtubule agent.
3683. The device of claim 3670 wherein the agent is paclitaxel.
3684. The device of claim 3670 wherein the agent is not paclitaxel.
3685. The device of claim 3670 wherein the agent is an analogue or derivative of paclitaxel.
3686. The device of claim 3670 wherein the agent is a vinca alkaloid.
3687. The device of claim 3670 wherein the agent is camptothecin or an analogue or derivative thereof.
3688. The device of claim 3670 wherein the agent is a podophyllotoxin.
3689. The device of claim 3670 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
3690. The device of claim 3670 wherein the agent is an anthracycline.
3691. The device of claim 3670 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
3692. The device of claim 3670 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
3693. The device of claim 3670 wherein the agent is a platinum compound.
3694. The device of claim 3670 wherein the agent is a nitrosourea.
3695. The device of claim 3670 wherein the agent is a nitroimidazole.
3696. The device of claim 3670 wherein the agent is a folic acid antagonist.
3697. The device of claim 3670 wherein the agent is a cytidine analogue.
3698. The device of claim 3670 wherein the agent is a pyrimidine analogue.
3699. The device of claim 3670 wherein the agent is a fluoropyrimidine analogue.
3700. The device of claim 3670 wherein the agent is a purine analogue.
3701. The device of claim 3670 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
3702. The device of claim 3670 wherein the agent is a hydroxyurea.
3703. The device of claim 3670 wherein the agent is a mytomicin or an analogue or derivative thereof.
3704. The device of claim 3670 wherein the agent is an alkyl sulfonate.
3705. The device of claim 3670 wherein the agent is a benzamide or an analogue or derivative thereof.
3706. The device of claim 3670 wherein the agent is a nicotinamide or an analogue or derivative thereof.
3707. The device of claim 3670 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
3708. The device of claim 3670 wherein the agent is a DNA
alkylating agent.
3709. The device of claim 3670 wherein the agent is an anti-microtubule agent.
3710. The device of claim 3670 wherein the agent is a topoisomerase inhibitor.
3711. The device of claim 3670 wherein the agent is a DNA
cleaving agent.
3712. The device of claim 3670 wherein the agent is an antimetabolite.
3713. The device of claim 3670 wherein the agent inhibits adenosine deaminase.
3714. The device of claim 3670 wherein the agent inhibits purine ring synthesis.
3715. The device of claim 3670 wherein the agent is a nucleotide interconversion inhibitor.
3716. The device of claim 3670 wherein the agent inhibits dihydrofolate reduction.
3717. The device of claim 3670 wherein the agent blocks thymidine monophosphate.
3713. The device of claim 3670 wherein the agent causes DNA
damage.
3719. The device of claim 3670 wherein the agent is a DNA
intercalation agent.
3720. The device of claim 3670 wherein the agent is a RNA
synthesis inhibitor.
3721. The device of claim 3670 wherein the agent is a pyrimidine synthesis inhibitor.
3722. The device of claim 3670 wherein the agent inhibits ribonucleotide synthesis or function.
3723. The device of claim 3670 wherein the agent inhibits thymidine monophosphate synthesis or function.
3724. The device of claim 3670 wherein the agent inhibits DNA
synthesis.
3725. The device of claim 3670 wherein the agent causes DNA
adduct formation.
3726. The device of claim 3670 wherein the agent inhibits protein synthesis.
3727. The device of claim 3670 wherein the agent inhibits microtubule function.
3728. The device of claim 3670 wherein the agent is a cyclin dependent protein kinase inhibitor.
3729. The device of claim 3670 wherein the agent is an epidermal growth factor kinase inhibitor.
3730. The device of claim 3670 wherein the agent is an elastase inhibitor.
3731. The device of claim 3670 wherein the agent is a factor Xa inhibitor.
3732. The device of claim 3670 wherein the agent is a farnesyltransferase inhibitor.
3733. The device of claim 3670 wherein the agent is a fibrinogen antagonist.
3734. The device of claim 3670 wherein the agent is a guanylate cyclase stimulant.
3735. The device of claim 3670 wherein the agent is a heat shock protein 90 antagonist.
3736. The device of claim 3670 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
3737. The device of claim 3670 wherein the agent is a guanylate cyclase stimulant.
3738. The device of claim 3670 wherein the agent is a HMGCoA
reductase inhibitor.
3739. The device of claim 3670 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
3740. The device of claim 3670 wherein the agent is a hydroorotate dehydrogenase inhibitor.
3741. The device of claim 3670 wherein the agent is an IKK2 inhibitor.
3742. The device of claim 3670 wherein the agent is an IL-1 antagonist.
3743. The device of claim 3670 wherein the agent is an ICE
antagonist.
3744. The device of claim 3670 wherein the agent is an IRAK
antagonist.
3745. The device of claim 3670 wherein the agent is an IL-4 agonist.
3746. The device of claim 3670 wherein the agent is an immunomodulatory agent.
3747. The device of claim 3670 wherein the agent is sirolimus or an analogue or derivative thereof.
3748. The device of claim 3670 wherein the agent is not sirolimus.
3749. The device of claim 3670 wherein the agent is everolimus or an analogue or derivative thereof.
3750. The device of claim 3670 wherein the agent is tacrolimus or an analogue or derivative thereof.
3751. The device of claim 3670 wherein the agent is not tacrolimus.
3752. The device of claim 3670 wherein the agent is biolmus or an analogue or derivative thereof.
3753. The device of claim 3670 wherein the agent is tresperimus or an analogue or derivative thereof.
3754. The device of claim 3670 wherein the agent is auranofin or an analogue or derivative thereof.
3755. The device of claim 3670 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
3756. The device of claim 3670 wherein the agent is gusperimus or an analogue or derivative thereof.
3757. The device of claim 3670 wherein the agent is pimecrolimus or an analogue or derivative thereof.
3758. The device of claim 3670 wherein the agent is ACT-578 or an analogue or derivative thereof.
3759. The device of claim 3670 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
3760. The device of claim 3670 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
3761. The device of claim 3670 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
3762. The device of claim 3670 wherein the agent is a leukotriene inhibitor.
3763. The device of claim 3670 wherein the agent is a MCP-1 antagonist.
3764. The device of claim 3670 wherein the agent is a MMP
inhibitor.
3765. The device of claim 3670 wherein the agent is an NF
kappa B inhibitor.
3766. The device of claim 3670 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
3767. The device of claim 3670 wherein the agent is an NO
agonist.
3768. The device of claim 3670 wherein the agent is a p38 MAP
kinase inhibitor.
3769. The device of claim 3670 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
3770. The device of claim 3670 wherein the agent is a phosphodiesterase inhibitor.
3771. The device of claim 3670 wherein the agent is a TGF beta inhibitor.
3772. The device of claim 3670 wherein the agent is a thromboxane A2 antagonist.
3773. The device of claim 3670 wherein the agent is a TNFa antagonist.
3774. The device of claim 3670 wherein the agent is a TACE
inhibitor.
3775. The device of claim 3670 wherein the agent is a tyrosine kinase inhibitor.
3776. The device of claim 3670 wherein the agent is a vitronectin inhibitor.
3777. The device of claim 3670 wherein the agent is a fibroblast growth factor inhibitor.
3778. The device of claim 3670 wherein the agent is a protein kinase inhibitor.
3779. The device of claim 3670 wherein the agent is a PDGF
receptor kinase inhibitor.
3780. The device of claim 3670 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
3781. The device of claim 3670 wherein the agent is a retinoic acid receptor antagonist.
3782. The device of claim 3670 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
3783. The device of claim 3670 wherein the agent is a fibronogin antagonist.
3784. The device of claim 3670 wherein the agent is an antimycotic agent.
3785. The device of claim 3670 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
3786. The device of claim 3670 wherein the agent is a bisphosphonate.
3787. The device of claim 3670 wherein the agent is a phospholipase A1 inhibitor.
3788. The device of claim 3670 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
3789. The device of claim 3670 wherein the agent is a macrolide antibiotic.
3790. The device of claim 3670 wherein the agent is a GPIIb/IIIa receptor antagonist.
3791. The device of claim 3670 wherein the agent is an endothelin receptor antagonist.
3792. The device of claim 3670 wherein the agent is a peroxisome proliferator-activated receptor agonist.
3793. The device of claim 3670 wherein the agent is an estrogen receptor agent.
3794. The device of claim 3670 wherein the agent is a somastostatin analogue.
3795. The device of claim 3670 wherein the agent is a neurokinin 1 antagonist.
3796. The device of claim 3670 wherein the agent is a neurokinin 3 antagonist.
3797. The device of claim 3670 wherein the agent is a VLA-4 antagonist.
3798. The device of claim 3670 wherein the agent is an osteoclast inhibitor.
3799. The device of claim 3670 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
3800. The device of claim 3670 wherein the agent is an angiotensin I converting enzyme inhibitor.
3801. The device of claim 3670 wherein the agent is an angiotensin II antagonist.
3802. The device of claim 3670 wherein the agent is an enkephalinase inhibitor.
3803. The device of claim 3670 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
3804. The device of claim 3670 wherein the agent is a protein kinase C inhibitor.
3805. The device of claim 3670 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
3806. The device of claim 3670 wherein the agent is a CXCR3 inhibitor.
3807. The device of claim 3670 wherein the agent is an Itk inhibitor.
3808. The device of claim 3670 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
3809. The device of claim 3670 wherein the agent is a PPAR
agonist.
3810. The device of claim 3670 wherein the agent is an immunosuppressant.
3811. The device of claim 3670 wherein the agent is an Erb inhibitor.
3812. The device of claim 3670 wherein the agent is an apoptosis agonist.
3813. The device of claim 3670 wherein the agent is a lipocortin agonist.
3814. The device of claim 3670 wherein the agent is a VCAM-1 antagonist.
3815. The device of claim 3670 wherein the agent is a collagen antagonist.
3816. The device of claim 3670 wherein the agent is an alpha 2 integrin antagonist.
3817. The device of claim 3670 wherein the agent is a TNF alpha inhibitor.
3818. The device of claim 3670 wherein the agent is a nitric oxide inhibitor
3819. The device of claim 3670 wherein the agent is a cathepsin inhibitor.
3820. The device of claim 3670 wherein the agent is not an anti-inflammatory agent.
3821. The device of claim 3670 wherein the agent is not a steroid.
3822. The device of claim 3670 wherein the agent is not a glucocorticosteroid.
3823. The device of claim 3670 wherein the agent is not dexamethasone.
3824. The device of claim 3670 wherein the agent is not an anti-infective agent.
3825. The device of claim 3670 wherein the agent is not an antibiotic.
3826. The device of claim 3670 wherein the agent is not an anti-fungal agent.
3827. The device of claim 3670, further comprising a polymer.
3828. The device of claim 3670, further comprising a polymeric carrier.
3829. The device of claim 3670 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3830. The device of claim 3670 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3831. The device of claim 3670, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3832. The device of claim 3670, further comprising a coating, wherein the coating is disposed on a surface of the device.
3833. The device of claim 3670, further comprising a coating, wherein the coating directly contacts the device.
3834. The device of claim 3670, further comprising a coating, wherein the coating indirectly contacts the device.
3835. The device of claim 3670, further comprising a coating, wherein the coating partially covers the device.
3836. The device of claim 3670, further comprising a coating, wherein the coating completely covers the device.
3837. The device of claim 3670, further comprising a coating, wherein the coating is a uniform coating.
3838. The device of claim 3670, further comprising a coating, wherein the coating is a non-uniform coating.
3839. The device of claim 3670, further comprising a coating, wherein the coating is a discontinuous coating.
3840. The device of claim 3670, further comprising a coating, wherein the coating is a patterned coating.
3841. The device of claim 3670, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
3842. The device of claim 3670, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
3843. The device of claim 3670, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3844. The device of claim 3670, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3845. The device of claim 3670, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
3846. The device of claim 3670, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
3847. The device of claim 3670, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3848. The device of claim 3670, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3849. The device of claim 3670, further comprising a coating, wherein the coating further comprises a polymer.
3850. The device of claim 3670, further comprising a first coating having a first composition and the second coating having a second composition.
3851. The device of claim 3670, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3852. The device of claim 3670, further comprising a polymer.
3853. The device of claim 3670, further comprising a polymeric carrier.
3854. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3855. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3856. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3857. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3858. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3859. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
3860. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3861. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3862. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3863. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3864. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3865. The device of claim 3670 further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3866. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3867. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3868. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3869. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
3870. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3871. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
3872. The device of claim 3670, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3873. The device of claim 3670, further comprising a lubricious coating.
3874. The device of claim 3670 wherein the anti-scarring agent is located within pores or holes of the device.
3875. The device of claim 3670 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3876. The device of claim 3670, further comprising a second pharmaceutically active agent.
3877. The device of claim 3670, further comprising an anti-inflammatory agent.
3878. The device of claim 3670, further comprising an agent that inhibits infection.
3879. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3880. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3881. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3882. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3883. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3884. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3885. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3886. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3887. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3888. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3889. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3890. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3891. The device of claim 3670, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3892. The device of claim 3670, further comprising an anti-thrombotic agent.
3893. The device of claim 3670, further comprising a visualization agent.
3894. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3895. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3896. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3897. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3898. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3899. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3900. The device of claim 3670, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3901. The device of claim 3670, further comprising an echogenic material.
3902. The device of claim 3670, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3903. The device of claim 3670 wherein the device is sterile.
3904. The device of claim 3670 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3905. The device of claim 3670 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
3906. The device of claim 3670 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3907. The device of claim 3670 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3908. The device of claim 3670 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3909. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3910. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3911. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3912. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3913. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
3914. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3915. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3916. The device of claim 3670 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3917. The device of claim 3670 wherein the device comprises about 0.01 µ to about 10 µg of the anti-scarring agent.
3918. The device of claim 3670 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
3919. The device of claim 3670 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3920. The device of claim 3670 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3921. The device of claim 3670 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3922. The device of claim 3670 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3923. The device of claim 3670 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3924. The device of claim 3670 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3925. The device of claim 3670 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3926. The device of claim 3670 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3927. The device of claim 3670 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3928. The device of any one of claims 3670-3927 wherein the implant is a GI tube for drainage.
3929. The device of any one of claims 3670-3927 wherein the implant is a GI tube for feeding.
3930. The device of any one of claims 3670-3927 wherein the implant is a portosystemic shunt.
3931. The device of any one of claims 3670-3927 wherein the implant is a shunt for ascites.
3932. The device of any one of claims 3670-3927 wherein the implant is a nasogastric tube.
3933. The device of any one of claims 3670-3927 wherein the implant is a nasoenteral tube.
3934. The device of any one of claims 3670-3927 wherein the implant is a gastrostomy feeding tube.
3935. The device of any one of claims 3670-3927 wherein the implant is a percutaneous feeding tube.
3936. The device of any one of claims 3670-3927 wherein the implant is a colostomy device.
3937. The device of any one of claims 3670-3927 wherein the implant is a biliary T-tube.
3938. The device of any one of claims 3670-3927 wherein the implant is a biliary stone removal device.
3939. The device of any one of claims 3670-3927 wherein the implant is a dilation balloon.
3940. The device of any one of claims 3670-3927 wherein the implant is a dilation catheter.
3941. The device of any one of claims 3670-3927 wherein the implant is an enteral feeding device.
3942. The device of any one of claims 3670-3927 wherein the implant is an esophageal stent.
3943. The device of any one of claims 3670-3927 wherein the implant is a biliary stent.
3944. The device of any one of claims 3670-3927 wherein the implant is a pancreatic stent.
3945. A device, comprising a spinal implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3946. The device of claim 3945 wherein the agent inhibits cell regeneration.
3947. The device of claim 3945 wherein the agent inhibits angiogenesis.
3948. The device of claim 3945 wherein the agent inhibits fibroblast migration.
3949. The device of claim 3945 wherein the agent inhibits fibroblast proliferation.
3950. The device of claim 3945 wherein the agent inhibits deposition of extracellular matrix.
3951. The device of claim 3945 wherein the agent inhibits tissue remodeling.
3952. The device of claim 3945 wherein the agent is an angiogenesis inhibitor.
3953. The device of claim 3945 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
3954. The device of claim 3945 wherein the agent is a chemokine receptor antagonist.
3955. The device of claim 3945 wherein the agent is a cell cycle inhibitor.
3956. The device of claim 3945 wherein the agent is a taxane.
3957. The device of claim 3945 wherein the agent is an anti-microtubule agent.
3958. The device of claim 3945 wherein the agent is paclitaxel.
3959. The device of claim 3945 wherein the agent is not paclitaxel.
3960. The device of claim 3945 wherein the agent is an analogue or derivative of paclitaxel.
3961. The device of claim 3945 wherein the agent is a vinca alkaloid.
3962. The device of claim 3945 wherein the agent is camptothecin or an analogue or derivative thereof.
3963. The device of claim 3945 wherein the agent is a podophyllotoxin.
3964. The device of claim 3945 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
3965. The device of claim 3945 wherein the agent is an anthracycline.
3966. The device of claim 3945 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
3967. The device of claim 3945 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
3968. The device of claim 3945 wherein the agent is a platinum compound.
3969. The device of claim 3945 wherein the agent is a nitrosourea.
3970. The device of claim 3945 wherein the agent is a nitroimidazole.
3971. The device of claim 3945 wherein the agent is a folic acid antagonist.
3972. The device of claim 3945 wherein the agent is a cytidine analogue.
3973. The device of claim 3945 wherein the agent is a pyrimidine analogue.
3974. The device of claim 3945 wherein the agent is a fluoropyrimidine analogue.
3975. The device of claim 3945 wherein the agent is a purine analogue.
3976. The device of claim 3945 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
3977. The device of claim 3945 wherein the agent is a hydroxyurea.
3978. The device of claim 3945 wherein the agent is a mytomicin or an analogue or derivative thereof.
3979. The device of claim 3945 wherein the agent is an alkyl sulfonate.
3980. The device of claim 3945 wherein the agent is a benzamide or an analogue or derivative thereof.
3981. The device of claim 3945 wherein the agent is a nicotinamide or an analogue or derivative thereof.
3982. The device of claim 3945 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
3983. The device of claim 3945 wherein the agent is a DNA
alkylating agent.
3984. The device of claim 3945 wherein the agent is an anti-microtubule agent.
3985. The device of claim 3945 wherein the agent is a topoisomerase inhibitor.
3986. The device of claim 3945 wherein the agent is a DNA
cleaving agent.
3987. The device of claim 3945 wherein the agent is an antimetabolite.
3988. The device of claim 3945 wherein the agent inhibits adenosine deaminase.
3989. The device of claim 3945 wherein the agent inhibits purine ring synthesis.
3990. The device of claim 3945 wherein the agent is a nucleotide interconversion inhibitor.
3991. The device of claim 3945 wherein the agent inhibits dihydrofolate reduction.
3992. The device of claim 3945 wherein the agent blocks thymidine monophosphate.
3993. The device of claim 3945 wherein the agent causes DNA
damage.
3994. The device of claim 3945 wherein the agent is a DNA
intercalation agent.
3995. The device of claim 3945 wherein the agent is a RNA
synthesis inhibitor.
3996. The device of claim 3945 wherein the agent is a pyrimidine synthesis inhibitor.
3997. The device of claim 3945 wherein the agent inhibits ribonucleotide synthesis or function.
3998. The device of claim 3945 wherein the agent inhibits thymidine monophosphate synthesis or function.
3999. The device of claim 3945 wherein the agent inhibits DNA
synthesis.
4000. The device of claim 3945 wherein the agent causes DNA
adduct formation.
4001. The device of claim 3945 wherein the agent inhibits protein synthesis.
4002. The device of claim 3945 wherein the agent inhibits microtubule function.
4003. The device of claim 3945 wherein the agent is a cyclin dependent protein kinase inhibitor.
4004. The device of claim 3945 wherein the agent is an epidermal growth factor kinase inhibitor.
4005. The device of claim 3945 wherein the agent is an elastase inhibitor.
4006. The device of claim 3945 wherein the agent is a factor Xa inhibitor.
4007. The device of claim 3945 wherein the agent is a farnesyltransferase inhibitor.
4008. The device of claim 3945 wherein the agent is a fibrinogen antagonist.
4009. The device of claim 3945 wherein the agent is a guanylate cyclase stimulant.
4010. The device of claim 3945 wherein the agent is a heat shock protein 90 antagonist.
4011. The device of claim 3945 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
4012. The device of claim 3945 wherein the agent is a guanylate cyclase stimulant.
4013. The device of claim 3945 wherein the agent is a HMGCoA
reductase inhibitor.
4014. The device of claim 3945 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
4015. The device of claim 3945 wherein the agent is a hydroorotate dehydrogenase inhibitor.
4016. The device of claim 3945 wherein the agent is an IKK2 inhibitor.
4017. The device of claim 3945 wherein the agent is an IL-1 antagonist.
4018. The device of claim 3945 wherein the agent is an ICE
antagonist.
4019. The device of claim 3945 wherein the agent is an IRAK
antagonist.
4020. The device of claim 3945 wherein the agent is an IL-4 agonist.
4021. The device of claim 3945 wherein the agent is an immunomodulatory agent.
4022. The device of claim 3945 wherein the agent is sirolimus or an analogue or derivative thereof.
4023. The device of claim 3945 wherein the agent is not sirolimus.
4024. The device of claim 3945 wherein the agent is everolimus or an analogue or derivative thereof.
4025. The device of claim 3945 wherein the agent is tacrolimus or an analogue or derivative thereof.
4026. The device of claim 3945 wherein the agent is not tacrolimus.
4027. The device of claim 3945 wherein the agent is biolmus or an analogue or derivative thereof.
4028. The device of claim 3945 wherein the agent is tresperimus or an analogue or derivative thereof.
4029. The device of claim 3945 wherein the agent is auranofin or an analogue or derivative thereof.
4030. The device of claim 3945 wherein the agent is 27-0-demethyirapamycin or an analogue or derivative thereof.
4031. The device of claim 3945 wherein the agent is gusperimus or an analogue or derivative thereof.
4032. The device of claim 3945 wherein the agent is pimecrolimus or an analogue or derivative thereof.
4033. The device of claim 3945 wherein the agent is ABT-578 or an analogue or derivative thereof.
4034. The device of claim 3945 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
4035. The device of claim 3945 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
4036. The device of claim 3945 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
4037. The device of claim 3945 wherein the agent is a leukotriene inhibitor.
4038. The device of claim 3945 wherein the agent is a MCP-1 antagonist.
4039. The device of claim 3945 wherein the agent is a MMP
inhibitor.
4040. The device of claim 3945 wherein the agent is an NF
kappa B inhibitor.
4041. The device of claim 3945 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
4042. The device of claim 3945 wherein the agent is an NO
agonist.
4043. The device of claim 3945 wherein the agent is a p38 MAP
kinase inhibitor.
4044. The device of claim 3945 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
4045. The device of claim 3945 wherein the agent is a phosphadiesterase inhibitor.
4046. The device of claim 3945 wherein the agent is a TGF beta inhibitor.
4047. The device of claim 3945 wherein the agent is a thromboxane A2 antagonist.
4048. The device of claim 3945 wherein the agent is a TNFa antagonist.
4049. The device of claim 3945 wherein the agent is a TACE
inhibitor.
4050. The device of claim 3945 wherein the agent is a tyrosine kinase inhibitor.
4051. The device of claim 3945 wherein the agent is a vitronectin inhibitor.
4052. The device of claim 3945 wherein the agent is a fibroblast growth factor inhibitor.
4053. The device of claim 3945 wherein the agent is a protein kinase inhibitor.
4054. The device of claim 3945 wherein the agent is a PDGF
receptor kinase inhibitor.
4055. The device of claim 3945 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
4056. The device of claim 3945 wherein the agent is a retinoic acid receptor antagonist.
4057. The device of claim 3945 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
4058. The device of claim 3945 wherein the agent is a fibronogin antagonist.
4059. The device of claim 3945 wherein the agent is an antimycotic agent.
4060. The device of claim 3945 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
4061. The device of claim 3945 wherein the agent is a bisphosphonate.
4062. The device of claim 3945 wherein the agent is a phospholipase A1 inhibitor.
4063. The device of claim 3945 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
4064. The device of claim 3945 wherein the agent is a macrolide antibiotic.
4065. The device of claim 3945 wherein the agent is a GPIIb/IIIa receptor antagonist.
4066. The device of claim 3945 wherein the agent is an endothelin receptor antagonist.
4067. The device of claim 3945 wherein the agent is a peroxisome proliferator-activated receptor agonist.
4068. The device of claim 3945 wherein the agent is an estrogen receptor agent.
4069. The device of claim 3945 wherein the agent is a somastostatin analogue.
4070. The device of claim 3945 wherein the agent is a neurokinin 1 antagonist.
4071. The device of claim 3945 wherein the agent is a neurokinin 3 antagonist.
4072. The device of claim 3945 wherein the agent is a VLA-4 antagonist.
4073. The device of claim 3945 wherein the agent is an osteoclast inhibitor.
4074. The device of claim 3945 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
4075. The device of claim 3945 wherein the agent is an angiotensin I converting enzyme inhibitor.
4076. The device of claim 3945 wherein the agent is an angiotensin II antagonist.
4077. The device of claim 3945 wherein the agent is an enkephalinase inhibitor.
4078. The device of claim 3945 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
4079. The device of claim 3945 wherein the agent is a protein kinase C inhibitor.
4080. The device of claim 3945 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
4081. The device of claim 3945 wherein the agent is a CXCR3 inhibitor.
4082. The device of claim 3945 wherein the agent is an Itk inhibitor.
4083. The device of claim 3945 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
4084. The device of claim 3945 wherein the agent is a PPAR
agonist.
4085. The device of claim 3945 wherein the agent is an immunosuppressant.
4086. The device of claim 3945 wherein the agent is an Erb inhibitor.
4087. The device of claim 3945 wherein the agent is an apoptosis agonist.
4088. The device of claim 3945 wherein the agent is a lipocortin agonist.
4089. The device of claim 3945 wherein the agent is a VCAM-1 antagonist.
4090. The device of claim 3945 wherein the agent is a collagen antagonist.
4091. The device of claim 3945 wherein the agent is an alpha 2 integrin antagonist.
4092. The device of claim 3945 wherein the agent is a TNF alpha inhibitor.
4093. The device of claim 3945 wherein the agent is a nitric oxide inhibitor
4094. The device of claim 3945 wherein the agent is a cathepsin inhibitor.
4095. The device of claim 3945 wherein the agent is not an anti-inflammatory agent.
4096. The device of claim 3945 wherein the agent is not a steroid.
4097. The device of claim 3945 wherein the agent is not a glucocorticosteroid.
4098. The device of claim 3945 wherein the agent is not dexamethasone.
4099. The device of claim 3945 wherein the agent is not an anti-infective agent.
4100. The device of claim 3945 wherein the agent is not an antibiotic.
4101. The device of claim 3945 wherein the agent is not an anti-fungal agent.
4102. The device of claim 3945, further comprising a polymer.
4103. The device of claim 3945, further comprising a polymeric carrier.
4104. The device of claim 3945 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
4105. The device of claim 3945 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4106. The device of claim 3945, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4107. The device of claim 3945, further comprising a coating, wherein the coating is disposed on a surface of the device.
4108. The device of claim 3945, further comprising a coating, wherein the coating directly contacts the device.
4109. The device of claim 3945, further comprising a coating, wherein the coating indirectly contacts the device.
4110. The device of claim 3945, further comprising a coating, wherein the coating partially covers the device.
4111. The device of claim 3945, further comprising a coating, wherein the coating completely covers the device.
4112. The device of claim 3945, further comprising a coating, wherein the coating is a uniform coating.
4113. The device of claim 3945, further comprising a coating, wherein the coating is a non-uniform coating.
4114. The device of claim 3945, further comprising a coating, wherein the coating is a discontinuous coating.
4115. The device of claim 3945, further comprising a coating, wherein the coating is a patterned coating.
4116. The device of claim 3945, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
4117. The device of claim 3945, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
4118. The device of claim 3945, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4119. The device of claim 3945, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4120. The device of claim 3945, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4121. The device of claim 3945, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4122. The device of claim 3945, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4123. The device of claim 3945, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4124. The device of claim 3945, further comprising a coating, wherein the coating further comprises a polymer.
4125. The device of claim 3945, further comprising a first coating having a first composition and the second coating having a second composition.
4126. The device of claim 3945, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4127. The device of claim 3945, further comprising a polymer.
4128. The device of claim 3945, further comprising a polymeric carver.
4129. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4130. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4131. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4132. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4133. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4134. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
4135. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4136. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4137. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4138. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4139. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4140. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4141. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4142. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4143. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4144. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
4145. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4146. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polyethylene glycol) polymer.
4147. The device of claim 3945, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4148. The device of claim 3945, further comprising a lubricious coating.
4149. The device of claim 3945 wherein the anti-scarring agent is located within pores or holes of the device.
4150. The device of claim 3945 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4151. The device of claim 3945, further comprising a second pharmaceutically active agent.
4152. The device of claim 3945, further comprising an anti-inflammatory agent.
4153. The device of claim 3945, further comprising an agent that inhibits infection.
4154. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
4155. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4156. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4157. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4153. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4159. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4160. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4161. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4162. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4163. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4164. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
4165. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4166. The device of claim 3945, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4167. The device of claim 3945, further comprising an anti-thrombotic agent.
4168. The device of claim 3945, further comprising a visualization agent.
4169. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4170. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4171. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4172. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
4173. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4174. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4175. The device of claim 3945, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4176. The device of claim 3945, further comprising an echogenic material.
4177. The device of claim 3945, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
4178. The device of claim 3945 wherein the device is sterile.
4179. The device of claim 3945 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4180. The device of claim 3945 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
4181. The device of claim 3945 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4182. The device of claim 3945 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4183. The device of claim 3945 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4184. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4185. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4186. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4187. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4188. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
4189. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4190. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4191. The device of claim 3945 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4192. The device of claim 3945 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
4193. The device of claim 3945 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
4194. The device of claim 3945 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4195. The device of claim 3945 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4196. The device of claim 3945 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4197. The device of claim 3945 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4198. The device of claim 3945 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4199. The device of claim 3945 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4200. The device of claim 3945 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4201. The device of claim 3945 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4202. The device of claim 3945 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4203. The device of any one of claims 3945-4202 wherein the implant is a spinal disc.
4204. The device of any one of claims 3945-4202 wherein the implant is a vertebral disc prosthesis.
4205. The device of any one of claims 3945-4202 wherein the implant is an intervertebral disc.
4206. The device of any one of claims 3945-4202 wherein the implant is a partial spinal prosthesis.
4207. The device of any one of claims 3945-4202 wherein the implant is a spinal nucleus implant.
4208. The device of any one of claims 3945-4202 wherein the implant is an intervertebral disc spacer.
4209. The device of any one of claims 3945-4202 wherein the implant is a fusion cage.
4210. The device of any one of claims 3945-4202 wherein the implant is a fusion basket.
4211. The device of any one of claims 3945-4202 wherein the implant is a fusion chamber.
4212. The device of any one of claims 3945-4202 wherein the implant is a spinal anchoring device.
4213. The device of any one of claims 3945-4202 wherein the implant is a bone fixation device.
4214. The device of any one of claims 3945-4202 wherein the implant is an anchoring bone plate for the spine.
4215. The device of any one of claims 3945-4202 wherein the implant is an anchoring screw for the spine.
4216. The device of any one of claims 3945-4202 wherein the implant is an implantable rod for the spine.
4217. The device of any one of claims 3945-4202 wherein the implant is an implantable dowel for the spine.
4218. The device of any one of claims 3945-4202 wherein the implant is an implantable hook for the spine.
4219. The device of any one of claims 3945-4202 wherein the implant is a wire for spinal binding.
4220. The device of any one of claims 3945-4202 wherein the implant is a wedge for spinal support.
4221. A device, comprising a pressure monitoring implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4222. The device of claim 4221 wherein the agent inhibits cell regeneration.
4223. The device of claim 4221 wherein the agent inhibits angiogenesis.
4224. The device of claim 4221 wherein the agent inhibits fibroblast migration.
4225. The device of claim 4221 wherein the agent inhibits fibroblast proliferation.
4226. The device of claim 4221 wherein the agent inhibits deposition of extracellular matrix.
4227. The device of claim 4221 wherein the agent inhibits tissue remodeling.
4228. The device of claim 4221 wherein the agent is an angiogenesis inhibitor.
4229. The device of claim 4221 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
4230. The device of claim 4221 wherein the agent is a chemokine receptor antagonist.
4231. The device of claim 4221 wherein the agent is a cell cycle inhibitor.
4232. The device of claim 4221 wherein the agent is a taxane.
4233. The device of claim 4221 wherein the agent is an anti-microtubule agent.
4234. The device of claim 4221 wherein the agent is paclitaxel.
4235. The device of claim 4221 wherein the agent is not paclitaxel.
4236. The device of claim 4221 wherein the agent is an analogue or derivative of paclitaxel.
4237. The device of claim 4221 wherein the agent is a vinca alkaloid.
4238. The device of claim 4221 wherein the agent is camptothecin or an analogue or derivative thereof.
4239. The device of claim 4221 wherein the agent is a podophyllotoxin.
4240. The device of claim 4221 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
4241. The device of claim 4221 wherein the agent is an anthracycline.
4242. The device of claim 4221 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
4243. The device of claim 4221 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
4244. The device of claim 4221 wherein the agent is a platinum compound.
4245. The device of claim 4221 wherein the agent is a nitrosourea.
4246. The device of claim 4221 wherein the agent is a nitroimidazole.
4247. The device of claim 4221 wherein the agent is a folic acid antagonist.
4248. The device of claim 4221 wherein the agent is a cytidine analogue.
4249. The device of claim 4221 wherein the agent is a pyrimidine analogue.
4250. The device of claim 4221 wherein the agent is a fluoropyrimidine analogue.
4251. The device of claim 4221 wherein the agent is a purine analogue.
4252. The device of claim 4221 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
4253. The device of claim 4221 wherein the agent is a hydroxyurea.
4254. The device of claim 4221 wherein the agent is a mytomicin or an analogue or derivative thereof.
4255. The device of claim 4221 wherein the agent is an alkyl sulfonate.
4256. The device of claim 4221 wherein the agent is a benzamide or an analogue or derivative thereof.
4257. The device of claim 4221 wherein the agent is a nicotinamide or an analogue or derivative thereof.
4258. The device of claim 4221 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
4259. The device of claim 4221 wherein the agent is a DNA
alkylating agent.
4260. The device of claim 4221 wherein the agent is an anti-microtubule agent.
4261. The device of claim 4221 wherein the agent is a topoisomerase inhibitor.
4262. The device of claim 4221 wherein the agent is a DNA
cleaving agent.
4263. The device of claim 4221 wherein the agent is an antimetabolite.
4264. The device of claim 4221 wherein the agent inhibits adenosine deaminase.
4265. The device of claim 4221 wherein the agent inhibits purine ring synthesis.
4266. The device of claim 4221 wherein the agent is a nucleotide interconversion inhibitor.
4267. The device of claim 4221 wherein the agent inhibits dihydrofolate reduction.
4268. The device of claim 4221 wherein the agent blocks thymidine monophosphate.
4269. The device of claim 4221 wherein the agent causes DNA
damage.
4270. The device of claim 4221 wherein the agent is a DNA
intercalation agent.
4271. The device of claim 4221 wherein the agent is a RNA
synthesis inhibitor.
4272. The device of claim 4221 wherein the agent is a pyrimidine synthesis inhibitor.
4273. The device of claim 4221 wherein the agent inhibits ribonucleotide synthesis or function.
4274. The device of claim 4221 wherein the agent inhibits thymidine monophosphate synthesis or function.
4275. The device of claim 4221 wherein the agent inhibits DNA
synthesis.
4276. The device of claim 4221 wherein the agent causes DNA
adduct formation.
4277. The device of claim 4221 wherein the agent inhibits protein synthesis.
4278. The device of claim 4221 wherein the agent inhibits microtubule function.
4279. The device of claim 4221 wherein the agent is a cyclin dependent protein kinase inhibitor.
4280. The device of claim 4221 wherein the agent is an epidermal growth factor kinase inhibitor.
4281. The device of claim 4221 wherein the agent is an elastase inhibitor.
4282. The device of claim 4221 wherein the agent is a factor Xa inhibitor.
4283. The device of claim 4221 wherein the agent is a farnesyltransferase inhibitor.
4284. The device of claim 4221 wherein the agent is a fibrinogen antagonist.
4285. The device of claim 4221 wherein the agent is a guanylate cyclase stimulant.
4286. The device of claim 4221 wherein the agent is a heat shock protein 90 antagonist.
2120 428. The device of claim 4221 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
4288. The device of claim 4221 wherein the agent is a guanylate cyclase stimulant.
4289. The device of claim 4221 wherein the agent is a HMGCoA
reductase inhibitor.
4290. The device of claim 4221 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
4291. The device. of claim 4221 wherein the agent is a hydroorotate dehydragenase inhibitor.
4292. The device of claim 4221 wherein the agent is an IKK2 inhibitor.
4293. The device of claim 4221 wherein the agent is an IL-1 antagonist.
4294. The device of claim 4221 wherein the agent is an ICE
antagonist.
4295. The device of claim 4221 wherein the agent is an IRAK
antagonist.
4296. The device of claim 4221 wherein the agent is an IL-4 agonist.
4297. The device of claim 4221 wherein the agent is an immunomodulatory agent.
4298. The device of claim 4221 wherein the agent is sirolimus or an analogue or derivative thereof.
4299. The device of claim 4221 wherein the agent is not sirolimus.
4300. The device of claim 4221 wherein the agent is everolimus or an analogue or derivative thereof.
4301. The device of claim 4221 wherein the agent is tacrolimus or an analogue or derivative thereof.
4302. The device of claim 4221 wherein the agent is not tacrolimus.
4303. The device of claim 4221 wherein the agent is biolmus or an analogue or derivative thereof.
4304. The device of claim 4221 wherein the agent is tresperimus or an analogue or derivative thereof.
4305. The device of claim 4221 wherein the agent is auranofin or an analogue or derivative thereof.
4306. The device of claim 4221 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
4307. The device of claim 4221 wherein the agent is gusperimus or an analogue or derivative thereof.
4308. The device of claim 4221 wherein the agent is pimecrolimus or an analogue or derivative thereof.
4309. The device of claim 4221 wherein the agent is ABT-578 or an analogue or derivative thereof.
4310. The device of claim 4221 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
4311. The device of claim 4221 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
4312. The device of claim 4221 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
4313. The device of claim 4221 wherein the agent is a leukotriene inhibitor.
4314. The device of claim 4221 wherein the agent is a MCP-1 antagonist.
4315. The device of claim 4221 wherein the agent is a MMP
inhibitor.
4316. The device of claim 4221 wherein the agent is an NF
kappa B inhibitor.
4317. The device of claim 4221 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
4318. The device of claim 4221 wherein the agent is an NO
agonist.
4319. The device of claim 4221 wherein the agent is a p38 MAP
kinase inhibitor.
4320. The device of claim 4221 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
4321. The device of claim 4221 wherein the agent is a phosphodiesterase inhibitor.
4322. The device of claim 4221 wherein the agent is a TGF beta inhibitor.
4323. The device of claim 4221 wherein the agent is a thromboxane A2 antagonist.
4324. The device of claim 4221 wherein the agent is a TNFa antagonist.
4325. The device of claim 4221 wherein the agent is a TACE
inhibitor.
4326. The device of claim 4221 wherein the agent is a tyrosine kinase inhibitor.
4327. The device of claim 4221 wherein the agent is a vitronectin inhibitor.
4323. The device of claim 4221 wherein the agent is a fibroblast growth factor inhibitor.
4329. The device of claim 4221 wherein the agent is a protein kinase inhibitor.
4330. The device of claim 4221 wherein the agent is a PDGF
receptor kinase inhibitor.
4331. The device of claim 4221 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
4332. The device of claim 4221 wherein the agent is a retinoic acid receptor antagonist.
4333. The device of claim 4221 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
4334. The device of claim 4221 wherein the agent is a fibronogin antagonist.
4335. The device of claim 4221 wherein the agent is an antimycotic agent.
4336. The device of claim 4221 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
4337. The device of claim 4221 wherein the agent is a bisphosphonate.
4338. The device of claim 4221 wherein the agent is a phospholipase A1 inhibitor.
4339. The device of claim 4221 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
4340. The device of claim 4221 wherein the agent is a macrolide antibiotic.
4341. The device of claim 4221 wherein the agent is a GPIIb/IIIa receptor antagonist.
4342. The device of claim 4221 wherein the agent is an endothelin receptor antagonist.
4343. The device of claim 4221 wherein the agent is a peroxisome proliferator-activated receptor agonist.
4344. The device of claim 4221 wherein the agent is an estrogen receptor agent.
4345. The device of claim 4221 wherein the agent is a somastostatin analogue.
4346. The device of claim 4221 wherein the agent is a neurokinin 1 antagonist.
4347. The device of claim 4221 wherein the agent is a neurokinin 3 antagonist.
4343. The device of claim 4221 wherein the agent is a VLA-4 antagonist.
4349. The device of claim 4221 wherein the agent is an osteoclast inhibitor.
4350. The device of claim 4221 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
4351. The device of claim 4221 wherein the agent is an angiotensin 1 converting enzyme inhibitor.
4352. The device of claim 4221 wherein the agent is an angiotensin II antagonist.
4353. The device of claim 4221 wherein the agent is an enkephalinase inhibitor.
4354. The device of claim 4221 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
4355. The device of claim 4221 wherein the agent is a protein kinase C inhibitor.
4356. The device of claim 4221 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
4357. The device of claim 4221 wherein the agent is a CXCR3 inhibitor.
4358. The device of claim 4221 wherein the agent is an Itk inhibitor.
4359. The device of claim 4221 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
4360. The device of claim 4221 wherein the agent is a PPAR
agonist.
4361. The device of claim 4221 wherein the agent is an immunosuppressant.
4362. The device of claim 4221 wherein the agent is an Erb inhibitor.
4363. The device of claim 4221 wherein the agent is an apoptosis agonist.
4364. The device of claim 4221 wherein the agent is a lipocortin agonist.
4365. The device of claim 4221 wherein the agent is a VCAM-1 antagonist.
4366. The device of claim 4221 wherein the agent is a collagen antagonist.
4367. The device of claim 4221 wherein the agent is an alpha 2 integrin antagonist.
4368. The device of claim 4221 wherein the agent is a TNF alpha inhibitor.
4369. The device of claim 4221 wherein the agent is a nitric oxide inhibitor
4370. The device of claim 4221 wherein the agent is a cathepsin inhibitor.
4371. The device of claim 4221 wherein the agent is not an anti-inflammatory agent.
4372. The device of claim 4221 wherein the agent is not a steroid.
4373. The device of claim 4221 wherein the agent is not a glucocorticosteroid.
4374. The device of claim 4221 wherein the agent is not dexamethasone.
4375. The device of claim 4221 wherein the agent is not an anti-infective agent.
4376. The device of claim 4221 wherein the agent is not an antibiotic.
4377. The device of claim 4221 wherein the agent is not an anti-fungal agent.
4378. The device of claim 4221, further comprising a polymer.
4379. The device of claim 4221, further comprising a polymeric carrier.
4380. The device of claim 4221 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
4381. The device of claim 4221 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4382. The device of claim 4221, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4383. The device of claim 4221, further comprising a coating, wherein the coating is disposed on a surface of the device.
4384. The device of claim 4221, further comprising a coating, wherein the coating directly contacts the device.
4385. The device of claim 4221, further comprising a coating, wherein the coating indirectly contacts the device.
4386. The device of claim 4221, further comprising a coating, wherein the coating partially covers the device.
4387. The device of claim 4221, further comprising a coating, wherein the coating completely covers the device.
4388. The device of claim 4221, further comprising a coating, wherein the coating is a uniform coating.
4389. The device of claim 4221, further comprising a coating, wherein the coating is a non-uniform coating.
4390. The device of claim 4221, further comprising a coating, wherein the coating is a discontinuous coating.
4391. The device of claim 4221, further comprising a coating, wherein the coating is a patterned coating.
4392. The device of claim 4221, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
4393. The device of claim 4221, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
4394. The device of claim 4221, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4395. The device of claim 4221, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4396. The device of claim 4221, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4397. The device of claim 4221, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4398. The device of claim 4221, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4399. The device of claim 4221, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4400. The device of claim 4221, further comprising a coating, wherein the coating further comprises a polymer.
4401. The device of claim 4221, further comprising a first coating having a first composition and the second coating having a second composition.
4402. The device of claim 4221, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4403. The device of claim 4221, further comprising a polymer.
4404. The device of claim 4221, further comprising a polymeric carrier.
4405. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4406. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4407. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4408. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4409. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4410. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
4411. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4412. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4413. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4414. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4415. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4416. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4417. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4418. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4419. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4420. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
4421. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4422. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4423. The device of claim 4221, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4424. The device of claim 4221, further comprising a lubricious coating.
4425. The device of claim 4221 wherein the anti-scarring agent is located within pores or holes of the device.
4426. The device of claim 4221 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4427. The device of claim 4221, further comprising a second pharmaceutically active agent.
4428. The device of claim 4221, further comprising an anti-inflammatory agent.
4429. The device of claim 4221, further comprising an agent that inhibits infection.
4430. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
4431. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4432. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4433. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4434. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4435. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4436. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4437. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2133 4433. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4439. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4440. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
4441. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4442. The device of claim 4221, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4443. The device of claim 4221, further comprising an anti-thrombotic agent.
4444. The device of claim 4221, further comprising a visualization agent.
4445. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4446. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4447. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4448. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
4449. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4450. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4451. The device of claim 4221, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4452. The device of claim 4221, further comprising an echogenic material.
4453. The device of claim 4221, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
4454. The device of claim 4221 wherein the device is sterile.
4455. The device of claim 4221 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4456. The device of claim 4221 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
4457. The device of claim 4221 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4458. The device of claim 4221 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4459. The device of claim 4221 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4460. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4461. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4462. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4463. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4464. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
4465. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4466. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4467. The device of claim 4221 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4468. The device of claim 4221 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
4469. The device of claim 4221 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
4470. The device of claim 4221 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4471. The device of claim 4221 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4472. The device of claim 4221 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4473. The device of claim 4221 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4474. The device of claim 4221 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4475. The device of claim 4221 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4476. The device of claim 4221 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4477. The device of claim 4221 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4478. The device of claim 4221 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4479. A device, comprising a tympanostomy tube implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4480. The device of claim 4479 wherein the agent inhibits cell regeneration.
4481. The device of claim 4479 wherein the agent inhibits angiogenesis.
4482. The device of claim 4479 wherein the agent inhibits fibroblast migration.
4483. The device of claim 4479 wherein the agent inhibits fibroblast proliferation.
4484. The device of claim 4479 wherein the agent inhibits deposition of extracellular matrix.
4485. The device of claim 4479 wherein the agent inhibits tissue remodeling.
4486. The device of claim 4479 wherein the agent is an angiogenesis inhibitor.
4487. The device of claim 4479 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
4488. The device of claim 4479 wherein the agent is a chemokine receptor antagonist.
4489. The device of claim 4479 wherein the agent is a cell cycle inhibitor.
4490. The device of claim 4479 wherein the agent is a taxane.
4491. The device of claim 4479 wherein the agent is an anti-microtubule agent.
4492. The device of claim 4479 wherein the agent is paclitaxel.
4493. The device of claim 4479 wherein the agent is not paclitaxel.
4494. The device of claim 4479 wherein the agent is an analogue or derivative of paclitaxel.
4495. The device of claim 4479 wherein the agent is a vinca alkaloid.
4496. The device of claim 4479 wherein the agent is camptothecin or an analogue or derivative thereof.
4497. The device of claim 4479 wherein the agent is a podophyllotoxin.
4498. The device of claim 4479 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
4499. The device of claim 4479 wherein the agent is an anthracycline.
4500. The device of claim 4479 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
4501. The device of claim 4479 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
4502. The device of claim 4479 wherein the agent is a platinum compound.
4503. The device of claim 4479 wherein the agent is a nitrosourea.
4504. The device of claim 4479 wherein the agent is a nitroimidazole.
4505. The device of claim 4479 wherein the agent is a folic acid antagonist.
4506. The device of claim 4479 wherein the agent is a cytidine analogue.
4507. The device of claim 4479 wherein the agent is a pyrimidine analogue.
4508. The device of claim 4479 wherein the agent is a fluoropyrimidine analogue.
4509. The device of claim 4479 wherein the agent is a purine analogue.
4510. The device of claim 4479 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
4511. The device of claim 4479 wherein the agent is a hydroxyurea.
4512. The device of claim 4479 wherein the agent is a mytomicin or an analogue or derivative thereof.
4513. The device of claim 4479 wherein the agent is an alkyl sulfonate.
4514. The device of claim 4479 wherein the agent is a benzamide or an analogue or derivative thereof.
4515. The device of claim 4479 wherein the agent is a nicotinamide or an analogue or derivative thereof.
4516. The device of claim 4479 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
4517. The device of claim 4479 wherein the agent is a DNA
alkylating agent.
4513. The device of claim 4479 wherein the agent is an anti-microtubule agent.
4519. The device of claim 4479 wherein the agent is a topoisomerase inhibitor.
4520. The device of claim 4479 wherein the agent is a DNA
cleaving agent.
4521. The device of claim 4479 wherein the agent is an antimetabolite.
4522. The device of claim 4479 wherein the agent inhibits adenosine deaminase.
4523. The device of claim 4479 wherein the agent inhibits purine ring synthesis.
4524. The device of claim 4479 wherein the agent is a nucleotide interconversion inhibitor.
4525. The device of claim 4479 wherein the agent inhibits dihydrofolate reduction.
4526. The device of claim 4479 wherein the agent blocks thymidine monophosphate.
4527. The device of claim 4479 wherein the agent causes DNA
damage.
4523. The device of claim 4479 wherein the agent is a DNA
intercalation agent.
4529. The device of claim 4479 wherein the agent is a RNA
synthesis inhibitor.
4530. The device of claim 4479 wherein the agent is a pyrimidine synthesis inhibitor.
4531. The device of claim 4479 wherein the agent inhibits ribonucleotide synthesis or function.
4532. The device of claim 4479 wherein the agent inhibits thymidine monophosphate synthesis or function.
4533. The device of claim 4479 wherein the agent inhibits DNA
synthesis.
4534. The device of claim 4479 wherein the agent causes DNA
adduct formation.
4535. The device of claim 4479 wherein the agent inhibits protein synthesis.
4536. The device of claim 4479 wherein the agent inhibits microtubule function.
4537. The device of claim 4479 wherein the agent is a cyclin dependent protein kinase inhibitor.
4538. The device of claim 4479 wherein the agent is an epidermal growth factor kinase inhibitor.
4539. The device of claim 4479 wherein the agent is an elastase inhibitor.
4540. The device of claim 4479 wherein the agent is a factor Xa inhibitor.
4541. The device of claim 4479 wherein the agent is a farnesyltransferase inhibitor.
4542. The device of claim 4479 wherein the agent is a fibrinogen antagonist.
4543. The device of claim 4479 wherein the agent is a guanylate cyclase stimulant.
4544. The device of claim 4479 wherein the agent is a heat shock protein 90 antagonist.
4545. The device of claim 4479 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
4546. The device of claim 4479 wherein the agent is a guanylate cyclase stimulant.
4547. The device of claim 4479 wherein the agent is a HMGCoA
reductase inhibitor.
4548. The device of claim 4479 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
4549. The device of claim 4479 wherein the agent is a hydroorotate dehydrogenase inhibitor.
4550. The device of claim 4479 wherein the agent is an IKK2 inhibitor.
4551. The device of claim 4479 wherein the agent is an IL-1 antagonist.
4552. The device of claim 4479 wherein the agent is an ICE
antagonist.
4553. The device of claim 4479 wherein the agent is an IRAK
antagonist.
4554. The device of claim 4479 wherein the agent is an IL-4 agonist.
4555. The device of claim 4479 wherein the agent is an immunomodulatory agent.
4556. The device of claim 4479 wherein the agent is sirolimus or an analogue or derivative thereof.
4557. The device of claim 4479 wherein the agent is not sirolimus.
4558. The device of claim 4479 wherein the agent is everolimus or an analogue or derivative thereof.
4559. The device of claim 4479 wherein the agent is tacrolimus or an analogue or derivative thereof.
4560. The device of claim 4479 wherein the agent is not tacrolimus.
4561. The device of claim 4479 wherein the agent is biolmus or an analogue or derivative thereof.
4562. The device of claim 4479 wherein the agent is tresperimus or an analogue or derivative thereof.
4563. The device of claim 4479 wherein the agent is auranofin or an analogue or derivative thereof.
4564. The device of claim 4479 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
4565. The device of claim 4479 wherein the agent is gusperimus or an analogue or derivative thereof.
4566. The device of claim 4479 wherein the agent is pimecrolimus or an analogue or derivative thereof.
4567. The device of claim 4479 wherein the agent is ABT-578 or an analogue or derivative thereof.
4568. The device of claim 4479 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
4569. The device of claim 4479 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
4570. The device of claim 4479 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
4571. The device of claim 4479 wherein the agent is a leukotriene inhibitor.
4572. The device of claim 4479 wherein the agent is a MCP-1 antagonist.
4573. The device of claim 4479 wherein the agent is a MMP
inhibitor.
4574. The device of claim 4479 wherein the agent is an NF
kappa B inhibitor.
4575. The device of claim 4479 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
4576. The device of claim 4479 wherein the agent is an NO
agonist.
4577. The device of claim 4479 wherein the agent is a p38 MAP
kinase inhibitor.
4578. The device of claim 4479 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
4579. The device of claim 4479 wherein the agent is a phosphodiesterase inhibitor.
4580. The device of claim 4479 wherein the agent is a TGF beta inhibitor.
4581. The device of claim 4479 wherein the agent is a thromboxane A2 antagonist.
4582. The device of claim 4479 wherein the agent is a TNFa antagonist.
4583. The device of claim 4479 wherein the agent is a TACE
inhibitor.
4584. The device of claim 4479 wherein the agent is a tyrosine kinase inhibitor.
4585. The device of claim 4479 wherein the agent is a vitronectin inhibitor.
4586. The device of claim 4479 wherein the agent is a fibroblast growth factor inhibitor.
4587. The device of claim 4479 wherein the agent is a protein kinase inhibitor.
4588. The device of claim 4479 wherein the agent is a PDGF
receptor kinase inhibitor.
4589. The device of claim 4479 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
4590. The device of claim 4479 wherein the agent is a retinoic acid receptor antagonist.
4591. The device of claim 4479 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
4592. The device of claim 4479 wherein the agent is a fibronogin antagonist.
4593. The device of claim 4479 wherein the agent is an antimycotic agent.
4594. The device of claim 4479 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
4595. The device of claim 4479 wherein the agent is a bisphosphonate.
4596. The device of claim 4479 wherein the agent is a phospholipase A1 inhibitor.
4597. The device of claim 4479 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
4598. The device of claim 4479 wherein the agent is a macrolide antibiotic.
4599. The device of claim 4479 wherein the agent is a GPIIb/IIIa receptor antagonist.
4600. The device of claim 4479 wherein the agent is an endothelin receptor antagonist.
4601. The device of claim 4479 wherein the agent is a peroxisome proliferator-activated receptor agonist.
4602. The device of claim 4479 wherein the agent is an estrogen receptor agent.
4603. The device of claim 4479 wherein the agent is a somastostatin analogue.
4604. The device of claim 4479 wherein the agent is a neurokinin 1 antagonist.
4605. The device of claim 4479 wherein the agent is a neurokinin 3 antagonist.
4606. The device of claim 4479 wherein the agent is a VLA-4 antagonist.
4607. The device of claim 4479 wherein the agent is an osteoclast inhibitor.
4608. The device of claim 4479 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
4609. The device of claim 4479 wherein the agent is an angiotensin I converting enzyme inhibitor.
4610. The device of claim 4479 wherein the agent is an angiotensin II antagonist.
4611. The device of claim 4479 wherein the agent is an enkephalinase inhibitor.
4612. The device of claim 4479 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
4613. The device of claim 4479 wherein the agent is a protein kinase C inhibitor.
4614. The device of claim 4479 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
4615. The device of claim 4479 wherein the agent is a CXCR3 inhibitor.
4616. The device of claim 4479 wherein the agent is an Itk inhibitor.
4617. The device of claim 4479 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
4618. The device of claim 4479 wherein the agent is a PPAR
agonist.
4619. The device of claim 4479 wherein the agent is an immunosuppressant.
4620. The device of claim 4479 wherein the agent is an Erb inhibitor.
4621. The device of claim 4479 wherein the agent is an apoptosis agonist.
4622. The device of claim 4479 wherein the agent is a lipocortin agonist.
4623. The device of claim 4479 wherein the agent is a VCAM-1 antagonist.
4624. The device of claim 4479 wherein the agent is a collagen antagonist.
4625. The device of claim 4479 wherein the agent is an alpha 2 integrin antagonist.
4626. The device of claim 4479 wherein the agent is a TNF alpha inhibitor.
4627. The device of claim 4479 wherein the agent is a nitric oxide inhibitor
4628. The device of claim 4479 wherein the agent is a cathepsin inhibitor.
4629. The device of claim 4479 wherein the agent is not an anti-inflammatory agent.
4630. The device of claim 4479 wherein the agent is not a steroid.
4631. The device of claim 4479 wherein the agent is not a glucocorticosteroid.
4632. The device of claim 4479 wherein the agent is not dexamethasone.
4633. The device of claim 4479 wherein the agent is not an anti-infective agent.
4634. The device of claim 4479 wherein the agent is not an antibiotic.
4635. The device of claim 4479 wherein the agent is not an anti-fungal agent.
4636. The device of claim 4479, further comprising a polymer.
4637. The device of claim 4479, further comprising a polymeric carrier.
4638. The device of claim 4479 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
4639. The device of claim 4479 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4640. The device of claim 4479, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4641. The device of claim 4479, further comprising a coating, wherein the coating is disposed on a surface of the device.
4642. The device of claim 4479, further comprising a coating, wherein the coating directly contacts the device.
4643. The device of claim 4479, further comprising a coating, wherein the coating indirectly contacts the device.
4644. The device of claim 4479, further comprising a coating, wherein the coating partially covers the device.
4645. The device of claim 4479, further comprising a coating, wherein the coating completely covers the device.
4646. The device of claim 4479, further comprising a coating, wherein the coating is a uniform coating.
4647. The device of claim 4479, further comprising a coating, wherein the coating is a non-uniform coating.
4643. The device of claim 4479, further comprising a coating, wherein the coating is a discontinuous coating.
4649. The device of claim 4479, further comprising a coating, wherein the coating is a patterned coating.
4650. The device of claim 4479, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
4651. The device of claim 4479, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
4652. The device of claim 4479, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4653. The device of claim 4479, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4654. The device of claim 4479, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
4655. The device of claim 4479, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
4656. The device of claim 4479, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4657. The device of claim 4479, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4658. The device of claim 4479, further comprising a coating, wherein the coating further comprises a polymer.
4659. The device of claim 4479, further comprising a first coating having a first composition and the second coating having a second composition.
4660. The device of claim 4479, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4661. The device of claim 4479, further comprising a polymer.
4662. The device of claim 4479, further comprising a polymeric carrier.
4663. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4664. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4665. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4666. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4667. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4668. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
4669. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4670. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4671. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4672. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4673. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4674. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4675. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4676. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4677. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4678. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
4679. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4680. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4681. The device of claim 4479, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4682. The device of claim 4479, further comprising a lubricious coating.
4683. The device of claim 4479 wherein the anti-scarring agent is located within pores or holes of the device.
4684. The device of claim 4479 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4685. The device of claim 4479, further comprising a second pharmaceutically active agent.
4686. The device of claim 4479, further comprising an anti-inflammatory agent.
4687. The device of claim 4479, further comprising an agent that inhibits infection.
4688. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
4689. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4690. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4691. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4692. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4693. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4694. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4695. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4696. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4697. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4698. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
4699. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4700. The device of claim 4479, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4701. The device of claim 4479, further comprising an anti-thrombotic agent.
4702. The device of claim 4479, further comprising a visualization agent.
4703. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4704. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4705. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4706. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
4707. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4708. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4709. The device of claim 4479, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4710. The device of claim 4479, further comprising an echogenic material.
4711. The device of claim 4479, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
4712. The device of claim 4479 wherein the device is sterile.
4713. The device of claim 4479 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4714. The device of claim 4479 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
4715. The device of claim 4479 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4716. The device of claim 4479 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4717. The device of claim 4479 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4718. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4719. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4720. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4721. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4722. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
4723. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4724. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4725. The device of claim 4479 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4726. The device of claim 4479 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
4727. The device of claim 4479 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
4723. The device of claim 4479 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4729. The device of claim 4479 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4730. The device of claim 4479 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4731. The device of claim 4479 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4732. The device of claim 4479 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4733. The device of claim 4479 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4734. The device of claim 4479 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4735. The device of claim 4479 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4736. The device of claim 4479 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4737. A device, comprising an implant that provides a surgical adhesion barrier and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4738. The device of claim 4737 wherein the agent inhibits cell regeneration.
4739. The device of claim 4737 wherein the agent inhibits angiogenesis.
4740. The device of claim 4737 wherein the agent inhibits fibroblast migration.
4741. The device of claim 4737 wherein the agent inhibits fibroblast proliferation.
4742. The device of claim 4737 wherein the agent inhibits deposition of extracellular matrix.
4743. The device of claim 4737 wherein the agent inhibits tissue remodeling.
4744. The device of claim 4737 wherein the agent is an angiogenesis inhibitor.
4745. The device of claim 4737 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
4746. The device of claim 4737 wherein the agent is a chemokine receptor antagonist.
4747. The device of claim 4737 wherein the agent is a cell cycle inhibitor.
4748. The device of claim 4737 wherein the agent is a taxane.
4749. The device of claim 4737 wherein the agent is an anti-microtubule agent.
4750. The device of claim 4737 wherein the agent is paclitaxel.
4751. The device of claim 4737 wherein the agent is not paclitaxel.
4752. The device of claim 4737 wherein the agent is an analogue or derivative of paclitaxel.
4753. The device of claim 4737 wherein the agent is a vinca alkaloid.
4754. The device of claim 4737 wherein the agent is camptothecin or an analogue or derivative thereof.
4755. The device of claim 4737 wherein the agent is a podophyllotoxin.
4756. The device of claim 4737 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
4757. The device of claim 4737 wherein the agent is an anthracycline.
4758. The device of claim 4737 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
4759. The device of claim 4737 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
compound.
nitrosourea.
4760. The device of claim 4737 wherein the agent is a platinum compound.
4761. The device of claim 4737 wherein the agent is a nitrosourea.
4762. The device of claim 4737 wherein the agent is a nitroimidazole.
4763. The device of claim 4737 wherein the agent is a folic acid antagonist.
4764. The device of claim 4737 wherein the agent is a cytidine analogue.
4765. The device of claim 4737 wherein the agent is a pyrimidine analogue.
4766. The device of claim 4737 wherein the agent is a fluoropyrimidine analogue.
4767. The device of claim 4737 wherein the agent is a purine analogue.
4763. The device of claim 4737 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
4769. The device of claim 4737 wherein the agent is a hydroxyurea.
4770. The device of claim 4737 wherein the agent is a mytomicin or an analogue or derivative thereof.
4771. The device of claim 4737 wherein the agent is an alkyl sulfonate.
4772. The device of claim 4737 wherein the agent is a benzamide or an analogue or derivative thereof.
4773. The device of claim 4737 wherein the agent is a nicotinamide or an analogue or derivative thereof.
4774. The device of claim 4737 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
4775. The device of claim 4737 wherein the agent is a DNA
alkylating agent.
4776. The device of claim 4737 wherein the agent is an anti-microtubule agent.
4777. The device of claim 4737 wherein the agent is a topoisomerase inhibitor.
4778. The device of claim 4737 wherein the agent is a DNA
cleaving agent.
4779. The device of claim 4737 wherein the agent is an antimetabolite.
4780. The device of claim 4737 wherein the agent inhibits adenosine deaminase.
4781. The device of claim 4737 wherein the agent inhibits purine ring synthesis.
4782. The device of claim 4737 wherein the agent is a nucleotide interconversion inhibitor.
4783. The device of claim 4737 wherein the agent inhibits dihydrofolate reduction.
4784. The device of claim 4737 wherein the agent blocks thymidine monophosphate.
4785. The device of claim 4737 wherein the agent causes DNA
damage.
4786. The device of claim 4737 wherein the agent is a DNA
intercalation agent.
4787. The device of claim 4737 wherein the agent is a RNA
synthesis inhibitor.
4788. The device of claim 4737 wherein the agent is a pyrimidine synthesis inhibitor.
4789. The device of claim 4737 wherein the agent inhibits ribonucleotide synthesis or function.
4790. The device of claim 4737 wherein the agent inhibits thymidine monophosphate synthesis or function.
4791. The device of claim 4737 wherein the agent inhibits DNA
synthesis.
4792. The device of claim 4737 wherein the agent causes DNA
adduct formation.
4793. The device of claim 4737 wherein the agent inhibits protein synthesis.
4794. The device of claim 4737 wherein the agent inhibits microtubule function.
4795. The device of claim 4737 wherein the agent is a cyclin dependent protein kinase inhibitor.
4796. The device of claim 4737 wherein the agent is an epidermal growth factor kinase inhibitor.
4797. The device of claim 4737 wherein the agent is an elastase inhibitor.
4798. The device of claim 4737 wherein the agent is a factor Xa inhibitor.
4799. The device of claim 4737 wherein the agent is a farnesyltransferase inhibitor.
4800. The device of claim 4737 wherein the agent is a fibrinogen antagonist.
4801. The device of claim 4737 wherein the agent is a guanylate cyclase stimulant.
4802. The device of claim 4737 wherein the agent is a heat shock protein 90 antagonist.
4803. The device of claim 4737 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
4804. The device of claim 4737 wherein the agent is a guanylate cyclase stimulant.
4805. The device of claim 4737 wherein the agent is a HMGCoA
reductase inhibitor.
4806. The device of claim 4737 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
4807. The device of claim 4737 wherein the agent is a hydroorotate dehydrogenase inhibitor.
4808. The device of claim 4737 wherein the agent is an IKK2 inhibitor.
4809. The device of claim 4737 wherein the agent is an IL-1 antagonist.
4810. The device of claim 4737 wherein the agent is an ICE
antagonist.
4811. The device of claim 4737 wherein the agent is an IRAK
antagonist.
4812. The device of claim 4737 wherein the agent is an IL-4 agonist.
4813. The device of claim 4737 wherein the agent is an immunomodulatory agent.
4814. The device of claim 4737 wherein the agent is sirolimus or an analogue or derivative thereof.
4815. The device of claim 4737 wherein the agent is not sirolimus.
4816. The device of claim 4737 wherein the agent is everolimus or an analogue or derivative thereof.
4817. The device of claim 4737 wherein the agent is tacrolimus or an analogue or derivative thereof.
4818. The device of claim 4737 wherein the agent is not tacrolimus.
4819. The device of claim 4737 wherein the agent is biolmus or an analogue or derivative thereof.
4820. The device of claim 4737 wherein the agent is tresperimus or an analogue or derivative thereof.
4821. The device of claim 4737 wherein the agent is auranofin or an analogue or derivative thereof.
4822. The device of claim 4737 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
4823. The device of claim 4737 wherein the agent is gusperimus or an analogue or derivative thereof.
4824. The device of claim 4737 wherein the agent is pimecrolimus or an analogue or derivative thereof.
4825. The device of claim 4737 wherein the agent is ABT-578 or an analogue or derivative thereof.
4826. The device of claim 4737 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
4827. The device of claim 4737 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
4828. The device of claim 4737 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
4829. The device of claim 4737 wherein the agent is a leukotriene inhibitor.
4830. The device of claim 4737 wherein the agent is a MCP-1 antagonist.
4831. The device of claim 4737 wherein the agent is a MMP
inhibitor.
4832. The device of claim 4737 wherein the agent is an NF
kappa B inhibitor.
4833. The device of claim 4737 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
4834. The device of claim 4737 wherein the agent is an NO
agonist.
4835. The device of claim 4737 wherein the agent is a p38 MAP
kinase inhibitor.
4836. The device of claim 4737 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
4837. The device of claim 4737 wherein the agent is a phosphodiesterase inhibitor.
4838. The device of claim 4737 wherein the agent is a TGF beta inhibitor.
4839. The device of claim 4737 wherein the agent is a thromboxane A2 antagonist.
4840. The device of claim 4737 wherein the agent is a TNFa antagonist.
4841. The device of claim 4737 wherein the agent is a TACE
inhibitor.
4842. The device of claim 4737 wherein the agent is a tyrosine kinase inhibitor.
4843. The device of claim 4737 wherein the agent is a vitronectin inhibitor.
4844. The device of claim 4737 wherein the agent is a fibroblast growth factor inhibitor.
4845. The device of claim 4737 wherein the agent is a protein kinase inhibitor.
4846. The device of claim 4737 wherein the agent is a PDGF
receptor kinase inhibitor.
4847. The device of claim 4737 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
4848. The device of claim 4737 wherein the agent is a retinoic acid receptor antagonist.
4849. The device of claim 4737 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
4850. The device of claim 4737 wherein the agent is a fibronogin antagonist.
4851. The device of claim 4737 wherein the agent is an antimycotic agent.
4852. The device of claim 4737 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
4853. The device of claim 4737 wherein the agent is a bisphosphonate.
4854. The device of claim 4737 wherein the agent is a phospholipase A1 inhibitor.
4855. The device of claim 4737 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
4856. The device of claim 4737 wherein the agent is a macrolide antibiotic.
4857. The device of claim 4737 wherein the agent is a GPIIb/IIIa receptor antagonist.
4858. The device of claim 4737 wherein the agent is an endothelin receptor antagonist.
4859. The device of claim 4737 wherein the agent is a peroxisome proliferator-activated receptor agonist.
4860. The device of claim 4737 wherein the agent is an estrogen receptor agent.
4861. The device of claim 4737 wherein the agent is a somastostatin analogue.
4862. The device of claim 4737 wherein the agent is a neurokinin 1 antagonist.
4863. The device of claim 4737 wherein the agent is a neurokinin 3 antagonist.
4864. The device of claim 4737 wherein the agent is a VLA-4 antagonist.
4865. The device of claim 4737 wherein the agent is an osteoclast inhibitor.
4866. The device of claim 4737 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
4867. The device of claim 4737 wherein the agent is an angiotensin I converting enzyme inhibitor.
4868. The device of claim 4737 wherein the agent is an angiotensin II antagonist.
4869. The device of claim 4737 wherein the agent is an enkephalinase inhibitor.
4870. The device of claim 4737 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
4871. The device of claim 4737 wherein the agent is a protein kinase C inhibitor.
4872. The device of claim 4737 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
4873. The device of claim 4737 wherein the agent is a CXCR3 inhibitor.
4874. The device of claim 4737 wherein the agent is an Itk inhibitor.
4875. The device of claim 4737 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
4876. The device of claim 4737 wherein the agent is a PPAR
agonist.
4877. The device of claim 4737 wherein the agent is an immunosuppressant.
4878. The device of claim 4737 wherein the agent is an Erb inhibitor.
4879. The device of claim 4737 wherein the agent is an apoptosis agonist.
4880. The device of claim 4737 wherein the agent is a lipocortin agonist.
4881. The device of claim 4737 wherein the agent is a VCAM-1 antagonist.
4882. The device of claim 4737 wherein the agent is a collagen antagonist.
4883. The device of claim 4737 wherein the agent is an alpha 2 integrin antagonist.
4884. The device of claim 4737 wherein the agent is a TNF alpha inhibitor.
4885. The device of claim 4737 wherein the agent is a nitric oxide inhibitor
4886. The device of claim 4737 wherein the agent is a cathepsin inhibitor.
4887. The device of claim 4737 wherein the agent is not an anti-inflammatory agent.
4888. The device of claim 4737 wherein the agent is not a steroid.
4889. The device of claim 4737 wherein the agent is not a glucocorticosteroid.
4890. The device of claim 4737 wherein the agent is not dexamethasone.
4891. The device of claim 4737 wherein the agent is not an anti-infective agent.
4892. The device of claim 4737 wherein the agent is not an antibiotic.
4893. The device of claim 4737 wherein the agent is not an anti-fungal agent.
4894. The device of claim 4737, further comprising a polymer.
4895. The device of claim 4737, further comprising a polymeric carrier.
4896. The device of claim 4737 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
4897. The device of claim 4737 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4898. The device of claim 4737, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4899. The device of claim 4737, further comprising a coating, wherein the coating is disposed on a surface of the device.
4900. The device of claim 4737, further comprising a coating, wherein the coating directly contacts the device.
4901. The device of claim 4737, further comprising a coating, wherein the coating indirectly contacts the device.
4902. The device of claim 4737, further comprising a coating, wherein the coating partially covers the device.
4903. The device of claim 4737, further comprising a coating, wherein the coating completely covers the device.
4904. The device of claim 4737, further comprising a coating, wherein the coating is a uniform coating.
4905. The device of claim 4737, further comprising a coating, wherein the coating is a non-uniform coating.
4906. The device of claim 4737, further comprising a coating, wherein the coating is a discontinuous coating.
4907. The device of claim 4737, further comprising a coating, wherein the coating is a patterned coating.
4903. The device of claim 4737, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
4909. The device of claim 4737, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
4910. The device of claim 4737, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4911. The device of claim 4737, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4912. The device of claim 4737, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
4913. The device of claim 4737, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4914. The device of claim 4737, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4915. The device of claim 4737, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4916. The device of claim 4737, further comprising a coating, wherein the coating further comprises a polymer.
4917. The device of claim 4737, further comprising a first coating having a first composition and the second coating having a second composition.
4918. The device of claim 4737, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4919. The device of claim 4737, further comprising a polymer.
4920. The device of claim 4737, further comprising a polymeric carrier.
4921. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4922. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4923. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4924. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4925. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4926. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
4927. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4928. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4929. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4930. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4931. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4932. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4933. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4934. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4935. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4936. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
4937. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4938. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4939. The device of claim 4737, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4940. The device of claim 4737, further comprising a lubricious coating.
4941. The device of claim 4737 wherein the anti-scarring agent is located within pores or holes of the device.
4942. The device of claim 4737 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4943. The device of claim 4737, further comprising a second pharmaceutically active agent.
4944. The device of claim 4737, further comprising an anti-inflammatory agent.
4945. The device of claim 4737, further comprising an agent that inhibits infection.
4946. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
4947. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4948. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4949. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4950. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4951. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4952. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4953. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4954. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4955. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4956. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
4957. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4958. The device of claim 4737, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4959. The device of claim 4737, further comprising an anti-thrombotic agent.
4960. The device of claim 4737, further comprising a visualization agent.
4961. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4962. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4963. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4964. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
4965. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4966. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4967. The device of claim 4737, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4968. The device of claim 4737, further comprising an echogenic material.
4969. The device of claim 4737, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
4970. The device of claim 4737 wherein the device is sterile.
4971. The device of claim 4737 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4972. The device of claim 4737 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
4973. The device of claim 4737 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4974. The device of claim 4737 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4975. The device of claim 4737 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4976. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4977. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4978. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4979. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4980. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
4981. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.~
4982. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4983. The device of claim 4737 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4984. The device of claim 4737 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
4985. The device of claim 4737 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
4986. The device of claim 4737 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4987. The device of claim 4737 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4988. The device of claim 4737 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4989. The device of claim 4737 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4990. The device of claim 4737 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4991. The device of claim 4737 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4992. The device of claim 4737 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4993. The device of claim 4737 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4994. The device of claim 4737 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4995. A device, comprising an implantable nonvascular stent or tube (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4996. The device of claim 4995 wherein the agent inhibits cell regeneration.
4997. The device of claim 4995 wherein the agent inhibits angiogenesis.
4998. The device of claim 4995 wherein the agent inhibits fibroblast migration.
4999. The device of claim 4995 wherein the agent inhibits fibroblast proliferation.
5000. The device of claim 4995 wherein the agent inhibits deposition of extracellular matrix.
5001. The device of claim 4995 wherein the agent inhibits tissue remodeling.
5002. The device of claim 4995 wherein the agent is an angiogenesis inhibitor.
5003. The device of claim 4995 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
5004. The device of claim 4995 wherein the agent is a chemokine receptor antagonist.
5005. The device of claim 4995 wherein the agent is a cell cycle inhibitor.
5006. The device of claim 4995 wherein the agent is a taxane.
5007. The device of claim 4995 wherein the agent is an anti-microtubule agent.
5008. The device of claim 4995 wherein the agent is paclitaxel.
5009. The device of claim 4995 wherein the agent is not paclitaxel.
5010. The device of claim 4995 wherein the agent is an analogue or derivative of paclitaxel.
5011. The device of claim 4995 wherein the agent is a vinca alkaloid.
5012. The device of claim 4995 wherein the agent is camptothecin or an analogue or derivative thereof.
5013. The device of claim 4995 wherein the agent is a podophyllotoxin.
5014. The device of claim 4995 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
5015. The device of claim 4995 wherein the agent is an anthracycline.
5016. The device of claim 4995 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
5017. The device of claim 4995 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
5018. The device of claim 4995 wherein the agent is a platinum compound.
5019. The device of claim 4995 wherein the agent is a nitrosourea.
5020. The device of claim 4995 wherein the agent is a nitroimidazole.
5021. The device of claim 4995 wherein the agent is a folic acid antagonist.
5022. The device of claim 4995 wherein the agent is a cytidine analogue.
5023. The device of claim 4995 wherein the agent is a pyrimidine analogue.
5024. The device of claim 4995 wherein the agent is a fluoropyrimidine analogue.
5025. The device of claim 4995 wherein the agent is a purine analogue.
5026. The device of claim 4995 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
5027. The device of claim 4995 wherein the agent is a hydroxyurea.
5028. The device of claim 4995 wherein the agent is a mytomicin or an analogue or derivative thereof.
5029. The device of claim 4995 wherein the agent is an alkyl sulfonate.
5030. The device of claim 4995 wherein the agent is a benzamide or an analogue or derivative thereof.
5031. The device of claim 4995 wherein the agent is a nicotinamide or an analogue or derivative thereof.
5032. The device of claim 4995 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
5033. The device of claim 4995 wherein the agent is a DNA
alkylating agent.
5034. The device of claim 4995 wherein the agent is an anti-microtubule agent.
5035. The device of claim 4995 wherein the agent is a topoisomerase inhibitor.
5036. The device of claim 4995 wherein the agent is a DNA
cleaving agent.
5037. The device of claim 4995 wherein the agent is an antimetabolite.
5038. The device of claim 4995 wherein the agent inhibits adenosine deaminase.
5039. The device of claim 4995 wherein the agent inhibits purine ring synthesis.
5040. The device of claim 4995 wherein the agent is a nucleotide interconversion inhibitor.
5041. The device of claim 4995 wherein the agent inhibits dihydrofolate reduction.
5042. The device of claim 4995 wherein the agent blocks thymidine monophosphate.
5043. The device of claim 4995 wherein the agent causes DNA
damage.
5044. The device of claim 4995 wherein the agent is a DNA
intercalation agent.
5045. The device of claim 4995 wherein the agent is a RNA
synthesis inhibitor.
5046. The device of claim 4995 wherein the agent is a pyrimidine synthesis inhibitor.
5047. The device of claim 4995 wherein the agent inhibits ribonucleotide synthesis or function.
5048. The device of claim 4995 wherein the agent inhibits thymidine monophosphate synthesis or function.~
5049. The device of claim 4995 wherein the agent inhibits DNA
synthesis.
5050. The device of claim 4995 wherein the agent causes DNA
adduct formation.
5051. The device of claim 4995 wherein the agent inhibits protein synthesis.
5052. The device of claim 4995 wherein the agent inhibits microtubule function.
5053. The device of claim 4995 wherein the agent is a cyclin dependent protein kinase inhibitor.
5054. The device of claim 4995 wherein the agent is an epidermal growth factor kinase inhibitor.
5055. The device of claim 4995 wherein the agent is an elastase inhibitor.
5056. The device of claim 4995 wherein the agent is a factor Xa inhibitor.
5057. The device of claim 4995 wherein the agent is a farnesyltransferase inhibitor.
5058. The device of claim 4995 wherein the agent is a fibrinogen antagonist.
5059. The device of claim 4995 wherein the agent is a guanylate cyclase stimulant.
5060. The device of claim 4995 wherein the agent is a heat shock protein 90 antagonist.
5061. The device of claim 4995 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
5062. The device of claim 4995 wherein the agent is a guanylate cyclase stimulant.
5063. The device of claim 4995 wherein the agent is a HMGCoA
reductase inhibitor.
5064. The device of claim 4995 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
5065. The device of claim 4995 wherein the agent is a hydroorotate dehydrogenase inhibitor.
5066. The device of claim 4995 wherein the agent is an IKK2 inhibitor.
5067. The device of claim 4995 wherein the agent is an IL-1 antagonist.
5068. The device of claim 4995 wherein the agent is an ICE
antagonist.
5069. The device of claim 4995 wherein the agent is an IRAK
antagonist.
5070. The device of claim 4995 wherein the agent is an IL-4 agonist.
5071. The device of claim 4995 wherein the agent is an immunomodulatory agent.
5072. The device of claim 4995 wherein the agent is sirolimus or an analogue or derivative thereof.
5073. The device of claim 4995 wherein the agent is not sirolimus.
5074. The device of claim 4995 wherein the agent is everolimus or an analogue or derivative thereof.
5075. The device of claim 4995 wherein the agent is tacrolimus or an analogue or derivative thereof.
5076. The device of claim 4995 wherein the agent is not tacrolimus.
5077. The device of claim 4995 wherein the agent is biolmus or an analogue or derivative thereof.
5078. The device of claim 4995 wherein the agent is tresperimus or an analogue or derivative thereof.
5079. The device of claim 4995 wherein the agent is auranofin or an analogue or derivative thereof.
5080. The device of claim 4995 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
5081. The device of claim 4995 wherein the agent is gusperimus or an analogue or derivative thereof.
5082. The device of claim 4995 wherein the agent is pimecrolimus or an analogue or derivative thereof.
5083. The device of claim 4995 wherein the agent is ABT-578 or an analogue or derivative thereof.
5084. The device of claim 4995 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
5085. The device of claim 4995 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
5086. The device of claim 4995 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
5087. The device of claim 4995 wherein the agent is a leukotriene inhibitor.
5088. The device of claim 4995 wherein the agent is a MCP-1 antagonist.
5089. The device of claim 4995 wherein the agent is a MMP
inhibitor.
5090. The device of claim 4995 wherein the agent is an NF
kappa B inhibitor.
5091. The device of claim 4995 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
5092. The device of claim 4995 wherein the agent is an NO
agonist.
5093. The device of claim 4995 wherein the agent is a p38 MAP
kinase inhibitor.
5094. The device of claim 4995 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
5095. The device of claim 4995 wherein the agent is a phosphodiesterase inhibitor.
5096. The device of claim 4995 wherein the agent is a TGF beta inhibitor.
5097. The device of claim 4995 wherein the agent is a thromboxane A2 antagonist.
5098. The device of claim 4995 wherein the agent is a TNFa antagonist.
5099. The device of claim 4995 wherein the agent is a TACE
inhibitor.
5100. The device of claim 4995 wherein the agent is a tyrosine kinase inhibitor.
5101. The device of claim 4995 wherein the agent is a vitronectin inhibitor.
5102. The device of claim 4995 wherein the agent is a fibroblast growth factor inhibitor.
5103. The device of claim 4995 wherein the agent is a protein kinase inhibitor.
5104. The device of claim 4995 wherein the agent is a PDGF
receptor kinase inhibitor.
5105. The device of claim 4995 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
5106. The device of claim 4995 wherein the agent is a retinoic acid receptor antagonist.
5107. The device of claim 4995 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
5108. The device of claim 4995 wherein the agent is a fibronogin antagonist.
5109. The device of claim 4995 wherein the agent is an antimycotic agent.
5110. The device of claim 4995 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
5111. The device of claim 4995 wherein the agent is a bisphosphonate.
5112. The device of claim 4995 wherein the agent is a phospholipase A1 inhibitor.
5113. The device of claim 4995 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
5114. The device of claim 4995 wherein the agent is a macrolide antibiotic.
5115. The device of claim 4995 wherein the agent is a GPIIb/IIIa receptor antagonist.
5116. The device of claim 4995 wherein the agent is an endothelin receptor antagonist.
5117. The device of claim 4995 wherein the agent is a peroxisome proliferator-activated receptor agonist.
5118. The device of claim 4995 wherein the agent is an estrogen receptor agent.
5119. The device of claim 4995 wherein the agent is a somastostatin analogue.
5120. The device of claim 4995 wherein the agent is a neurokinin 1 antagonist.
5121. The device of claim 4995 wherein the agent is a neurokinin 3 antagonist.
5122. The device of claim 4995 wherein the agent is a VLA-4 antagonist.
5123. The device of claim 4995 wherein the agent is an osteoclast inhibitor.
5124. The device of claim 4995 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
5125. The device of claim 4995 wherein the agent is an angiotensin I converting enzyme inhibitor.
5126. The device of claim 4995 wherein the agent is an angiotensin II antagonist.
5127. The device of claim 4995 wherein the agent is an enkephalinase inhibitor.
5128. The device of claim 4995 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
5129. The device of claim 4995 wherein the agent is a protein kinase C inhibitor.
5130. The device of claim 4995 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
5131. The device of claim 4995 wherein the agent is a CXCR3 inhibitor.
5132. The device of claim 4995 wherein the agent is an Itk inhibitor.
5133. The device of claim 4995 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
5134. The device of claim 4995 wherein the agent is a PPAR
agonist.
5135. The device of claim 4995 wherein the agent is an immunosuppressant.
5136. The device of claim 4995 wherein the agent is an Erb inhibitor.
5137. The device of claim 4995 wherein the agent is an apoptosis agonist.
5133. The device of claim 4995 wherein the agent is a lipocortin agonist.
5139. The device of claim 4995 wherein the agent is a VCAM-1 antagonist.
5140. The device of claim 4995 wherein the agent is a collagen antagonist.
5141. The device of claim 4995 wherein the agent is an alpha 2 integrin antagonist.
5142. The device of claim 4995 wherein the agent is a TNF alpha inhibitor.
5143. The device of claim 4995 wherein the agent is a nitric oxide inhibitor
5144. The device of claim 4995 wherein the agent is a cathepsin inhibitor.
5145. The device of claim 4995 wherein the agent is not an anti-inflammatory agent.
5146. The device of claim 4995 wherein the agent is not a steroid.
5147. The device of claim 4995 wherein the agent is not a glucocorticosteroid.
5148. The device of claim 4995 wherein the agent is not dexamethasone.
5149. The device of claim 4995 wherein the agent is not an anti-infective agent.
5150. The device of claim 4995 wherein the agent is not an antibiotic.
5151. The device of claim 4995 wherein the agent is not an anti-fungal agent.
5152. The device of claim 4995, further comprising a polymer.
5153. The device of claim 4995, further comprising a polymeric carrier.
5154. The device of claim 4995 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
5155. The device of claim 4995 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
5156. The device of claim 4995, further comprising a coating, wherein the coating comprises the anti-scarring agent.
5157. The device of claim 4995, further comprising a coating, wherein the coating is disposed on a surface of the device.
5158. The device of claim 4995, further comprising a coating, wherein the coating directly contacts the device.
5159. The device of claim 4995, further comprising a coating, wherein the coating indirectly contacts the device.
5160. The device of claim 4995, further comprising a coating, wherein the coating partially covers the device.
5161. The device of claim 4995, further comprising a coating, wherein the coating completely covers the device.
5162. The device of claim 4995, further comprising a coating, wherein the coating is a uniform coating.
5163. The device of claim 4995, further comprising a coating, wherein the coating is a non-uniform coating.
5164. The device of claim 4995, further comprising a coating, wherein the coating is a discontinuous coating.
5165. The device of claim 4995, further comprising a coating, wherein the coating is a patterned coating.
5166. The device of claim 4995, further comprising a coating, wherein the coating has a thickness of 100 µm or less.
5167. The device of claim 4995, further comprising a coating, wherein the coating has a thickness of 10 µm or less.
5168. The device of claim 4995, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
5169. The device of claim 4995, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
5170. The device of claim 4995, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5171. The device of claim 4995, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5172. The device of claim 4995, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5173. The device of claim 4995, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5174. The device of claim 4995, further comprising a coating, wherein the coating further comprises a polymer.
5175. The device of claim 4995, further comprising a first coating having a first composition and the second coating having a second composition.
5176. The device of claim 4995, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
5177. The device of claim 4995, further comprising a polymer.
5178. The device of claim 4995, further comprising a polymeric carrier.
5179. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
5180. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
5181. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
5182. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
5183. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
5184. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
5185. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
5186. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
5187. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
5188. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
5189. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
5190. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
5191. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
5192. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
5193. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
5194. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
5195. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
5196. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
5197. The device of claim 4995, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
5198. The device of claim 4995, further comprising a lubricious coating.
5199. The device of claim 4995 wherein the anti-scarring agent is located within pores or holes of the device.
5200. The device of claim 4995 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
5201. The device of claim 4995, further comprising a second pharmaceutically active agent.
5202. The device of claim 4995, further comprising an anti-inflammatory agent.
5203. The device of claim 4995, further comprising an agent that inhibits infection.
5204. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
5205. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
5206. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
5207. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
5208. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
5209. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
5210. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
5211. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
5212. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is etoposide.
5213. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
5214. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
5215. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
5216. The device of claim 4995, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
5217. The device of claim 4995, further comprising an anti-thrombotic agent.
5218. The device of claim 4995, further comprising a visualization agent.
5219. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5220. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
5221. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
5222. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
5223. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
5224. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
5225. The device of claim 4995, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
5226. The device of claim 4995, further comprising an echogenic material.
5227. The device of claim 4995, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
5228. The device of claim 4995 wherein the device is sterile.
5229. The device of claim 4995 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
5230. The device of claim 4995 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
5231. The device of claim 4995 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
5232. The device of claim 4995 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
5233. The device of claim 4995 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
5234. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
5235. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
5236. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
5237. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
5238. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
5239. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
5240. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
5241. The device of claim 4995 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
5242. The device of claim 4995 wherein the device comprises about 0.01 µg to about 10 µg of the anti-scarring agent.
5243. The device of claim 4995 wherein the device comprises about 10 µg to about 10 mg of the anti-scarring agent.
5244. The device of claim 4995 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
5245. The device of claim 4995 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
5246. The device of claim 4995 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
5247. The device of claim 4995 wherein a surface of the device comprises less than 0.01 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5248. The device of claim 4995 wherein a surface of the device comprises about 0.01 µg to about 1 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5249. The device of claim 4995 wherein a surface of the device comprises about 1 µg to about 10 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5250. The device of claim 4995 wherein a surface of the device comprises about 10 µg to about 250 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5251. The device of claim 4995 wherein a surface of the device comprises about 250 µg to about 1000 µg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5252. The device of claim 4995 wherein a surface of the device comprises about 1000 µg to about 2500 µg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5253. A method for inhibiting scarring comprising placing an intravascular implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5254. The method of claim 5253 wherein the agent inhibits cell regeneration.
5255. The method of claim 5253 wherein the agent inhibits angiogenesis.
5256. The method of claim 5253 wherein the agent inhibits fibroblast migration.
5257. The method of claim 5253 wherein the agent inhibits fibroblast proliferation.
5258. The method of claim 5253 wherein the agent inhibits deposition of extracellular matrix.
5259. The method of claim 5253 wherein the agent inhibits tissue remodeling.
5260. The method of claim 5253 wherein the agent is an angiogenesis inhibitor.
5261. The method of claim 5253 wherein the agent is a 5-lipoxygenase inhibitor or antagonist.
5262. The method of claim 5253 wherein the agent is a chemokine receptor antagonist.
5263. The method of claim 5253 wherein the agent is a cell cycle inhibitor.
5264. The method of claim 5253 wherein the agent is a taxane.
5265. The method of claim 5253 wherein the agent is an anti-microtubule agent.
5266. The method of claim 5253 wherein the agent is paclitaxel.
5267. The method of claim 5253 wherein the agent is not paclitaxel.
5263. The method of claim 5253 wherein the agent is an analogue or derivative of paclitaxel.
5269. The method of claim 5253 wherein the agent is a vinca alkaloid.
5270. The method of claim 5253 wherein the agent is camptothecin or an analogue or derivative thereof.
5271. The method of claim 5253 wherein the agent is a podophyllotoxin.
5272. The method of claim 5253 wherein the agent is a podophyllotoxin, wherein the podophyllotoxin is etoposide or an analogue or derivative thereof.
5273. The method of claim 5253 wherein the agent is an anthracycline.
5274. The method of claim 5253 wherein the agent is an anthracycline, wherein the anthracycline is doxorubicin or an analogue or derivative thereof.
5275. The method of claim 5253 wherein the agent is an anthracycline, wherein the anthracycline is mitoxantrone or an analogue or derivative thereof.
5276. The method of claim 5253 wherein the agent is a platinum compound.
5277. The method of claim 5253 wherein the agent is a nitrosourea.
5278. The method of claim 5253 wherein the agent is a nitroimidazole.
5279. The method of claim 5253 wherein the agent is a folic acid antagonist.
5280. The method of claim 5253 wherein the agent is a cytidine analogue.
5281. The method of claim 5253 wherein the agent is a pyrimidine analogue.
5282. The method of claim 5253 wherein the agent is a fluoropyrimidine analogue.
5283. The method of claim 5253 wherein the agent is a purine analogue.
5284. The method of claim 5253 wherein the agent is a nitrogen mustard or an analogue or derivative thereof.
5285. The method of claim 5253 wherein the agent is a hydroxyurea.
5286. The method of claim 5253 wherein the agent is a mytomicin or an analogue or derivative thereof.
5287. The method of claim 5253 wherein the agent is an alkyl sulfonate.
5288. The method of claim 5253 wherein the agent is a benzamide or an analogue or derivative thereof.
5289. The method of claim 5253 wherein the agent is a nicotinamide or an analogue or derivative thereof.
5290. The method of claim 5253 wherein the agent is a halogenated sugar or an analogue or derivative thereof.
5291. The method of claim 5253 wherein the agent is a DNA
alkylating agent.
5292. The method of claim 5253 wherein the agent is an anti-microtubule agent.
5293. The method of claim 5253 wherein the agent is a topoisomerase inhibitor.
5294. The method of claim 5253 wherein the agent is a DNA
cleaving agent.
5295. The method of claim 5253 wherein the agent is an antimetabolite.
5296. The method of claim 5253 wherein the agent inhibits adenosine deaminase.
5297. The method of claim 5253 wherein the agent inhibits purine ring synthesis.
5298. The method of claim 5253 wherein the agent is a nucleotide interconversion inhibitor.
5299. The method of claim 5253 wherein the agent inhibits dihydrofolate reduction.
5300. The method of claim 5253 wherein the agent blocks thymidine monophosphate.
5301. The method of claim 5253 wherein the agent causes DNA
damage.
5302. The method of claim 5253 wherein the agent is a DNA
intercalation agent.
5303. The method of claim 5253 wherein the agent is a RNA
synthesis inhibitor.
5304. The method of claim 5253 wherein the agent is a pyrimidine synthesis inhibitor.
5305. The method of claim 5253 wherein the agent inhibits ribonucleotide synthesis or function.
5306. The method of claim 5253 wherein the agent inhibits thymidine monophosphate synthesis or function.
5307. The method of claim 5253 wherein the agent inhibits DNA
synthesis.
5308. The method of claim 5253 wherein the agent causes DNA
adduct formation.
5309. The method of claim 5253 wherein the agent inhibits protein synthesis.
5310. The method of claim 5253 wherein the agent inhibits microtubule function.
5311. The method of claim 5253 wherein the agent is a cyclin dependent protein kinase inhibitor.
5312. The method of claim 5253 wherein the agent is an epidermal growth factor kinase inhibitor.
5313. The method of claim 5253 wherein the agent is an elastase inhibitor.
5314. The method of claim 5253 wherein the agent is a factor Xa inhibitor.
5315. The method of claim 5253 wherein the agent is a farnesyltransferase inhibitor.
5316. The method of claim 5253 wherein the agent is a fibrinogen antagonist.
5317. The method of claim 5253 wherein the agent is a guanylate cyclase stimulant.
5318. The method of claim 5253 wherein the agent is a heat shock protein 90 antagonist.
5319. The method of claim 5253 wherein the agent is a heat shock protein 90 antagonist, wherein the heat shock protein 90 antagonist is geldanamycin or an analogue or derivative thereof.
5320. The method of claim 5253 wherein the agent is a guanylate cyclase stimulant.
5321. The method of claim 5253 wherein the agent is a HMGCoA
reductase inhibitor.
5322. The method of claim 5253 wherein the agent is a HMGCoA
reductase inhibitor, wherein the HMGCoA reductase inhibitor is simvastatin or an analogue or derivative thereof.
5323. The method of claim 5253 wherein the agent is a hydroorotate dehydrogenase inhibitor.
5324. The method of claim 5253 wherein the agent is an IKK2 inhibitor.
5325. The method of claim 5253 wherein the agent is an IL-1 antagonist.
5326. The method of claim 5253 wherein the agent is an ICE
antagonist.
5327. The method of claim 5253 wherein the agent is an IRAK
antagonist.
5328. The method of claim 5253 wherein the agent is an IL-4 agonist.
5329. The method of claim 5253 wherein the agent is an immunomodulatory agent.
5330. The method of claim 5253 wherein the agent is sirolimus or an analogue or derivative thereof.
5331. The method of claim 5253 wherein the agent is not sirolimus.
5332. The method of claim 5253 wherein the agent is everolimus or an analogue or derivative thereof.
5333. The method of claim 5253 wherein the agent is tacrolimus or an analogue or derivative thereof.
5334. The method of claim 5253 wherein the agent is not tacrolimus.
5335. The method of claim 5253 wherein the agent is biolmus or an analogue or derivative thereof.
5336. The method of claim 5253 wherein the agent is tresperimus or an analogue or derivative thereof.
5337. The method of claim 5253 wherein the agent is auranofin or an analogue or derivative thereof.
5338. The method of claim 5253 wherein the agent is 27-0-demethylrapamycin or an analogue or derivative thereof.
5339. The method of claim 5253 wherein the agent is gusperimus or an analogue or derivative thereof.
5340. The method of claim 5253 wherein the agent is pimecrolimus or an analogue or derivative thereof.
5341. The method of claim 5253 wherein the agent is ABT-578 or an analogue or derivative thereof.
5342. The method of claim 5253 wherein the agent is an inosine monophosphate dehydrogenase (IMPDH) inhibitor.
5343. The method of claim 5253 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is mycophenolic acid or an analogue or derivative thereof.
5344. The method of claim 5253 wherein the agent is an IMPDH
inhibitor, wherein the IMPDH inhibitor is 1-alpha-25 dihydroxy vitamin D3 or an analogue or derivative thereof.
5345. The method of claim 5253 wherein the agent is a leukotriene inhibitor.
5346. The method of claim 5253 wherein the agent is a MCP-1 antagonist.
5347. The method of claim 5253 wherein the agent is a MMP
inhibitor.
5348. The method of claim 5253 wherein the agent is an NF
kappa B inhibitor.
5349. The method of claim 5253 wherein the agent is an NF
kappa B inhibitor, wherein the NF kappa B inhibitor is Bay 11-7082.
5350. The method of claim 5253 wherein the agent is an NO
agonist.
5351. The method of claim 5253 wherein the agent is a p38 MAP
kinase inhibitor.
5352. The method of claim 5253 wherein the agent is a p38 MAP
kinase inhibitor, wherein the p38 MAP kinase inhibitor is SB 202190.
5353. The method of claim 5253 wherein the agent is a phosphodiesterase inhibitor.
5354. The method of claim 5253 wherein the agent is a TGF beta inhibitor.
5355. The method of claim 5253 wherein the agent is a thromboxane A2 antagonist.
5356. The method of claim 5253 wherein the agent is a TNFa antagonist.
5357. The method of claim 5253 wherein the agent is a TACE
inhibitor.
5358. The method of claim 5253 wherein the agent is a tyrosine kinase inhibitor.
5359. The method of claim 5253 wherein the agent is a vitronectin inhibitor.
5360. The method of claim 5253 wherein the agent is a fibroblast growth factor inhibitor.
5361. The method of claim 5253 wherein the agent is a protein kinase inhibitor.
5362. The method of claim 5253 wherein the agent is a PDGF
receptor kinase inhibitor.
5363. The method of claim 5253 wherein the agent is an endothelial growth factor receptor kinase inhibitor.
5364. The method of claim 5253 wherein the agent is a retinoic acid receptor antagonist.
5365. The method of claim 5253 wherein the agent is a platelet derived growth factor receptor kinase inhibitor.
5366. The method of claim 5253 wherein the agent is a fibronogin antagonist.
5367. The method of claim 5253 wherein the agent is an antimycotic agent.
5368. The method of claim 5253 wherein the agent is an antimycotic agent, wherein the antimycotic agent is sulconizole.
5369. The method of claim 5253 wherein the agent is a bisphosphonate.
5370. The method of claim 5253 wherein the agent is a phospholipase A1 inhibitor.
5371. The method of claim 5253 wherein the agent is a histamine H1/H2/H3 receptor antagonist.
5372. The method of claim 5253 wherein the agent is a macrolide antibiotic.
5373. The method of claim 5253 wherein the agent is a GPIIb/IIIa receptor antagonist.
5374. The method of claim 5253 wherein the agent is an endothelin receptor antagonist.
5375. The method of claim 5253 wherein the agent is a peroxisome proliferator-activated receptor agonist.
5376. The method of claim 5253 wherein the agent is an estrogen receptor agent.
5377. The method of claim 5253 wherein the agent is a somastostatin analogue.
5373. The method of claim 5253 wherein the agent is a neurokinin 1 antagonist.
5379. The method of claim 5253 wherein the agent is a neurokinin 3 antagonist.
5380. The method of claim 5253 wherein the agent is a VLA-4 antagonist.
5381. The method of claim 5253 wherein the agent is an osteoclast inhibitor.
5382. The method of claim 5253 wherein the agent is a DNA
topoisomerase ATP hydrolyzing inhibitor.
5383. The method of claim 5253 wherein the agent is an angiotensin I converting enzyme inhibitor.
5384. The method of claim 5253 wherein the agent is an angiotensin II antagonist.
5385. The method of claim 5253 wherein the agent is an enkephalinase inhibitor.
5386. The method of claim 5253 wherein the agent is a peroxisome proliferator-activated receptor gamma agonist insulin sensitizer.
5387. The method of claim 5253 wherein the agent is a protein kinase C inhibitor.
5388. The method of claim 5253 wherein the agent is a ROCK
(rho-associated kinase) inhibitor.
5389. The method of claim 5253 wherein the agent is a CXCR3 inhibitor.
5390. The method of claim 5253 wherein the agent is an Itk inhibitor.
5391. The method of claim 5253 wherein the agent is a cytosolic phospholipase A2-alpha inhibitor.
5392. The method of claim 5253 wherein the agent is a PPAR
agonist.
5393. The method of claim 5253 wherein the agent is an immunosuppressant.
5394. The method of claim 5253 wherein the agent is an Erb inhibitor.
5395. The method of claim 5253 wherein the agent is an apoptosis agonist.
5396. The method of claim 5253 wherein the agent is a lipocortin agonist.
5397. The method of claim 5253 wherein the agent is a VCAM-1 antagonist.
5398. The method of claim 5253 wherein the agent is a collagen antagonist.
5399. The method of claim 5253 wherein the agent is an alpha 2 integrin antagonist.
5400. The method of claim 5253 wherein the agent is a TNF
alpha inhibitor.
5401. The method of claim 5253 wherein the agent is a nitric oxide inhibitor
5402. The method of claim 5253 wherein the agent is a cathepsin inhibitor.
5403. The method of claim 5253 wherein the agent is not an anti-inflammatory agent.
5404. The method of claim 5253 wherein the agent is not a steroid.
5405. The method of claim 5253 wherein the agent is not a glucocorticosteroid.
5406. The method of claim 5253 wherein the agent is not dexamethasone.
5407. The method of claim 5253 wherein the agent is not an anti-infective agent.
5408. The method of claim 5253 wherein the agent is not an antibiotic.
5409. The method of claim 5253 wherein the agent is not an anti-fungal agent.
5410. The method of claim 5253, wherein the composition comprises a polymer.
5411. The method of claim 5253, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
5412. The method of claim 5253, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5413. The method of claim 5253, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5414. The method of claim 5253, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
5415. The method of claim 5253, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-biodegradable polymer.
5416. The method of claim 5253, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
CA002536042A 2003-11-10 2004-11-10 Medical implants and anti-scarring agents Abandoned CA2536042A1 (en)

Applications Claiming Priority (15)

Application Number Priority Date Filing Date Title
US51878503P 2003-11-10 2003-11-10
US60/518,785 2003-11-10
US52390803P 2003-11-20 2003-11-20
US52402303P 2003-11-20 2003-11-20
US60/523,908 2003-11-20
US60/524,023 2003-11-20
US52522603P 2003-11-24 2003-11-24
US60/525,226 2003-11-24
US52654103P 2003-12-03 2003-12-03
US60/526,541 2003-12-03
US57847104P 2004-06-09 2004-06-09
US60/578,471 2004-06-09
US58686104P 2004-07-09 2004-07-09
US60/586,861 2004-07-09
PCT/US2004/037426 WO2005049105A2 (en) 2003-11-10 2004-11-10 Medical implants and anti-scarring agents

Publications (1)

Publication Number Publication Date
CA2536042A1 true CA2536042A1 (en) 2005-06-02

Family

ID=34596276

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002536042A Abandoned CA2536042A1 (en) 2003-11-10 2004-11-10 Medical implants and anti-scarring agents

Country Status (7)

Country Link
US (12) US20050181977A1 (en)
EP (1) EP1682196A2 (en)
JP (1) JP2007516740A (en)
AU (1) AU2004291062A1 (en)
CA (1) CA2536042A1 (en)
IL (1) IL174636A0 (en)
WO (2) WO2005049105A2 (en)

Families Citing this family (472)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8603511B2 (en) 1996-08-27 2013-12-10 Baxter International, Inc. Fragmented polymeric compositions and methods for their use
US7435425B2 (en) * 2001-07-17 2008-10-14 Baxter International, Inc. Dry hemostatic compositions and methods for their preparation
US6066325A (en) 1996-08-27 2000-05-23 Fusion Medical Technologies, Inc. Fragmented polymeric compositions and methods for their use
US8303981B2 (en) 1996-08-27 2012-11-06 Baxter International Inc. Fragmented polymeric compositions and methods for their use
US6309384B1 (en) 1999-02-01 2001-10-30 Adiana, Inc. Method and apparatus for tubal occlusion
US8702727B1 (en) 1999-02-01 2014-04-22 Hologic, Inc. Delivery catheter with implant ejection mechanism
AU2001253479A1 (en) * 2000-04-13 2001-10-30 Sts Biopolymers, Inc. Targeted therapeutic agent release devices and methods of making and using the same
US7416559B2 (en) * 2000-10-27 2008-08-26 Poly-Med, Inc. Micromantled drug-eluting stent
US8299205B2 (en) * 2000-10-27 2012-10-30 Poly-Med, Inc. Acetone-soluble, absorbable, crystalline polyaxial copolymers and applications thereof
US8038708B2 (en) 2001-02-05 2011-10-18 Cook Medical Technologies Llc Implantable device with remodelable material and covering material
US7771468B2 (en) * 2001-03-16 2010-08-10 Angiotech Biocoatings Corp. Medicated stent having multi-layer polymer coating
US8741335B2 (en) 2002-06-14 2014-06-03 Hemcon Medical Technologies, Inc. Hemostatic compositions, assemblies, systems, and methods employing particulate hemostatic agents formed from hydrophilic polymer foam such as Chitosan
DE10212815A1 (en) * 2002-03-22 2003-10-02 Ernst Fuchs Distraction device for osteogenesis
US8347891B2 (en) 2002-04-08 2013-01-08 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for performing a non-continuous circumferential treatment of a body lumen
US7756583B2 (en) 2002-04-08 2010-07-13 Ardian, Inc. Methods and apparatus for intravascularly-induced neuromodulation
US20060216267A1 (en) * 2002-08-20 2006-09-28 Kovacs Stephen G Hydrophobic elastomeric polymer chemistry device for inhibiting the growth of onychomycosis and urushiol-induced allergic contact dermatitis
US7744651B2 (en) 2002-09-18 2010-06-29 Warsaw Orthopedic, Inc Compositions and methods for treating intervertebral discs with collagen-based materials
US20040054414A1 (en) 2002-09-18 2004-03-18 Trieu Hai H. Collagen-based materials and methods for augmenting intervertebral discs
CN100394989C (en) 2002-11-15 2008-06-18 华沙整形外科股份有限公司 Collagen-based materials and methods for augmenting intervertebral discs
CA2518960C (en) 2003-03-14 2013-08-27 Sinexus, Inc. Sinus delivery of sustained release therapeutics
US20040225250A1 (en) 2003-05-05 2004-11-11 Michael Yablonski Internal shunt and method for treating glaucoma
US8834864B2 (en) * 2003-06-05 2014-09-16 Baxter International Inc. Methods for repairing and regenerating human dura mater
US7704225B2 (en) * 2005-07-29 2010-04-27 L-Vad Technology, Inc. Percutaneous access device system facilitating cell growth thereon
US7976452B2 (en) * 2003-06-11 2011-07-12 L.Vad Technology, Inc. Long term ambulatory intro-aortic balloon pump with percutaneous access device
US7927626B2 (en) 2003-08-07 2011-04-19 Ethicon, Inc. Process of making flowable hemostatic compositions and devices containing such compositions
DE202004021944U1 (en) 2003-09-12 2013-07-16 Vessix Vascular, Inc. Selectable eccentric remodeling and / or ablation of atherosclerotic material
US7291125B2 (en) 2003-11-14 2007-11-06 Transcend Medical, Inc. Ocular pressure regulation
US7377939B2 (en) * 2003-11-19 2008-05-27 Synecor, Llc Highly convertible endolumenal prostheses and methods of manufacture
WO2005057272A2 (en) * 2003-12-05 2005-06-23 Innfocus, Llc Improved ocular lens
US8871224B2 (en) 2003-12-09 2014-10-28 Allergan, Inc. Botulinum toxin therapy for skin disorders
US20050149174A1 (en) * 2003-12-18 2005-07-07 Medtronic Vascular, Inc. Medical devices to treat or inhibit restenosis
US20050152942A1 (en) * 2003-12-23 2005-07-14 Medtronic Vascular, Inc. Medical devices to treat or inhibit restenosis
US7901447B2 (en) 2004-12-29 2011-03-08 Boston Scientific Scimed, Inc. Medical devices including a metallic film and at least one filament
US8591568B2 (en) 2004-03-02 2013-11-26 Boston Scientific Scimed, Inc. Medical devices including metallic films and methods for making same
US8998973B2 (en) * 2004-03-02 2015-04-07 Boston Scientific Scimed, Inc. Medical devices including metallic films
US8632580B2 (en) * 2004-12-29 2014-01-21 Boston Scientific Scimed, Inc. Flexible medical devices including metallic films
US20050197687A1 (en) * 2004-03-02 2005-09-08 Masoud Molaei Medical devices including metallic films and methods for making same
US8992592B2 (en) * 2004-12-29 2015-03-31 Boston Scientific Scimed, Inc. Medical devices including metallic films
US20050220734A1 (en) * 2004-04-02 2005-10-06 Allergan, Inc. Therapy for melanin related afflictions
US8057815B2 (en) * 2004-04-19 2011-11-15 Portola Pharmaceuticals, Inc. Methods of treatment with Syk inhibitors
US20050267596A1 (en) * 2004-05-03 2005-12-01 Fulfillium, Inc. A Delaware Corporation Devices and systems for gastric volume control
EP1765204B1 (en) 2004-06-07 2018-12-26 Synthes GmbH Orthopaedic implant with sensors
US8999364B2 (en) 2004-06-15 2015-04-07 Nanyang Technological University Implantable article, method of forming same and method for reducing thrombogenicity
CA2571304A1 (en) 2004-06-23 2006-01-05 Bioprotect Ltd. Device system and method for tissue displacement or separation
WO2006017275A1 (en) 2004-07-13 2006-02-16 The University Of Tennessee Research Foundation Adhesive composition for carrying therapeutic agents as delivery vehicle on coatings applied to vascular grafts
CA2573671A1 (en) * 2004-07-21 2006-02-23 Medtronic, Inc. Methods for reducing or preventing localized fibrosis using sirna
CA2574086A1 (en) * 2004-07-21 2006-02-23 Medtronic, Inc. Medical devices and methods for reducing localized fibrosis
US20060040894A1 (en) * 2004-08-13 2006-02-23 Angiotech International Ag Compositions and methods using hyaluronic acid
US8396548B2 (en) 2008-11-14 2013-03-12 Vessix Vascular, Inc. Selective drug delivery in a lumen
WO2006031899A2 (en) * 2004-09-10 2006-03-23 The Cleveland Clinic Foundation Intraluminal electrode assembly
US9713730B2 (en) 2004-09-10 2017-07-25 Boston Scientific Scimed, Inc. Apparatus and method for treatment of in-stent restenosis
US9974607B2 (en) 2006-10-18 2018-05-22 Vessix Vascular, Inc. Inducing desirable temperature effects on body tissue
US7807192B2 (en) * 2004-10-22 2010-10-05 Collagen Matrix, Inc. Biopolymeric membranes
US9387010B2 (en) 2004-10-28 2016-07-12 Nico Corporation Surgical access assembly and method of using same
US9186175B2 (en) 2004-10-28 2015-11-17 Nico Corporation Surgical access assembly and method of using same
US9456915B2 (en) 2004-11-19 2016-10-04 Fulfilium, Inc. Methods, devices, and systems for obesity treatment
US8070807B2 (en) 2004-11-19 2011-12-06 Fulfillium, Inc. Wireless breach detection
US20080118561A1 (en) * 2004-12-08 2008-05-22 Helen Marie Nugent Materials and Methods for Minimally-Invasive Administration of a Cell-Containing Flowable Composition
US7270655B2 (en) * 2005-01-14 2007-09-18 Haidukewych George J Autologous material delivery apparatus and method
US8190236B2 (en) * 2005-01-24 2012-05-29 Prince Martin R Tourniquet for magnetic resonance angiography, and method of using same
USH2260H1 (en) 2005-02-17 2011-07-05 Angiotech International Ag Stents combined with paclitaxel derivatives
US9452001B2 (en) * 2005-02-22 2016-09-27 Tecres S.P.A. Disposable device for treatment of infections of human limbs
US9204957B2 (en) * 2005-03-17 2015-12-08 Hemcon Medical Technologies, Inc. Systems and methods for hemorrhage control and or tissue repair
US20060224170A1 (en) * 2005-03-30 2006-10-05 Michael Duff Surgical marker clip and method for cholangiography
US20060222627A1 (en) * 2005-03-30 2006-10-05 Andrew Carter Optimizing pharmacodynamics of therapeutic agents for treating vascular tissue
CA2603081C (en) 2005-04-04 2013-09-03 Sinexus, Inc. Device and methods for treating paranasal sinus conditions
US20080221135A1 (en) * 2005-05-13 2008-09-11 Bristol-Myers Squibb Company Combination therapy
US7854760B2 (en) 2005-05-16 2010-12-21 Boston Scientific Scimed, Inc. Medical devices including metallic films
EP1726256A1 (en) * 2005-05-27 2006-11-29 Nederlandse Organisatie voor toegepast-natuurwetenschappelijk Onderzoek TNO Phantom device
US20120143341A1 (en) * 2006-12-12 2012-06-07 Arthrodisc, L.L.C. Devices and methods for visual differentiation of intervertebral spaces
WO2007016251A2 (en) * 2005-07-28 2007-02-08 Cook Incorporated Implantable thromboresistant valve
WO2007033152A2 (en) * 2005-09-12 2007-03-22 Conor Medsystems, Inc. Composition, system and method for modulating release kinetics in implantable drug delivery devices by modifying drug solubility
JP5226517B2 (en) * 2005-09-21 2013-07-03 サーモディクス,インコーポレイティド In vivo formed matrices containing natural biodegradable polysaccharides and their ocular use
CA2625264C (en) 2005-10-13 2015-12-15 Synthes (U.S.A.) Drug-impregnated sleeve for a medical implant
US20070093907A1 (en) * 2005-10-26 2007-04-26 Goupil Dennis W Hydrogel spinal disc implants with swellable articles
US20090035348A1 (en) * 2005-11-22 2009-02-05 Z & Z Medical Holdings, Inc. Dissolution of arterial plaque
US8304383B2 (en) * 2005-11-22 2012-11-06 Atheronova Operations, Inc. Dissolution of arterial plaque
WO2007084549A2 (en) * 2006-01-20 2007-07-26 Filiberto Zadini Drug-eluting stent with atherosclerotic plaques dissolving pharmacological preparation
US20070123994A1 (en) * 2005-11-29 2007-05-31 Ethicon Endo-Surgery, Inc. Internally Placed Gastric Restriction Device
US7638156B1 (en) * 2005-12-19 2009-12-29 Advanced Cardiovascular Systems, Inc. Apparatus and method for selectively coating a medical article
US7513865B2 (en) * 2005-12-20 2009-04-07 Boston Scientific Scimed, Inc. Flattened tubular mesh sling and related methods
US8025915B2 (en) * 2006-01-11 2011-09-27 Schott Ag Method of preparing a macromolecule deterrent surface on a pharmaceutical package
EP3005996B1 (en) 2006-01-17 2019-12-04 Novartis Ag Glaucoma treatment device
CA2637602C (en) 2006-01-17 2014-09-16 Forsight Labs, Llc Drug delivery treatment device
US8636753B2 (en) * 2006-02-08 2014-01-28 Tyrx, Inc. Temporarily stiffened mesh prostheses
US8591531B2 (en) * 2006-02-08 2013-11-26 Tyrx, Inc. Mesh pouches for implantable medical devices
US20070218038A1 (en) 2006-03-17 2007-09-20 Pegasus Biologics, Inc. Stabilized, sterilized collagen scaffolds with active adjuncts attached
US20080183237A1 (en) * 2006-04-18 2008-07-31 Electrocore, Inc. Methods And Apparatus For Treating Ileus Condition Using Electrical Signals
US20100057178A1 (en) * 2006-04-18 2010-03-04 Electrocore, Inc. Methods and apparatus for spinal cord stimulation using expandable electrode
US8019435B2 (en) 2006-05-02 2011-09-13 Boston Scientific Scimed, Inc. Control of arterial smooth muscle tone
WO2007139845A2 (en) * 2006-05-23 2007-12-06 Providence Health System-Oregon D/B/A Providence St. Vincent Medical Center Systems and methods for introducing and applying a bandage structure within a body lumen or hollow body organ
EP2021045B1 (en) * 2006-05-31 2016-03-16 Baxter International Inc. Collagen for use in prevention of peridural fibrosis formation after spinal surgery
US20070289596A1 (en) * 2006-06-14 2007-12-20 Campbell Shannon E Endotracheal cuff and technique for using the same
US7833284B2 (en) * 2006-06-28 2010-11-16 The Cleveland Clinic Foundation Anti-adhesion membrane
US8399619B2 (en) 2006-06-30 2013-03-19 Warsaw Orthopedic, Inc. Injectable collagen material
US8118779B2 (en) 2006-06-30 2012-02-21 Warsaw Orthopedic, Inc. Collagen delivery device
US7722665B2 (en) * 2006-07-07 2010-05-25 Graft Technologies, Inc. System and method for providing a graft in a vascular environment
US8535707B2 (en) 2006-07-10 2013-09-17 Intersect Ent, Inc. Devices and methods for delivering active agents to the osteomeatal complex
WO2008008007A1 (en) * 2006-07-13 2008-01-17 St. Jude Medical Ab An implantable cardiac stimulation drug releasing electrode
US7846361B2 (en) 2006-07-20 2010-12-07 Orbusneich Medical, Inc. Bioabsorbable polymeric composition for a medical device
US10675298B2 (en) 2006-07-27 2020-06-09 Boston Scientific Scimed Inc. Particles
TWI436793B (en) 2006-08-02 2014-05-11 Baxter Int Rapidly acting dry sealant and methods for use and manufacture
US20080039362A1 (en) * 2006-08-09 2008-02-14 Afmedica, Inc. Combination drug therapy for reducing scar tissue formation
US8868211B2 (en) 2006-08-15 2014-10-21 Case Western Reserve University Nerve cuff for implantable electrode
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
CN101528293A (en) 2006-08-30 2009-09-09 戴维·W·史密斯 Method of imparting a mono-axial or multiaxial stiffness to extruded materials and products resulting therefrom
US7780730B2 (en) 2006-09-25 2010-08-24 Iyad Saidi Nasal implant introduced through a non-surgical injection technique
US20080082170A1 (en) * 2006-09-29 2008-04-03 Peterman Marc M Apparatus and methods for surgical repair
US7578842B2 (en) * 2006-10-03 2009-08-25 St. Jude Medical, Inc. Prosthetic heart valves
US20080091228A1 (en) * 2006-10-06 2008-04-17 Vascular Technology Inc. Doppler enabled soft tissue dissector
US7846728B2 (en) * 2006-10-13 2010-12-07 BioStruxs, LLC Tissue engineering in vivo with vascularized scaffolds
CA2666663C (en) 2006-10-18 2016-02-09 Minnow Medical, Inc. System for inducing desirable temperature effects on body tissue
EP2455036B1 (en) 2006-10-18 2015-07-15 Vessix Vascular, Inc. Tuned RF energy and electrical tissue characterization for selective treatment of target tissues
EP2073754A4 (en) 2006-10-20 2012-09-26 Orbusneich Medical Inc Bioabsorbable polymeric composition and medical device background
US7959942B2 (en) 2006-10-20 2011-06-14 Orbusneich Medical, Inc. Bioabsorbable medical device with coating
US20080097517A1 (en) * 2006-10-23 2008-04-24 Webtec Converting, Llc. External Nasal Dilator and Methods of Manufacture
US8088147B2 (en) * 2006-10-24 2012-01-03 Trans1 Inc. Multi-membrane prosthetic nucleus
US9023114B2 (en) 2006-11-06 2015-05-05 Tyrx, Inc. Resorbable pouches for implantable medical devices
US20100063360A1 (en) * 2006-11-28 2010-03-11 Adiana, Inc. Side-arm Port Introducer
US8668703B2 (en) * 2006-12-01 2014-03-11 Wake Forest University Health Sciences Medical devices incorporating collagen inhibitors
US7980000B2 (en) 2006-12-29 2011-07-19 Applied Materials, Inc. Vapor dryer having hydrophilic end effector
US8125498B2 (en) * 2007-01-03 2012-02-28 Siemens Medical Solutions Usa, Inc. Generating a 3D volumetric mask from a closed surface mesh
US8886280B2 (en) 2007-01-23 2014-11-11 The Magstim Company Limited Nerve monitoring device
US20100145178A1 (en) * 2007-01-23 2010-06-10 Kartush Jack M Nerve monitoring device
EP2120693A4 (en) * 2007-01-23 2011-04-27 Jack M Kartush Nerve monitoring device
UY30883A1 (en) * 2007-01-31 2008-05-31 Alcon Res PUNCTURAL PLUGS AND METHODS OF RELEASE OF THERAPEUTIC AGENTS
ES2530862T3 (en) * 2007-02-07 2015-03-06 Cook Medical Technologies Llc Coatings of medical devices to release a therapeutic agent at different rates
US7892595B2 (en) * 2007-02-09 2011-02-22 Medtronic Vascular, Inc Implantable device coating system and method
DE102007007865A1 (en) 2007-02-14 2008-08-21 Jennissen, Herbert, Prof. Dr. Process for the preparation of storable implants with an ultrahydrophilic surface
DE102007010274B4 (en) * 2007-03-02 2010-11-18 Siemens Ag Method for automatic coil selection of a magnetic resonance apparatus, magnetic resonance system, computer program product and electronically readable data carrier
US20080226693A1 (en) * 2007-03-14 2008-09-18 Vipul Bhupendra Dave Apparatus and Method for Making a Polymeric Structure
CA2681090C (en) * 2007-03-15 2013-07-16 The Polymer Technology Group Incorporated Novel pdms-pvp block copolymers
CA2680814A1 (en) * 2007-03-15 2008-09-18 Bioprotect Ltd. Soft tissue fixation devices
ES2593085T3 (en) * 2007-03-15 2016-12-05 Ortho-Space Ltd. Prosthetic devices
DE102007013564B4 (en) * 2007-03-21 2017-11-09 Siemens Healthcare Gmbh Method and device for automatic determination of radiation-attenuating objects by means of a magnetic resonance system
US8642067B2 (en) 2007-04-02 2014-02-04 Allergen, Inc. Methods and compositions for intraocular administration to treat ocular conditions
DE102007016314B3 (en) * 2007-04-04 2009-01-02 Siemens Ag Local coil arrangement for placing on a patient bed and patient bed for receiving a local coil arrangement and magnetic resonance apparatus with a patient bed
WO2008137874A1 (en) * 2007-05-04 2008-11-13 Personics Holdings Inc. Earguard sealing system ii: single chamber systems
US20080279833A1 (en) 2007-05-10 2008-11-13 Matheny Robert G Laminate sheet articles for tissue regeneration
US9283302B2 (en) 2011-12-16 2016-03-15 Cormatrix Cardiovascular, Inc. Extracellular matrix encasement structures and methods
JP2010526635A (en) 2007-05-14 2010-08-05 バイオプロテクト リミテッド Delivery device for delivering biologically active agents to internal tissues of the body
FR2916345B1 (en) * 2007-05-22 2009-08-28 Optisoins Sa DEVICE FOR THE TREATMENT OF THE REAGONMENT OF THE EYE
US9101357B2 (en) * 2007-06-08 2015-08-11 Board Of Trustees Of The University Of Arkansas Physiologic abdominal closure
US7994786B2 (en) * 2007-06-19 2011-08-09 Mary Hitchcock Memorial Hospital System and method for use of nanoparticles in imaging and temperature measurement
US7524512B2 (en) 2007-07-13 2009-04-28 Di Bartolomeo Joseph R Composition and method for the prevention and relief of the symptoms of an incompetent or patulous Eustachian tube
EP2173289A4 (en) 2007-07-17 2010-11-24 Transcend Medical Inc Ocular implant with hydrogel expansion capabilities
US8092541B2 (en) * 2007-08-03 2012-01-10 Warsaw Orthopedic, Inc. Method of using an anti-growth matrix as a barrier for cell attachment and osteo-inductive factors
US8162924B2 (en) * 2007-08-17 2012-04-24 The Invention Science Fund I, Llc System, devices, and methods including actively-controllable superoxide water generating systems
US8702640B2 (en) 2007-08-17 2014-04-22 The Invention Science Fund I, Llc System, devices, and methods including catheters configured to monitor and inhibit biofilm formation
US8647292B2 (en) 2007-08-17 2014-02-11 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having components that are actively controllable between two or more wettability states
US8734718B2 (en) 2007-08-17 2014-05-27 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having an actively controllable therapeutic agent delivery component
US8460229B2 (en) * 2007-08-17 2013-06-11 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having components that are actively controllable between transmissive and reflective states
US8706211B2 (en) 2007-08-17 2014-04-22 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having self-cleaning surfaces
US8753304B2 (en) 2007-08-17 2014-06-17 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having acoustically actuatable waveguide components for delivering a sterilizing stimulus to a region proximate a surface of the catheter
US8366652B2 (en) 2007-08-17 2013-02-05 The Invention Science Fund I, Llc Systems, devices, and methods including infection-fighting and monitoring shunts
US8075908B2 (en) * 2007-08-17 2011-12-13 Anhese Llc Apparatus and method for reducing the occurrence of post-surgical adhesions
US20090048648A1 (en) * 2007-08-17 2009-02-19 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Self-sterilizing device
CA2736663C (en) 2007-09-07 2018-01-02 Surgical Biologics, Llc. Placental tissue grafts and improved methods of preparing and using the same
US20090064999A1 (en) * 2007-09-12 2009-03-12 E. Benson Hood Laboratories Tracheostomy tube with inner cannula and obturator for regular and extra-long stomas
US8771725B2 (en) 2007-10-12 2014-07-08 Chesson Laboratory Associates, Inc. Poly(urea-urethane) compositions useful as topical medicaments and methods of using the same
US8703180B1 (en) * 2007-10-23 2014-04-22 Abbott Cardiovascular Systems Inc. Multiple growth factor compositions, methods of fabrication, and methods of treatment
AR069109A1 (en) * 2007-10-30 2009-12-30 Baxter Int USE OF A REGENERATIVE BIOFUNCTIONAL COLOGEN BIOMATRIZ TO TREAT VISCERAL OR PARIETAL DEFECTS
US20090149958A1 (en) * 2007-11-01 2009-06-11 Ann Prewett Structurally reinforced spinal nucleus implants
WO2009064806A1 (en) * 2007-11-12 2009-05-22 Endologix, Inc. Method and agent for in-situ stabilization of vascular tissue
EP3791826A1 (en) * 2007-12-18 2021-03-17 Intersect ENT, Inc. Self-expanding devices
US8299316B2 (en) * 2007-12-18 2012-10-30 Ethicon, Inc. Hemostatic device
US8629314B2 (en) * 2007-12-18 2014-01-14 Ethicon, Inc. Surgical barriers having adhesion inhibiting properties
US8414646B2 (en) 2007-12-27 2013-04-09 Forsight Labs, Llc Intraocular, accommodating lens and methods of use
WO2009088448A2 (en) 2008-01-03 2009-07-16 Forsight Labs, Llc Intraocular, accomodating lens and methods of use
US9855370B2 (en) * 2008-01-08 2018-01-02 Yale University Compositions and methods for promoting patency of vascular grafts
GB2490834B (en) * 2008-02-06 2013-05-29 Hmicro Inc Wireless communications systems using multiple radios
US9034002B2 (en) 2008-02-18 2015-05-19 Covidien Lp Lock bar spring and clip for implant deployment device
US8317808B2 (en) 2008-02-18 2012-11-27 Covidien Lp Device and method for rolling and inserting a prosthetic patch into a body cavity
US8808314B2 (en) 2008-02-18 2014-08-19 Covidien Lp Device and method for deploying and attaching an implant to a biological tissue
US9393002B2 (en) 2008-02-18 2016-07-19 Covidien Lp Clip for implant deployment device
US9833240B2 (en) 2008-02-18 2017-12-05 Covidien Lp Lock bar spring and clip for implant deployment device
US8758373B2 (en) 2008-02-18 2014-06-24 Covidien Lp Means and method for reversibly connecting a patch to a patch deployment device
US9301826B2 (en) 2008-02-18 2016-04-05 Covidien Lp Lock bar spring and clip for implant deployment device
US9398944B2 (en) 2008-02-18 2016-07-26 Covidien Lp Lock bar spring and clip for implant deployment device
EP2247245B1 (en) 2008-02-18 2017-06-28 Covidien LP A device for deploying and attaching a patch to a biological tissue
US9044235B2 (en) 2008-02-18 2015-06-02 Covidien Lp Magnetic clip for implant deployment device
US9393093B2 (en) 2008-02-18 2016-07-19 Covidien Lp Clip for implant deployment device
US8053020B2 (en) * 2008-02-28 2011-11-08 Cook Medical Technologies Llc Process for coating a portion of an implantable medical device
US8324292B2 (en) * 2008-02-29 2012-12-04 Ethicon, Inc. Medically acceptable formulation of a diisocyanate terminated macromer for use as an internal adhesive or sealant
US8071663B2 (en) * 2008-02-29 2011-12-06 Ethicon, Inc. Medically acceptable formulation of a diisocyanate terminated macromer for use as an internal adhesive or sealant
CA2716872C (en) 2008-02-29 2015-02-10 Ferrosan Medical Devices A/S Device for promotion of hemostasis and/or wound healing
US11254926B2 (en) 2008-04-29 2022-02-22 Virginia Tech Intellectual Properties, Inc. Devices and methods for high frequency electroporation
US9198733B2 (en) 2008-04-29 2015-12-01 Virginia Tech Intellectual Properties, Inc. Treatment planning for electroporation-based therapies
US9283051B2 (en) 2008-04-29 2016-03-15 Virginia Tech Intellectual Properties, Inc. System and method for estimating a treatment volume for administering electrical-energy based therapies
US10272178B2 (en) 2008-04-29 2019-04-30 Virginia Tech Intellectual Properties Inc. Methods for blood-brain barrier disruption using electrical energy
US10702326B2 (en) 2011-07-15 2020-07-07 Virginia Tech Intellectual Properties, Inc. Device and method for electroporation based treatment of stenosis of a tubular body part
US11272979B2 (en) 2008-04-29 2022-03-15 Virginia Tech Intellectual Properties, Inc. System and method for estimating tissue heating of a target ablation zone for electrical-energy based therapies
US10448989B2 (en) 2009-04-09 2019-10-22 Virginia Tech Intellectual Properties, Inc. High-frequency electroporation for cancer therapy
US9867652B2 (en) 2008-04-29 2018-01-16 Virginia Tech Intellectual Properties, Inc. Irreversible electroporation using tissue vasculature to treat aberrant cell masses or create tissue scaffolds
US10245098B2 (en) 2008-04-29 2019-04-02 Virginia Tech Intellectual Properties, Inc. Acute blood-brain barrier disruption using electrical energy based therapy
US8992517B2 (en) 2008-04-29 2015-03-31 Virginia Tech Intellectual Properties Inc. Irreversible electroporation to treat aberrant cell masses
WO2009134876A1 (en) 2008-04-29 2009-11-05 Virginia Tech Intellectual Properties, Inc. Irreversible electroporation to create tissue scaffolds
US10117707B2 (en) 2008-04-29 2018-11-06 Virginia Tech Intellectual Properties, Inc. System and method for estimating tissue heating of a target ablation zone for electrical-energy based therapies
US10238447B2 (en) 2008-04-29 2019-03-26 Virginia Tech Intellectual Properties, Inc. System and method for ablating a tissue site by electroporation with real-time monitoring of treatment progress
US9205170B2 (en) 2008-05-02 2015-12-08 Hemcon Medical Technologies, Inc. Wound dressing devices and methods
WO2009143309A2 (en) * 2008-05-21 2009-11-26 Trustees Of Dartmouth College Female reproductive tract and anal prophylaxes
US8206635B2 (en) 2008-06-20 2012-06-26 Amaranth Medical Pte. Stent fabrication via tubular casting processes
US10898620B2 (en) 2008-06-20 2021-01-26 Razmodics Llc Composite stent having multi-axial flexibility and method of manufacture thereof
US9173704B2 (en) * 2008-06-20 2015-11-03 Angiodynamics, Inc. Device and method for the ablation of fibrin sheath formation on a venous catheter
US8206636B2 (en) 2008-06-20 2012-06-26 Amaranth Medical Pte. Stent fabrication via tubular casting processes
US8617139B2 (en) 2008-06-25 2013-12-31 Transcend Medical, Inc. Ocular implant with shape change capabilities
US20100010519A1 (en) * 2008-07-09 2010-01-14 Joshua Stopek Anastomosis Sheath And Method Of Use
CA2732355A1 (en) 2008-08-01 2010-02-04 Intersect Ent, Inc. Methods and devices for crimping self-expanding devices
US8039433B2 (en) * 2008-08-19 2011-10-18 Warsaw Orthopedic, Inc. Osteogenic compositions containing a coloring agent
US8945142B2 (en) * 2008-08-27 2015-02-03 Cook Medical Technologies Llc Delivery system for implanting nasal ventilation tube
US8353943B2 (en) * 2008-08-29 2013-01-15 Cook Medical Technologies Llc Variable weave graft with metal strand reinforcement for in situ fenestration
GB0816365D0 (en) * 2008-09-08 2008-10-15 Univ Belfast Polymeric material
ES2859624T3 (en) 2008-10-03 2021-10-04 Bard Inc C R Implantable prosthesis
WO2010042540A1 (en) 2008-10-06 2010-04-15 Providence Health System - Oregon Foam medical devices and methods
KR101726885B1 (en) * 2008-10-17 2017-04-26 내셔널 유니버시티 오브 싱가포르 Resorbable scaffolds for bone repair and long bone tissue engineering
EP2792307B1 (en) 2008-10-20 2017-10-04 Covidien LP A device for attaching a patch to a biological tissue
WO2010048189A2 (en) 2008-10-20 2010-04-29 IMDS, Inc. Systems and methods for cerebrospinal fluid repair
FR2937857B1 (en) * 2008-10-30 2015-04-03 Brothier Lab SURGICAL MEMBRANE ANTIADHERENCE
US8996134B2 (en) * 2008-11-07 2015-03-31 W. L. Gore & Associates, Inc. Implantable lead
WO2010056745A1 (en) 2008-11-17 2010-05-20 Minnow Medical, Inc. Selective accumulation of energy with or without knowledge of tissue topography
US9597220B2 (en) 2008-11-19 2017-03-21 Spirox, Inc. Apparatus and methods for correcting nasal valve collapse
US20110208026A1 (en) * 2008-12-04 2011-08-25 Goodall Eleanor V Systems, devices, and methods including implantable devices with anti-microbial properties
WO2010065135A1 (en) 2008-12-04 2010-06-10 Searete, Llc System, devices, and methods including actively-controllable sterilizing excitation delivery implants
US20120041285A1 (en) 2008-12-04 2012-02-16 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems, devices, and methods including implantable devices with anti-microbial properties
US8585627B2 (en) 2008-12-04 2013-11-19 The Invention Science Fund I, Llc Systems, devices, and methods including catheters configured to monitor biofilm formation having biofilm spectral information configured as a data structure
US20110208023A1 (en) * 2008-12-04 2011-08-25 Goodall Eleanor V Systems, devices, and methods including implantable devices with anti-microbial properties
US20110160681A1 (en) * 2008-12-04 2011-06-30 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems, devices, and methods including catheters having light removable coatings based on a sensed condition
US20110208021A1 (en) * 2008-12-04 2011-08-25 Goodall Eleanor V Systems, devices, and methods including implantable devices with anti-microbial properties
US8377122B2 (en) 2009-01-28 2013-02-19 Transcend Medical, Inc. Ocular implant with stiffness qualities, methods of implantation and system
US8883208B2 (en) * 2009-04-08 2014-11-11 Surmodics, Inc. Particles for delivery of nucleic acids and related devices and methods
US11382681B2 (en) 2009-04-09 2022-07-12 Virginia Tech Intellectual Properties, Inc. Device and methods for delivery of high frequency electrical pulses for non-thermal ablation
US11638603B2 (en) 2009-04-09 2023-05-02 Virginia Tech Intellectual Properties, Inc. Selective modulation of intracellular effects of cells using pulsed electric fields
PL2424473T3 (en) * 2009-04-28 2017-10-31 Ostomycure As Manufacturing method for percutaneous implant
US20100280600A1 (en) * 2009-04-30 2010-11-04 Vipul Bhupendra Dave Dual drug stent
CN102573981B (en) 2009-05-15 2016-06-22 因特尔赛克特耳鼻喉公司 Extendable device and using method thereof
US10206813B2 (en) 2009-05-18 2019-02-19 Dose Medical Corporation Implants with controlled drug delivery features and methods of using same
US9039783B2 (en) * 2009-05-18 2015-05-26 Baxter International, Inc. Method for the improvement of mesh implant biocompatibility
US9265633B2 (en) 2009-05-20 2016-02-23 480 Biomedical, Inc. Drug-eluting medical implants
US8992601B2 (en) 2009-05-20 2015-03-31 480 Biomedical, Inc. Medical implants
US8888840B2 (en) * 2009-05-20 2014-11-18 Boston Scientific Scimed, Inc. Drug eluting medical implant
JP5820370B2 (en) * 2009-05-20 2015-11-24 アーセナル メディカル, インコーポレイテッド Medical implant
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
US8903488B2 (en) 2009-05-28 2014-12-02 Angiodynamics, Inc. System and method for synchronizing energy delivery to the cardiac rhythm
WO2010141667A1 (en) * 2009-06-03 2010-12-09 Case Western Reserve University Therapeutic agent delivery system and method
KR101699992B1 (en) * 2009-06-16 2017-01-26 백스터 인터내셔널 인코포레이티드 Hemostatic sponge
US9895189B2 (en) 2009-06-19 2018-02-20 Angiodynamics, Inc. Methods of sterilization and treating infection using irreversible electroporation
ES2435618T3 (en) * 2009-06-25 2013-12-20 Optonol Ltd. Fiber matrix to maintain a soft tissue space
US8709467B2 (en) 2009-06-26 2014-04-29 Cute Lovable Teddy Bear, Llc. Para-xylylene based microfilm elution devices
US8424516B2 (en) * 2009-07-06 2013-04-23 Terence Kernan Gray Ventilating tube and stylet system
US20110033515A1 (en) * 2009-08-04 2011-02-10 Rst Implanted Cell Technology Tissue contacting material
EP2464406B1 (en) * 2009-08-10 2016-10-26 Proviflo, LLC Catheter lock solutions utilizing tocopherol and mid-chain fatty acids
DE102009036939A1 (en) * 2009-08-11 2011-02-24 Siemens Aktiengesellschaft Head coil for a magnetic resonance tomography device with exchangeable operating elements
US8361591B2 (en) * 2009-08-12 2013-01-29 Medos International Sarl Packaging with active protection layer
US8313811B2 (en) * 2009-08-12 2012-11-20 Medos International S.A.R.L. Plasma enhanced polymer ultra-thin multi-layer packaging
US8313819B2 (en) 2009-08-12 2012-11-20 Medos International S.A.R.L. Ultra-thin multi-layer packaging
AU2010286117B9 (en) 2009-08-17 2014-07-10 Covidien Lp Articulating patch deployment device and method of use
AU2010286116B2 (en) 2009-08-17 2014-06-26 Covidien Lp Means and method for reversibly connecting an implant to a deployment device
WO2011021712A1 (en) * 2009-08-21 2011-02-24 独立行政法人農業生物資源研究所 Substratum for supplying cells/tissues, cell/tissue supply material and process for production thereof, method for regeneration of tissues, and process for production of porous material
US9821094B2 (en) 2014-06-11 2017-11-21 Silver Bullet Therapeutics, Inc. Coatings for the controllable release of antimicrobial metal ions
US8927004B1 (en) 2014-06-11 2015-01-06 Silver Bullet Therapeutics, Inc. Bioabsorbable substrates and systems that controllably release antimicrobial metal ions
US8221396B2 (en) 2009-08-27 2012-07-17 Silver Bullet Therapeutics, Inc. Bone implants for the treatment of infection
US9114197B1 (en) 2014-06-11 2015-08-25 Silver Bullett Therapeutics, Inc. Coatings for the controllable release of antimicrobial metal ions
US10265435B2 (en) 2009-08-27 2019-04-23 Silver Bullet Therapeutics, Inc. Bone implant and systems and coatings for the controllable release of antimicrobial metal ions
US20110118650A1 (en) * 2009-11-18 2011-05-19 Anteromed, Inc. Method and apparatus for treating obesity and controlling weight gain and absorption of glucose in mammals
KR101811070B1 (en) 2009-12-16 2017-12-20 백스터 인터내셔널 인코포레이티드 Hemostatic sponge
US8666471B2 (en) 2010-03-17 2014-03-04 The Board Of Trustees Of The University Of Illinois Implantable biomedical devices on bioresorbable substrates
US8529492B2 (en) 2009-12-23 2013-09-10 Trascend Medical, Inc. Drug delivery devices and methods
WO2011080588A2 (en) 2009-12-30 2011-07-07 Vivasure Medical Limited Closure system and uses thereof
EP2521586A4 (en) 2010-01-07 2013-06-19 Bioprotect Ltd Controlled tissue dissection systems and methods
US8231619B2 (en) 2010-01-22 2012-07-31 Cytyc Corporation Sterilization device and method
US9138308B2 (en) 2010-02-03 2015-09-22 Apollo Endosurgery, Inc. Mucosal tissue adhesion via textured surface
CN102844074B (en) 2010-02-17 2016-06-08 弗洛福沃德医药股份有限公司 It is used for increasing the system and method for vein overall diameter
US9259334B2 (en) * 2012-02-13 2016-02-16 Board Of Regents Of The University Of Texas System Scaffold system for tissue repair
US8641761B2 (en) * 2010-03-18 2014-02-04 Mark Bauman Creation and implantation of an artificial nail for the treatment of deformed or missing nails
SA111320355B1 (en) 2010-04-07 2015-01-08 Baxter Heathcare S A Hemostatic sponge
KR20130108067A (en) 2010-04-09 2013-10-02 베식스 바스큘라 인코포레이티드 Power generating and control apparatus for the treatment of tissue
US9192790B2 (en) 2010-04-14 2015-11-24 Boston Scientific Scimed, Inc. Focused ultrasonic renal denervation
US8604265B2 (en) * 2010-04-16 2013-12-10 Kci Licensing, Inc. Dressings and methods for treating a tissue site on a patient
AU2011245522A1 (en) * 2010-04-27 2012-12-06 Allergan, Inc. Foam-like materials and methods for producing same
US8550086B2 (en) * 2010-05-04 2013-10-08 Hologic, Inc. Radiopaque implant
EP2569473B1 (en) 2010-05-10 2019-10-16 Allergan, Inc. Porous materials, methods of making and uses
KR101854481B1 (en) 2010-05-11 2018-05-03 알러간, 인코포레이티드 Porogen compositions, methods of making and uses
AU2011252023B2 (en) 2010-05-11 2015-05-21 Allergan, Inc. Porous materials, methods of making and uses
CA2801116C (en) 2010-06-01 2019-02-12 Baxter International Inc. Process for making dry and stable hemostatic compositions
CA2801118C (en) 2010-06-01 2016-01-05 Baxter International Inc. Process for making dry and stable hemostatic compositions
EP2575776B1 (en) 2010-06-01 2018-05-30 Baxter International Inc Process for making dry and stable hemostatic compositions
US8577477B2 (en) 2010-06-11 2013-11-05 ProNerve, LLC Endotracheal tube with a selectively positional electrode
US8473067B2 (en) 2010-06-11 2013-06-25 Boston Scientific Scimed, Inc. Renal denervation and stimulation employing wireless vascular energy transfer arrangement
EP2582412A1 (en) 2010-06-16 2013-04-24 Allergan, Inc. Open-cell surface foam materials
EP2585516B1 (en) * 2010-06-25 2023-01-25 Nanovis Inc. Method for producing nanosurfaces with nano, micron, and/or submicron structures on a polymer
US9084609B2 (en) 2010-07-30 2015-07-21 Boston Scientific Scime, Inc. Spiral balloon catheter for renal nerve ablation
US9408661B2 (en) 2010-07-30 2016-08-09 Patrick A. Haverkost RF electrodes on multiple flexible wires for renal nerve ablation
US9463062B2 (en) 2010-07-30 2016-10-11 Boston Scientific Scimed, Inc. Cooled conductive balloon RF catheter for renal nerve ablation
US9155589B2 (en) 2010-07-30 2015-10-13 Boston Scientific Scimed, Inc. Sequential activation RF electrode set for renal nerve ablation
US9358365B2 (en) 2010-07-30 2016-06-07 Boston Scientific Scimed, Inc. Precision electrode movement control for renal nerve ablation
US20130116794A1 (en) 2010-08-04 2013-05-09 Shaul Shohat Shoulder implant
EP3489313A1 (en) 2010-08-25 2019-05-29 Tyrx, Inc. Novel medical device coatings
WO2012051433A2 (en) 2010-10-13 2012-04-19 Angiodynamics, Inc. System and method for electrically ablating tissue of a patient
US8974451B2 (en) 2010-10-25 2015-03-10 Boston Scientific Scimed, Inc. Renal nerve ablation using conductive fluid jet and RF energy
US9220558B2 (en) 2010-10-27 2015-12-29 Boston Scientific Scimed, Inc. RF renal denervation catheter with multiple independent electrodes
WO2012064632A1 (en) 2010-11-12 2012-05-18 Wake Forest University Health Sciences Methods of treating cancer and other disorders
EP2637608B1 (en) 2010-11-12 2016-03-02 Silver Bullet Therapeutics Inc. Bone implant and systems that controllably releases silver
JP6397190B2 (en) 2010-11-12 2018-09-26 タイレックス・インコーポレイテッドTyrx Inc. Fixing device containing active pharmaceutical ingredients
US9028485B2 (en) 2010-11-15 2015-05-12 Boston Scientific Scimed, Inc. Self-expanding cooling electrode for renal nerve ablation
US9089350B2 (en) 2010-11-16 2015-07-28 Boston Scientific Scimed, Inc. Renal denervation catheter with RF electrode and integral contrast dye injection arrangement
US9668811B2 (en) 2010-11-16 2017-06-06 Boston Scientific Scimed, Inc. Minimally invasive access for renal nerve ablation
US9326751B2 (en) 2010-11-17 2016-05-03 Boston Scientific Scimed, Inc. Catheter guidance of external energy for renal denervation
US9060761B2 (en) 2010-11-18 2015-06-23 Boston Scientific Scime, Inc. Catheter-focused magnetic field induced renal nerve ablation
US9023034B2 (en) 2010-11-22 2015-05-05 Boston Scientific Scimed, Inc. Renal ablation electrode with force-activatable conduction apparatus
US9192435B2 (en) 2010-11-22 2015-11-24 Boston Scientific Scimed, Inc. Renal denervation catheter with cooled RF electrode
US20120157993A1 (en) 2010-12-15 2012-06-21 Jenson Mark L Bipolar Off-Wall Electrode Device for Renal Nerve Ablation
US9220561B2 (en) 2011-01-19 2015-12-29 Boston Scientific Scimed, Inc. Guide-compatible large-electrode catheter for renal nerve ablation with reduced arterial injury
WO2012161875A1 (en) 2011-04-08 2012-11-29 Tyco Healthcare Group Lp Iontophoresis drug delivery system and method for denervation of the renal sympathetic nerve and iontophoretic drug delivery
WO2012148969A2 (en) 2011-04-25 2012-11-01 Brian Kelly Apparatus and methods related to constrained deployment of cryogenic balloons for limited cryogenic ablation of vessel walls
RU2454190C1 (en) * 2011-05-31 2012-06-27 Государственное бюджетное образовательное учреждение высшего профессионального образования "Ростовский государственный медицинский университет" Министерства здравоохранения и социального развития Российской Федерации (ГБОУ ВПО РостГМУ Минздравсоцразвития России) Method of treating atresia of esophagus
WO2013006176A1 (en) * 2011-07-07 2013-01-10 Empire Technology Development Llc Microwave induced photodynamic therapy
WO2013011511A1 (en) 2011-07-18 2013-01-24 Mor Research Applications Ltd. A device for adjusting the intraocular pressure
WO2013013156A2 (en) 2011-07-20 2013-01-24 Boston Scientific Scimed, Inc. Percutaneous devices and methods to visualize, target and ablate nerves
JP6106669B2 (en) 2011-07-22 2017-04-05 ボストン サイエンティフィック サイムド,インコーポレイテッドBoston Scientific Scimed,Inc. A neuromodulation system having a neuromodulation element that can be placed in a helical guide
US20130041406A1 (en) * 2011-08-10 2013-02-14 Brian W. Bear Surgical staple with localized adjunct coating
EP2741809B1 (en) 2011-08-12 2018-08-01 Cardiac Pacemakers, Inc. Method for coating devices using electrospinning and melt blowing
KR102062132B1 (en) 2011-08-17 2020-01-03 플로우 포워드 메디컬, 인크. Blood pump systems and methods
GB201115625D0 (en) 2011-09-09 2011-10-26 Magstim Company The Ltd Nerve snesing/monitoring device
US9078665B2 (en) 2011-09-28 2015-07-14 Angiodynamics, Inc. Multiple treatment zone ablation probe
EP2765942B1 (en) 2011-10-10 2016-02-24 Boston Scientific Scimed, Inc. Medical devices including ablation electrodes
US9420955B2 (en) 2011-10-11 2016-08-23 Boston Scientific Scimed, Inc. Intravascular temperature monitoring system and method
AU2012318258B2 (en) 2011-10-11 2015-07-09 Baxter Healthcare S.A. Hemostatic compositions
EP2765940B1 (en) 2011-10-11 2015-08-26 Boston Scientific Scimed, Inc. Off-wall electrode device for nerve modulation
AU2012318257B2 (en) 2011-10-11 2015-10-01 Baxter Healthcare S.A. Hemostatic compositions
US9364284B2 (en) 2011-10-12 2016-06-14 Boston Scientific Scimed, Inc. Method of making an off-wall spacer cage
EP2768563B1 (en) 2011-10-18 2016-11-09 Boston Scientific Scimed, Inc. Deflectable medical devices
EP2768568B1 (en) 2011-10-18 2020-05-06 Boston Scientific Scimed, Inc. Integrated crossing balloon catheter
US9289307B2 (en) 2011-10-18 2016-03-22 Ortho-Space Ltd. Prosthetic devices and methods for using same
US9622779B2 (en) 2011-10-27 2017-04-18 DePuy Synthes Products, Inc. Method and devices for a sub-splenius / supra-levator scapulae surgical access technique
AR088531A1 (en) 2011-10-27 2014-06-18 Baxter Int HEMOSTATIC COMPOSITIONS
WO2013070724A1 (en) 2011-11-08 2013-05-16 Boston Scientific Scimed, Inc. Ostial renal nerve ablation
EP2779929A1 (en) 2011-11-15 2014-09-24 Boston Scientific Scimed, Inc. Device and methods for renal nerve modulation monitoring
US9119632B2 (en) 2011-11-21 2015-09-01 Boston Scientific Scimed, Inc. Deflectable renal nerve ablation catheter
US9691873B2 (en) 2011-12-01 2017-06-27 The Board Of Trustees Of The University Of Illinois Transient devices designed to undergo programmable transformations
US9265969B2 (en) 2011-12-21 2016-02-23 Cardiac Pacemakers, Inc. Methods for modulating cell function
WO2013096920A1 (en) 2011-12-23 2013-06-27 Vessix Vascular, Inc. Methods and apparatuses for remodeling tissue of or adjacent to a body passage
WO2013101452A1 (en) 2011-12-28 2013-07-04 Boston Scientific Scimed, Inc. Device and methods for nerve modulation using a novel ablation catheter with polymeric ablative elements
TWI590843B (en) 2011-12-28 2017-07-11 信迪思有限公司 Films and methods of manufacture
US9050106B2 (en) 2011-12-29 2015-06-09 Boston Scientific Scimed, Inc. Off-wall electrode device and methods for nerve modulation
US20150031772A1 (en) * 2012-02-17 2015-01-29 Bacterin International, Inc. Adjustable bioactive agent dispersion within a polymeric coating
EP2822474B1 (en) 2012-03-06 2018-05-02 Ferrosan Medical Devices A/S Pressurized container containing haemostatic paste
US9220584B2 (en) 2012-03-30 2015-12-29 Abbott Cardiovascular Systems Inc. Treatment of diabetic patients with a stent and locally administered adjunctive therapy
EP3842580B1 (en) 2012-04-06 2023-10-18 Poly-Med Inc. Polymeric mesh products and method of making thereof
US8858980B2 (en) 2012-04-12 2014-10-14 Poly-Med, Inc. Synthetic mechanical hemostatic composition, method of making and use thereof
US10085633B2 (en) 2012-04-19 2018-10-02 Novartis Ag Direct visualization system for glaucoma treatment
US9241832B2 (en) 2012-04-24 2016-01-26 Transcend Medical, Inc. Delivery system for ocular implant
BR102012010543A2 (en) * 2012-05-04 2014-04-08 Bioactive Biomateriais Ltda BIOACTIVE BIOACTIVE POROSA MEMBRANE AND ITS OBTAINING PROCESS
US20130297023A1 (en) * 2012-05-07 2013-11-07 Hee-Jeong Im Sampen Methods and Devices For Treating Intervertebral Disc Disease
US10660703B2 (en) 2012-05-08 2020-05-26 Boston Scientific Scimed, Inc. Renal nerve modulation devices
BR102012011209A2 (en) * 2012-05-11 2014-03-25 Bioactive Biomateriais Ltda BIODEGRADABLE THREE-DIMENSIONAL MATERIAL AND BIODEGRADABLE THREE-DIMENSIONAL MATERIAL PREPARATION PROCESS
CN104349797B (en) 2012-06-12 2017-10-27 弗罗桑医疗设备公司 Dry hemostatic composition
WO2014010759A1 (en) * 2012-07-10 2014-01-16 주식회사 엠아이텍 Cylindrical structure having lumen inserted into human body
WO2014022657A1 (en) 2012-08-02 2014-02-06 Allergan, Inc. Mucosal tissue adhesion via textured surface
US10258730B2 (en) 2012-08-17 2019-04-16 Flow Forward Medical, Inc. Blood pump systems and methods
US10321946B2 (en) 2012-08-24 2019-06-18 Boston Scientific Scimed, Inc. Renal nerve modulation devices with weeping RF ablation balloons
EP3228286A1 (en) 2012-09-17 2017-10-11 Novartis AG Expanding ocular impant devices
CN104780859B (en) 2012-09-17 2017-07-25 波士顿科学西美德公司 Self-positioning electrode system and method for renal regulation
US10549127B2 (en) 2012-09-21 2020-02-04 Boston Scientific Scimed, Inc. Self-cooling ultrasound ablation catheter
US10398464B2 (en) 2012-09-21 2019-09-03 Boston Scientific Scimed, Inc. System for nerve modulation and innocuous thermal gradient nerve block
EP2897658A1 (en) 2012-09-24 2015-07-29 Allergan, Inc. Porous materials, methods of making and uses
WO2014052724A1 (en) 2012-09-28 2014-04-03 Allergan, Inc. Porogen compositions, methods of making and uses
CN104869930B (en) 2012-10-10 2020-12-25 波士顿科学国际有限公司 Renal neuromodulation apparatus and methods
AU2013338051C1 (en) 2012-10-29 2017-08-10 Ariste Medical, Llc. Polymer coating compositions and coated products
WO2014078288A1 (en) 2012-11-14 2014-05-22 Transcend Medical, Inc. Flow promoting ocular implant
US9949692B2 (en) 2012-12-21 2018-04-24 Canary Medical Inc. Stent graft monitoring assembly and method of use thereof
US20160298866A1 (en) * 2013-01-08 2016-10-13 Polymerium, Llc Evaporative Cooling System
US9888956B2 (en) 2013-01-22 2018-02-13 Angiodynamics, Inc. Integrated pump and generator device and method of use
PL2961350T3 (en) 2013-02-27 2018-07-31 Spirox, Inc. Nasal implants and systems
US10973618B2 (en) * 2013-03-01 2021-04-13 St. Jude Medical, Cardiology Division, Inc. Embolic protection device
US9693821B2 (en) 2013-03-11 2017-07-04 Boston Scientific Scimed, Inc. Medical devices for modulating nerves
WO2014163987A1 (en) 2013-03-11 2014-10-09 Boston Scientific Scimed, Inc. Medical devices for modulating nerves
US9808311B2 (en) 2013-03-13 2017-11-07 Boston Scientific Scimed, Inc. Deflectable medical devices
JP6399663B2 (en) 2013-03-14 2018-10-03 インターセクト エント, インコーポレイテッド System, device and method for treating sinus conditions
US10265122B2 (en) 2013-03-15 2019-04-23 Boston Scientific Scimed, Inc. Nerve ablation devices and related methods of use
CN105283152A (en) 2013-03-15 2016-01-27 威廉·L·亨特 Stent monitoring assembly and method of use thereof
WO2014149690A2 (en) 2013-03-15 2014-09-25 Boston Scientific Scimed, Inc. Medical devices and methods for treatment of hypertension that utilize impedance compensation
EP2967734B1 (en) 2013-03-15 2019-05-15 Boston Scientific Scimed, Inc. Methods and apparatuses for remodeling tissue of or adjacent to a body passage
US9731051B2 (en) 2013-03-26 2017-08-15 Innovia Llc Pacemaker lead and other medical implant devices
WO2014169298A1 (en) * 2013-04-12 2014-10-16 Bui The Duy Timing controlled in-situ cross-linking ofhalyuronic acid during injection
US9987163B2 (en) 2013-04-16 2018-06-05 Novartis Ag Device for dispensing intraocular substances
US9320645B2 (en) * 2013-05-29 2016-04-26 Terry Glasser Approach to administering ocular medication
JP2016523147A (en) 2013-06-21 2016-08-08 ボストン サイエンティフィック サイムド,インコーポレイテッドBoston Scientific Scimed,Inc. Renal denervation balloon catheter with a riding-type electrode support
CN105473092B (en) 2013-06-21 2019-05-17 波士顿科学国际有限公司 The medical instrument for renal nerve ablation with rotatable shaft
US9724078B2 (en) 2013-06-21 2017-08-08 Ferrosan Medical Devices A/S Vacuum expanded dry composition and syringe for retaining same
CN105555328B (en) 2013-06-21 2019-01-11 德普伊新特斯产品公司 film and manufacturing method
US9707036B2 (en) 2013-06-25 2017-07-18 Boston Scientific Scimed, Inc. Devices and methods for nerve modulation using localized indifferent electrodes
AU2014284558B2 (en) 2013-07-01 2017-08-17 Boston Scientific Scimed, Inc. Medical devices for renal nerve ablation
US10245355B2 (en) * 2013-07-10 2019-04-02 President And Fellows Of Harvard College Modification of surfaces for fluid and solid repellency
WO2015006573A1 (en) 2013-07-11 2015-01-15 Boston Scientific Scimed, Inc. Medical device with stretchable electrode assemblies
US10660698B2 (en) 2013-07-11 2020-05-26 Boston Scientific Scimed, Inc. Devices and methods for nerve modulation
CN105682594B (en) 2013-07-19 2018-06-22 波士顿科学国际有限公司 Helical bipolar electrodes renal denervation dominates air bag
US20150025608A1 (en) 2013-07-22 2015-01-22 Cardiac Pacemakers, Inc. Lubricious, biocompatible hydrophilic thermoset coating using interpenetrating hydrogel networks
EP3024406B1 (en) 2013-07-22 2019-06-19 Boston Scientific Scimed, Inc. Medical devices for renal nerve ablation
JP6122217B2 (en) 2013-07-22 2017-04-26 ボストン サイエンティフィック サイムド,インコーポレイテッドBoston Scientific Scimed,Inc. Renal nerve ablation medical device
US10722300B2 (en) 2013-08-22 2020-07-28 Boston Scientific Scimed, Inc. Flexible circuit having improved adhesion to a renal nerve modulation balloon
EP3041425B1 (en) 2013-09-04 2022-04-13 Boston Scientific Scimed, Inc. Radio frequency (rf) balloon catheter having flushing and cooling capability
WO2015038947A1 (en) 2013-09-13 2015-03-19 Boston Scientific Scimed, Inc. Ablation balloon with vapor deposited cover layer
US11246654B2 (en) 2013-10-14 2022-02-15 Boston Scientific Scimed, Inc. Flexible renal nerve ablation devices and related methods of use and manufacture
CN105592778B (en) 2013-10-14 2019-07-23 波士顿科学医学有限公司 High-resolution cardiac mapping electrod-array conduit
US9770606B2 (en) 2013-10-15 2017-09-26 Boston Scientific Scimed, Inc. Ultrasound ablation catheter with cooling infusion and centering basket
CN105636537B (en) 2013-10-15 2018-08-17 波士顿科学国际有限公司 Medical instrument sacculus
WO2015057961A1 (en) 2013-10-18 2015-04-23 Boston Scientific Scimed, Inc. Balloon catheters with flexible conducting wires and related methods of use and manufacture
WO2015061457A1 (en) 2013-10-25 2015-04-30 Boston Scientific Scimed, Inc. Embedded thermocouple in denervation flex circuit
US9161680B2 (en) 2013-11-26 2015-10-20 Bracco Diagnostics Inc. Disposable air/water valve for an endoscopic device
CA2928963C (en) 2013-12-11 2020-10-27 Ferrosan Medical Devices A/S Dry composition comprising an extrusion enhancer
JP6382989B2 (en) 2014-01-06 2018-08-29 ボストン サイエンティフィック サイムド,インコーポレイテッドBoston Scientific Scimed,Inc. Medical device with tear resistant flexible circuit assembly
US10166321B2 (en) 2014-01-09 2019-01-01 Angiodynamics, Inc. High-flow port and infusion needle systems
US9956384B2 (en) 2014-01-24 2018-05-01 Cook Medical Technologies Llc Articulating balloon catheter and method for using the same
US20170167054A1 (en) 2014-01-30 2017-06-15 Poly-Med, Inc. Thermally and dimensionally stabilized electrospun compositions and methods of making same
WO2015116917A1 (en) 2014-01-30 2015-08-06 Poly-Med, Inc. Time-dependent synthetic biological barrier material
CN106572881B (en) 2014-02-04 2019-07-26 波士顿科学国际有限公司 Substitution of the heat sensor on bipolar electrode is placed
US11000679B2 (en) 2014-02-04 2021-05-11 Boston Scientific Scimed, Inc. Balloon protection and rewrapping devices and related methods of use
GB2528421B (en) 2014-04-22 2016-12-14 Ariste Medical Inc Methods and processes for application of drug delivery polymeric coatings
US10709490B2 (en) 2014-05-07 2020-07-14 Medtronic Ardian Luxembourg S.A.R.L. Catheter assemblies comprising a direct heating element for renal neuromodulation and associated systems and methods
AU2015259303B2 (en) 2014-05-12 2021-10-28 Arena, Christopher B. Selective modulation of intracellular effects of cells using pulsed electric fields
US11141314B2 (en) 2014-05-15 2021-10-12 Biopierce Technologies, Llc Bioabsorbable tissue support adjunctive to tissue piercing
WO2015175744A1 (en) * 2014-05-15 2015-11-19 Nathan Mark David Bioabsorbable tissue support adjunctive to tissue piercing
US9452242B2 (en) 2014-06-11 2016-09-27 Silver Bullet Therapeutics, Inc. Enhancement of antimicrobial silver, silver coatings, or silver platings
CA3187647A1 (en) 2014-06-15 2015-12-23 Yeda Research And Development Co. Ltd. Surface treatment by water-soluble polymers and lipids/liposomes
US10524694B2 (en) 2014-06-25 2020-01-07 Canaray Medical Inc. Devices, systems and methods for using and monitoring tubes in body passageways
WO2016033196A1 (en) * 2014-08-26 2016-03-03 Spirox, Inc. Nasal implants and systems and method of use
AU2015333206B2 (en) 2014-10-13 2019-07-11 Ferrosan Medical Devices A/S. Dry composition for use in haemostasis and wound healing
WO2016062763A1 (en) * 2014-10-23 2016-04-28 Biotronik Se & Co. Kg Method for coating a medical implant
WO2016100325A1 (en) 2014-12-15 2016-06-23 Virginia Tech Intellectual Properties, Inc. Devices, systems, and methods for real-time monitoring of electrophysical effects during tissue treatment
US10433826B2 (en) 2014-12-15 2019-10-08 Vivasure Medical Limited Closure apparatus with flexible sealable member and flexible support member
EP3232939B1 (en) * 2014-12-15 2020-09-23 Vivasure Medical Limited Implantable sealable member with mesh layer
RU2705905C2 (en) 2014-12-24 2019-11-12 Ферросан Медикал Дивайсиз А/С Syringe for holding and mixing first and second substances
EP3307374A4 (en) 2015-06-11 2019-04-10 Proviflo, LLC Graft-port hemodialysis systems, devices and methods
JP6595232B2 (en) 2015-07-02 2019-10-23 ソニー・オリンパスメディカルソリューションズ株式会社 Endoscope imaging apparatus, endoscope apparatus, and endoscope cable
RU2717356C2 (en) 2015-07-03 2020-03-23 Ферросан Медикал Дивайсиз А/С Syringe for holding vacuum in storage state
EP3115031B1 (en) 2015-07-06 2021-12-22 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Intraocular device and method for preparing the same
EP3325084B1 (en) 2015-07-25 2019-08-21 Cardiac Pacemakers, Inc. Medical electrical lead with biostable pvdf-based materials
CN113349985A (en) 2015-09-25 2021-09-07 斯贝洛克斯公司 Nasal implants and systems and methods of use
US10925543B2 (en) 2015-11-11 2021-02-23 The Board Of Trustees Of The University Of Illinois Bioresorbable silicon electronics for transient implants
EP3386556A4 (en) 2015-12-11 2019-07-31 Research Institute at Nationwide Children's Hospital Systems and methods for optimized patient specific tissue engineering vascular grafts
WO2017102941A1 (en) 2015-12-15 2017-06-22 Vivasure Medical Limited Arteriotomy closure apparatus with slotted shoe for advantageous pressure distribution
CA3016675A1 (en) * 2016-03-11 2017-09-14 The Regents Of The University Of California Engineered scaffolds for vascularized tissue repair
EP3223181B1 (en) * 2016-03-24 2019-12-18 Sofradim Production System and method of generating a model and simulating an effect on a surgical repair site
US10498940B2 (en) * 2016-04-29 2019-12-03 Deuk Soo Jang Endoscope
AU2017257508B2 (en) 2016-04-29 2021-10-14 Artio Medical, Inc. Conduit tips and systems and methods for use
JP6934889B2 (en) 2016-05-02 2021-09-15 エンテラス メディカル インコーポレイテッドEntellus Medical,Inc. Nasal flap implant and its transplant method
TWI794171B (en) 2016-05-11 2023-03-01 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-l1 inhibitors
TWI808055B (en) 2016-05-11 2023-07-11 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-1 inhibitors
GB2565028B (en) * 2016-05-17 2021-09-15 Monarch Biosciences Inc Thin-film transcatheter heart valve
US10905492B2 (en) 2016-11-17 2021-02-02 Angiodynamics, Inc. Techniques for irreversible electroporation using a single-pole tine-style internal device communicating with an external surface electrode
CN110087726A (en) * 2016-12-18 2019-08-02 心脏起搏器股份公司 Anti-infectives elute lead casing
US10465318B2 (en) 2016-12-27 2019-11-05 Boston Scientific Scimed Inc Degradable scaffolding for electrospinning
CN110418660B (en) * 2017-03-02 2023-05-30 美敦力公司 Medical device, method for producing same, and use thereof
US20180353175A1 (en) * 2017-06-13 2018-12-13 Ethicon Llc Surgical Fastener with Broad Spectrum MMP Inhibitors
US11058804B2 (en) 2017-06-13 2021-07-13 Ethicon Llc Surgical fastener device for the prevention of ECM degradation
US10939911B2 (en) 2017-06-13 2021-03-09 Ethicon Llc Surgical stapler with end effector coating
US11045589B2 (en) 2017-09-22 2021-06-29 Becton, Dickinson And Company 4% trisodium citrate solution for use as a catheter lock solution
US11607537B2 (en) 2017-12-05 2023-03-21 Virginia Tech Intellectual Properties, Inc. Method for treating neurological disorders, including tumors, with electroporation
US11717372B2 (en) * 2018-02-22 2023-08-08 Phoenix Spine Holdings, Inc. Shape memory surgical sponge for retracting the dura during a laminectomy procedure
US11076902B2 (en) 2018-02-22 2021-08-03 Phoenix Spine Holdings, Inc. Locking screw assembly for facilitating direct lateral interbody fusion procedures
US11925405B2 (en) 2018-03-13 2024-03-12 Virginia Tech Intellectual Properties, Inc. Treatment planning system for immunotherapy enhancement via non-thermal ablation
US11311329B2 (en) 2018-03-13 2022-04-26 Virginia Tech Intellectual Properties, Inc. Treatment planning for immunotherapy based treatments using non-thermal ablation techniques
US20190300724A1 (en) * 2018-04-02 2019-10-03 Medical Surface Inc. Covalently Attached Antioxidant Coatings
WO2019204061A1 (en) 2018-04-19 2019-10-24 Poly-Med, Inc. Macromers and compositions for photocuring processes
JP7395113B2 (en) 2018-05-09 2023-12-11 フェロサン メディカル デバイシーズ エイ/エス Method of preparing a hemostatic composition
EP3877175A4 (en) 2018-11-09 2022-08-10 Sofresh, Inc. Blown film materials and processes for manufacturing thereof and uses thereof
CN109901213B (en) * 2019-03-05 2022-06-07 中国辐射防护研究院 Method and system for generating gamma scanning scheme based on Router grid
CN110331124B (en) * 2019-06-14 2022-03-22 浙江大学 Conductive polypyrrole/extracellular matrix composite film and preparation method thereof
US11752020B2 (en) * 2019-06-19 2023-09-12 Michael J. Spearman Tool for placement of degradable ostial stent
CN111494716B (en) * 2020-04-15 2021-12-21 青岛市妇女儿童医院(青岛市妇幼保健院、青岛市残疾儿童医疗康复中心、青岛市新生儿疾病筛查中心) Heart surgery autologous pericardium patch processor and rapid anti-calcification treatment method
CN114848541B (en) * 2022-04-11 2023-06-27 广东丸美生物技术股份有限公司 Anti-aging wrinkle-removing composition, preparation method thereof, cosmetic and preparation method thereof

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6053031B2 (en) * 1978-03-31 1985-11-22 武田薬品工業株式会社 Spiro compounds and their production methods
US5028339A (en) * 1989-01-23 1991-07-02 Clark Iii William T Polymer matrix and method for retaining reactants in a polymer matrix
US6326017B1 (en) * 1989-10-02 2001-12-04 University Of Washington Methods for the localized delivery of agents to blood vessels
US5527532A (en) * 1989-11-13 1996-06-18 President And Fellows Of Harvard College Extraluminal regulation of the growth and repair of tubular structures in vivo
US6515009B1 (en) * 1991-09-27 2003-02-04 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5811447A (en) * 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6306421B1 (en) * 1992-09-25 2001-10-23 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
US6663881B2 (en) * 1993-01-28 2003-12-16 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6491938B2 (en) * 1993-05-13 2002-12-10 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
US5886026A (en) * 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
PT711158E (en) * 1993-07-29 2004-04-30 Us Gov Health & Human Serv METHOD FOR TREATING ATHEROSCLEROSIS OR RESTENING USING A MICROTUBLE STABILIZING AGENT
US6063116A (en) * 1994-10-26 2000-05-16 Medarex, Inc. Modulation of cell proliferation and wound healing
US20030083733A1 (en) * 1997-10-10 2003-05-01 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
DK0914102T3 (en) * 1996-05-24 2006-01-09 Angiotech Pharm Inc Preparations and methods for treating or preventing diseases of the body canals
US6495579B1 (en) * 1996-12-02 2002-12-17 Angiotech Pharmaceuticals, Inc. Method for treating multiple sclerosis
US6515016B2 (en) * 1996-12-02 2003-02-04 Angiotech Pharmaceuticals, Inc. Composition and methods of paclitaxel for treating psoriasis
US5893839A (en) * 1997-03-13 1999-04-13 Advanced Research And Technology Institute, Inc. Timed-release localized drug delivery by percutaneous administration
US5985592A (en) * 1997-06-05 1999-11-16 Dalhousie University Uses for pentoxifylline or functional derivatives/metabolites thereof
US6203536B1 (en) * 1997-06-17 2001-03-20 Medtronic, Inc. Medical device for delivering a therapeutic substance and method therefor
EP1011743B1 (en) * 1997-08-13 2011-07-27 Boston Scientific Limited Loading and release of water-insoluble drugs
US7985415B2 (en) * 1997-09-10 2011-07-26 Rutgers, The State University Of New Jersey Medical devices employing novel polymers
DE19740211A1 (en) * 1997-09-12 1999-03-18 Bodenseewerk Perkin Elmer Co Photoelectric detector arrangement for nuclear absorption spectroscopy
AU4679499A (en) * 1998-06-10 1999-12-30 Advanced Bypass Technologies, Inc. Thermal securing anastomosis systems
EP1100494A1 (en) * 1998-07-30 2001-05-23 Novopharm Biotech, Inc. Pharmaceutically composition comprising an aqueous solution of paclitaxel and albumin
US5916913A (en) * 1998-08-03 1999-06-29 Joseph; Hazel L. Inhibition of wound contraction with paclitaxel, colchicine and penicillamine
CA2340652C (en) * 1998-08-20 2013-09-24 Cook Incorporated Coated implantable medical device comprising paclitaxel
US6335029B1 (en) * 1998-08-28 2002-01-01 Scimed Life Systems, Inc. Polymeric coatings for controlled delivery of active agents
CN1378445B (en) * 1999-08-06 2013-02-06 得克萨斯系统大学评议会 Drug releasing biodegradable fiber implant
JP2003520830A (en) * 2000-01-25 2003-07-08 エドワーズ ライフサイエンシーズ コーポレイション Delivery system for treatment of restenosis and anastomotic intimal hyperplasia
US6596292B2 (en) * 2000-06-22 2003-07-22 Sumitomo Chemical Company, Limited Solid pesticidal formulation
ATE343969T1 (en) * 2000-09-29 2006-11-15 Cordis Corp COATED MEDICAL DEVICES
US20040018228A1 (en) * 2000-11-06 2004-01-29 Afmedica, Inc. Compositions and methods for reducing scar tissue formation
US20050084514A1 (en) * 2000-11-06 2005-04-21 Afmedica, Inc. Combination drug therapy for reducing scar tissue formation
US20040241211A9 (en) * 2000-11-06 2004-12-02 Fischell Robert E. Devices and methods for reducing scar tissue formation
US6534693B2 (en) * 2000-11-06 2003-03-18 Afmedica, Inc. Surgically implanted devices having reduced scar tissue formation
JP4584537B2 (en) * 2001-01-16 2010-11-24 ヴァスキュラー セラピーズ リミテッド ライアビリティ カンパニー Implantable devices and methods for preventing or treating hemodialysis vascular access and other vascular graft dysfunctions, including resorbable matrix materials and antiproliferative agents
US8182527B2 (en) * 2001-05-07 2012-05-22 Cordis Corporation Heparin barrier coating for controlled drug release
US6626920B2 (en) * 2001-07-05 2003-09-30 Converge Medical, Inc. Distal anastomosis system
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
US20030065345A1 (en) * 2001-09-28 2003-04-03 Kevin Weadock Anastomosis devices and methods for treating anastomotic sites
US20030065344A1 (en) * 2001-10-03 2003-04-03 Kirsch Wolff M. Method and device for creating microvascular anastomoses
JP4549059B2 (en) * 2001-10-15 2010-09-22 ヘモテック アーゲー Stent coating to prevent restenosis
AU2003225882A1 (en) * 2002-03-20 2003-10-08 Advanced Cardiovascular Systems, Inc. Biodegradable hydrophobic polymer for stents
US20030207907A1 (en) * 2002-05-03 2003-11-06 Iversen Patrick L. Delivery of microparticle-conjugated drugs for inhibition of stenosis
EP1501566B1 (en) * 2002-05-09 2008-08-13 Hemoteq AG Medical products comprising a haemocompatible coating, production and use thereof
KR20050010827A (en) * 2002-05-20 2005-01-28 오르버스 메디칼 테크놀로지즈 인코포레이티드 Drug eluting implantable medical device
MXPA04011651A (en) * 2002-05-24 2005-03-07 Angiotech Pharm Inc Compositions and methods for coating medical implants.
US7438925B2 (en) * 2002-08-26 2008-10-21 Biovention Holdings Ltd. Drug eluting coatings for medical implants
US20040167572A1 (en) * 2003-02-20 2004-08-26 Roth Noah M. Coated medical devices
US7527632B2 (en) * 2003-03-31 2009-05-05 Cordis Corporation Modified delivery device for coated medical devices
US20040236416A1 (en) * 2003-05-20 2004-11-25 Robert Falotico Increased biocompatibility of implantable medical devices
CA2526033A1 (en) * 2003-05-23 2005-03-03 Angiotech International Ag Anastomotic connector devices
US20050033417A1 (en) * 2003-07-31 2005-02-10 John Borges Coating for controlled release of a therapeutic agent
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
WO2005051452A2 (en) * 2003-11-20 2005-06-09 Angiotech International Ag Polymer compositions and methods for their use
US8652502B2 (en) * 2003-12-19 2014-02-18 Cordis Corporation Local vascular delivery of trichostatin A alone or in combination with sirolimus to prevent restenosis following vascular injury

Also Published As

Publication number Publication date
US20060147492A1 (en) 2006-07-06
WO2005046516A3 (en) 2006-06-01
IL174636A0 (en) 2006-08-20
US20050177225A1 (en) 2005-08-11
US20050181011A1 (en) 2005-08-18
US20050143817A1 (en) 2005-06-30
WO2005049105A2 (en) 2005-06-02
US20050183728A1 (en) 2005-08-25
EP1682196A2 (en) 2006-07-26
US20050149158A1 (en) 2005-07-07
US20050149080A1 (en) 2005-07-07
US20050175663A1 (en) 2005-08-11
US20050181008A1 (en) 2005-08-18
US20050181977A1 (en) 2005-08-18
AU2004291062A1 (en) 2005-06-02
WO2005046516A2 (en) 2005-05-26
WO2005049105A8 (en) 2005-10-13
JP2007516740A (en) 2007-06-28
US20050191331A1 (en) 2005-09-01
US20050165488A1 (en) 2005-07-28

Similar Documents

Publication Publication Date Title
US20050143817A1 (en) Medical implants and anti-scarring agents
US20040260318A1 (en) Anastomotic connector devices
US20050196421A1 (en) Polymer compositions and methods for their use
US20050181010A1 (en) Implantable sensors and implantable pumps and anti-scarring agents
WO2005051452A2 (en) Polymer compositions and methods for their use

Legal Events

Date Code Title Description
FZDE Discontinued